Page 65«..1020..64656667..7080..»

Archive for the ‘Bone Marrow Stem Cells’ Category

Human Primary Cells – Stemcell Technologies

Request customized products to meet your specific research needs:

Please contact Technical Support for further details: techsupport@stemcell.com

Cryopreserved Leuko Pak and Whole Blood Products:

Donor Screening:Donors are screened for HIV (1 & 2), Hepatitis B, and Hepatitis C.

Cryopreserved products are shipped with negative test results from donor screening that is done within 90 days of collection.

Fresh Leuko Pak and Whole Blood Products:

Donor Screening:Donors are screened for HIV (1 & 2), Hepatitis B, and Hepatitis C.

If the donor was screened within 90 days of donation the product will be shipped with negative test results from donor screening.

If the donor was not screened within 90 days of collection, a test sample will be taken at the time of donation and the product will be shipped before the screening results are available. In the unlikely event that a test result is positive, the customer will be contacted as soon as possible (usually within 24-72 hours from the time of shipment).

Cryopreserved Cord Blood Products:

Donor Screening:Cord blood is only collected from mothers that have tested negative for HIV (1 & 2) and Hepatitis B during their pregnancy. Hepatitis C is tested for at the time of collection.

Cryopreserved products are shipped with negative test results from donor screening.

Fresh Cord Blood Products:

Donor Screening: Cord blood is only collected from mothers that have tested negative for HIV (1 & 2) and Hepatitis B during their pregnancy. Hepatitis C is tested for at the time of collection.

Fresh cord blood products are shipped with negative test results for HIV (1 & 2) and Hepatitis B donor screening. Hepatitis C test results are not available at the time of shipment. In the unlikely event that the Hepatitis C test result is positive, the customer will be contacted as soon as possible (usually within 24-72 hours from the time of shipment).

STEMCELL does not test for infectious diseases other than those listed above and the testing that is done cannot completely guarantee that the donor was virus-free. Therefore THESE PRODUCTS SHOULD BE TREATED AS POTENTIALLY INFECTIOUS and only used following appropriate handling precautions such as those described in biological safety level 2. When handling these products do not use sharps such as needles and syringes.

See the original post here:
Human Primary Cells - Stemcell Technologies

Bone Marrow and Blood Stem Cell Transplants – City of Hope

What are hematopoietic cell transplantation (HCT) and peripheral blood stem cell transplantation (PBSCT)?

HCT and PBSCT are procedures that use stem cells to treat a patient's malignancy or to repair diseased or defective bone marrow. A patient receives intensive chemotherapy with or without total body irradiation therapy in an attempt to kill all cancerous cells, but which also destroy his/her own bone marrow function. This therapy also causes immunosuppression, which prevents rejection of the newly transplanted stem cells from a related or unrelated donor.

There is little risk of rejection of a patient's own stem cells following autologous transplant. After transplantation, the new stem cells replace the damaged bone marrow and cells of the immune system.

How do HCT and PBSCT help patients?

HCT and PBSCT allow a patient to receive very high doses of chemotherapy and radiation designed to kill cancer cells. The high doses of therapy lead to the destruction of a patient's own marrow and immune system, which is then replaced by marrow from a donor or from peripheral blood stem cells that have been harvested before therapy.

How many HCTs and PBSCTs are performed at City of Hope?

City of Hope has performed more than 12,000 transplants for patients from virtually every state as well as from numerous countries. HCT and PBSCT patients at City of Hope have ranged in age from less than 1 year old to 79 years old. City of Hope's HCT program is one of America's largest, dedicated solely to the traditional and newer uses of this procedure.

Which diseases are HCT and PBSCT most frequently used to treat?

What is the difference between autologous and allogeneic HCT?

How are donors for allogeneic transplantations found?

About 30 percent of patients needing a transplant get one from a family member whose HLA testing has identified compatibility between a patient and donor. This matching of donor and recipient reduces the chance of marrow rejection and greatly increases the likelihood of a successful transplant. The remaining 70 percent of patients must find an unrelated donor whose marrow is compatible.

Currently, there are nearly 5 million volunteer donors in the National Marrow Donor Program (NMDP) Be The Match Registry. Almost 50 percent of patients searching the registry have at least one identically matched, unrelated donor. The NMDP is conducting a major effort at the 97 donor centers around the United States (of which City of Hope is one) to increase minority registration.

Because HLA types vary greatly between people of different ethnic backgrounds, increasing minority and ethnic representation will increase minority patients' chances of finding matches.

What is a mini-HCT?

Mini-HCT is a procedure that allows successful transplant of bone marrow without the use of high-dose chemo and radiation therapy. It is less intensive but allows transplant to be utilized in the treatment of older patients who may not be able to endure the intensity of traditional HCT transplant regimens.

Because many diseases, such as leukemia, lymphoma, myeloma and myelodysplasia, are more common in older patients, mini-HCTs allow these patients to potentially benefit from transplant.

What is bone marrow?

Bone marrow is the soft, spongy material found inside bones. Bone marrow contains stem cells that give rise to white blood cells (to fight infections), red blood cells (for oxygenation) and platelets (to prevent hemorrhaging). The chief function of bone marrow is to produce blood cells.

What are platelets?

Platelets are critical in the clotting process and to help control bleeding. Platelets are commonly used to treat leukemia and cancer patients undergoing chemotherapy and bone marrow transplants. Platelets are also used for trauma patients.

What are stem cells?

All blood cells develop from very immature cells called stem cells. Most stem cells are found in the bone marrow, although some, called peripheral blood stem cells, circulate in blood vessels throughout the body. Stem cells can divide to form more stem cells, or they can go through a series of cell divisions by which they become fully mature blood cells.

Who can donate bone marrow or peripheral blood stem cells?

Donating bone marrow or stem cell to someone suffering from a life-threatening disease is one of the greatest gifts you can provide, the gift of life. The first step is to join the National Marrow Donor Program (NMDP) Be The Match Registry. The NMDP maintains the registry of potential donors and searches this when people need a match. To join the registry, you need to complete a brief health questionnaire, sign a consent form, and provide a small blood sample to determine your tissue type.

At City of Hope, we ask that you donate a unit of blood or platelets to help offset the cost of a tissue-type test. Your tissue type will be compared to the tissue types of thousands of patients awaiting a bone marrow transplant. If you are ever a potential match, the City of Hope Donor Center will notify you to see if you are still interested in continuing with the process. If you are, a City of Hope staff member will request an additional blood sample. This sample will determine if the donor matches well enough to continue with the process.

Will patients need blood and platelet donations?

Blood donations from friends and family are a great source of encouragement and support for a patient needing transfusions. If your blood type is compatible with the patient, your donated blood can be given directly to your loved one. If your blood is not the same type, it is still important that you donate to help other City of Hope patients who are a blood type match and seriously in need of your help.

In most circumstances, platelet donations do not need be the same blood type. Therefore, most friends and family members can direct their platelet donations to their loved one. Because platelets can only be stored for 3-5 days, consistent support for our patients is crucial. You can help rally friends and family members by sponsoring blood drives for patients as well as arranging for group donations in our Donor Center.

Encourage friends and family members to call the CityofHopeBloodDonorCenter at 626- 471-7171 and schedule an appointment to donate blood and/or platelets or make arrangements for a blood drive in your community. To find a blood drive in your community, please call 626-301-8385.

Why do patients need platelets?

Before a patient receives a donor's marrow, his or her own marrow must be destroyed by a rigorous treatment of chemotherapy and/or radiation. Once the patient receives the donated marrow, it takes about 4 to 8 weeks for the new marrow to produce platelets. During that time period, the patient needs transfusions of platelets to help his/her blood to clot. City of Hope patients sometimes receive platelet transfusion on a daily basis.

What are the risks to marrow donors?

Virtually none. Bone marrow is extracted under general anesthesia in a procedure that takes less than an hour. Donors have commented that their buttocks felt sore for several days after aspiration. Contrary to organ donations, marrow is completely replenished by the body within a couple of weeks. There are no increased risks to the donor during this period. Historically, at HCT centers around the world, marrow has been donated by individuals less than 1 year old to 60 or 70 years old.

What are the possible complications associated with HCT and PBSCT?

Immediately following allogeneic transplantation, patients are immunosuppressed and unable to fight infection. Different drugs are administered during this critical period and isolation is sometimes necessary for the patient.

Another possible complication for patients receiving allogeneic transplantation is known as graft-versus-host disease (GvHD). Despite the close match between patient and donor, in GvHD, the donated marrow may recognize its new home as foreign and react against the host.

In addition, patients can acquire post-transplant cytomegalovirus (CMV) pneumonia. City of Hope has pioneered several outstanding advances for the prevention and treatment of this potentially fatal complication. Recurrent disease also is possible if the pre-transplant chemotherapy and irradiation therapy were not successful in killing all malignant cells.

In autologous transplantations there are few complications once the patient leaves the hospital, and the only risk is whether the disease will return, causing relapse.

Read more from the original source:
Bone Marrow and Blood Stem Cell Transplants - City of Hope

Mobilization of hematopoietic stem cells from the bone …

Stem Cell Research & Therapy20112:13

DOI: 10.1186/scrt54

BioMed Central Ltd.2011

Published: 14March2011

The vast majority of hematopoietic stem cells (HSCs) reside in specialized niches within the bone marrow during steady state, maintaining lifelong blood cell production. A small number of HSCs normally traffic throughout the body; however, exogenous stimuli can enhance their release from the niche and entry into the peripheral circulation. This process, termed mobilization, has become the primary means to acquire a stem cell graft for hematopoietic transplant at most transplant centers. Currently, the preferred method of HSC mobilization for subsequent transplantation is treatment of the donor with granulocyte colony-stimulating factor. The mobilizing effect of granulocyte colony-stimulating factor is not completely understood, but recent studies suggest that its capacity to mobilize HSCs, at least in part, is a consequence of alterations to the hematopoietic niche. The present article reviews some of the key mechanisms mediating HSC mobilization, highlighting recent advances and controversies in the field.

The online version of this article (doi:10.1186/scrt54) contains supplementary material, which is available to authorized users.

Higher organisms have the remarkable capacity to produce and maintain adequate numbers of blood cells throughout their entire lifespan to meet the normal physiological requirements of blood cell turnover, as well as to respond to needs for increased blood cell demand as a consequence of injury or infection. At the center of lifelong blood cell production is the hematopoietic stem cell (HSC), with the capacity to give rise to all mature circulating blood cell types. Regulation of HSC function is a highly complex process involving not only intrinsic cues within the HSC themselves, but signaling from the surrounding microenvironment in which they reside. It was first postulated by Schofield that defined local microenvironments created specialized stem cell niches that regulated HSCs [1]. Bone marrow is the primary HSC niche in mammals and is composed of stromal cells and an extracellular matrix of collagens, fibronectin, proteoglycans [2], and endosteal lining osteoblasts [36]. HSCs are thought to be tethered to osteoblasts, other stromal cells, and the extracellular matrix in this stem cell niche through a variety of adhesion molecule inter-actions, many of which are probably redundant systems.

Disruption of one or more of these niche interactions can result in release of HSCs from the niche and their trafficking from the bone marrow to the peripheral circulation, a process termed peripheral blood stem cell mobilization. Mobilization can be achieved through administration of chemotherapy [79], hematopoietic growth factors, chemokines and small-molecule chemokine receptor inhibitors or antibodies against HSC niche interactions [1012].

The process of mobilization has been exploited for collection of hematopoietic stem and progenitor cells (HSPCs) and is widely used for hematopoietic trans-plantation in both the autologous and allogeneic settings. Mobilized peripheral blood hematopoietic stem cell grafts are associated with more rapid engraftment, reduction in infectious complications and, in patients with advanced malignancies, lower regimen-related mor-tality [1315] compared with bone marrow grafts. In many transplantation centers, mobilized HSC grafts are now the preferred hematopoietic stem cell source used for human leukocyte antigen-identical sibling transplants as well as for matched related and unrelated donor transplants [16, 17]. Granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor and - more recently, for patients who fail to mobilize with a G-CSF or granulocyte-macrophage colony-stimulating factor - plerixafor (AMD3100) are the only US Food and Drug Administration-approved agents for mobilizing HSCs. Despite the clinical prevalence of peripheral blood stem and progenitor cell mobilization, the mechanisms orchestrating the release of these cells from the hematopoietic niche are still not completely understood. In the following sections, we highlight some of the key mechanistic findings concerning HSPC mobilization, with an emphasis on the effects of mobilizing agents on bone marrow niche interactions.

The most explored HSC niche interaction is between the CXC4 chemokine receptor (CXCR4) and its ligand, stromal cell-derived factor 1 (SDF-1). SDF-1 is produced by osteoblasts [18], a specialized set of reticular cells found in endosteal and vascular niches [19], endothelial cells and bone itself [20, 21], and high levels of SDF-1 were observed recently in nestin-positive mesenchymal stem cells [22]. HSPCs express CXCR4 and are chemoattracted to and retained within the bone marrow by SDF-1 [2325]. Genetic knockout of either CXCR4 [26] or SDF-1 [27] in mice is embryonically lethal, with a failure of HSPCs to tracffic to the bone marrow niche during development. In addition, conditional CXCR4 knockout in mice results in a substantial egress of hematopoietic cells from the bone marrow [28] and impaired ability of CXCR4 knockout HSPCs to be retained within the bone marrow after transplantation [29].

Many agents reported to mobilize HSCs have been shown to disrupt the CXCR4/SDF-1 axis. Most notably, the CXCR4 antagonist AMD3100 (Plerixafor; Mozobil, Genzyme Corporation, Cambridge, MA, USA) mobilizes HSPCs [3035]; and similarly, the CXCR4 antagonists T140 [36] and T134 [37] are both capable of mobilization. Partially agonizing CXCR4 with SDF-1 mimetics including (met)-SDF-1 [38], CTCE-0214 [39], and CTCE-0021 [35] also mobilizes HSCs through CXCR4 receptor desensitization and/or downregulation of surface CXCR4 expression. Intriguingly, these agents that directly disrupt the CXCR4/SDF-1 axis lead to rapid mobilization of HSPCs - that is, hours after treatment - in contrast to other mobilization agents like G-CSF, which take several days to maximally mobilize HSPCs.

Despite the abundance of evidence supporting a key role for the CXCR4/SDF-1 axis in HSPC retention/trafficking/mobilization, it is still not clear which population of cells within the bone marrow niche is the pre-dominate source of SDF-1. Some studies have demonstrated that SDF-1 production by osteoblasts is reduced after G-CSF treatment [21, 40, 41], and seminal work by Katayama and colleagues suggests that this reduction in osteoblast SDF-1 is at least partly mediated by the sympathetic nervous system [21]. Notwithstanding the fact that decreased levels of SDF-1 production by osteoblasts are routinely seen following G-CSF administration, however, other studies have questioned the relative importance of osteoblast-derived SDF-1 in HSC maintenance and mobilization [19, 22, 42]. A recent study by Christopher and colleagues indicated that reduction in osteoblast production of SDF-1 is a common mechanism of cytokine-induced HSC mobilization and showed a specific reduction in SDF-1 production in Col2.3-expressing osteoblasts with no reduction in Col2.3-negative stromal cells [43]. Mendez-Ferrer and colleagues, however, showed, using a similar approach, a substantial decrease in SDF-1 in a novel population of nestin-expressing mesenchymal stem cells [22], relative to a similar population of stromal cells described by Christopher and colleagues [43], although a direct comparison with defined osteoblasts was not made. Future studies are clearly required in order to define the specific niche cells responsible for SDF-1 production and HSC retention, and may identify specific targets for future HSC therapies.

Osteoblasts are important HSC regulators [36], and express numerous signaling molecules in addition to SDF-1 that regulate HSC function and retention in the bone marrow niche. Osteoblasts express vascular cell adhesion molecule 1 (VCAM-1), and targeting the inter-action between very late antigen 4 (VLA-4) and VCAM-1 with either antibodies against VLA-4 [44, 45], antibodies against VCAM-1 [46, 47], or a small molecule inhibitor of VLA-4 (BIO5192) [48] results in HPSC mobilization. In addition, the Eph-ephrin A3 signaling axis increases adhesion to fibronectin and VCAM-1, and disruption of this signaling axis in vivo with a soluble EphA3-Fc fusion protein mobilizes HSPCs [49].

Osteoblasts also express significant amounts of osteo-pontin, and HSPCs adhere to osteopontin via 1 integrins, such as VLA-4 [50]. Osteopontin is a negative regulator of HSC pool size within the bone marrow niche [50, 51], and knockout of osteopontin in mice results in endoge-nous HSPC mobilization and increases the mobilization response to G-CSF [52]. Future therapies that target osteopontin may not only increase the HSC pool size available for hematopoietic mobilization, but may also act to untether the expanded HSCs from the bone marrow niche, resulting in significantly enhanced HSC mobilization.

Mobilizing regimens of G-CSF are associated with suppression of niche osteoblasts [21, 41, 53], with increased osteoblast apoptosis [41] and osteoblast flattening [21], resulting in significant decreases in endosteal niche expression of many of the above-mentioned retention molecules. This suppression has been reported to be the result of altered sympathetic nervous system signaling to osteoblasts [21]. A recent report by Winkler and colleagues demonstrated that G-CSF treatment results in the reduction of endosteal-lining osteomacs, which results in suppression of osteoblasts [53]. This osteomac population of cells is F4/80+ Ly-6G+ CD11b+ and provides a yet to be determined positive supporting role for osteoblasts. When osteomacs are depleted using Mafia transgenic mice or by treatment of mice with clodronate-loaded liposomes, significant mobilization of HSPCs was observed. These findings support a mechanistic role for osteoblasts in mediating G-CSF-induced mobilization, independent of the sympathetic nervous system, and highlight that multiple mechanisms may be responsible for the mobilizing effects of G-CSF.

Osteoblasts and osteoclasts regulate/coordinate bone formation and bone resorption, respectively, within the bone marrow niche. A report from Kollet and colleagues suggested that osteoclasts can mediate HSPC mobilization [54], and proposed a model where the balance between osteoblasts and osteoclasts is required for homeostatic maintenance of the stem cell niche and HSPC pool size. In their model, increased osteoblasts - for example, after parathyroid hormone administration [3] - increase the stem cell pool size and adherence in the niche, whereas increased osteoclasts degrade the niche - facilitating release and egress of HSPCs.

A role for osteoclasts in mobilization was shown by treating mice with RANK ligand, which increased osteoclast activity that correlated with a moderate increase in hematopoietic progenitor cell (HPC) mobilization [54]. Similarly, bleeding mice or treating them with lipopoly-saccharide, two models of physiological stress, resulted in an increase in the number of bone marrow niche osteoclasts as well as HPC mobilization. Inhibition of osteoclasts, either by treatment with calcitonin or using a genetic knockout model of PTP in female mice, resulted in a reduced HPC mobilization response to G-CSF compared with controls, further suggesting that osteoclasts were involved in G-CSF-mediated mobilization. The authors proposed that osteoclast-derived proteolytic enzymes, such as cathepsin K, degraded important niche interaction components including SDF-1 and osteopontin, thereby facilitating mobilization [54]. A more recent study by the same laboratory demonstrated reduced osteoclast maturation and activity in CD45 knockout mice, which correlated with reduced mobilization to RANK ligand and G-CSF [55], providing an additional link between osteoclast activity and HSPC mobilization.

In contrast to studies showing that increased osteoclasts enhance HPC mobilization, an earlier report by Takamatsu and colleagues demonstrated that while G-CSF treatment increases osteoclast number and bone resorption in both BALB/c mice and humans, the increase in osteoclasts did not occur until 10 to 15 days or 6 to 8 days, respectively, after treatment with G-CSF [56] - a finding that has also been observed by other groups using similar systems [40, 57]. Since HSPC mobilization by G-CSF is typically evaluated after 4 to 5 days, the importance of osteoclasts to HSPC mobilization in response to G-CSF treatment remains unclear. Furthermore, treatment of mice with bisphosphonates, which inhibit osteoclast activity and/or number, prior to G-CSF administration does not result in an impaired HSPC mobilization response [53, 56]; in fact, in one case, bisphosphonate treatment increased mobilization by G-CSF [53]. These studies suggest that while osteoclasts elicit mechanisms that can induce hematopoietic stem and progenitor mobilization, their role in clinical HSC mobilization with G-CSF is not sufficiently defined and may not be a primary mechanism of mobilization.

The endosteal surface of bone, particularly at the site of resorbing osteoclasts, is a significant source of soluble extracellular calcium within the bone marrow niche. Studies by Adams and colleagues demonstrated that HSCs express calcium-sensing receptors and are chemo-attracted to soluble Ca2+ [58]. When the gene for the calcium-sensing receptor was knocked out, mice had reduced HSC content within the bone marrow niche and increased HSCs in peripheral blood. Moreover, calcium-sensing receptor-knockout HSCs failed to engraft in hematopoietic transplantation experiments. These results suggest that Ca2+ at the endosteal surface is an important retention signal within the hematopoietic niche and that pharmacologic antagonism of the HSC calcium-sensing receptor may represent a possible strategy for HSPC mobilization.

The bone marrow hematopoietic niche has been shown to be hypoxic [59, 60]. HSCs that reside in hypoxic niches have also been shown to have greater hematopoietic-repopulating ability than those that do not [61]. A known physiological response to hypoxia is stabilization of the transcription factor hypoxia inducible factor 1 (HIF-1). HIF-1 has been shown to upregulate erythropoietin production [62], numerous cell proliferation and survival genes [6365], the angiogenic vascular endothelial growth factor [66], and other genes. It has also been suggested that the hypoxic bone marrow niche maintains HIF-1 activity, thereby maintaining stem cells [67] - a hypothesis supported by the fact that hypoxic conditions expand human HSCs [68] and HPC populations [6971] in vitro. In response to G-CSF, both the hypoxic environment and HIF-1 expand within the bone marrow compartment [72] and increase production of vascular endothelial growth factor A; however, bone marrow vascular density and permeability are not increased [61]. HIF-1 also increases production of SDF-1 [73] and CXCR4 receptor expression [74], suggesting that hypoxia may be a physiological regulator of this important signaling axis within the hematopoietic niche.

HIF-1 has recently been reported to prevent hematopoietic cell damage caused by overproduction of reactive oxygen species [75], suggesting that the hypoxic niche helps maintain the long lifespan of HSCs. However, some small degree of reactive oxygen species signaling may be necessary for HSC mobilization. A recent report demonstrated that enhanced c-Met activity promotes HSPC mobilization by activating mTOR and increasing reactive oxygen species production in HSPCs [76], while inhibition of mTOR with rapamycin reduced HSC mobilization [76, 77]. Genetic knockout of the gene for thioredoxin-interacting protein also results in increased HSPC mobilization under stress conditions [78], suggesting a role for oxygen tension and reactive oxygen species in regulation of hematopoietic stem and progenitor mobilization. These findings clearly warrant additional exploration.

It has been known for some time that there is dynamic interaction between the bone marrow niche and the nervous system. Studies by Katayama and colleagues demonstrated that HSPC mobilization by G-CSF requires peripheral 2-adrenergic signals [21], showing that G-CSF mobilization was reduced in chemically sympathectomized mice treated with 6-hydroxydopamine, in mice treated with the -blocker propanolol, or in mice genetically deficient in the gene for dopamine -hydroxylase (Dbh), an enzyme that converts dopamine into norepinephrine. They also showed that treatment with the 2-adrenergic agonist clenbuterol reversed the phenotype of Dbh knockout mice [21]. Intriguingly, G-CSF attenuated osteoblast function via the sympathetic nervous system resulting in osteoblasts having a marked flattened appearance. The effects of nervous system signaling can also be mediated directly on HSCs, as human CD34+ hematopoietic cells express 2-adrenergic and dopamine receptors that are upregulated after G-CSF treatment [79]. Neurotransmitters serve as direct chemo-attractants to HSPCs, and treatment with norepinephrine results in HSC mobilization [79]. Norepinephrine treatment of mice has also been shown to increase CXCR4 receptor expression [80], perhaps suggesting that adrenergic signaling could directly affect CXCR4/SDF-1 signaling in HSPCs. Additional studies directly assessing effects of neurotransmitter signaling in HSPCs will help to further define the role of the nervous system in hematopoietic regulation.

Not only does the sympathetic nervous system affect HSC mobilization during stress situations, but it also regulates HSC trafficking via a circadian rhythm [81, 82]. 3-Adrenergic stimulations demonstrate regular oscillations controlling norepinephrine release, CXCR4 expression, and SDF-1 production, leading to rhythmic release of HSPCs from the bone marrow niche. Intriguingly, while optimal mobilization occurs in the morning in mice (Zeitgeber time 5), HSC mobilization circadian control is inverted in humans, with peak mobilization occurring later in the evening [81]. Mobilization by both G-CSF and AMD3100 is affected by circadian control of the CXCR4/SDF-1 axis. Recently, it was demonstrated that 2-adrenergic signaling upregulates the vitamin D receptor on osteoblasts; that expression of this receptor is necessary for the G-CSF-induced suppression of osteoblast function; and that vitamin D receptor knockout mice have reduced HSC mobilization [83]. Intriguingly, vitamin D receptor is an important regulator of extracellular calcium and HSPC localization [84] and the receptor is also regulated by circadian rhythms [85], possibly suggesting additional interconnected mobilization mechanisms. Further assessment of the role of nervous system signaling and vitamin D receptor signaling on other niche cells, particularly mesenchymal stem cells, should be performed.

There has been significant progress in understanding the mechanisms of action of G-CSF and other stimuli that increase HSPC trafficking/mobilization. As described in the present review, however, there is currently an abundance of proposed mechanisms that may be responsible for mobilization. This raises the question of whether the proposed mechanisms, be they HSPC intrinsic or manifested through the bone marrow niche, truly represent alternate and independent means to mobilize or enhance egress of HSPCs from bone marrow to the circulation, or whether we have not yet found the unifying mechanism.

Intriguingly, many of the proposed mechanisms of mobilization converge on the CXCR4/SDF-1 pathway (Figure

). Alterations of the osteoblast/osteoclast balance result in a reduction of SDF-1 production and/or degradation of SDF-1 by proteases. Signaling from the sympathetic nervous system, stimulated by G-CSF, can alter the osteoblast/osteoclast balance leading to reduced CXCR4/SDF-1 signaling and HSPC mobilization. Circadian rhythms act to reduce niche SDF-1 production and HSPC CXCR4 expression in an oscillating manner, suggesting that clinical mobilization should be performed at the trough of SDF-1 and CXCR4 expression (early night for humans) and perhaps suggesting that clinical transplantation should be performed at the peak of expression (early morning in humans). The hypoxic nature of the hematopoietic bone marrow niche may itself regulate the CXCR4/SDF-1 signaling axis, perhaps further identifying this axis as a unifying mobilization mechanism. The importance of CXCR4 signaling in HSPC retention and mobilization is certainly supported by the abundance of agents that directly antagonize, or compete with SDF-1 and partially agonize, the CXCR4 receptor and result in HSPC mobilization. Even a rapid mobilizing agent such as GRO (CXCR2 agonist) may function by increasing proteolytic cleavage of SDF-1 [

,

], or altering a homeostatic balance between the CXCR4 and CXCR2 signaling pathways [

].

Hematopoietic stem and progenitor mobilization converges on the CXCR4/SDF-1 signaling axis within the hematopoietic niche. Many of the proposed mechanisms for hematopoietic stem and progenitor mobilization function by altering the marrow microenvironmental CXC4 chemokine receptor (CXCR4)/stromal cell-derived factor 1 (SDF-1) signaling axis. Shown are representative mobilization mechanisms and their relationship to the CXCR4/SDF-1 axis. Question marks denote hypothetical linkage to the CXCR4/SDF-1 axis. G-CSF, granulocyte colony-stimulating factor; HSC, hematopoietic stem cell; HSPC, hematopoietic stem and progenitor cell; ROS, reactive oxygen species.

While perhaps connecting many of the proposed mechanistic pathways for HSPC mobilization, however, the CXCR4/SDF-1 pathway does not appear to be an exclusive target for HSPC mobilization. Continued investigation of the molecular mechanism(s) for action of G-CSF and other HSPC mobilizers is warranted and may define new molecular targets that can be used to enhance the magnitude and/or ease of HSPC collection for hematopoietic transplant.

This article is part of a review series on Stem cell niche. Other articles in the series can be found online at http://stemcellres.com/series/ stemcellniche

CXC4 chemokine receptor

granulocyte colony-stimulating factor

hypoxia inducible factor 1

hematopoietic progenitor cell

hematopoietic stem cell

hematopoietic stem and progenitor cell

mammalian target of rapamycin

receptor activator NF-B

stromal cell-derived factor 1

vascular cell adhesion molecule 1

late antigen 4.

The present work was supported by NIH grants HL069669 and HL096305 (to LMP). JH is supported by training grant HL007910.

Below are the links to the authors original submitted files for images.

The authors declare that they have no competing interests.

Read the original:
Mobilization of hematopoietic stem cells from the bone ...

Comparison Between Bone Marrow or Peripheral Blood Stem …

Comparison Between Bone Marrow or Peripheral Blood Stem Cells and Cord Blood Donated for Transplantation

Cord blood transplants, as all unrelated hematopoietic stem cell transplants, can be associated with serious complications, severe organ toxicity, and in some cases, death.

A transplant requires donation of a quart or more of bone marrow (mixed with blood).

After a formal search is started, it usually takes 2 or more months to transplant, if a donor is available.

When a match is found, it can take only a few days for confirmatory and special testing for shipment to the Transplant Center (less than 24 hours in an emergency).

Donor may be available to give a second transplant or to donate blood for T-cells if necessary.

Patient must begin conditioning before the bone marrow or peripheral bloods harvest. Coordination between donation and transplant is critical and complex.

Cord blood graft can be shipped to the transplant center before the patient enters the hospital and begins conditioning for transplantation. Coordination is simple. Cord blood units are shipped on demand.

No risk of transplanting a genetic disease.

There is a small probability that a rare, unrecognized genetic disease affecting the blood or immune system of the baby may be given with the cord blood transplant.

Generally requires a perfect match between donor and recipient for 8/8 HLA-A, -B, -C and -DRB1 antigens. Additional HLA factors (HLA-DQ and -DP) increasingly used to improve prognosis.

HLA-mismatched cord blood transplants are possible, making it easier to find a suitable match. Role of HLA-C, -DQ and -DP are not yet known.

Link:
Comparison Between Bone Marrow or Peripheral Blood Stem ...

Bone Marrow Stromal Stem Cells: Nature, Biology, and …

Introduction

The post-natal bone marrow has traditionally been seen as an organ composed of two main systems rooted in distinct lineagesthe hematopoietic tissue proper and the associated supporting stroma. The evidence pointing to a putative stem cell upstream of the diverse lineages and cell phenotypes comprising the bone marrow stromal system has made marrow the only known organ in which two separate and distinct stem cells and dependent tissue systems not only coexist, but functionally cooperate. Originally examined because of their critical role in the formation of the hematopoietic microenvironment (HME), marrow stromal cells later came to center stage with the recognition that they are the stem/progenitor cells of skeletal tissues. More recent data pointing to the unexpected differentiation potential of marrow stromal cells into neural tissue or muscle grant them membership in the diverse family of putative somatic stem cells. These cells exist in a number of post-natal tissues that display transgermal plasticity; that is, the ability to differentiate into cell types phenotypically unrelated to the cells in their tissue of origin.

The increasing recognition of the properties of marrow stromal cells has spawned a major switch in our perception of their nature, and ramifications of their potential therapeutic application have been envisioned and implemented. Yet, several aspects of marrow stromal cell biology remain in question and unsettled throughout this evolution both in general perspective and in detail, and have gained further appeal and interest along the way. These include the identity, nature, developmental origin and in vivo function of marrow stromal cells, and their amenability to ex vivo manipulation and in vivo use for therapy. Just as with other current members of the growing list of somatic stem cells, imagination is required to put a finger on the seemingly unlikely properties of marrow stromal cells, many of which directly confront established dogmas or premature inferences made from other more extensively studied stem cell systems.

Alexander Friedenstein, Maureen Owen, and their coworkers were the first to utilize in vitro culture and transplantation in laboratory animals, either in closed systems (diffusion chambers) or open systems (under the renal capsule, or subcutaneously) to characterize cells that compose the physical stroma of bone marrow [1-3]. Because there is very little extracellular matrix present in marrow, gentle mechanical disruption (usually by pipetting and passage through syringe needles of decreasing sizes) can readily dissociate stroma and hematopoietic cells into a single-cell suspension. When these cells are plated at low density, bone marrow stromal cells (BMSCs) rapidly adhere and can be easily separated from the nonadherent hematopoietic cells by repeated washing. With appropriate culture conditions, distinct colonies are formed, each of which is derived from a single precursor cell, the CFU-F.

The ratio of CFU-F in nucleated marrow cells, as determined by the colony-forming efficiency (CFE) assay [4], is highly dependent on the culture conditions, and there is a great deal of variability in the requirements from one animal species to another. In rodents, irradiated marrow feeder cells are absolutely required in addition to selected lots of serum in order to obtain the maximum number of assayable CFU-F (100% CFE), whereas CFE is feeder cell-independent in humans [5]. The mitogenic factors that are required to stimulate the proliferation of CFU-F are not completely known at this time, but do at least include platelet-derived growth factor (PDGF), epidermal growth factor (EGF), basic fibroblast growth factor, transforming growth factor-, and insulin-like growth factor-1 [6, 7]. Under optimal conditions, multi-colony-derived strains (where all colonies are combined by trypsinization) can undergo over 25 passages in vitro (more than 50 cell doublings), demonstrating a high capacity for self-replication. Therefore, billions of BMSCs can be generated from a limited amount of starting material, such as 1 ml of a bone marrow aspirate. Thus, the in vitro definition of BMSCs is that they are rapidly adherent and clonogenic, and capable of extended proliferation.

The heterogeneous nature of the BMSC population is immediately apparent upon examination of individual colonies. Typically this is exemplified by a broad range of colony sizes, representing varying growth rates, and different cell morphologies, ranging from fibroblast-like spindle-shaped cells to large flat cells. Furthermore, if such cultures are allowed to develop for up to 20 days, phenotypic heterogeneity is also noted. Some colonies are highly positive for alkaline phosphatase (ALP), while others are negative, and a third type is positive in the central region, and negative in the periphery [8]. Some colonies form nodules (the initiation of matrix mineralization) which can be identified by alizarin red or von Kossa staining for calcium. Yet others accumulate fat, identified by oil red O staining [9], and occasionally, some colonies form cartilage as identified by alcian blue staining [10].

Upon transplantation into a host animal, multi-colony-derived strains form an ectopic ossicle, complete with a reticular stroma supportive of myelopoiesis and adipocytes, and occasionally, cartilage [8, 11]. When single colony-derived BMSC strains (isolated using cloning cylinders) are transplanted, a proportion of them have the ability to completely regenerate a bone/marrow organ in which bone cells, myelosupportive stroma, and adipocytes are clonal and of donor origin, whereas hematopoiesis and the vasculature are of recipient origin [7] (Fig. 1). These results define the stem cell nature of the original CFU-F from which the clonal strain was derived. However, they also confirm that not all of the clonogenic cells (those cells able to proliferate to form a colony) are in fact multipotent stem cells. It must also be noted that it is the behavior of clonal strains upon transplantation, and not their in vitro phenotype, that provides the most reliable information on the actual differentiation potential of individual clones. Expression of osteogenic, chondrogenic, or adipogenic phenotypic markers in culture (detected either by mRNA expression or histochemical techniques), and even the production of mineralized matrix, does not reflect the degree of pluripotency of a selected clone in vivo [12]. Therefore, the identification of stem cells among stromal cells is only done a posteriori and only by using the appropriate assay. In this respect, chondrogenesis requires an additional comment. It is seldom observed in open transplantation assays, whereas it is commonly seen in closed systems such as diffusion chambers [11], or in micromass cultures of stromal cells in vitro [13], where locally low oxygen tensions, per se, permissive for chondrogenesis, are attained [14]. Thus, the conditions for transplantation or even in vitro assays are critical determinants of the range of differentiation characteristics that can be assessed.

FigureFigure 1.. Transplantation of ex vivo-expanded human BMSC into the subcutis of immunocompromised mice.A) Multi-colony and some single colony-derived strains attached to particles of hydroxyapatite/tricalcium phosphate ceramic (HA) form a complete bone/marrow organ composed of bone (B) encasing hematopoietic marrow (HP). B) The bone (B) and the stroma (S) are of human origin as determined by in situ hybridization using a human specific alu sequence as probe, while the hematopoietic cells are of recipient origin.

Download figure to PowerPoint

The ability to isolate the subset of marrow stromal cells with the most extensive replication and differentiation potential would naturally be of utmost importance for both theoretical and applicative reasons. This requires definitive linkage of the multipotency displayed in transplantation assays with a phenotypic trait that could be assessed prior to, and independently of, any subsequent assays. Several laboratories have developed monoclonal antibodies using BMSCs as immunogen in order to identify one or more markers suitable for identification and sorting of stromal cell preparations [15-18]. To date, however, the isolation of a pure population of multipotent marrow stromal stem cells remains elusive. The nearest approximation has been the production of a monoclonal antibody, Stro-1, which is highly expressed by stromal cells that are clonogenic (Stro-1+bright), although a certain percentage of hematopoietic cells express low levels of the antigen (Stro-1+dull) [19]. In principle, the use of the same reagent in tissue sections would be valuable in establishing in vivo-in vitro correlation, and in pursuing the potential microanatomical niches, if not anatomical identity, of the cells that are clonogenic. The Stro-1 reagent has limited application in fixed and paraffin-embedded tissue. However, preliminary data using frozen sections suggest that the walls of the microvasculature in a variety of tissues are the main site of immunoreactivity (Fig. 2), a finding of potentially high significance (see below).

FigureFigure 2.. Immunolocalization of the Stro-1 epitope in the microvasculature of human thymus.A) CD34 localizes to endothelial cells (E) forming the lumen (L) of the blood vessel. B) Stro-1 localizes not only to endothelial cells, but also the perivascular cells of the blood vessel wall (BVW).

Download figure to PowerPoint

Freshly isolated Stro-1+bright cells and multi-colony-derived BMSC strains, both of which contain but are not limited to multipotent stromal stem cells, have been extensively characterized for a long list of markers expressed by fibroblasts, myofibroblasts, endothelial cells, and hematopoietic cells in several different laboratories [20-24]. From these studies, it is apparent that the BMSC population at large shares many, but not all, properties of fibroblastic cells such as expression of matrix proteins, and interestingly, some markers of myofibroblastic cells, notably, the expression of -smooth muscle actin (-SMA) and some characteristics of endothelial cells such as endoglin and MUC-18. It has been claimed that the true mesenchymal stem cell can be isolated using rather standard procedures, and characterized using a long list of indeterminate markers [23]. However, in spite of this putative purification and extensive characterization, the resulting population was no more pure than multi-colony-derived strains isolated by simple, short-term adherence to plastic; the resulting clones displayed varying degrees of multipotentiality. Furthermore, the pattern of expressed markers in even clonal strains that are able to completely regenerate a bone/marrow organ in vivo is not identical, and changes as a function of time in culture. These results indicate that identifying the phenotypic fingerprint of a stromal stem cell may well be like shooting at a moving target, in that they seem to be constantly changing in response to their microenvironment, both in vitro and in vivo.

The primitive marrow stroma is established in development through a complex series of events that takes place following the differentiation of primitive osteogenic cells, the formation of the first bone, and the vascular invasion of bone rudiments [25]. This intimate relationship of the stromal cells with the marrow vascularity is also found in the adult marrow. In the post-natal skeleton, bone and bone marrow share a significant proportion of their respective vascular bed [26]. The medullary vascular network, much like the circulatory system of other organs, is lined by a continuous layer of endothelial cells and subendothelial pericytes [27]. In the arterial and capillary sections of this network, pericytes express both ALP (Fig. 3B, C, D, F, G) and -SMA (Fig. 3E), both of which are useful markers for their visualization in tissue sections. In the venous portion, cells residing on the abluminal side of the endothelium display a reticular morphology, with long processes emanating from the sinus wall into the adjacent hematopoietic cords where they establish close cell-cell contacts, that convey microenvironmental cues to maturing blood cells. These particular adventitial reticular cells express ALP (Fig. 3G) but not -SMA under normal steady-state conditions (Fig. 3H). In spite of this, but in view of their specific position along with the known diversity of pericytes in different sites, organs and tissues [28], reticular cells can be seen as bona fide specialized pericytes of venous sinusoids in the marrow. Hence, phenotypic properties of marrow pericytes vary along the different sections of the marrow microvascular network (arterial/capillary versus post-capillary venous sinusoids). In addition, adventitial reticular cells of venous sinusoids can accumulate lipid and convert to adipocytes, and they do so mainly under two circumstances: A) during growth of an individual skeletal segment when the expansion of the total marrow cavity makes available space in excess of what is required by hematopoietic cells, or B) independent of growth, when there is an abnormal or age-related numerical reduction of hematopoietic cells thereby making space redundant [29-31].

FigureFigure 3.. Anatomical and immunohistological relationship of marrow stromal cells to marrow pericytes.A) Marrow vascular structures as seen in a histological section of human adult bone marrow. hc = hematopoietic cells; ad = adipocytes; a = artery; VS = venous sinusoid; PCA = pre-capillary arteriole. Note the thin wall of the venous sinusoid. B) Semi-thin section from low-temperature processed glycol-methacrylate embedded human adult bone marrow reacted for ALP. Arrows point to three arterioles emerging from a parent artery (A). Note that while there is no ALP activity in the wall of the large size parent artery, a strong reaction is noted in the arteriolar walls. C, D) Details of the arterioles shown in A and B. Note that ALP activity is associated with pericytes (P). E) Section of human adult bone marrow immunolabeled for -SMA. Note the reactivity of an arteriolar wall, and the complete absence of reactivity in the hematopoietic cords (hc) interspersed between adipocytes (ad). F) Detail of the wall of a marrow venous sinusoid lined by thin processes of adventitial reticular cells (venous pericytes). Note the extension of cell processes apparently away from the wall of the venous sinusoid (vs) and into the adjacent hematopoietic cord ALP reaction. G, H) High power views of hematopoietic cords in sections reacted for ALP (G) and -SMA (H). Note the presence of ALP activity identifying reticular cells, and the absence of labeling for -SMA.

Download figure to PowerPoint

The ability of reticular cells to convert to adipocytes makes them a unique and specialized pericyte. Production of a basement membrane by adipocytes endows the sinus with a more substantial basement membrane, likely reducing the overall permeability of the vessel. Furthermore, the dramatic increase in cell volume through the accumulation of lipid during adipose conversion collapses the lumen of the sinus. This may exclude an individual sinus from the circulation without causing its irreversible loss. In general, the loss of pericyte coating on a microvessel is associated with vessel regression by apoptosis, while a normal pericyte coating is thought to stabilize them and prevent vessel pruning [32]. Adipose conversion is thus a mechanism whereby the size and permeability of the overall sinusoidal system is reversibly regulated in the bone marrow. Not surprisingly, regions of bone marrow that are hematopoietically inactive are filled with fat.

Given the similar location of pericytes and stromal cells, the significance of -SMA expression, a marker of smooth muscle cells, in marrow stromal cells takes on new meaning, although its expression is variable, both in vitro and in vivo. -SMA expression is commonly observed in nonclonal, and some clonal cultures of marrow stromal cells [33], where it appears to be related to phases of active cell growth [34], and may reflect a myoid differentiation event, at least in vitro [35]. However, the phenotype of -SMA-expressing stromal cells in culture resembles that of pericytes and subintimal myoid cells rather than that of true smooth muscle cells [35]. In the steady-state normal bone marrow, -SMA expressing stromal cells other than those forming the pericyte/smooth muscle coats of arteries and capillaries are not seen. In contrast, -SMA+ stromal cells not associated with the vasculature are commonly observed in the fetal bone marrow [36, 37], that physically grows together with the bone encasing it. -SMA+ marrow stromal cells are likewise seen in conjunction with a host of hematological diseases [37], and in some bone diseases, such as hyperparathyroidism [37] and fibrous dysplasia (FD) of bone (Riminucci and Bianco, unpublished results). In some of these conditions, these cells have been interpreted as myofibroblasts [34, 37]. More interestingly, at least some of these conditions also feature an increased vascularity, possibly related to angiogenesis [38], and an increased number of CFU-F, quantitated as discussed above (Bianco, Kuznetsov, Robey, unpublished results). Taken together, these observations seem to indicate that -SMA expression in extravascular marrow stromal cells (other than arterial/ capillary pericytes) is related to growth or regeneration events in the marrow environment, which is in turn associated with angiogenesis.

Angiogenesis in all tissues involves the coordinated growth of endothelial cells and pericytes. Nascent endothelial tubes produce EGF and PDGF-B, which stimulate the growth and migration of pericytes away from the subintimal myoid cell layer of the vascular section. A precise ligand-receptor expression loop of PDGF-B produced by endothelial cells and expression of the cognate receptor on pericytes regulates the formation of a pericyte coating and its occurrence in physical continuity with the nascent vascular network [39]. Interestingly, PDGF-receptor beta and EGF receptor are two of the most abundant tyrosine kinase growth factor receptors in BMSCs, and PDGF-B and EGF have been found to stimulate proliferation of BMSCs [6, 40], indicating a physiological similarity between pericytes and BMSCs.

In bone, as in any other organ, angiogenesis is normally restricted to phases of developmentally programmed tissue growth, but may reappear in tissue repair and regeneration or proliferative/neoplastic diseases. During normal bone growth, endothelial cell growth, pericyte coverage, and bone formation by newly generated bone-forming cells occur in a precise spatial and temporal sequence, best visualized in metaphyseal growth plates. Growing endothelial tubes devoid of pericytes occupy the foremost 200 microns of the developing metaphysis [41]. Actively dividing abluminal pericytes and bone-forming osteoblasts are next in line. Progression of endochondral bone formation is dependent on efficient angiogenesis, and is blocked if angiogenesis is blocked, as illustrated by both experimental and pathological conditions. Experimentally, inhibition of VEGF signaling initiated by chondrocytes with blocking antibodies to the cognate receptor on growing blood vessels in the metaphysis results in a blockade not only of bone growth, but also of the related activities in the adjacent cartilage growth plates [42]. A remarkably similar event occurs naturally in rickets, and can be mimicked by microsurgical ablation of the metaphyseal vasculature [41].

Taking into account the similarities in their physical relationship to the vasculature, the cellular response to growth factors, and expression of similar markers lead one to suspect that marrow pericytes and marrow stromal cells are the same entity. Pericytes are perhaps one of the most elusive cell types in the body, and their significance as potential progenitor cells has been repeatedly surmised or postulated [28, 43-46]. Elegant as much as unconventional, experimental proof of their ability to generate cartilage and bone in vivo, for example, has been given in the past [47, 48]. Likewise, it has been shown that retinal pericytes form cartilage and bone (and express Stro-1) in vitro [49]. But, there has been little definitive understanding of the origin of this elusive cell type. Current evidence suggests that there is most likely more than one source of pericytes throughout development and growth. First, during development, pericytes may be recruited during angiogenesis or vasculogenesis from neighboring resident mesenchymal cells [50]. Secondly, as recently shown, pericytes may arise directly from endothelial cells or their progenitors [51, 52]. Third, they can be generated during angiogenesis, either pre- or post-natally, through replication, migration and differentiation of other pericytes downstream of the growing vascular bud [32, 39, 53, 54]. With regards to bone marrow, this implies that marrow pericytes might also be heterogeneous in their mode of development and origin. Some may be recruited during blood vessel formation from resident, preexisting osteogenic cells; others may originate from endothelial cells; still others may grow from preexisting pericytes during vascular growth. Interestingly, it would be predicted from this model that a hierarchy of marrow stromal/progenitor cells exists. Some would be osteogenic in nature, while others would not. If so, one would expect to find multipotent cells with markers of osteogenic commitment, and multipotent cells with endothelial/pericytic markers. With respect to the phenotypic characterization of clonal stromal cells, evidence supporting a dual origin is indeed available.

As described above, stromal cells can take on many forms such as cartilage, bone, myelosupportive stroma, or fat. This behavior of marrow stromal cells, both in vitro and in vivo, has perhaps offered the first glimpse of the property now widely referred to as plasticity. It was shown, for example, that clonal strains of marrow adipocytes could be directed to an osteogenic differentiation and form genuine bone in an in vivo assay [55, 56]. Earlier, the ability of marrow reticular cells to convert to adipocytes in vivo had been noted [29, 57]. A number of different studies have claimed that fully differentiated chondrocytes can dedifferentiate in culture and then shift to an osteogenic phenotype [58, 59], and that similar or correlated events can be detected in vivo [60]. All of these data highlight the non-irreversible nature of the differentiation of several cell types otherwise seen as end points of various pathways/lineages (i.e., reticular cells, osteoblasts, chondrocytes, and adipocytes). The primary implication of these findings has remained largely unnoticed until recently. Commitment and differentiation are not usually thought of as reversible, but rather as multistep, unidirectional and terminal processes. This concept is reflected in the basic layout of virtually every scheme in every textbook depicting the organization of a multilineage system dependent on a stem cell. Here, a hierarchy of progenitors of progressively restricted differentiation potential is recognized or postulated. Lineages are segregated, leaving no room for switching phenotype at a late stage of differentiation, no way of turning red blood cells into white blood cells, for example. In contrast, it seems that one can turn an adipocyte or a chondrocyte into an osteoblast, and nature itself seems to do this under specific circumstances. If so, then some kind of reversible commitment is maintained until very late in the history of a single cell of the stromal systema notable and yet unnoticed singularity of the system, with broad biological significance.

There is a real physiological need for plasticity of connective tissue cells, namely the need to adapt different tissues that reside next to one another during organ growth, for example [30, 61], and it is likely that nature has evolved mechanisms for maintaining plasticity which remain to be fully elucidated. One example may be the key transcription factor controlling osteogenic commitment, cbfa1 [62, 63], which is commonly if not constitutively expressed in stromal cells derived in culture from the post-natal marrow [12], and maintained during differentiation towards other cell types such as adipocytes. This is perhaps the most stringent proof that a cell committed to osteogenesis (as demonstrated by expression of the key gene of commitment) may still enter other pathways of differentiation that were thought to be alternative ones [61]. Whether one can isolate a multipotent cbfa1-negative (non-osteogenically committed) stromal cell is at present unclear. However, freshly isolated stromal cells sorted as Stro-1bright have been shown to be cbfa1-negative by reverse transcriptase-polymerase chain reaction (Gronthos and Simmons, unpublished results). Interestingly, these cells also exhibit several endothelial markers, although never a true endothelial phenotype [21, 22].

The fact that chondrocytes, osteoblasts, reticular cells, and adipocytes come from a single precursor cell carrying a marker of osteogenic commitment is consistent with the fact that all of these cell types are members of the same organ, even though of different tissues. A single skeletal segment contains all of these cell types either at different stages of its own organogenesis or simultaneously. Although heretical to some and novel to others, even the notion that each of these cell phenotypes can switch to another within the same family under specific circumstances is consistent with the development and maintenance of the organ from which they were derived. This kind of plasticity is thus orthodox, meaning that it remains within the context of the organ system.

Over the past 2 years, several studies have indicated or implied that progenitors can be found in a host of different post-natal tissues with the apparently unorthodox potential of differentiating into unrelated tissues. First, it was shown that the bone marrow contained systemically transplantable myogenic progenitors [64]. Second, it was shown that neural stem cells could reestablish hematopoiesis in irradiated mice [65]; third, that bone marrow cells could generate neural cells [66], and hepatocytes [67]; and fourth, that a neurogenic potential could be ascribed to marrow stromal cells [68, 69]. What is striking about these data is the developmentally distant nature of the source of these progenitors and their ultimate destination. Differentiation across germ layers violates a consolidated law of developmental biology. Although consolidated laws are not dogmas (which elicited the comment that germ layers are more important to embryologists than to embryos), it is still indisputable and remarkable that even in embryos, cells with transgermal potential only exist under strict temporal and spatial constraints, with the notable exception of neural crest cells, which in spite of their neuroectodermal nature generate a number of craniofacial mesodermal tissues including bone. Cells grown in culture from the inner cell mass self-renew and maintain totipotency in culture for extended periods of time. However, this is in a way an artifact, of which we know some whys and wherefores (feeder cell layers, leukemia inhibitory factor). Embryonic stem (ES) cells only remain multipotent and self-renewing in the embryo itself for a very short period of time, after which totipotent cells only exist in the germline.

Consequently, the first key question iswhere do the multipotent cells of post-natal organisms come from? All answers at this time are hypothetical at best. However, if marrow stromal cells are indeed members of a diffuse system of post-natal multipotent stem cells and they are at the same time vascular/pericytic in nature/origin, then a natural corollary would read that perhaps the microvasculature is a repository of multipotent cells in many, if not all, tissues [70]a hypothesis that is currently being tested.

A second question is that if multipotent cells are everywhere, or almost everywhere, then what are the mechanisms by which differentiated cells keep their multipotency from making every organ a teratoma? Phrased in another way, adult tissues must retain some kind of organizing ability previously thought of as specific to embryonic organizers. If indeed cells in the bone marrow are able to become muscle or liver or brain, then there must be mechanisms ensuring that there is no liver or brain or muscle in the marrow. Hence, signals for maintenance of a tissue's self must exist and be accomplished by differentiated cells. (That is, of course, if stem cells are not differentiated cells themselves).

A third question ishow much of the stemness (self-renewal and multipotency) observed in experimental systems is inherent to the cells that we manipulate, and how much is due to the manipulation? Are we discovering unknown and unexpected cells, or rather unknown and unexpected effects of manipulation of cells in culture? To what extent do cell culture conditions mimic the effects of an enucleated oocyte cytoplasm, which permits a somatic cell nucleus to generate an organism such as Dolly, the cloned sheep? For sure, a new definition of what a stem cell isa timely, and biotechnologically correct, oneshould incorporate the conditions under which phenomena are recorded, rather than guessing from ex vivo performance what the true in vivo properties are. This exercise also has important implications for understanding where and when stem cells come into action in physiology. Even for the mother of all stem cells, the ES cell, self-renewal and multipotency are limited to specific times and events in vivo, and are much less limited ex vivo. Are similar constraints operating for other stem cells? Marrow stromal stem cells for example, can be expanded extensively in culture, but the majority of them likely never divide in vivo once skeletal growth has ceased (except the few that participate in bone turnover, and perhaps in response to injury or disease). What physiological mechanism calls for resumption of a stem cell behavior in vivo in the skeleton and other systems?

All of these questions are important not only for philosophical or esoteric reasons, but also for applicative purposes. Knowing even a few of the answers will undoubtedly enable biotechnology to better harness the magical properties of stem cells for clinical applications.

In vivo transplantation under defined experimental conditions has been the gold standard for defining the differentiation potential of marrow stromal cells, and a cardinal element of their very discovery. Historically, studies on the transplantability of marrow stromal cells are inscribed into the general problem of bone marrow transplantation (BMT). The HME is created by transplantation of marrow stromal cell strains and allows for the ectopic development of a hematopoietic tissue at the site of transplantation. The donor origin of the microenvironment and the host origin of hematopoiesis make the ectopic ossicle a true reverse BMT.

Local transplantation of marrow stromal cells for therapeutic applications permits the efficient reconstruction of bone defects larger than those that would spontaneously heal (critical size). A number of preclinical studies in animal models have convincingly shown the feasibility of marrow stromal cell grafts for orthopedic purposes [71-77], even though extensive work lies ahead in order to optimize the procedures, even in their simplest applications. For example, the ideal ex vivo expansion conditions have yet to be determined, or the composition and structure of the ideal carrier, or the numbers of cells that are required for regeneration of a volume of bone.

In addition to utilizing ex vivo-expanded BMSCs for regeneration of bone and associated tissues, evidence of the unorthodox plasticity of marrow stromal cells has suggested their potential use for unorthodox transplantation; that is, for example, to regenerate neural cells or deliver required gene products at unorthodox sites such as the central nervous system (CNS) [78]. This could simplify an approach to cell therapy of the nervous system by eliminating the need for harvesting autologous human neural stem cells, an admittedly difficult procedure, although it is currently believed that heterologous cells may be used for the CNS, given the immune tolerance of the brain. Moreover, if indeed marrow stromal cells represent just a special case of post-natal multipotent stem cells, there is little doubt that they represent one of the most accessible sources of such cells for therapeutic use. The ease with which they are harvested (a simple marrow aspirate), and the simplicity of the procedures required for their culture and expansion in vitro may make them ideal candidates. For applicative purposes, understanding the actual differentiation spectrum of stromal stem cells requires further investigation. Besides neural cells, cardiomyocytes have been reported to represent another possible target of stromal cell manipulation and transplantation [79]. It also remains to be determined whether the myogenic progenitors found in the marrow [64] are indeed stromal (as some recent data would suggest, [80]) or non-stromal in nature [81], or both.

Given their residency in the marrow, and the prevailing view that marrow stromal cells fit into the hematopoietic paradigm, it was unavoidable that systemic transplantation of marrow stromal cells would be attempted [82] in order to cure more generalized skeletal diseases based on the successes of hematopoietic reconstitution by BMT. Yet major uncertainties remain in this area. Undoubtedly, the marrow stromal cell is the entity responsible for conveying genetic alterations into diseases of the skeleton. This is illustrated very well by the ability of these cells to recapitulate natural or targeted genetic abnormalities into abnormal bone formation in animal transplantation assays [83-85]. As such, they also represent a potential repository for therapy to alleviate bone disease. However, a significant rationale for the ability of stromal cells to colonize the skeleton once infused into the circulation is still missing.

The stroma is not transplanted along with hematopoiesis in standard BMT performed for hematological or oncological purposes [86-88]. Infusion of larger numbers of stromal cells than those present in cell preparations used for hematological BMT should be investigated further, as it might result, in principle, in limited engraftment. Stringent criteria must be adopted when assessing successful engraftment of systemically infused stromal cells [61]. The detection of reporter genes in tissue extracts or the isolation in culture of cells of donor origin does not prove cell engraftment; it proves cell survival. In this respect, it should be noted that even intra-arterial infusion of marrow stromal cells in a mouse limb may result in virtually no engraftment, even though abundant cells of donor origin are found impacted within the marrow microvascular network. Of note, these nonengrafted cells would routinely be described as engrafted by the use of any reporter gene or ex vivo culture procedure. Less than stringent definitions of stromal cells (for example, their identification by generic or nonspecific markers) must be avoided when attempting their detection in the recipient's marrow. Clear-cut evidence for the sustained integration in the target tissue of differentiated cells of donor origin must be provided. This is rarely the case in current studies claiming engraftment of marrow stromal cells to the skeleton. Some evidence for a limited engraftment of skeletal progenitors following systemic infusion has, however, been obtained in animal models [89, 90]. These data match similar evidence for the possible delivery of marrow-derived myogenic progenitors to muscle via the systemic circulation [64]. It should be kept in mind that both skeletal and muscle tissues are normally formed during development and growth by extravascular cells that exploit migratory processes not involving the circulation. Is there an independent circulatory route for delivery of progenitors to solid phase tissues, and if so, are there physiologically circulating mesodermal progenitors? From where would these cells originate, both in development and post-natal organisms, and how would they negotiate the vessel wall? Addressing these questions is mandatory and requires extensive preclinical work.

Even once these issues are addressed, kinetic considerations regarding skeletal growth and turnover represent another major hurdle that must be overcome in order to cure systemic skeletal diseases via systemic infusion of skeletal progenitors. Yet there is broad opportunity for the treatment of single clinical episodes within the context of skeletal disease. While curing osteogenesis imperfecta by replacing the entire population of mutated skeletal progenitors with normal ones may remain an unattainable goal, individual fractures or deformity in osteogenesis imperfecta or FD of bone could be successfully treated with ex vivo repaired stromal cells, for example. Towards this end, future work must focus on the feasibility of transducing or otherwise genetically correcting autologous mutated osteoprogenitors ex vivo, and studies are beginning to move in this direction.

Molecular engineering of cells, either transiently or permanently, has become a mainstay in cell and molecular biology, leading to many exciting insights into the role of a given protein in cell metabolism both in vitro and in vivo. Application of these techniques for correcting human deficiencies and disease is a challenge that is currently receiving much attention. BMSCs offer a unique opportunity to establish transplantation schemes to correct genetic diseases of the skeleton. They may be easily obtained from the future recipient, manipulated genetically and expanded in number before reintroduction. This eliminates not only the complications of xenografts, but also bypasses the limitations and risks connected with delivery of genetic repair material directly to the patient via pathogen-associated vectors. While a similar strategy may be applied to ES cells, the use of post-natal BMSCs is preferable considering that they can be used autologously, thereby avoiding possible immunological complications from a xenograft. Furthermore, there is far less concern of inappropriate differentiation as may occur with ES cell transplantation. Finally, ES cell transplantation is highly controversial, and it is likely that the ethical debate surrounding their usage will continue for quite some time.

Depending on the situation, there are several approaches that can be envisioned. If a short-lived effect is the goal, such as in speeding up bone regeneration, transient transduction would be the desired outcome, utilizing methods such as electroporation, chemical methods including calcium phosphate precipitation and lipofection, and plasmids and viral constructs such as adenovirus. Transducing BMSCs with adenoviral constructs containing BMP-2 has demonstrated at least partial efficacy of this approach in hastening bone regeneration in animal models [75, 91, 92]. Adenoviral techniques are attractive due to the lack of toxicity; however, the level at which BMSCs are transfected is variable, and problematic. It has been reported that normal, non-transformed BMSCs require 10 more infective agent than other cell types [93], which is often associated with cellular toxicity. Clearly, further optimization is needed for full implementation of this approach.

For treatment of recessive diseases in which a biological activity is either missing or diminished, long-lasting or permanent transduction is required, and has depended on the use of adeno-associated viruses, retroviruses, lentiviruses (a subclass of retrovirus), and more recently, adeno-retroviral chimeras [94]. These viruses are able to accommodate large constructs of DNA (up to 8 kb), and while retroviruses require active proliferation for efficient transfection, lentiviruses do not. Exogenous biological activity in BMSCs by transduction with retroviral constructs directing the synthesis of reporter molecules, interleukin 3, CD-2, Factor VIII, or the enzymes that synthesize L-DOPA has been reported [78, 95-102]. However, these studies also highlight some of the hurdles that must be overcome before this technology will become practical. The first hurdle is optimization of ex vivo transfection. It has been reported that lengthy ex vivo expansion (3-4 weeks) to increase cell numbers reduces transfectability of BMSCs, whereas short-term culture (10-12 days) does not [98]. Furthermore, high levels of transduction may require multiple rounds of transfection [95, 101]. The second hurdle relates to the durability of the desired gene expression. No reported study has extended beyond 4 months post-transplantation of transduced cells [99] (Gronthos, unpublished results), and in most instances, it has been reported that expression decreases with time [96], due to promoter inactivation [102] and/or loss of transduced cells (Mankani and Robey, unpublished results). While promising, these results point to the need for careful consideration of the ex vivo methods, choice of promoter to drive the desired biological activity, and assessment of the ability of the transduced BMSCs to retain their ability to self-maintain upon in vivo transplantation. It must also be pointed out that using retrovirally transduced BMSCs for this type of application, providing a missing or decreased biological activity, does not necessarily require that they truly engraft, as defined above. They may be able to perform this function by remaining resident without actually physically incorporating and functioning within a connective tissue. In this case, they can be envisioned as forming an in vivo biological mini-pump as a means of introducing a required factor, as opposed to standard means of oral or systemic administration.

Use of transduced BMSCs for the treatment of a dominant negative disease, in which there is actual expression of misfunctioning or inappropriate biological activity, is far more problematic, independent of whether we are able to deliver BMSCs systemically or orthotopically. In this case, an activity must be silenced such that it does not interfere with any normal activity that is present, or reintroduced by any other means. The most direct approach would be the application of homologous recombination, as applied to ES cells and generation of transgenic animals. The almost vanishing low rate of homologous recombination in current methodology, coupled with issues of the identification, separation, and expansion of such recombinants does not make this seem feasible in the near future. However, new techniques for increasing the rate of homologous recombinations are under development [103] which may make this approach more feasible. Another approach to gene therapy is based on the processes whereby mismatches in DNA heteroduplexes that arise sporadically during normal cell activity are automatically corrected. Genetic mutations could be targeted by introducing exogenous DNA with the desired sequence (either short DNA oligonucleotides or chimeric RNA/DNA oligonucleotides) which binds to homologous sequences in the genome forming a heteroduplex that is then rectified by a number of naturally occurring repair processes [104]. A third option exists using a specially constructed oligonucleotide that binds to the gene in question to form a triple helical structure, thereby disallowing gene transcription [105].

While it would be highly desirable to correct a genetic disease at the genomic level, mRNA represents another very significant target, and perhaps a more accessible one, to silence the activity of a dominant negative gene. Methods for inhibiting mRNA translation and/or increasing its degradation have been employed through the use of protein decoys to prevent association of a particular mRNA to the biosynthetic machinery and antisense sequences (either oligonucleotides or full-length sequences). Double-stranded RNA also induces rapid degradation of mRNA (termed RNA interference, RNAi) by a process that is not well understood [105]. However, eliminating mRNAs transcribed from a mutant allele with short or single-base mutations by these approaches would most likely not maintain mRNA from a normal allele. For this reason, hammerhead and hairpin ribozymes represent yet another alternative, based on their ability to bind to very specific sequences, and to cleave them and inactivate them from subsequent translation. Consequently, incorporating a mutant sequence, even one that transcribes a single base mutation, can direct a hammerhead or hairpin ribozyme to inactivate a very specific mRNA. This approach is currently being probed for its possible use in the treatment of osteogenesis imperfecta [106]. Taking this technology one step further, DNAzymes that mimic the enzymatic activity of ribozymes, which would be far more stable than ribozymes, are also being developed. Regardless of whether genomic or cytoplasmic sequences are the target of gene therapy, the efficacy of all of these new technologies will depend on: A) the efficiency at which the reagents are incorporated into BMSCs in the ex vivo environment; B) the selection of specific targets, and C) the maintenance of the ability of BMSCs to function appropriately in vitro.

In conclusion, the isolation of post-natal stem cells from a variety of tissues along with discovery of their unexpected capabilities has provided us with a new conceptual framework in which to both view them and use them. However, even with this new perspective, there is much to be done to better understand them: their origins, their relationships to one another, their ability to differentiate or re-differentiate, their physiological role during development, growth, and maturity, and in disease. These types of studies will most certainly require a great deal of interdisciplinary crosstalk between investigators in the areas of natal and post-natal development, and in different organ systems. Clearly, as these studies progress, open mindedness will be needed to better understand the nature of this exciting family of cells, as well as to better understand the full utilization of stem cells with or without genetic manipulation. Much to be learned. Much to be gained.

The rest is here:
Bone Marrow Stromal Stem Cells: Nature, Biology, and ...

Bone marrow stem-cells – ScienceDaily

Bone marrow is the tissue comprising the center of large bones.

It is the place where new blood cells are produced.

Bone marrow contains two types of stem cells: hemopoietic (which can produce blood cells) and stromal (which can produce fat, cartilage and bone).

There are two types of bone marrow: red marrow (also known as myeloid tissue) and yellow marrow.

Red blood cells, platelets and most white blood cells arise in red marrow; some white blood cells develop in yellow marrow.

The color of yellow marrow is due to the much higher number of fat cells.

Both types of bone marrow contain numerous blood vessels and capillaries.

At birth, all bone marrow is red.

With age, more and more of it is converted to the yellow type.

Adults have on average about 2.6kg (5.7lbs) of bone marrow, with about half of it being red.

Red marrow is found mainly in the flat bones such as hip bone, breast bone, skull, ribs, vertebrae and shoulder blades, and in the cancellous ("spongy") material at the proximal ends of the long bones femur and humerus.

Pink Marrow is found in the hollow interior of the middle portion of long bones.

There are several serious diseases involving bone marrow.

In cases of severe blood loss, the body can convert yellow marrow back to red marrow in order to increase blood cell production.

The normal bone marrow architecture can be displaced by malignancies or infections such as tuberculosis, leading to a decrease in the production of blood cells and blood platelets.

In addition, cancers of the hematologic progenitor cells in the bone marrow can arise; these are the leukemias.

Read more:
Bone marrow stem-cells - ScienceDaily

Stem | Treatments | DBA | NCBDDD | CDC

In Diamond Blackfan anemia (DBA), the bone marrow (the center of the bone where blood cells are made) does not make enough red blood cells. Red blood cells carry oxygen to all of the organs in the body. When the number of red blood cells is low, the organs in the body may not get the oxygen they need.

A stem cell transplant can help restore the marrows ability to make red blood cells, and it is currently the only known cure for DBA.

However, physical problems associated with DBA but not related to the bone marrow, such as a cleft palate or a heart defect, will not change. In addition, the persons genes will still have DBA, so there is still a 50 percent chance of passing the disorder to any future children, if fertility is retained.

Stem cell transplant is an expensive and potentially dangerous procedure that can lead to death or severe chronic illness in some patients. For this reason, it typically is not a first line treatment. Other treatments, such as steroid medicine (corticosteroid) therapy and blood transfusion therapy, tend to be used first, if possible. Before deciding to have a transplant, people with DBA should discuss the pros and cons of this procedure with their medical team.

All of the blood cells in the body start out as immature cells called blood-forming stem cells. Stem cells are able to grow into other blood cells that mature and function as needed in the body. Stem cells create the three main types of blood cells: red blood cells that carry oxygen throughout the body, white blood cells that fight infection, and platelets that help the blood to clot and prevent abnormal bleeding.

Stem cells are located in three placesbone marrow (the spongy center of the bone where blood cells are made), peripheral blood (found in blood vessels throughout the body), and cord blood (found in the umbilical cord and collected after a babys birth). Stem cells for transplantation are obtained from any of these three places.

A stem cell transplant (also commonly referred to as a bone marrow transplant), takes healthy stem cells from a donor and gives them to the patient through a central line in a vein in the chest. The bag of stem cells usually looks similar to a bag of blood used for blood transfusion. This is because it contains red blood cells. The goal of a stem cell transplant is to replace unhealthy stem cells with new healthy ones. If all goes well, these healthy stem cells find their way to the bone marrow and begin to function and produce blood cells normally (called an engraft). It often takes several weeks for this to happen.

For a person to be a donor, the donated stem cells must closely match the patients Human Leukocyte Antigen (HLA) type. HLA markers are special proteins found on most cells in the body. The immune system uses these proteins or markers to recognize which cells belong in the body and which do not. These markers are inherited from both parents. Special tests called HLA typing or HLA tissue typing determines whether the patient and the donor cells match.

Close family members such as brothers and sisters (but rarely parents) are often used as donors because they are most likely to match the patients tissue type. Each sibling who has the same parents has a 25 percent chance of matching the patients tissue type. However, if a sibling also has one of the DBA genes, it will be passed to the recipient during the transplant. It is important to screen potential donors for DBA genes because there is a risk of transfer from a sibling who has the gene for DBA, but who has no symptoms.

If there is not a brother or sister or other family member who is a match for the patient, the transplant center can check the National Marrow Donor Program (NMDP) registry for an unrelated matching donor. In some instances, unrelated donors may be adequately matched and able to donate. However, the rate of successful transplant from matched unrelated donors (MUDs) is lower. The best scenario is an identically matched, sibling who does not have DBA. The National Marrow Donor Program (NMDP) is a database containing the tissue types of more than six million potential volunteer donors. Visit the program online to learn more: http://www.marrow.org/index.html.

For DBA patients, a stem cell transplant is intended to restore the marrows ability to make red blood cells. Once the body starts producing red blood cells, the patient may experience a decrease in signs and symptoms of anemia, such as tiredness and paleness. Often times, stem cell transplant may result in a cure of DBA and, when successful, may often extend a persons life and improve the quality of life they are able to enjoy. The person will no longer require long-term steroid medicine or blood transfusions. The persons blood type will actually change to that of the donor.

A stem cell transplant is a complex procedure with risks. Although some people with DBA experience few problems with transplant, others experience many problems and must endure frequent tests and hospitalizations. Before a stem cell transplant, the patient receives chemotherapy and occasionally radiation therapy to destroy their unhealthy stem cells. This is called a preparative regimen. Some side effects, such as nausea, vomiting, fatigue, loss of appetite, mouth sores, hair loss, and skin reactions may be due to the preparative regimen.

Several complications, some potentially fatal, can occur as a result of a stem cell transplant:

After the transplant, before the new marrow has started to grow, the number of white blood cells is low and the immune system (how the body fights infection and stays healthy) is very weak. During this time, the body is susceptible to infections, sometimes from the bacteria that live in the patients own body. Therefore, infections that normally would not be harmful can be very serious, and patients can die of them. Bacterial, viral, and fungal infections are often seen following transplant.

Graft-versus-host disease (GVHD) occurs when the new stem cells (from the donor) do not recognize the patients cells and attacks them, leading to skin rashes, diarrhea, or liver abnormalities. GVHD can be acute or chronic and range in severity from mild to moderate to severe. Medicines are given to prevent GVHD. Mild and moderate GVHD can be treated successfully with drugs and does not increase the risk of the patient dying. The most severe degree of GVHD is less frequent, but very serious, and patients can die of this complication. A close match between the donor and recipient will reduce the risk for GVHD, thereby allowing a greater chance for the donor stem cells to produce normal blood cells without complications.

Some of the more common long-term risks of stem cell transplant include infertility (the inability to produce children) and cataracts (clouding of the lens of the eye, which can be fixed with surgery). Less common effects include long-term damage to organs such as the liver, kidneys, lungs, or heart, and the occurrence of cancers.

After the transplant regular check-ups are needed to identify and take care of any problems that may arise after a patient has a stem cell transplant. Initially, follow-up care involves clinic visits once or twice a week with platelet or blood transfusions, as needed. Long-term follow-up is necessary to maintain a healthy lifestyle, ensure that the DBA continues to be in remission, and ensure that any late effects of the transplant or DBA are caught early. During long-term follow up, growth and development, immunizations, fertility, and mental and physical health are monitored.

See the original post here:
Stem | Treatments | DBA | NCBDDD | CDC

An improved protocol for isolation and culture of …

Summary

Mesenchymal stem cells (MSCs) from bone marrow are main cell source for tissue repair and engineering, and vehicles of cell-based gene therapy. Unlike other species, mouse bone marrow derived MSCs (BM-MSCs) are difficult to harvest and grow due to the low MSCs yield. We report here a standardised, reliable, and easy-to-perform protocol for isolation and culture of mouse BM-MSCs. There are five main features of this protocol. (1) After flushing bone marrow out of the marrow cavity, we cultured the cells with fat mass without filtering and washing them. Our method is simply keeping the MSCs in their initial niche with minimal disturbance. (2) Our culture medium is not supplemented with any additional growth factor. (3) Our method does not need to separate cells using flow cytometry or immunomagnetic sorting techniques. (4) Our method has been carefully tested in several mouse strains and the results are reproducible. (5) We have optimised this protocol, and list detailed potential problems and trouble-shooting tricks. Using our protocol, the isolated mouse BM-MSCs were strongly positive for CD44 and CD90, negative CD45 and CD31, and exhibited tri-lineage differentiation potentials. Compared with the commonly used protocol, our protocol had higher success rate of establishing the mouse BM-MSCs in culture. Our protocol may be a simple, reliable, and alternative method for culturing MSCs from mouse bone marrow tissues.

Mesenchymal stem cells (MSCs) are multipotent stem cells that have the potential to self-renew and differentiate into a variety of specialised cell types such as osteoblasts, chondrocytes, adipocytes, and neurons [1]and[2]. MSCs are easily accessible, expandable, immunosuppressive and they do not elicit immediate immune responses [3]and[4]. Therefore, MSCs are an attractive cell source for tissue engineering and vehicles of cell therapy.

MSCs can be isolated from various sources such as adipose tissue, tendon, peripheral blood, and cord blood [5], [6]and[7]. Bone marrow (BM) is the most common source of MSCs. MSCs have been successfully isolated and characterised from many species including mouse, rat, rabbit, dog, sheep, pig, and human [8], [9], [10], [11]and[12]. Mice are one of the most commonly used experimental animals in biology and medicine primarily because they are mammals, small, inexpensive, easily maintained, can reproduce quickly, and share a high degree of homology with humans [13]. However, the isolation and purification of MSCs from mouse bone marrow is more difficult than other species due to their heterogeneity and low percentage in the bone marrow [1], [14]and[15].

Two main stem cell populations and their progenies, haematopoietic stem cells and BM-MSCs, are the main residents of bone marrow [1]and[15]. BM-MSCs are usually isolated and purified through their physical adherence to the plastic cell culture plate [16]. Several techniques have been used to purify or enrich MSCs including antibody-based cell sorting [17], low and high-density culture techniques [18]and[19], positive and negative selection method [20], frequent medium changes [21], and enzymatic digestion approach [22]. However, they all had some short falls: the standard MSCs culture method based on plastic adherence has been confirmed to have lower successful rate [23]; whereas the cell sorting approach reduced the osteogenic potentials of MSCs [17]. Negative selection method leads to granulocytemonocyte lineage cells reappearing after 1 week of culture [24]. Cells obtained using a positive selection method show higher proliferation ability compared with the negative selection method, but the method was only repeated in the C57B1/6 mice and failed to repeat in other strains of mice [25]. Frequent medium change method is inconvenient because it is required to change the culture medium every 8 hours during the first 72 hours of the initial culture [21]. Therefore, an easy and effective protocol for isolation of mouse BM-MSCs is needed.

Reagents used included: 0.25% trypsinEDTA (1) with phenol red; penicillinstreptomycin neomycin (PSN; Life Technologies, Carlsbad, CA, USA) antibiotic mixture; foetal bovine serum, qualified, heat-inactivated (Life Technologies); minimal essential medium (MEM) , nucleosides, powder (Life Technologies); and NaHCO3 (SigmaAldrich, St Louis, MO, USA).

Stock of -MEM was made up with 1 bag of -MEM powder (1L) and 2.2g NaHCO3 in 1000mL of Milli-Q water, adjusted to pH 7.2, filtered to sterilise, and stored for 12 weeks at 4C. Complete -MEM medium was -MEM medium stock supplemented with 15% foetal bovine serum and 1% PSN, stored at 4C. Phosphate-buffered saline (PBS) included: NaCl 8.0g, KCl 0.2g, KH2PO4 0.24g, and Na2HPO4 1.44g in 1L Milli-Q water (pH 7.4, sterilised and stored at 4C).

In this study, two mouse strains (ICR and C57) with different ages (4 weeks and 8 weeks, males and females) were tested using our protocol. All mice were purchased from and housed in a designated and government approved animal facility at The Chinese University Hong Kong, Hong Kong SAR, China, in according to The Chinese University Hong Kong's animal experimental regulations. All efforts were made to minimise animal suffering.

Mice aged 4 weeks or 8 weeks are terminated by cervical dislocation and placed in a 100-mm cell culture dish (Becton Dickinson, Franklin Lakes, NJ, USA), where the whole body is soaked in 70% (v/v) ethanol for 2 minutes, and then the mouse is transferred to a new dish (Fig.1A). Four claws are dissected at the ankle and carpal joints, and incisions made around the connection between hindlimbs and trunk, forelimbs, and trunk. The whole skin is then removed from the hind limbs and forelimbs by pulling toward the cutting site of the claw. Muscles, ligaments, and tendons are carefully disassociated from tibias, femurs, and humeri using microdissecting scissors and surgical scalpel. Tibias, femurs, and humeri are dissected by cutting at the joints, and the bones are transferred onto sterile gauze. Bones are carefully scrubbed to remove the residual soft tissues (Fig.1B), and transferred to a 100-mm sterile culture dish with 10mL complete -MEM medium on ice (Fig.1C). All samples are processed within 30 minutes following animal death to ensure high cell viability. The soft tissues are completely dissociated from the bones to avoid contamination.

Here is the original post:
An improved protocol for isolation and culture of ...

Bone marrow transplant – NHS Choices

Introduction

A bone marrow transplant, alsoknown as a haemopoietic stem cell transplant, replaces damaged bone marrow with healthy bone marrow stem cells.

Bone marrow is aspongytissue found in the hollow centres of some bones. It contains specialist stem cells, which produce the body's blood cells.

Stem cells in bone marrow produce three important types of blood cells:

Bone marrow transplants are often needed to treat conditions thatdamage bone marrow. If bone marrow is damaged, it is no longer able to produce normal blood cells. The new stem cells take over blood cellproduction.

Conditions that bone marrow transplants are used to treat include:

Read more about why a bone marrow transplantis needed.

A bone marrow transplant involves taking healthy stem cells from the bone marrow of one person and transferring them to the bone marrow of another person.

In some cases, it may be possible to take the bone marrow from your own body. This is known as an autologous transplantation. Before it is returned, the bone marrow is cleared of any damaged or diseased cells.

A bone marrowtransplant has five stages. These are:

Having a bone marrow transplant can be an intensive and challenging experience. Many people take up to a year to fully recover from the procedure.

Read more about what happens during a bone marrow transplant.

Bone marrow transplants are usually only recommended if:

Read more about who can have a bone marrow transplant.

Bone marrow transplants arecomplicated procedures with significant risks.

In some cases, the transplanted cells (graft cells) recognise the recipient's cells as "foreign"and try to attack them. This is known as graft versus host disease (GvHD).

The risk of infectionis alsoincreased because your immune system is weakened when you're conditioned (prepared) for the transplant.

Read more about the risks of having a bone marrow transplant.

It's nowpossible to harvest stem cells from sources other than bone marrow.

Peripheral blood stem cell donation involves injectinga medicine into the donor's blood thatcauses the stem cells to moveout of the bone marrow and into the bloodstream where theycan be harvested (collected).

The advantage of this type of stem cell donation is that the donor doesn't needa general anaesthetic.

Stem cells can also be collectedfrom the placenta and umbilical cord of a newborn baby and stored in a laboratory until they're needed.

Cord blood stem cells are very usefulbecause they don't need to be as closely matched as bone marrow or peripheral blood stem cells for a successful outcome.

Find out more about theNHS Cord Blood Bank(external link).

Page last reviewed: 18/02/2014

Next review due: 18/02/2016

Originally posted here:
Bone marrow transplant - NHS Choices

Hematopoietic stem cell transplantation – Wikipedia, the …

Hematopoietic stem cell transplantation (HSCT) is the transplantation of multipotent hematopoietic stem cells, usually derived from bone marrow, peripheral blood, or umbilical cord blood. It may be autologous (the patient's own stem cells are used) or allogeneic (the stem cells come from a donor). It is a medical procedure in the field of hematology, most often performed for patients with certain cancers of the blood or bone marrow, such as multiple myeloma or leukemia. In these cases, the recipient's immune system is usually destroyed with radiation or chemotherapy before the transplantation. Infection and graft-versus-host disease are major complications of allogeneic HSCT.

Hematopoietic stem cell transplantation remains a dangerous procedure with many possible complications; it is reserved for patients with life-threatening diseases. As survival following the procedure has increased, its use has expanded beyond cancer, such as autoimmune diseases.[1][2]

Indications for stem cell transplantation are as follows:

Many recipients of HSCTs are multiple myeloma[3] or leukemia patients[4] who would not benefit from prolonged treatment with, or are already resistant to, chemotherapy. Candidates for HSCTs include pediatric cases where the patient has an inborn defect such as severe combined immunodeficiency or congenital neutropenia with defective stem cells, and also children or adults with aplastic anemia[5] who have lost their stem cells after birth. Other conditions[6] treated with stem cell transplants include sickle-cell disease, myelodysplastic syndrome, neuroblastoma, lymphoma, Ewing's sarcoma, desmoplastic small round cell tumor, chronic granulomatous disease and Hodgkin's disease. More recently non-myeloablative, "mini transplant(microtransplantation)," procedures have been developed that require smaller doses of preparative chemo and radiation. This has allowed HSCT to be conducted in the elderly and other patients who would otherwise be considered too weak to withstand a conventional treatment regimen.

A total of 50,417 first hematopoietic stem cell transplants were reported as taking place worldwide in 2006, according to a global survey of 1327 centers in 71 countries conducted by the Worldwide Network for Blood and Marrow Transplantation. Of these, 28,901 (57 percent) were autologous and 21,516 (43 percent) were allogeneic (11,928 from family donors and 9,588 from unrelated donors). The main indications for transplant were lymphoproliferative disorders (54.5 percent) and leukemias (33.8 percent), and the majority took place in either Europe (48 percent) or the Americas (36 percent).[7] In 2009, according to the World Marrow Donor Association, stem cell products provided for unrelated transplantation worldwide had increased to 15,399 (3,445 bone marrow donations, 8,162 peripheral blood stem cell donations, and 3,792 cord blood units).[8]

Autologous HSCT requires the extraction (apheresis) of haematopoietic stem cells (HSC) from the patient and storage of the harvested cells in a freezer. The patient is then treated with high-dose chemotherapy with or without radiotherapy with the intention of eradicating the patient's malignant cell population at the cost of partial or complete bone marrow ablation (destruction of patient's bone marrow function to grow new blood cells). The patient's own stored stem cells are then transfused into his/her bloodstream, where they replace destroyed tissue and resume the patient's normal blood cell production. Autologous transplants have the advantage of lower risk of infection during the immune-compromised portion of the treatment since the recovery of immune function is rapid. Also, the incidence of patients experiencing rejection (graft-versus-host disease) is very rare due to the donor and recipient being the same individual. These advantages have established autologous HSCT as one of the standard second-line treatments for such diseases as lymphoma.[9]

However, for others cancers such as acute myeloid leukemia, the reduced mortality of the autogenous relative to allogeneic HSCT may be outweighed by an increased likelihood of cancer relapse and related mortality, and therefore the allogeneic treatment may be preferred for those conditions.[10] Researchers have conducted small studies using non-myeloablative hematopoietic stem cell transplantation as a possible treatment for type I (insulin dependent) diabetes in children and adults. Results have been promising; however, as of 2009[update] it was premature to speculate whether these experiments will lead to effective treatments for diabetes.[11]

Allogeneics HSCT involves two people: the (healthy) donor and the (patient) recipient. Allogeneic HSC donors must have a tissue (HLA) type that matches the recipient. Matching is performed on the basis of variability at three or more loci of the HLA gene, and a perfect match at these loci is preferred. Even if there is a good match at these critical alleles, the recipient will require immunosuppressive medications to mitigate graft-versus-host disease. Allogeneic transplant donors may be related (usually a closely HLA matched sibling), syngeneic (a monozygotic or 'identical' twin of the patient - necessarily extremely rare since few patients have an identical twin, but offering a source of perfectly HLA matched stem cells) or unrelated (donor who is not related and found to have very close degree of HLA matching). Unrelated donors may be found through a registry of bone marrow donors such as the National Marrow Donor Program. People who would like to be tested for a specific family member or friend without joining any of the bone marrow registry data banks may contact a private HLA testing laboratory and be tested with a mouth swab to see if they are a potential match.[12] A "savior sibling" may be intentionally selected by preimplantation genetic diagnosis in order to match a child both regarding HLA type and being free of any obvious inheritable disorder. Allogeneic transplants are also performed using umbilical cord blood as the source of stem cells. In general, by transfusing healthy stem cells to the recipient's bloodstream to reform a healthy immune system, allogeneic HSCTs appear to improve chances for cure or long-term remission once the immediate transplant-related complications are resolved.[13][14][15]

A compatible donor is found by doing additional HLA-testing from the blood of potential donors. The HLA genes fall in two categories (Type I and Type II). In general, mismatches of the Type-I genes (i.e. HLA-A, HLA-B, or HLA-C) increase the risk of graft rejection. A mismatch of an HLA Type II gene (i.e. HLA-DR, or HLA-DQB1) increases the risk of graft-versus-host disease. In addition a genetic mismatch as small as a single DNA base pair is significant so perfect matches require knowledge of the exact DNA sequence of these genes for both donor and recipient. Leading transplant centers currently perform testing for all five of these HLA genes before declaring that a donor and recipient are HLA-identical.

Race and ethnicity are known to play a major role in donor recruitment drives, as members of the same ethnic group are more likely to have matching genes, including the genes for HLA.[16]

As of 2013[update], there were at least two commercialized allogeneic cell therapies, Prochymal and Cartistem.[17]

To limit the risks of transplanted stem cell rejection or of severe graft-versus-host disease in allogeneic HSCT, the donor should preferably have the same human leukocyte antigens (HLA) as the recipient. About 25 to 30 percent of allogeneic HSCT recipients have an HLA-identical sibling. Even so-called "perfect matches" may have mismatched minor alleles that contribute to graft-versus-host disease.

In the case of a bone marrow transplant, the HSC are removed from a large bone of the donor, typically the pelvis, through a large needle that reaches the center of the bone. The technique is referred to as a bone marrow harvest and is performed under general anesthesia.

Peripheral blood stem cells[18] are now the most common source of stem cells for allogeneic HSCT. They are collected from the blood through a process known as apheresis. The donor's blood is withdrawn through a sterile needle in one arm and passed through a machine that removes white blood cells. The red blood cells are returned to the donor. The peripheral stem cell yield is boosted with daily subcutaneous injections of Granulocyte-colony stimulating factor, serving to mobilize stem cells from the donor's bone marrow into the peripheral circulation.

It is also possible to extract stem cells from amniotic fluid for both autologous or heterologous use at the time of childbirth.

Umbilical cord blood is obtained when a mother donates her infant's umbilical cord and placenta after birth. Cord blood has a higher concentration of HSC than is normally found in adult blood. However, the small quantity of blood obtained from an Umbilical Cord (typically about 50 mL) makes it more suitable for transplantation into small children than into adults. Newer techniques using ex-vivo expansion of cord blood units or the use of two cord blood units from different donors allow cord blood transplants to be used in adults.

Cord blood can be harvested from the Umbilical Cord of a child being born after preimplantation genetic diagnosis (PGD) for human leucocyte antigen (HLA) matching (see PGD for HLA matching) in order to donate to an ill sibling requiring HSCT.

Unlike other organs, bone marrow cells can be frozen (cryopreserved) for prolonged periods without damaging too many cells. This is a necessity with autologous HSC because the cells must be harvested from the recipient months in advance of the transplant treatment. In the case of allogeneic transplants, fresh HSC are preferred in order to avoid cell loss that might occur during the freezing and thawing process. Allogeneic cord blood is stored frozen at a cord blood bank because it is only obtainable at the time of childbirth. To cryopreserve HSC, a preservative, DMSO, must be added, and the cells must be cooled very slowly in a controlled-rate freezer to prevent osmotic cellular injury during ice crystal formation. HSC may be stored for years in a cryofreezer, which typically uses liquid nitrogen.

The chemotherapy or irradiation given immediately prior to a transplant is called the conditioning regimen, the purpose of which is to help eradicate the patient's disease prior to the infusion of HSC and to suppress immune reactions. The bone marrow can be ablated (destroyed) with dose-levels that cause minimal injury to other tissues. In allogeneic transplants a combination of cyclophosphamide with total body irradiation is conventionally employed. This treatment also has an immunosuppressive effect that prevents rejection of the HSC by the recipient's immune system. The post-transplant prognosis often includes acute and chronic graft-versus-host disease that may be life-threatening. However, in certain leukemias this can coincide with protection against cancer relapse owing to the graft versus tumor effect.[19]Autologous transplants may also use similar conditioning regimens, but many other chemotherapy combinations can be used depending on the type of disease.

A newer treatment approach, non-myeloablative allogeneic transplantation, also termed reduced-intensity conditioning (RIC), uses doses of chemotherapy and radiation too low to eradicate all the bone marrow cells of the recipient.[20]:320321 Instead, non-myeloablative transplants run lower risks of serious infections and transplant-related mortality while relying upon the graft versus tumor effect to resist the inherent increased risk of cancer relapse.[21][22] Also significantly, while requiring high doses of immunosuppressive agents in the early stages of treatment, these doses are less than for conventional transplants.[23] This leads to a state of mixed chimerism early after transplant where both recipient and donor HSC coexist in the bone marrow space.

Decreasing doses of immunosuppressive therapy then allows donor T-cells to eradicate the remaining recipient HSC and to induce the graft versus tumor effect. This effect is often accompanied by mild graft-versus-host disease, the appearance of which is often a surrogate marker for the emergence of the desirable graft versus tumor effect, and also serves as a signal to establish an appropriate dosage level for sustained treatment with low levels of immunosuppressive agents.

Because of their gentler conditioning regimens, these transplants are associated with a lower risk of transplant-related mortality and therefore allow patients who are considered too high-risk for conventional allogeneic HSCT to undergo potentially curative therapy for their disease. The optimal conditioning strategy for each disease and recipient has not been fully established, but RIC can be used in elderly patients unfit for myeloablative regimens, for whom a higher risk of cancer relapse may be acceptable.[20][22]

After several weeks of growth in the bone marrow, expansion of HSC and their progeny is sufficient to normalize the blood cell counts and re-initiate the immune system. The offspring of donor-derived hematopoietic stem cells have been documented to populate many different organs of the recipient, including the heart, liver, and muscle, and these cells had been suggested to have the abilities of regenerating injured tissue in these organs. However, recent research has shown that such lineage infidelity does not occur as a normal phenomenon[citation needed].

HSCT is associated with a high treatment-related mortality in the recipient (1 percent or higher)[citation needed], which limits its use to conditions that are themselves life-threatening. Major complications are veno-occlusive disease, mucositis, infections (sepsis), graft-versus-host disease and the development of new malignancies.

Bone marrow transplantation usually requires that the recipient's own bone marrow be destroyed ("myeloablation"). Prior to "engraftment" patients may go for several weeks without appreciable numbers of white blood cells to help fight infection. This puts a patient at high risk of infections, sepsis and septic shock, despite prophylactic antibiotics. However, antiviral medications, such as acyclovir and valacyclovir, are quite effective in prevention of HSCT-related outbreak of herpetic infection in seropositive patients.[24] The immunosuppressive agents employed in allogeneic transplants for the prevention or treatment of graft-versus-host disease further increase the risk of opportunistic infection. Immunosuppressive drugs are given for a minimum of 6-months after a transplantation, or much longer if required for the treatment of graft-versus-host disease. Transplant patients lose their acquired immunity, for example immunity to childhood diseases such as measles or polio. For this reason transplant patients must be re-vaccinated with childhood vaccines once they are off immunosuppressive medications.

Severe liver injury can result from hepatic veno-occlusive disease (VOD). Elevated levels of bilirubin, hepatomegaly and fluid retention are clinical hallmarks of this condition. There is now a greater appreciation of the generalized cellular injury and obstruction in hepatic vein sinuses, and hepatic VOD has lately been referred to as sinusoidal obstruction syndrome (SOS). Severe cases of SOS are associated with a high mortality rate. Anticoagulants or defibrotide may be effective in reducing the severity of VOD but may also increase bleeding complications. Ursodiol has been shown to help prevent VOD, presumably by facilitating the flow of bile.

The injury of the mucosal lining of the mouth and throat is a common regimen-related toxicity following ablative HSCT regimens. It is usually not life-threatening but is very painful, and prevents eating and drinking. Mucositis is treated with pain medications plus intravenous infusions to prevent dehydration and malnutrition.

Graft-versus-host disease (GVHD) is an inflammatory disease that is unique to allogeneic transplantation. It is an attack of the "new" bone marrow's immune cells against the recipient's tissues. This can occur even if the donor and recipient are HLA-identical because the immune system can still recognize other differences between their tissues. It is aptly named graft-versus-host disease because bone marrow transplantation is the only transplant procedure in which the transplanted cells must accept the body rather than the body accepting the new cells. Acute graft-versus-host disease typically occurs in the first 3 months after transplantation and may involve the skin, intestine, or the liver. High-dose corticosteroids such as prednisone are a standard treatment; however this immuno-suppressive treatment often leads to deadly infections. Chronic graft-versus-host disease may also develop after allogeneic transplant. It is the major source of late treatment-related complications, although it less often results in death. In addition to inflammation, chronic graft-versus-host disease may lead to the development of fibrosis, or scar tissue, similar to scleroderma; it may cause functional disability and require prolonged immunosuppressive therapy. Graft-versus-host disease is usually mediated by T cells, which react to foreign peptides presented on the MHC of the host[citation needed].

Graft versus tumor effect (GVT) or "graft versus leukemia" effect is the beneficial aspect of the Graft-versus-Host phenomenon. For example, HSCT patients with either acute, or in particular chronic, graft-versus-host disease after an allogeneic transplant tend to have a lower risk of cancer relapse.[25][26] This is due to a therapeutic immune reaction of the grafted donor T lymphocytes against the diseased bone marrow of the recipient. This lower rate of relapse accounts for the increased success rate of allogeneic transplants, compared to transplants from identical twins, and indicates that allogeneic HSCT is a form of immunotherapy. GVT is the major benefit of transplants that do not employ the highest immuno-suppressive regimens.

Graft versus tumor is mainly beneficial in diseases with slow progress, e.g. chronic leukemia, low-grade lymphoma, and some cases multiple myeloma. However, it is less effective in rapidly growing acute leukemias.[27]

If cancer relapses after HSCT, another transplant can be performed, infusing the patient with a greater quantity of donor white blood cells (Donor lymphocyte infusion).[27]

Patients after HSCT are at a higher risk for oral carcinoma. Post-HSCT oral cancer may have more aggressive behavior with poorer prognosis, when compared to oral cancer in non-HSCT patients.[28]

Prognosis in HSCT varies widely dependent upon disease type, stage, stem cell source, HLA-matched status (for allogeneic HCST) and conditioning regimen. A transplant offers a chance for cure or long-term remission if the inherent complications of graft versus host disease, immuno-suppressive treatments and the spectrum of opportunistic infections can be survived.[13][14] In recent years, survival rates have been gradually improving across almost all populations and sub-populations receiving transplants.[29]

Mortality for allogeneic stem cell transplantation can be estimated using the prediction model created by Sorror et al.,[30] using the Hematopoietic Cell Transplantation-Specific Comorbidity Index (HCT-CI). The HCT-CI was derived and validated by investigators at the Fred Hutchinson Cancer Research Center (Seattle, WA). The HCT-CI modifies and adds to a well-validated comorbidity index, the Charlson Comorbidity Index (CCI) (Charlson et al.[31]) The CCI was previously applied to patients undergoing allogeneic HCT but appears to provide less survival prediction and discrimination than the HCT-CI scoring system.

The risks of a complication depend on patient characteristics, health care providers and the apheresis procedure, and the colony-stimulating factor used (G-CSF). G-CSF drugs include filgrastim (Neupogen, Neulasta), and lenograstim (Graslopin).

Filgrastim is typically dosed in the 10 microgram/kg level for 45 days during the harvesting of stem cells. The documented adverse effects of filgrastim include splenic rupture (indicated by left upper abdominal or shoulder pain, risk 1 in 40000), Adult respiratory distress syndrome (ARDS), alveolar hemorrage, and allergic reactions (usually expressed in first 30 minutes, risk 1 in 300).[32][33][34] In addition, platelet and hemoglobin levels dip post-procedure, not returning to normal until one month.[34]

The question of whether geriatrics (patients over 65) react the same as patients under 65 has not been sufficiently examined. Coagulation issues and inflammation of atherosclerotic plaques are known to occur as a result of G-CSF injection.[33] G-CSF has also been described to induce genetic changes in mononuclear cells of normal donors.[33] There is evidence that myelodysplasia (MDS) or acute myeloid leukaemia (AML) can be induced by GCSF in susceptible individuals.[35]

Blood was drawn peripherally in a majority of patients, but a central line to jugular/subclavian/femoral veins may be used in 16 percent of women and 4 percent of men. Adverse reactions during apheresis were experienced in 20 percent of women and 8 percent of men, these adverse events primarily consisted of numbness/tingling, multiple line attempts, and nausea.[34]

A study involving 2408 donors (1860 years) indicated that bone pain (primarily back and hips) as a result of filgrastim treatment is observed in 80 percent of donors by day 4 post-injection.[34] This pain responded to acetaminophen or ibuprofen in 65 percent of donors and was characterized as mild to moderate in 80 percent of donors and severe in 10 percent.[34] Bone pain receded post-donation to 26 percent of patients 2 days post-donation, 6 percent of patients one week post-donation, and <2 percent 1 year post-donation. Donation is not recommended for those with a history of back pain.[34] Other symptoms observed in more than 40 percent of donors include myalgia, headache, fatigue, and insomnia.[34] These symptoms all returned to baseline 1 month post-donation, except for some cases of persistent fatigue in 3 percent of donors.[34]

In one metastudy that incorporated data from 377 donors, 44 percent of patients reported having adverse side effects after peripheral blood HSCT.[35] Side effects included pain prior to the collection procedure as a result of GCSF injections, post-procedural generalized skeletal pain, fatigue and reduced energy.[35]

A study that surveyed 2408 donors found that serious adverse events (requiring prolonged hospitalization) occurred in 15 donors (at a rate of 0.6 percent), although none of these events were fatal.[34] Donors were not observed to have higher than normal rates of cancer with up to 48 years of follow up.[34] One study based on a survey of medical teams covered approximately 24,000 peripheral blood HSCT cases between 1993 and 2005, and found a serious cardiovascular adverse reaction rate of about 1 in 1500.[33] This study reported a cardiovascular-related fatality risk within the first 30 days HSCT of about 2 in 10000. For this same group, severe cardiovascular events were observed with a rate of about 1 in 1500. The most common severe adverse reactions were pulmonary edema/deep vein thrombosis, splenic rupture, and myocardial infarction. Haematological malignancy induction was comparable to that observed in the general population, with only 15 reported cases within 4 years.[33]

Georges Math, a French oncologist, performed the first European bone marrow transplant in November 1958 on five Yugoslavian nuclear workers whose own marrow had been damaged by irradiation caused by a criticality accident at the Vina Nuclear Institute, but all of these transplants were rejected.[36][37][38][39][40] Math later pioneered the use of bone marrow transplants in the treatment of leukemia.[40]

Stem cell transplantation was pioneered using bone-marrow-derived stem cells by a team at the Fred Hutchinson Cancer Research Center from the 1950s through the 1970s led by E. Donnall Thomas, whose work was later recognized with a Nobel Prize in Physiology or Medicine. Thomas' work showed that bone marrow cells infused intravenously could repopulate the bone marrow and produce new blood cells. His work also reduced the likelihood of developing a life-threatening complication called graft-versus-host disease.[41]

The first physician to perform a successful human bone marrow transplant on a disease other than cancer was Robert A. Good at the University of Minnesota in 1968.[42] In 1975, John Kersey, M.D., also of the University of Minnesota, performed the first successful bone marrow transplant to cure lymphoma. His patient, a 16-year-old-boy, is today the longest-living lymphoma transplant survivor.[43]

At the end of 2012, 20.2 million people had registered their willingness to be a bone marrow donor with one of the 67 registries from 49 countries participating in Bone Marrow Donors Worldwide. 17.9 million of these registered donors had been ABDR typed, allowing easy matching. A further 561,000 cord blood units had been received by one of 46 cord blood banks from 30 countries participating. The highest total number of bone marrow donors registered were those from the USA (8.0 million), and the highest number per capita were those from Cyprus (15.4 percent of the population).[44]

Within the United States, racial minority groups are the least likely to be registered and therefore the least likely to find a potentially life-saving match. In 1990, only six African-Americans were able to find a bone marrow match, and all six had common European genetic signatures.[45]

Africans are more genetically diverse than people of European descent, which means that more registrations are needed to find a match. Bone marrow and cord blood banks exist in South Africa, and a new program is beginning in Nigeria.[45] Many people belonging to different races are requested to donate as there is a shortage of donors in African, Mixed race, Latino, Aboriginal, and many other communities.

In 2007, a team of doctors in Berlin, Germany, including Gero Htter, performed a stem cell transplant for leukemia patient Timothy Ray Brown, who was also HIV-positive.[46] From 60 matching donors, they selected a [CCR5]-32 homozygous individual with two genetic copies of a rare variant of a cell surface receptor. This genetic trait confers resistance to HIV infection by blocking attachment of HIV to the cell. Roughly one in 1000 people of European ancestry have this inherited mutation, but it is rarer in other populations.[47][48] The transplant was repeated a year later after a leukemia relapse. Over three years after the initial transplant, and despite discontinuing antiretroviral therapy, researchers cannot detect HIV in the transplant recipient's blood or in various biopsies of his tissues.[49] Levels of HIV-specific antibodies have also declined, leading to speculation that the patient may have been functionally cured of HIV. However, scientists emphasise that this is an unusual case.[50] Potentially fatal transplant complications (the "Berlin patient" suffered from graft-versus-host disease and leukoencephalopathy) mean that the procedure could not be performed in others with HIV, even if sufficient numbers of suitable donors were found.[51][52]

In 2012, Daniel Kuritzkes reported results of two stem cell transplants in patients with HIV. They did not, however, use donors with the 32 deletion. After their transplant procedures, both were put on antiretroviral therapies, during which neither showed traces of HIV in their blood plasma and purified CD4 T cells using a sensitive culture method (less than 3 copies/mL). However, the virus was once again detected in both patients some time after the discontinuation of therapy.[53]

Since McAllister's 1997 report on a patient with multiple sclerosis (MS) who received a bone marrow transplant for CML,[54] there have been over 600 reports of HSCTs performed primarily for MS.[55] These have been shown to "reduce or eliminate ongoing clinical relapses, halt further progression, and reduce the burden of disability in some patients" that have aggressive highly active MS, "in the absence of chronic treatment with disease-modifying agents".[55]

Read more:
Hematopoietic stem cell transplantation - Wikipedia, the ...

JAMA | Comparison of Allogeneic vs Autologous Bone Marrow …

Corresponding Author: Joshua M. Hare, MD, The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Bldg/Room 908, PO Box 016960 (R-125), Miami, FL 33101 (jhare@med.miami.edu).

Published Online: November 6, 2012. doi:10.1001/jama.2012.25321

Author Contributions:Dr Hare had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Study concept and design: Hare, Gerstenblith, DiFede Velazquez, George, Mendizabal, McNiece, Heldman.

Acquisition of data: Hare, Fishman, Gerstenblith, DiFede Velazquez, Zambrano, Suncion, Tracy, Johnston, Brinker, Breton, Davis-Sproul, Byrnes, George, Lardo, Mendizabal, Lowery, Wong Po Foo, Ruiz, Amador, Da Silva, McNiece, Heldman.

Analysis and interpretation of data: Hare, Fishman, Zambrano, Suncion, Tracy, Ghersin, Lardo, Schulman, Mendizabal, Altman, Ruiz, Amador, Da Silva, McNiece, Heldman.

Drafting of the manuscript: Hare, Fishman, Ghersin, Mendizabal, Ruiz, Amador, Heldman.

Critical revision of the manuscript for important intellectual content: Hare, Fishman, Gerstenblith, DiFede Velazquez, Suncion, Tracy, Johnston, Brinker, Breton, Davis-Sproul, Schulman, Byrnes, Geroge, Lardo, Mendizabal, Lowery, Rouy, Altman, Wong Po Foo, Ruiz, Da Silva, McNiece, Heldman.

Statistical analysis: Hare, Mendizabal, McNiece, Heldman.

Obtained funding: Hare, Lardo.

Administrative, technical, or material support: Hare, DiFede Velazquez, Zambrano, Suncion, Ghersin, Johnston, Breton, Davis-Sproul, Schulman, Byrnes, Lowery, Rouy, Altman, Wong Po Foo, Da Silva, McNiece, Heldman.

Study supervision: Hare, Fishman, Gerstenblith, Tracy, George, Schulman, Altman, Da Silva, McNiece, Heldman.

Conflict of Interest Disclosures: All authors have completed and submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Dr Hare reported having a patent for cardiac cell-based therapy, receiving research support from and being a board member of Biocardia, having equity interest in Vestion Inc, and being a consultant for Kardia. Dr George reported serving on the board of GE Healthcare, consulting for ICON Medical Imaging, and receiving trademark royalties for fluoroperfusion imaging. Mr Mendizabal is an employee of EMMES Corporation. Drs Rouy, Altman, and Wong Po Foo are employees of Biocardia Inc. Dr McNiece reported being a consultant and board member of Proteonomix Inc. Dr Heldman reported having a patent for cardiac cell-based therapy, receiving research support from and being a board member of Biocardia, and having equity interest in Vestion Inc. No other authors reported any financial disclosures.

Funding/Support: This study was funded by the US National Heart, Lung, and Blood Institute (NHLBI) as part of the Specialized Centers for Cell-Based Therapy U54 grant (U54HL081028-01). Dr Hare is also supported by National Institutes of Health (NIH) grants RO1 HL094849, P20 HL101443, RO1 HL084275, RO1 HL107110, RO1 HL110737, and UM1HL113460. The NHLBI provided oversight of the clinical trial through the independent Gene and Cell Therapy Data and Safety Monitoring Board (DSMB). Biocardia Inc provided the Helical Infusion Catheters for the conduct of POSEIDON.

Role of the Sponsors: The NHLBI, NIH, and Biocardia Inc had no role in the design and conduct of the study; in the collection, management, analysis, and interpretation of the data; or in the preparation, review, or approval of the manuscript.

Additional Contributions: We thank the NHLBI Gene and Cell Therapy DSMB, the patients who participated in this trial, the bone marrow donors, the staff of the cardiac catheterization laboratories at the University of Miami Hospital and The Johns Hopkins Hospital. Erica Anderson, MA (EMMES Corporation), provided data management and Hongwei Tang, MD (TeraRecon Inc), provided consultation regarding CT imaging analysis. Ms Anderson received compensation for her contribution via the Specialized Centers for Cell-Based Therapy grant. Dr Tang did not receive any compensation for his contribution.

This article was corrected for errors on July 19, 2013.

Read more here:
JAMA | Comparison of Allogeneic vs Autologous Bone Marrow ...

Characterization of bone marrow derived mesenchymal stem …

Abstract Introduction

Bone marrow mesenchymal stem cells (BMMSCs) are a heterogeneous population of postnatal precursor cells with the capacity of adhering to culture dishes generating colony-forming unit-fibroblasts (CFU-F). Here we identify a new subset of BMMSCs that fail to adhere to plastic culture dishes and remain in culture suspension (S-BMMSCs).

To catch S-BMMSCs, we used BMMSCs-produced extracellular cell matrix (ECM)-coated dishes. Isolated S-BMMSCs were analyzed by in vitro stem cell analysis approaches, including flow cytometry, inductive multiple differentiation, western blot and in vivo implantation to assess the bone regeneration ability of S-BMMSCs. Furthermore, we performed systemic S-BMMSCs transplantation to treat systemic lupus erythematosus (SLE)-like MRL/lpr mice.

S-BMMSCs are capable of adhering to ECM-coated dishes and showing mesenchymal stem cell characteristics with distinction from hematopoietic cells as evidenced by co-expression of CD73 or Oct-4 with CD34, forming a single colony cluster on ECM, and failure to differentiate into hematopoietic cell lineage. Moreover, we found that culture-expanded S-BMMSCs exhibited significantly increased immunomodulatory capacities in vitro and an efficacious treatment for SLE-like MRL/lpr mice by rebalancing regulatory T cells (Tregs) and T helper 17 cells (Th17) through high NO production.

These data suggest that it is feasible to improve immunotherapy by identifying a new subset BMMSCs.

Bone marrow mesenchymal stem cells (BMMSCs) are hierarchical postnatal stem/progenitor cells capable of self-renewing and differentiating into osteoblasts, chondrocytes, adipocytes, and neural cells [1,2]. BMMSCs express a unique surface molecule profile, including expression of STRO-1, CD29, CD73, CD90, CD105, CD146, Octamer-4 (Oct4), and stage-specific embryonic antigen-4 (SSEA4) [3,4]. It is generally believed that BMMSCs are negative for hematopoietic cell markers such as CD14 and CD34 [5-13]. BMMSCs have been widely used for tissue engineering [14-16]. Recently, a growing body of evidence has indicated that BMMSCs produce a variety of cytokines and display profound immunomodulatory properties [17-19], perhaps by inhibiting the proliferation and function of several major immune cells, such as natural killer cells, dendritic cells, and T and B lymphocytes [17-20]. These unique properties make BMMSCs of great interest for clinical applications in the treatment of different immune disorders [17,21-24].

BMMSCs are thought to be derived from the bone marrow stromal compartment, initially appearing as adherent, single colony clusters (colony-forming unit-fibroblasts [CFU-F]), and subsequently proliferating on culture dishes [25]. To date, the CFU-F assay has been considered one of the gold standards for determining the incidence of clonogenic BMMSC [26,27]. Since BMMSC are a heterogeneous population of stem cells, it is critical to identify whether BMMSC contain unique cell subsets with distinctive functions, analogous to the hematopoietic stem/progenitor cell system. In this study, we identified a subset of mouse BMMSCs in culture suspension and determined their immunomodulatory characteristics.

Female C3H/HeJ, C57BL/6J, and C3MRL-Faslpr/J mice were purchased from Jackson Laboratory (Bar Harbor, ME, USA). Female immunocompromised mice (Beige nude/nude XIDIII) were purchased from Harlan (Indianapolis, IN, USA). All animal experiments were performed under the institutionally approved protocols for the use of animal research (USC #10874 and 10941).

Anti Oct4, SSEA4, Runx2, OCN, active catenin and catenin were purchased from Millipore (Billerica, MA, USA). Anti alkaline phosphatase (ALP) antibody was purchased from Abcam (Cambridge, MA, USA). Anti Sca-1-PE, CD34-PE, CD34-FITC, CD45-PE, CD73-PE, CD4-PerCP, CD8-FITC, CD25-APC, CD3 and CD28 antibodies were purchased from BD Bioscience (San Jose, CA, USA). Anti Foxp3-PE, IL17-PE, and IFN-APC antibodies were purchased from eBioscience (San Diego, CA, USA). Unconjugated anti CD34, CD73, and CD105, NOS2 were purchased from Santa Cruz Biosciences (Santa Cruz, CA, USA). Anti actin antibody was purchased from Sigma (St. Louis, MO, USA).

The single suspension of bone marrow derived all nucleated cells (ANCs) from femurs and tibias were seeded at a density of 15 106 into 100 mm culture dishes (Corning, NY, USA) at 37C and 5% CO2. Non-adherent cells were removed after two days and attached cells were maintained for 16 days in alpha minimum essential medium (-MEM, Invitrogen, Grand Island, NY, USA) supplemented with 20% fetal bovine serum (FBS, Equitech-bio, Kerrville, TX, USA), 2 mM L-glutamine, 55 M 2-mercaptoethanol, 100 U/ml penicillin, and 100 g/ml streptomycin (Invitrogen). Colony-forming attached cells were passed once for further experimental use.

ECM coated dishes were prepared as described previously [28]. Briefly, 100% confluence of BMMSCs was cultured in medium with 100 nM L-ascorbic acid phosphate (Wako Pure Chemical, Richmond, VA, USA). After two weeks, cultures were washed with PBS and incubated with 0.005% Triton X-100 (Sigma) for 15 minutes at room temperature to remove cells. The ECM was treated with DNase I (100 units/ml; Sigma) for 1 hour at 37C. The ECM was washed with PBS three times and stored in 2 ml of PBS containing 100 U/ml penicillin, 100 g/ml streptomycin and 0.25 g/ml fungizone (Invitrogen) at 4C.

Bone marrow-derived ANCs (15 106) were seeded into 100 mm culture dishes and cultured for two days. The culture supernatant with floating cells was collected and centrifuged to obtain putative non-attached BMMSCs. The cells were re-seeded at indicated numbers on ECM-coated dishes. After 2 days, the floating cells in the cultures were removed with PBS and the attached cells on ECM were maintained for an additional 14 days. Colony-forming attached cells were passed once and sub-cultured on regular plastic culture dishes for further experiments. For some stem cell characterization analyses, we collected SSEA4 positive S-BMMSCs using the MACS magnetic separation system (Milteny Biotech, Auburn, CA, USA) and expanded in the cultures.

One million cells of ANCs from bone marrow were seeded on a T-25 cell culture flask (Nunc, Rochester, NY, USA). After 16 days, the cultures were washed with PBS and stained with 1% toluidine blue solution in 2% paraformaldehyde (PFA). A cell cluster that had more than 50 cells was counted as a colony under microscopy. The colony number was counted in five independent samples per each experimental group.

The proliferation of BMMSCs and S-BMMSCs was performed using the bromodeoxyuridine (BrdU) incorporation assay. Each cell population (1 104 cells/well) was seeded on two-well chamber slides (Nunc) and cultured for two to three days. The cultures were incubated with BrdU solution (1:100) (Invitrogen) for 20 hours, and stained with a BrdU staining kit (Invitrogen). BrdU-positive and total cell numbers were counted in ten images per subject. The BrdU assay was repeated in five independent samples for each experimental group.

A total of 0.5 106 cells of BMMSCs and S-BMMSCs was seeded on 60 mm culture dishes at the first passage. Upon reaching confluence, the cells were passaged at the same cell density. The population doubling was calculated at every passage according to the equation: log2 (number of harvested cells/number of seeded cells). The finite population doublings were determined by cumulative addition of total numbers generated from each passage until the cells ceased dividing.

BMMSCs or S-BMMSCs (0.2 106 cells) were incubated with 1 g of R-Phycoerythrin (PE). (PE)-conjugated antibodies or isotype-matched control immunoglobulin Gs (IgGs) (Southern Biotech, Birmingham, AL, USA) at 4C for 45 minutes. Samples were analyzed by a fluorescence-activated cell sorting (FACS)Calibur flow cytometer (BD Bioscience). For dual color analysis, the cells were treated with PE-conjugated and fluorescein isothiocyanate (FITC)-conjugated antibodies or isotype-matched control IgGs (1 g each). The cells were analyzed on FACSCalibur (BD Bioscience).

The cells subcultured on eight-well chamber slides (Nunc) (2 103/well) were fixed with 4% PFA. The samples were incubated with the specific or isotype-matched mouse antibodies (1:200) overnight at 4C, and treated with Rhodamine-conjugated secondary antibodies (1:400, Jackson ImmunoResearch, West Grove, PA, USA; Southern Biotechnology, Birmingham, AL, USA). Finally, chamber slides were mounted using Vectashield mounting medium containing 4', 6-diamidino-2-phenylindole (DAPI) (Vector Laboratories, Burlingame, CA, USA).

A total of 4.0 106 cells was mixed with hydroxyapatite/tricalcium phosphate (HA/TCP) ceramic powders (40 mg, Zimmer Inc., Warsaw, IN, USA) and subcutaneously transplanted into eight-week-old immunocompromised mice. After eight weeks, the transplants were harvested, fixed in 4% PFA and then decalcified with 5% ethylenediaminetetraacetic acid (EDTA; pH 7.4), followed by paraffin embedding. The paraffin sections were stained with H & E and analyzed by an NIH Image-J. The newly-formed mineralized tissue area from five fields was calculated and shown as a percentage to total tissue area.

BMMSCs and S-BMMSCs were cultured under osteogenic culture conditions containing 2 mM -glycerophosphate (Sigma), 100 M L-ascorbic acid 2-phosphate and 10 nM dexamethasone (Sigma). After induction, the cultures were stained with alizarin red or alkaline phosphatase.

For adipogenic induction, 500 nM isobutylmethylxanthine, 60 M indomethacin, 500 nM hydrocortisone, 10 g/ml insulin (Sigma), 100 nM L-ascorbic acid phosphate were added to the culture medium. After 10 days, the cultured cells were stained with Oil Red-O and positive cells were quantified by using an NIH Image-J. Total RNA was also isolated from cultures after 10 days induction for further experiments.

For chondrogenic induction, 1 106 cell pellets were cultured under chondrogenic medium containing 15% FBS, 1% ITS (BD), 100 nM dexamethasone, 2 mM pyruvate (SIGMA), and 10 ng/ml transforming growth factor beta 1 (TGF1) in (D)MEM (Invitrogen) for threeweeks. Cell pellets were harvested at three weeks post induction, fixed overnight with 4% PFA and then, sections were prepared for staining.

Extraction of total RNA and RT-PCR were performed according to standard procedures. Primer information is described in Additional materials and methods [see Additional file 1].

Additional file 1. Figures S1 to S8 and Additional materials and methods. Figure S1. ECM coated dish could capture a greater number of CFU-F. CFU-f number in ECM coated dish compared to regular dish. Figure S2. CD45-CD34-BMMSCs showed similar property with S-BMMSCs. (A) CFU-f number. (B) Flow cytometric analysis. Figure S3. S-BMMSCs extended survival rate of lethal dose of irradiated mice. The life span of irradiated mice. Figure S4. Osteoclast activity in S-BMMSC-treated MRL/lpr mice. (A) Osteoclast number. (B) sRANKL level. (C) CTX level. Figure S5. L-NMMA pre-treated BMMSC transplantation failed to ameliorate disease phenotype of MRL/lpr mice. (A) Anti dsDNA (IgG) level. (B) Anti dsDNA (IgM) level. (C) Urine protein level. (D) Tregs level. (E) Th17 level. (F) Ratio between Tregs/Th17. Figure S6. Inhibition of NO production in BMMSCs. (A) NO level with inhibitors. (B) iNOS level by western blot. Figure S7. Endogenous S-BMMSCs in mice bone marrow. (A) Cell sorting result. (B) CFU-f number. (C) Osteogenic differentiation in vitro. (D) NO level. Figure S8. Human bone marrow contains S-BMMSCs (hS-BMMSCs). (A) NO level. (B) Kynurenine production. (C) Kynurenine production in co-culture system. (D) T cell apoptosis induction by hS-BMMSCs. Additional materials and methods describe about TRAP staining, Histomotry, Rescue lethal dose irradiated mice, and Isolation of CD34+CD73+ double positive cells.

Format: DOC Size: 2.6MB Download file

This file can be viewed with: Microsoft Word Viewer

A total of 20 g of protein was used and SDS-PAGE and western blotting were performed according to standard procedures. Detailed procedures are described in Additional materials and methods [see Additional file 1]. -actin on the same membrane served as the loading control.

BMMSCs and S-BMMSCs were cultured onto 35 mm low attach culture dishes (2 104/dish, STEMCELL Technologies, Vancouver, BC, V5Z 1B3, Canada) under hematopoietic differentiation medium (STEMCELL Technologies) with or without erythropoietin (EPO; 3 U/mL) for seven days. Whole bone marrow cells and linage negative bone marrow cells (Linage-cells) were used as positive controls. The results are representative of five independent experiments.

S-BMMSCs and BMMSCs were treated with 1 mM L-NG-monomethyl-arginine (L-NMMA) (Cayman Chemical, Ann Arbor, MI, USA) or 0.2 mM 1400 W (Cayman Chemical) to inhibit total nitric oxide synthase (NOS) or inducible nitric oxide synthase (iNOS), respectively.

BMMSCs (0.2 106/well) were cultured on 24-well plates with or without cytokines (IFN, 25 ng/ml; IL-1, 5 ng/ml, R&D Systems, Minneapolis, MN, USA) and chemicals (L-NMMA, 1 mM; 1400 W, 0.2 mM) at the indicated concentration and days. The supernatant from each culture was collected and nitric oxide concentration measured using a Total Nitric Oxide and Nitrate/Nitrite Parameter Assay kit (R&D Systems) according to the manufacturer's instruction.

The transwell system (Corning) was used for co-culture experiments. A total of 0.2 106 of S-BMMSCs or BMMSCs was seeded on each lower chamber. Activated spleen cells (1 106/chamber), which were pre-stimulated with plate-bound anti CD3 antibody (3 g/ml) and soluble anti CD28 antibody (2 g/ml) for two days, were loaded in the upper chambers. Both chambers were filled with a complete medium containing (D)MEM (Lonza, CH-4002 Basel, Switzerland) with 10% heat-inactivated FBS, 50 M 2-mercaptoethanol, 10 mM HEPES, 1 mM sodium pyruvate (Sigma), 1% non-essential amino acid (Cambrex, East Rutherford, NJ, USA), 2 mM L-glutamine, 100 U/ml penicillin and 100 mg/ml streptomycin. To measure the spleen cells viability, cell counting kit-8 (Dojindo Molecular Technologies, Rockville, MD, USA) was used. For apoptosis of spleen cells analyses, Annexin V-PE apoptosis detection kits I (BD Bioscience) were used and analyzed on FACSCalibur (BD Bioscience).

CD4+CD25- T-lymphocytes (1 106/well), collected using a CD4+CD25+ Treg isolation kit (Miltenyi Biotec), were pre-stimulated with plate-bound anti CD3 antibody (3 g/ml) and soluble anti CD28 antibody (2 g/ml) for two days. These activated T-lymphocytes were loaded on 0.2 106 BMMSCs or S-BMMSCs cultures with recombinant human TFG1 (2 ng/ml) (R&D Systems) and recombinant mouse IL2 (2 ng/ml) (R&D Systems). For Th17 induction, recombinant human TFG1 (2 ng/ml) and recombinant mouse IL6 (50 ng/ml) (Biolegend, San Diego, CA, USA) were added. After three days, cells in suspension were collected and stained with anti CD4-PerCP, anti CD8a-FITC, anti CD25-APC antibodies (each 1 g) for 45 minutes on ice under dark conditions. The cells were then stained with anti Foxp3-PE antibody (1 g) using a Foxp3 staining buffer kit (eBioscience) for cell fixation and permeabilization. For Th17, cells in suspension were stained with anti CD4-FITC (1g, Biolegend) for 45 minutes on ice under dark conditions followed by intercellular staining with anti-IL 17 antibody (1g, Biolegend) using a Foxp3 staining buffer kit. The cells were analyzed on FACSCalibur.

Under general anesthesia, C3H/HeJ-derived BMMSCs, S-BMMSCs, L-NMMA pre-treated BMMSCs (1 mM for five days), or CD34+/CD73+ double sorted cells (0.1 106 cells/10 g body weight) were infused into MRL/lpr mice via the tail vein at 10 weeks of age (n = 6 each group). In the control group, MRL/lpr mice received PBS (n = 5). All mice were sacrificed at two weeks post transplantation for further analysis. The protein concentration in urine was measured using a Bio-Rad Protein Assay (Bio-Rad, Hercules, CA, USA).

Peripheral blood serum samples were collected from mice. Autoantibodies, sRANKL and CTX were analyzed by ELISA using commercially available kits (anti-dsDNA antibodies and ANA; alpha diagnostics, albumin and sRANKL; R&D Systems, CTX; Nordic Bioscience Diagnostics, Herlev, Rigion Hovedstaden, Denmark) according to their manufactures' instructions. The results were averaged in each group. The intra-group differences were calculated between the mean values.

To detect Tregs, peripheral blood mononuclear cells (PBMNCs) (1 106) were treated with PerCP-conjugated anti-CD4, FITC-conjugated anti-CD8a, APC-conjugated anti-CD25 antibodies, and stained with R-PE-conjugated anti-Foxp3 antibody using a Foxp3 staining buffer kit (eBioscience). To measure Th17 cells, PBMNCs (1 106) were incubated with PerCP-conjugated anti-CD4, FITC-conjugated anti-CD8a, followed by treatment with R-PE-conjugated anti-IL-17 and APC-conjugated anti-IFN antibodies using a Foxp3 staining buffer kit. The cells were then analyzed on FACSCalibur .

Student's t-test was used to analyze statistical difference. P values less than 0.05 were considered significant.

To determine whether a subset of BMMSCs remain in culture suspension, ANCs (15 106 cells) from bone marrow were plated onto regular plastic culture dishes for two days and all non-attached cells were subsequently transplanted into immunocompromised mice subcutaneously using HA/TCP as a carrier. At eight weeks post-transplantation, newly formed bone was identified in the transplants by H & E staining (Figure 1A), suggesting that the BMMSC culture suspension may contain cells with a capacity to differentiate into bone forming cells. In vitro studies indicated that ECM produced by culture-expanded BMMSCs (BMMSC-ECM) could capture higher numbers of CFU-Fs when compared to plastic cultures [see Additional file 1, Figure S1] [28]. Thus, we collected culture supernatant with floating cells at two days post CFU-F culture and re-loaded it onto BMMSC-ECM-coated dishes (Figure 1B). A subset of BMMSCs in the suspension (S-BMMSCs) was able to adhere to the BMMSC-ECM and form CFU-F (Figure 1B), at a lower incidence compared to the number of CFU-F generated from regular BMMSCs (Figure 1C). In order to characterize the stem cell properties of S-BMMSCs, we collected SSEA4-positive S-BMMSCs and assessed their proliferation rate by BrdU incorporation. We found that S-BMMSCs had a significantly elevated BrdU uptake rate compared to regular BMMSCs (Figure 1D). In addition, we used a continuous cell culture assay to indicate that SSEA4-positive S-BMMSCs acquired a significantly increased number of population doublings (Figure 1E). These data imply that S-BMMSCs are distinct from regular BMMSCs in terms of attachment, proliferation, and self-renewal.

Figure 1. Identification of suspension BMMSCs (S-BMMSCs). (A) Hypothetical model indicates that bone marrow all nucleated cells (ANCs) were seeded at 15 106 into 100 mm culture dishes and incubated for two days at 37C with 5% CO2, and subsequently non-attached cells from culture suspension were transplanted into immunocompromised mice subcutaneously using hydroxyapatite tricalcium phosphate (HA) as a carrier for eight weeks. Newly formed bone (B) by osteoblasts (arrow heads) and associated connective tissue (C) were detected in this non-attached cell transplants by H & E staining. Bar = 100 m. (B) Hypothetical model of isolating S-BMMSCs. BMMSCs usually attach on culture dishes within two days; however, a small portion of BMMSCs in ANCs failed to attach to the dishes and remained in the suspension. The suspensions containing putative non-attached BMMSCs were collected and transferred to the extracellular matrix (ECM) coated dish with generating single colony clusters (CFU-F). These ECM-attached BMMSCs (S-BMMSCs) were sub-cultured on regular plastic culture dishes for additional experiments. (C) The number of plastic attached CFU-F from ANCs (1.5 106 cells) is more than seven-fold higher than that derived from BMMSC-ECM adherent S-BMMSCs. (D) Proliferation rates of S-BMMSCs and BMMSCs were assessed by BrdU incorporation for 24 hours. The percentage of positive cells is significantly increased in S-BMMSCs when compared to BMMSCs. (E) S-BMMSCs exhibit a significant increase in population doublings when compared to BMMSCs. The results are representative of five independent experiments. Scale bars = 50 m. ***P <0.001. The graph bar represents mean SD. BMMSCs, bone marrow mesenchymal stem cells; BrdU, bromodeoxyuridine; S-BMMSCs, BMMSCs in suspension; SD, standard deviation.

To examine the multipotent differentiation potential, we showed that S-BMMSCs are analogous to BMMSCs in their expression of alkaline phosphatase (ALP), mineralized nodule accumulation under the osteogenic inductive cultures, and bone regeneration when transplanted into immunocompromised mice using HA/TCP as a carrier (Figures 2A and 2B). Furthermore, we showed that S-BMMSCs were similar to regular BMMSCs in forming Oil red-O positive fat cells under adipogenic inductive conditions, which was associated with expression of the adipogenic genes, peroxisome proliferator-activated receptor gamma 2 (ppar2) and lipoprotein lipase (lpl) (Figures 2C and 2D). Parallel studies showed a similar capacity between S-BMMSCs and regular BMMSCs to differentiate into chondrocytes under chondrogenic inductive conditions, associated with the expression of proteoglycan, trichrome positive collagen, and type II collagen (Figure 2E). Collectively, these data confirm that S-BMMSCs are a subset of BMMSCs.

Figure 2. Multipotent differentiation of S-BMMSCs. (A) Alizarin Red S and alkaline phosphatase (ALP) staining showed that S-BMMSCs were similar to regular BMMSCs in osteogenic differentiation in vitro. (B) S-BMMSCs or regular BMMSCs (4 106 cells/transplant) were transplanted into immunocompromised mice using HA/TCP (HA) as a carrier for eight weeks. Bone formation was detected in S-BMMSC and BMMSC transplants, evidenced by H & E staining. HA, hydroxyapatite tricalcium phosphate; B, bone; M, bone marrow; CT, connective tissue. Bar = 50 m. (C-D) S-BMMSCs are capable of forming Oil Red O positive cells (C) and expression of ppar2 and lpl mRNA as seen in regular BMMSCs (D). Glyceraldehyde 3-phosphate dehydrogenase (gapdh) was used as an internal control. The results are representative of five independent experiments. Scale bars = 100 m. (E) Chondrogenic differentiation was assessed by Alcian blue staining for acidic sulfated mucosubstances, Pollak's Trichrome staining for collagen, and immunohistochemical staining for collagen type II. S-BMMSCs were able to differentiate into chondrocytes as observed in regular BMMSCs. Bar = 50 m. The results are representative of three independent experiments. The graph bar represents mean SD. BMMSCs, bone marrow mesenchymal stem cells; S-BMMSCs, BMMSCs in suspension; SD, standard deviation.

By flow cytometric analysis, S-BMMSCs expressed mesenchymal stem cell markers at the same level as regular BMMSCs (Figure 3A). Interestingly, 23.4% of S-BMMSCs expressed CD34, a hematopoietic stem cell (HSC) and endothelial cell marker, whereas 0.2% of BMMSCs expressed CD34 (Figure 3A). BMMSCs (21.4%) and S-BMMSCs (31.2%) expressed CD45, another hematopoietic marker, at passage 2 (Figure 3A). Both BMMSCs and S-BMMSCs were negative to CD11b antibody staining (data not shown), excluding the possibility that S-BMMSCs are derived from monocyte/macrophage lineage cells. Importantly, CD34+ S-BMMSCs co-expressed BMMSC-associated markers CD73 or Octamer-4 (Oct4), as evidenced by flow cytometric analysis (Figure 3B). Western blot analysis confirmed that S-BMMSCs expressed CD34, CD73, and CD105 (Figure 3C), and regular BMMSCs expressed CD73 and CD105 but lacked CD34 expression (Figure 3C). Whole bone marrow cells (BMC) were used as positive control. S-BMMSCs also showed a continued expression of CD34 from passage one to five; however, the expression levels appear reduced after passage three (Figure 3D). In order to further verify CD34 expression in S-BMMSCs, immunocytostaining analyses were performed to show co-expression of CD34 with mesenchymal markers CD73 (Figure 3E) in contrast to regular BMMSCs that were negative for anti-CD34 antibody staining (Figure 3E).

Figure 3. S-BMMSCs express CD34. (A) Flow cytometric analysis showed that regular BMMSCs fail to express CD34, but are positive for CD45 antibody staining (21.4%). However, S-BMMSCs express both CD34 (23.4%) and CD45 (31.2%). (B) Flow cytometric analysis also showed that CD34+ S-BMMSCs were positive for anti CD73 (13.8%) and Oct4 (13.4%) antibody staining. IgG isotype staining groups were used as negative controls. (C, D) Western blot analysis indicated that S-BMMSCs express CD34 and mesenchymal surface molecules CD73 and CD105. In contrast, regular BMMSCs only express CD73 and CD105 (C). S-BMMSCs express CD34 at passage one to five (D). -actin was used as a sample loading control. BMC, whole bone marrow ANC. (E) Immunocytostaining confirmed that S-BMMSCs are double positive for CD34/CD73 (triangle). Regular BMMSCs are negative for CD34 antibody staining and only positive for anti CD73 antibody staining. Bar = 100 m. (F) Both BMMSCs and S-BMMSCs failed to differentiate into hematopoietic lineage under hematopoietic inductive conditions with EPO (upper panel) or without EPO (lower panel). Whole bone marrow cells and lineage negative cells were used as positive (yellow arrowheads) control. Bar = 100 m. ANC, all nucleated cells; BMMSCs, bone marrow mesenchymal stem cells; EPO, erythropoietin; S-BMMSCs, BMMSCs in suspension.

It is generally believed that CD34 expression is associated with HSCs and endothelial populations. HSCs can differentiate into all the blood cell lineages and rescue lethally irradiated subjects. Thus, we cultured S-BMMSCs and regular BMMSCs in hematopoietic differentiation medium and determined that these mesenchymal cells failed to differentiate into a hematopoietic cell lineage compare to bone marrow cells that formed myeloid and erythroid colony forming clusters (Figure 3F). In addition, CD45-CD34-BMMSCs showed an ability similar to that of S-BMMSCs in colony forming and expressing surface marker as MSC [see Additional file 1, Figure S2]. Furthermore, we infused S-BMMSCs systemically to rescue lethally irradiated mice and found that S-BMMSCs, but not regular BMMSCs, could extend the lifespan of lethally irradiated mice [see Additional file 1, Figure S3]. However, S-BMMSCs failed to rescue lethally irradiated mice, as shown in the whole bone marrow cell group [see Additional file 1, Figure S3]. These data provid further evidence that CD34 expression in S-BMMSCs is not due to HSC contamination.

Since the immunomodulation property of MSCs is one of the essential factors for MSC characterization, allogenic S-BMMSC transplantation into MRL/lpr mice was performed (Figure 4A). Two weeks after transplantation, both S-BMMSCs and BMMSCs were capable of ameliorating SLE-induced glomerular basal membrane disorder (yellow arrow, Figure 4B) and reducing the urine protein level (Figure 4C). It appeared that S-BMMSCs were superior compared to BMMSCs in terms of reducing the overall urine protein levels (Figure 4C). As expected, MRL/lpr mice showed remarkably increased levels of autoantibodies, including anti-double strand DNA (dsDNA) IgG and IgM antibodies (Figures 4D and 4E) and anti-nuclear antibody (ANA; Figure 4F) in the peripheral blood serum. Although S-BMMSC and BMMSC infusion showed significantly decreased serum levels of anti-dsDNA IgG, IgM antibodies and ANA in peripheral blood (Figures 4D-F), S-BMMSCs showed a superior therapeutic effect in reducing anti-dsDNA IgG antibody and ANA levels when compared to BMMSCs (Figures 4D and 4F). Additionally, decreased serum albumin levels in MRL/lpr mice were recovered by S-BMMSC and BMMSC infusion (Figure 4G) but S-BMMSC treatment resulted in a more significant recovery than BMMSC treatment (Figure 4G). Next, flow cytometric analysis revealed that S-BMMSC showed more effectiveness in recovering the decreased level of CD4+CD25+Foxp3+ Tregs and increased the number of CD4+IL17+IFN- T-lymphocytes (Th17 cells) in peripheral blood when compared to BMMSCs (Figures 4H, 4I). In addition, highly passaged mouse S-BMMSCs failed to inhibit Th17 differentiation in vitro (data not shown) suggesting that mouse S-BMMSCs probably lose their immunomodulation property under long culture expansion.

Figure 4. S-BMMSCs showed superior therapeutic effect on SLE-like MRL/lpr mice. (A) Schema of BMMSC transplantation into MRL/lpr mice. (B) S-BMMSC and BMMSC treatment recover basal membrane disorder and mesangium cell over-growth in glomerular (G) (H&E staining). (C) S-BMMSC and BMMSC transplantation could reduce urine protein levels at two weeks post transplantation compared to the MRL/lpr group. S-BMMSCs offered a more significant reduction compared to BMMSCs. (D, E) The serum levels of anti-dsDNA IgG and IgM antibodies were significantly increased in MRL/lpr mice compared to controls (C3H). S-BMMSC and BMMSC treatments could reduce antibody levels but S-BMMSCs showed a superior treatment effect than BMMSC in reducing anti-dsDNA IgG antibody (D). (F) S-BMMSC and BMMSC treatments could reduce increased levels of anti nuclear antibody (ANA) in MRL/lpr mice. S-BMMSC showed a better effect in ANA reduction compared to BMMSC. (G) S-BMMSC and BMMSC treatments could increase the albumin level in MRL/lpr mice, which was decreased in controls. S-BMMSC treatments were more effective in elevating the albumin level compared to BMMSC treatment. (H) Flow cytometric analysis showed a reduced number of Tregs in MRL/lpr peripheral blood compared to control. BMMSC and S-BMMSC treatments elevated the number of Tregs. S-BMMSCs induced a more significant elevation of the Tregs level than BMMSCs. (I) Flow cytometric analysis showed an increased number of Th17 in MRL/lpr mice peripheral blood compared to control. Th17 were markedly decreased in BMMSC and S-BMMSC treated groups. S-BMMSC treatment induced a more significant reduction of Th17 cells than treatment with BMMSCs. *P <0.05; ** P <0.01; ***P <0.001. The graph bar represents mean SD. BMMSCs, bone marrow mesenchymal stem cells; Ig, immunoglobulin; S-BMMSCs, BMMSCs in suspension; SD, standard deviation; SLE, systemic lupus erythematosus; Tregs, regulatory T cells.

Furthermore, we showed that S-BMMSCs were superior to BMMSCs in terms of reducing increased numbers of tartrate-resistant acid phosphatase (TRAP) positive osteoclasts in the distal femur epiphysis of MRL/lpr mice [see Additional file 1, Figure S4A], elevated serum levels of sRANKL, a critical factor for osteoclastogenesis [see Additional file 1, Figure S4B] and bone resorption marker CTX [see Additional file 1, Figure S4C]. These data suggest that S-BMMSCs exhibit a superior therapeutic effect for SLE disorders compared to regular BMMSCs.

Recently, immunomodulatory properties were identified as an important stem cell characteristic of BMMSCs, leading to the utilization of systemic infused BMMSCs to treat a variety of immune diseases [19-21]. Here, we found that S-BMMSCs exhibited a significantly increased capacity for NO production compared to regular BMMSCs when treated with IFN and IL-1 (Figure 5A). It is known that NO plays a critical role in BMMSC-mediated immunosuppression [see Additional file 1, Figures S5A-F] [29]. Therefore, we assessed the functional role of high NO production in S-BMMSC-associated immunomodulatory properties. Spleen (SP) cells were activated by anti-CD3 and anti-CD28 antibodies for three days and then co-cultured with S-BMMSCs or regular BMMSCs in the presence of the general NOS inhibitor, L-NMMA or the iNOS inhibitor, 1400 W, using a Transwell culture system. The efficacy of L-NMMA and 1400 W to inhibit NO production in BMMSCs was verified [see Additional file 1, Figures S6A and 6B]. Although both S-BMMSCs and regular BMMSCs were capable of inhibiting cell viability of activated SP cells, S-BMMSCs showed a marked inhibition of SP cell viability over that of regular BMMSCs (Figure 5B). Moreover, both BMMSCs and S-BMMSCs induced SP cell apoptosis (Figure 5C). However, S-BMMSCs showed an elevated capacity in inducing activated SP cell apoptosis compared to regular BMMSCs (Figure 5C). Interestingly, when L-NMMA and 1400 W were added to the cultures, the number of apoptotic SP cells was significantly reduced in both S-BMMSC and regular BMMSC groups (Figure 5D and 5E). These in vitro experimental data suggested that NO production is an essential factor for BMMSC-mediated immunomodulation.

Figure 5. S-BMMSCs show up-regulated immunomodulatory properties through nitric oxide (NO) production. (A) NO levels in the supernatant of S-BMMSC and BMMSC culture were significantly higher in the INF-/IL-1 treated S-BMMSC group than in BMMSCs. (B-C) S-BMMSCs showed a significant reduction in the cell viability of activated SP cells compared to the cells cultured without BMMSCs (SP cell) and with BMMSCs (B). Both BMMSCs and S-BMMSCs showed a significantly increased rate of SP cell apoptosis compared to the SP cell only group but S-BMMSCs could induce higher SP cell apoptosis (C). (D-E) The induction of SP cell apoptosis by BMMSCs or S-BMMSCs was abolished in general NOS inhibitor L-NMMA-treated (D) and iNOS specific inhibitor 1400 W-treated (E) group. (F-H) Activated CD4+CD25- T-cells and S-BMMSCs or BMMSCs were co-cultured in the presence of TGF1 and IL-2 with or without NOS inhibitor for three days. The floating cells were stained for CD4+CD25+FoxP3+ regulatory T cells (Tregs). Both BMMSCs and S-BMMSC up-regulated Tregs but S-BMMSCs showed a significant effect in up-regulating Tregs. (F). Interestingly, L-NMMA and 1400 W treatments resulted in an abolishing of S-BMMSC-induced up-regulation of Tregs (G, H). (I) BMMSCs and S-BMMSCs could inhibit Th17 differentiation in vitro. S-BMMSC could inhibit it more effectively. (J, K) L-NMMA (J) or 1400 W (K) could abolish the inhibition of Th17 differentiation by BMMSCs or S-BMMSCs. The results are representative of at least three independent experiments. *P <0.05; **P <0.01; ***P <0.001. The graph bar represents mean SD. BMMSCs, bone marrow mesenchymal stem cells; iNOS, inducible nitric oxide synthase; L-NMMA, L-NG-monomethyl-arginine; NOS, nitric oxide synthase; S-BMMSCs, BMMSCs in suspension; SD, standard deviation; SP, spleen; Tregs, regulatory T cells.

Since up-regulation of CD4+CD25+Foxp3+ Tregs is required for immunotolerance [30], we tested Tregs up-regulation property of S-BMMSCs and BMMSCs in an in vitro co-culture system. When nave-T-cells were co-cultured with S-BMMSCs or regular BMMSCs in the presence of IL-2 and TGF-1, S-BMMSCs showed a significant up-regulation of Treg levels compared to regular BMMSCs (Figure 5F). Both L-NMMA and 1400 W were able to inhibit BMMSC- and S-BMMSC-induced up-regulation of Tregs, as shown by flow cytometric analysis (Figures 5G and 5H). Interestingly, the regulation effect on Tregs was more significant in the S-BMMSC group compared to the BMMSC group (Figure 5G and 5H). Moreover, both BMMSCs and S-BMMSCs could inhibit differentiation of Th17 in vitro, with a more prominent effect observed with S-BMMSC (Figure 5I). These inhibitions of Th17 differentiation were abolished by L-NMMA (Figure 5J) and 1400 W (Figure 5K). These data further verified the functional role of NO in S-BMMSC-induced immunomodulatory effect.

In order to identify whether there are functional endogenous S-BMMSCs, we used fluorescence activated cell sorting (FACS) to isolate CD34 and CD73 double-positive cells from bone marrow ANCs which resulted in the recovery of 3.77% double-positive cells [see Additional file 1, Figure S7A]. These CD34 and CD73 double-positive cells exhibited mesenchymal stem cell characteristics, including the capacity to form single colony clusters of fibroblast-like cells [see Additional file 1, Figure S7B], which could differentiate into osteogenic cells in vitro [see Additional file 1, Figure S7C]. These data indicated the feasibility of this approach to isolate S-BMMSC-like cells directly from bone marrow. We found that CD34+/CD73+ BMMSCs were analogous to S-BMMSCs in terms of having higher levels of NO production when compared to regular BMMSCs [see Additional file 1, Figure S7D] and reducing levels of urine protein, serum anti-dsDNA IgG and IgM antibodies in MRL/lpr mice (data not shown). These data indicate that endogenous S-BMMSCs could be isolated from bone marrow using CD34 and CD73 antibodies double sorting.

Additionally, we used the same BMMSC-ECM isolation approach to reveal the existence of human S-BMMSCs (hS-BMMSC) that possess stem cell properties including multipotent differentiation and self-renewal but lack expression of CD34 (data not shown). hS-BMMSCs showed elevated NO and kynurenine production which indicate high indoleamine 2,3-dioxygenase (IDO) activity when compared to regular BMMSCs [see Additional file 1, Figures S8A-C]. Thus, when activated T cells were co-cultured with hS-BMMSCs, AnnexinV-7 aminoactinomycinD (7AAD) double positive apoptotic SP cells were significantly elevated compared to BMMSCs [see Additional file 1, Figure S8D].

Adherent BMMSCs are able to proliferate and undergo osteogenic differentiation, providing the first evidence of CFU-F as precursors for osteoblastic lineage [25]. For over a few decades, the adherent CFU-F assay has been used as an effective approach to identify and select BMMSCs. In the current study, we showed that the adherent CFU-F assay collects the majority of clonogenic BMMSCs, but a subpopulation of BMMSCs is sustained in the culture suspension. This newly identified subpopulation of BMMSCs may be lost in the standard CFU-F assay for BMMSC isolation.

Due to the heterogeneity of the BMMSCs, there is no single, unique marker allowing for BMMSC isolation, rather an array of cell molecules are utilized to profile BMMSCs. It is widely accepted that BMMSCs express SH2 (CD105), SH3/SH4 (CD73), integrin 1 (CD29), CD44, Thy-1 (CD90), CD71, vascular cell adhesion molecule-1 (CD106), activated leukocyte cell adhesion molecule (CD166), STRO-1, GD2, and melanoma cell adhesion molecule (CD146) [5,7-13,31,32]. Nevertheless, it is believed that BMMSCs lack expression of hematopoietic surface molecules including CD34, integrin M (CD11b) and CD14. However, recent studies have implied that mouse BMMSCs might express the hematopoietic surface molecules, CD45 [28] and CD34 [33]. To ensure purity of S-BMMSCs, we used immune FACS to collect SSEA4+ S-BMMSCs for proliferation and differentiation assays in this study. Interestingly, previous experimental evidence appeared to support a notion that HSCs are capable of differentiating into mesenchymal cells [34] and osteoblastic lineage in vivo [35]. Thus, it is critical to clarify whether BMMSCs express hematopoietic associated surface molecules.

In this study, we have identified a novel subset of S-BMMSCs that failed to form adherent CFU-F in regular culture dishes, but were capable of adhering on mesenchymal stem cell-produced ECM and differentiating into osteoblasts, adipocytes and chondrocytes from both C3H/HeJ and C57BL/6J mice. S-BMMSCs co-expressed the HSC marker CD34 with the MSC markers CD73 and Oct4, excluding the potential of HSC contamination. Furthermore, S-BMMSCs were found to be distinct from HSC because they lacked the ability to differentiate into hematopoietic cell lineages in vitro and failed to rescue lethally-irradiated mice. The mechanism that may contribute to the up-regulated immunomodulatory function was associated with high NO production in S-BMMSCs and a NO-driven high Tregs level [36]. NO is a gaseous biological mediator with important roles in affecting T cell function [37].

This is the reason that S-BMMSCs showed a superior therapeutic effect in treating SLE mice.

One successful approach is to isolate cells that express specific molecules on their cell surfaces using monoclonal antibodies and cell sorting technologies. Enriched populations of BMMSCs have been isolated from human bone marrow aspirates using a STRO-1 monoclonal antibody in conjunction with antibodies against VCAM-1/CD106 [32], CD146 [11], low affinity nerve growth factor receptor/CD271, PDGR-R, EGF-R and IGF-1-R [38], fibroblast cell marker/D7-Fib [39] and integrin alpha 1/CD49a [40]. A more recent study has also identified molecules co-expressed by a CD271+ mesenchymal stem cell population including platelet derived growth factor receptor- (CD140b), human epidermal growth factor 2/ErbB2 (CD340) and frizzled-9 (CD349) [41]. Further cell separation based upon multi-parameter FACS identified a population of proposed mouse mesenchymal precursors with the composite phenotype Lin-CD45-CD31-Sca-1+[42]. Another recent study also identified and characterized an alternate population of primitive mesenchymal cells derived from adult mouse bone marrow, based upon their expression of the SSEA-1 [43]. All approaches used for BMMSC purification and isolation will undergo ex vivo expansion to enrich cell numbers for tissue regeneration or systemic therapies by plastic adherent assay. In addition to identifying a novel sub-population of BMMSCs that possess enhanced immunomodulatory properties when compared to regular BMMSCs, we showed that CD34+/CD73+ BMMSCs could be isolated directly from whole bone marrow and that CD34+/CD73+ BMMSCs are endogenous S-BMMSCs with higher NO production, and are superior in treating SLE-like mice when compared to regular BMMSCs.

Recently, non-adherent bone marrow cells (NA-BMCs) were identified [44,45]. The NA-BMSCs could be expanded in suspension and gave rise to multiple mesenchymal phenotypes, including osteoblasts, chondrocytes, and adipocytes in vitro, suggesting the presence of non-adherent BMMSCs in primary CFU-F cultures [45]. Although it has been reported that the NA-BMCs can rescue lethally-irradiated mouse recipients, our data indicated that S-BMMSCs only showed improved survival lifespan without a complete rescue of lethally-irradiated mice, compared to whole bone marrow transplantation. While the mechanism of S-BMMSC-mediated lifespan extension in lethally-irradiated mice is unknown, it is possible that S-BMMSCs have a more active interplay with hematopoietic cells than regular BMMSCs. It has been reported that granulocyte colony stimulating factor might promote BMMSCs into the circulation in humans [46], suggesting that non-attached BMMSCs may exist in vivo for specific functional needs. Added evidence indicated that osteocalcin-positive cells in circulation were able to differentiate into osteoblastic cells when cultured in the presence of TGF [47]. However, it is unknown whether S-BMMSCs are associated with circulating mesenchymal stem cells initially identified in mice, and this is very rare in humans.

A new subset of BMMSCs (S-BMMSCs) which failed to adhere to culture dishes possesses similar stem cell properties as those seen in BMMSCs, including CFU-F, stem cell markers, osto-, adipo-, and chondro-genic differentiation. However, S-BMMSC showed distinct features including expression of CD34 and a superior immunomodulation property through high NO production. These findings suggest that it is feasible to improve immunotherapy by identifying new subset BMMSCs.

7AAD: 7aminoactinomycineD; ALP: alkaline phosphatase; ANCs: all nucleated cells; BMMSCs: bone marrow mesenchymal stem cells; BrdU: bromodeoxyuridine; CFU-F: colony forming unit fibroblastic; CTX: C-terminal telopeptides of type I collagen; DAPI: 4', 6-diamidino-2-phenylindole; (D)MEM: (Dulbecco's) modified Eagle's medium; ECM: extracellular cell matrix; ELISA: enzyme-linked immunosorbent assay; EPO: erythropoietin; FACS: fluorescence-activated cell sorting; FBS: fetal bovine serums; FITC: fluorescein isothiocyanate; H & E: hematoxylin and eosin; HA/TCP: hydroxyapatite/tricalcium phosphate; HSC: hematopoietic stem cell; IDO: indoleamine 2,3-dioxygenase; IFN: interferon gamma; IgG: immunoglobulin G; IL-1: interleukin-1 beta; iNOS: inducible NOS; L-NMMA: L-NG-monomethyl-arginine; lpl: lipoprotein lipase; NF-B: nuclear factor-kappa B; NOS: nitric oxide synthase; PBMNCs: peripheral blood mononuclear cells; PBS: phosphate-buffered saline; PE: phycoerythrin; PFA: paraformaldehyde; ppar2: peroxisome proliferator-activated receptor gamma 2; RT-PCR: reverse transcriptase polymerase chain reaction; S-BMMSC: BMMSCs in suspension; SLE: systemic lupus erythematosus; SP: spleen; sRANKL: soluble runt-related NF-B ligand; SSEA: stage-specific embryonic antigen; TGF: transforming growth factor beta; Th17: T helper 17 cells; TRAP: tartrate-resistant acid phosphatase; Tregs: regulatory T cells.

The authors declare that they have no competing interests.

KA and YY: contributions to conception and design of experiments, acquisition of data, analysis and interpretation of data. TY, CC, LT, and YJ: contributions to acquisition of data, analysis and interpretation of data. XC and SG: contributions to drafting the manuscript and revising critically. SS: contributions to conception and design, drafting the manuscript, and giving final approval of the version to be published. All authors have read and approved the manuscript for publication.

We thank Dr. Tao Cai from NIH for discussions and critical reading of the manuscript. This work was supported by grants from the National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services (R01DE017449 and R01 DE019932 to S.S.).

Friedenstein AJ, Chailakhyan RK, Latsinik NV, Panasyuk AF, Keiliss-Borok IV: Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo.

Transplantation 1974, 17:331-340. PubMedAbstract | PublisherFullText

Prockop DJ: Marrow stromal cells as stem cells for nonhematopoietic tissues.

Science 1997, 276:71-74. PubMedAbstract | PublisherFullText

Gang EJ, Bosnakovski D, Figueiredo CA, Visser JW, Perlingeiro RC: SSEA-4 identifies mesenchymal stem cells from bone marrow.

Blood 2007, 109:1743-1751. PubMedAbstract | PublisherFullText

Greco SJ, Liu K, Rameshwar P: Functional similarities among genes regulated by OCT4 in human mesenchymal and embryonic stem cells.

Stem Cells 2007, 25:3143-3154. PubMedAbstract | PublisherFullText

Conget PA, Minguell JJ: Phenotypical and functional properties of human bone marrow mesenchymal progenitor cells.

J Cell Physiol 1999, 181:67-73. PubMedAbstract | PublisherFullText

Covas DT, Panepucci RA, Fontes AM, Silva WA Jr, Orellana MD, Freitas MC, Neder L, Santos AR, Peres LC, Jamur MC, Zago MA: Multipotent mesenchymal stromal cells obtained from diverse human tissues share functional properties and gene-expression profiles with CD146+ perivascular cells and fibroblasts.

Exp Hematol 2008, 36:642-654. PubMedAbstract | PublisherFullText

Galmiche MC, Koteliansky VE, Brire J, Herv P, Charbord P: Stromal cells from human long-term marrow cultures are mesenchymal cells that differentiate following a vascular smooth muscle differentiation pathway.

Blood 1993, 82:66-76. PubMedAbstract | PublisherFullText

Haynesworth SE, Baber MA, Caplan AI: Cell surface antigens on human marrow-derived mesenchymal cells are detected by monoclonal antibodies.

Bone 1992, 13:69-80. PubMedAbstract | PublisherFullText

Martinez C, Hofmann TJ, Marino R, Dominici M, Horwitz EM: Human bone marrow mesenchymal stromal cells express the neural ganglioside GD2: a novel surface marker for the identification of MSCs.

Blood 2007, 109:4245-4248. PubMedAbstract | PublisherFullText | PubMedCentralFullText

Sacchetti B, Funari A, Michienzi S, Di Cesare S, Piersanti S, Saggio I, Tagliafico E, Ferrari S, Robey PG, Riminucci M, Bianco P: Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment.

Cell 2007, 131:324-336. PubMedAbstract | PublisherFullText

Shi S, Gronthos S: Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp.

J Bone Miner Res 2003, 18:696-704. PubMedAbstract | PublisherFullText

Shi S, Gronthos S, Chen S, Counter CM, Robey PG, Wang C-Y: Bone formation by human postnatal bone marrow stromal stem cells is enhanced by telomerase expression.

Nat Biotechnol 2002, 20:587-591. PubMedAbstract | PublisherFullText

Sordi V, Malosio ML, Marchesi F, Mercalli A, Melzi R, Giordano T, Belmonte N, Ferrari G, Leone BE, Bertuzzi F, Zerbini G, Allavena P, Bonifacio E, Piemonti L: Bone marrow mesenchymal stem cells express a restricted set of functionally active chemokine receptors capable of promoting migration to pancreatic islets.

Blood 2005, 106:419-427. PubMedAbstract | PublisherFullText

Kwan MD, Slater BJ, Wan DC, Longaker MT: Cell-based therapies for skeletal regenerative medicine.

Hum Mol Genet 2002, 17(R1):R93-98.

Panetta NJ, Gupta DM, Quarto N, Longaker MT: Mesenchymal cells for skeletal tissue engineering.

Panminerva Med 2009, 51:25-41. PubMedAbstract | PublisherFullText

Liu Y, Wang L, Kikuiri T, Akiyama K, Chen CD, Xu XT, Yang RL, Chen WJ, Wang SL, Shi S: Mesenchymal stem cell-based tissue regeneration is governed by recipient T lymphocytes via IFN-Gamma and TNF-Alpha.

Nat Medicine 2011, 17:1594-1601. PublisherFullText

Nauta AJ, Fibbe WE: Immunomodulatory properties of mesenchymal stromal cells.

Blood 2007, 110:3499-3506. PubMedAbstract | PublisherFullText

Uccelli A, Pistoia V, Moretta L: Mesenchymal stem cells: a new strategy for immunosuppression?

Trends Immunol 2007, 28:219-226. PubMedAbstract | PublisherFullText

Uccelli A, Moretta L, Pistoia V: Mesenchymal stem cells in health and disease.

Nat Rev Immunol 2008, 8:726-736. PubMedAbstract | PublisherFullText

Aggarwal S, Pittenger MF: Human mesenchymal stem cells modulate allogeneic immune cell responses.

Read more here:
Characterization of bone marrow derived mesenchymal stem ...

Whats it like to donate stem cells?

People usually volunteer to donate stem cells for an allogeneic transplant either because they have a loved one or friend who needs a match or because they want to help people. Some people give their stem cells so they can get them back later for an autologous transplant.

People who want to donate stem cells or join a volunteer registry can speak with their doctors or contact the National Marrow Donor Program to find the nearest donor center. Potential donors are asked questions to make sure they are healthy enough to donate and dont pose a risk of infection to the recipient. For more information about donor eligibility guidelines, contact the National Marrow Donor Program or the donor center in your area (see the To learn more section for contact information).

A simple blood test is done to learn the potential donors HLA type. There may be a one-time, tax-deductible fee of about $75 to $100 for this test. People who join a volunteer donor registry will most likely have their tissue type kept on file until they reach age 60.

Pregnant women who want to donate their babys cord blood should make arrangements for it early in the pregnancy, at least before the third trimester. Donation is safe, free, and does not affect the birth process. For more, see the section called How umbilical cord blood is collected.

If a possible stem cell donor is a good match for a recipient, steps are taken to teach the donor about the transplant process and make sure he or she is making an informed decision. If a person decides to donate, a consent form must be signed after the risks of donating are fully discussed. The donor is not pressured take part. Its always a choice.

If a person decides to donate, a medical exam and blood tests will be done to make sure the donor is in good health.

This process is often called bone marrow harvest, and its done in an operating room. The donor is put under general anesthesia (given medicine to put them into a deep sleep so they dont feel pain) while bone marrow is taken. The marrow cells are taken from the back of the pelvic (hip) bone. A large needle is put through the skin and into the back of the hip bone. Its pushed through the bone to the center and the thick, liquid marrow is pulled out through the needle. This is repeated several times until enough marrow has been taken out (harvested). The amount taken depends on the donors weight. Often, about 10% of the donors marrow, or about 2 pints, are collected. This takes about 1 to 2 hours. The body will replace these cells within 4 to 6 weeks. If blood was taken from the donor before the marrow donation, its often given back to the donor at this time.

After the bone marrow is harvested, the donor is taken to the recovery room while the anesthesia wears off. The donor may then be taken to a hospital room and watched until fully alert and able to eat and drink. In most cases, the donor is free to leave the hospital within a few hours or by the next morning.

The donor may have soreness, bruising, and aching at the back of the hips and lower back for a few days. Over-the-counter acetaminophen (Tylenol) or non-steroidal anti-inflammatory drugs (such as aspirin, ibuprofen, or naproxen) are helpful. Some people may feel tired or weak, and have trouble walking for a few days. The donor might be told to take iron supplements until the number of red blood cells returns to normal. Most donors are back to their usual schedule in 2 to 3 days. But it could take 2 or 3 weeks before they feel completely back to normal.

There are few risks for donors and serious complications are rare. But bone marrow donation is a surgical procedure. Rare complications could include anesthesia reactions, infection, transfusion reactions (if a blood transfusion of someone elses blood is needed this doesnt happen if you get your own blood), or injury at the needle insertion sites. Problems such as sore throat or nausea may be caused by anesthesia.

Allogeneic stem cell donors do not have to pay for the harvesting because the recipients insurance company usually covers the cost.

Once the cells are collected, they are filtered through fine mesh screens. This prevents bone or fat particles from being given to the recipient. For an allogeneic or syngeneic transplant, the cells may be given to the recipient through a vein soon after they are harvested. Sometimes they are frozen, such as when the donor lives far away from the recipient.

For several days before starting the donation process, the donor is given a daily injection (shot) of filgrastim (Neupogen). This is a growth-factor drug that causes the bone marrow to make and release stem cells into the blood. Filgrastim can cause some side effects, the most common being bone pain and headaches. These may be helped by over-the-counter acetaminophen (Tylenol) or nonsteroidal anti-inflammatory drugs (like aspirin or ibuprofen). Nausea, sleeping problems, low-grade (mild) fevers, and tiredness are other possible effects. These go away once the injections are finished and collection is completed.

Blood is removed through a catheter (a thin, flexible plastic tube) that is put in a large vein in the arm or chest. Its then cycled through a machine that separates the stem cells from the other blood cells. The stem cells are kept while the rest of the blood is returned to the donor through the same catheter. This process is called apheresis (a-fur-REE-sis). It takes about 2 to 4 hours and is done as an outpatient procedure. Often the process needs to be repeated daily for a few days, until enough stem cells have been collected.

Possible side effects of the catheter can include trouble placing the catheter in the vein, a collapsed lung from catheter placement, blockage of the catheter, or infection of the catheter or at the area where it enters the vein. Blood clots are another possible side effect. During the apheresis procedure donors may have problems caused by low calcium levels from the anti-coagulant drug used to keep the blood from clotting in the machine. These can include feeling lightheaded or tingly, and having chills or muscle cramps. These go away after donation is complete, but may be treated by giving the donor calcium supplements.

The process of donating cells for yourself (autologous stem cell donation) is pretty much the same as when someone donates them for someone else (allogeneic donation). Its just that in autologous stem cell donation the donor is also the recipient, giving stem cells for his or her own use later on. For some people, there are a few differences. For instance, sometimes chemotherapy (chemo) is given before the filgrastim is used to tell the body to make stem cells. Also, sometimes it can be hard to get enough stem cells from a person with cancer. Even after several days of apheresis, there may not be enough for the transplant. This is more likely to be a problem if the patient has had certain kinds of chemo in the past, or if they have an illness that affects their bone marrow.

Sometimes a second drug called plerixafor (Mozobil) is used along with filgrastim in people with non-Hodgkin lymphoma or multiple myeloma. This boosts the stem cell numbers in the blood, and helps reduce the number of apheresis sessions needed to get enough stem cells. It may cause nausea, diarrhea, and sometimes, vomiting. There are medicines to help if these symptoms become a problem. Rarely the spleen can enlarge and even rupture. This can cause severe internal bleeding and requires emergency medical care. The patient should tell the doctor right away if they have any pain in their left shoulder or under their left rib cage which can be symptoms of this emergency.

Parents can donate their newborns cord blood to volunteer or public cord blood banks at no cost. This process does not pose any health risk to the infant. Cord blood transplants use blood that would otherwise be thrown away.

After the umbilical cord is clamped and cut, the placenta and umbilical cord are cleaned. The cord blood is put into a sterile container, mixed with a preservative, and frozen until needed.

Remember that if you want to donate or bank (save) your childs cord blood, you will need to arrange it before the baby is born. Some banks require you to set it up before the 28th week of pregnancy, although others accept later setups. Among other things, you will be asked to answer health questions and sign a consent form.

Many hospitals collect cord blood for donation, which makes it easier for parents to donate. For more about donating your newborns cord blood, call 1-800-MARROW2 (1-800-627-7692) or visit Be the Match.

Privately storing a babys cord blood for future use is not the same as donating cord blood. Its covered in the section called Other transplant issues.

Read the original here:
Whats it like to donate stem cells?

Bone Marrow and Stem Cell Transplants Lymphoma Info

Surgeon performs bone marrow harvest

The terms "Hodgkin's Disease," "Hodgkin's Lymphoma," and "Hodgkin Lymphoma" are used interchangeably throughout this site.

Bone Marrow Transplants (BMT) and Peripheral Blood Stem Cell Transplants (PBSCT) are emerging as mainstream treatment for many cancers, including Hodgkin's Disease and Medium/High grade aggressive)Non-Hodgkin's lymphoma.

BMTs have been used to treat lymphoma for more than 10 years, but until recently they were used mostly within clinical trials. Now BMTs are being used in conjunction with high doses of chemotherapy as a mainstream treatment.

When high doses of chemotherapy are planned, which can destroy the patients bone marrow, physicians will typically remove marrow from the patients bone before treatment and freeze it. After chemotherapy, the marrow is thawed and injected into a vein to replace destroyed marrow. This type of transplant is called an autologous transplant. If the transplanted marrow is from another person, it is called an allogeneic transplant.

In PBSCTs, another type of autologous transplant, the patient's blood is passed through a machine that removes the stem cells the immature cells from which all blood cells develop. This procedure is called apheresis and usually takes three or four hours over one or more days. After treatment to kill any cancer cells, the stem cells are frozen until they are transplanted back to the patient. Studies have shown that PBSCTs result in shorter hospital stays and are safer and more cost effective than BMTs.

Read more here:
Bone Marrow and Stem Cell Transplants Lymphoma Info

New blood cancer drug reaches cells hiding in bone marrow …

SAN DIEGO, July 28 (UPI) -- A new drug aimed at dormant cancer stem cells that hide in the hypoxic zones of bone marrow, where most drugs can't reach, is currently entering 5 Phase II clinical trials after it was shown to make blood cancer treatment more effective.

Researchers in a Phase I clinical trial, the results of which are published in The Lancet Haematology, found that the drug vismodegib was effective against three types of blood cancer -- refractory or resistant myeloid leukemia, myelodysplastic syndrome and myelofibrosis.

Vismodegib inhibits the Hedgehog signaling pathway, which is essential to both vertebrate embryonic development and has been implicated in the development of some cancers. The drug, trade name Erivedge, is already approved in the U.S. and Europe for treatment of metastatic or locally advanced basal cell carcinoma.

"Our hope is that this drug will enable more effective treatment to begin earlier and that with earlier intervention, we can alter the course of disease and remove the need for, or improve the chances of success with, bone marrow transplantation," said Dr. Catriona Jamieson, chief of the Division of Regenerative Medicine in the School of Medicine at the University of California San Diego, in a press release. "It's all about reducing the burden of disease by intervening early."

Preclinical research showed the drug could "coax" dormant cancer stem cells in hypoxic zones to begin differentiating and enter the bloodstream, where they can be attacked by the chemotherapy and the immune system.

In the study, researchers treated 47 adults with blood and marrow cancers with with the drug in 28-day cycles. Treatment cycles were continued with escalating doses until a participant experienced adverse effects with no improvement in their condition. The participants who did not have adverse reactions or serious side effects continued to receive treatment cycles of the drug.

Serious adverse effects were seen in only 3 of the participants, though 60 percent of the group experienced treatment-related problems. Nearly half the people in the study saw positive clinical activity as a result of treatment with vismedogib, the researchers said, and 5 Phase II clinical trials are being scheduled for the drug for use with blood cancer.

"This drug gets that unwanted house guests to leave and never come back," Jamieson said. "It's a significant step forward in treating people with refractory or resistant myeloid leukemia, myelodysplastic syndrome and myelofibrosis. It's a bonus that the drug can be administered as easily as an aspirin, in a single, daily oral tablet."

Related UPI Stories

Visit link:
New blood cancer drug reaches cells hiding in bone marrow ...

Effects of Tanshinone IIA on osteogenic differentiation of …

Date: 01 Aug 2015

Rent the article at a discount

* Final gross prices may vary according to local VAT.

Tanshinone IIA (TSA) is a lipophilic diterpene purified from the Chinese herb Danshen, which exhibits potent antioxidant and anti-inflammatory properties. Effect of TSA remains largely uninvestigated on the osteogenic differentiation of bone marrow mesenchymal stem cells (BM-MSCs), which are widely used in cell-based therapy of bone diseases. In the present study, both ALP activity at day 7 and calcium content at day 24 were upregulated during the osteogenesis of mouse BM-MSCs treated with TSA (1 and 5M), demonstrating that it promoted the osteogenesis at both early and late stages. We found that TSA promoted osteogenesis and inhibited osteoclastogenesis, evident by RT-PCR analysis of osteogenic marker gene expressions. However, osteogenesis was inhibited by TSA at 20M. We further revealed that TSA (1 and 5M) upregulated BMP and Wnt signaling. Co-treatment with Wnt inhibitor DKK-1 or BMP inhibitor noggin significantly decreased the TSA-promoted osteogenesis, indicating that upregulation of BMP and Wnt signaling plays a significant role and contributes to the TSA-promoted osteogenesis. Of clinical interest, our study suggests TSA as a promising therapeutic strategy during implantation of BM-MSCs for a more effective treatment of bone diseases.

Read this article:
Effects of Tanshinone IIA on osteogenic differentiation of ...

Bone Marrow/Stem Cell Transplant | UCLA Transplantation …

The UCLA Program is a combined program caring for patients with Hematologic Malignancies receiving chemotherapy and those patients for whom Stem Cell Transplantation is the therapy of choice. The treatmentof blood and marrow cancers includecurrently available therapies, investigational drugs and treatments, as well as stem cell transplantation. Our physicians meet weekly to discussindividual treatment approachesas part of developing a coordinated treatment recommendation.

Bone Marrow Transplantation was first performed at UCLA in 1968 using a related allogeneic transplant to treat an 18 month old child with severe combined immunodeficiency syndrome. The UCLA Marrow Transplantation Program was formally initiated in 1973. Unrelated donor marrow transplants have been carried out at UCLA since 1987, and Cord Blood Transplants have been performed at UCLA since 1996. Autologous transplants have been performed at our program since 1977. Since 1992 most of the Autologous Transplants have utilized Peripheral Blood Stem Cells. Since 1998 an increasing number of the Allogenic Transplants have utilized Peripheral Blood Stem Cells. From inception to the completion of 2007 we have performed 3726 transplants (3080 transplants in the adult population and 646 in the pediatric population).

For decades, this comprehensive program has provided a full range of services as a local, regional, national, and international referral center for transplantations for selected malignancies:

Our goals include finding new and innovative treatments for malignancies and expanding the effectiveness and applicability of bone marrow transplantation through such means as biologic response modifiers, growth factors, and chemotherapeutic agents.

Protocols involving chemotherapy with or without radiation therapy for patients in remission or relapse are available using bone marrow or peripheral blood stem cells from allogeneic, autologous and unrelated donors.

A bone marrow transplant is a procedure that transplant healthy bone marrow into a patient whose bone marrow is not working properly. A bone marrow transplant may be done for several conditions including hereditary blood diseases, hereditary metabolic diseases, hereditary immune deficiencies, and various forms of cancer.

Visit our Health Library to learn more:

Bone MarrowTransplant

How to Schedule Your Evaluation Appointment at UCLA

The United Network for Organ Sharing (UNOS) provides a toll-free patient services lines to help transplant candidates, recipients, and family members understand organ allocation practices and transplantation data. You may also call this number to discuss problems you may be experiencing with your transplant center or the transplantation system in general. The toll-free patient services line number is 1-888-894-6361

Read this article:
Bone Marrow/Stem Cell Transplant | UCLA Transplantation ...

Mesenchymal stem cell – Wikipedia, the free encyclopedia

Mesenchymal stem cells, or MSCs, are multipotent stromal cells that can differentiate into a variety of cell types,[1] including: osteoblasts (bone cells),[2]chondrocytes (cartilage cells),[3]myocytes (muscle cells)[4] and adipocytes (fat cells). This phenomenon has been documented in specific cells and tissues in living animals and their counterparts growing in tissue culture.

While the terms mesenchymal stem cell and marrow stromal cell have been used interchangeably, neither term is sufficiently descriptive:

The youngest, most primitive MSCs can be obtained from the umbilical cord tissue, namely Wharton's jelly and the umbilical cord blood. However the MSCs are found in much higher concentration in the Whartons jelly compared to the umbilical cord blood, which is a rich source of hematopoietic stem cells. The umbilical cord is easily obtained after the birth of the newborn, is normally thrown away, and poses no risk for collection. The umbilical cord MSCs have more primitive properties than other adult MSCs obtained later in life, which might make them a useful source of MSCs for clinical applications.

An extremely rich source for mesenchymal stem cells is the developing tooth bud of the mandibular third molar. While considered multipotent, they may prove to be pluripotent. The stem cells eventually form enamel, dentin, blood vessels, dental pulp, and nervous tissues, including a minimum of 29 different unique end organs. Because of extreme ease in collection at 810 years of age before calcification, and minimal to no morbidity, they will probably constitute a major source for personal banking, research, and multiple therapies. These stem cells have been shown capable of producing hepatocytes.

Additionally, amniotic fluid has been shown to be a rich source of stem cells. As many as 1 in 100 cells collected during amniocentesis has been shown to be a pluripotent mesenchymal stem cell.[9]

Adipose tissue is one of the richest sources of MSCs. There are more than 500 times more stem cells in 1 gram of fat than in 1 gram of aspirated bone marrow. Adipose stem cells are actively being researched in clinical trials for treatment of a variety of diseases.

The presence of MSCs in peripheral blood has been controversial. However, a few groups have successfully isolated MSCs from human peripheral blood and been able to expand them in culture.[10] Australian company Cynata also claims the ability to mass-produce MSCs from induced pluripotent stem cells obtained from blood cells using the method of K. Hu et al.[11][12]

Mesenchymal stem cells are characterized morphologically by a small cell body with a few cell processes that are long and thin. The cell body contains a large, round nucleus with a prominent nucleolus, which is surrounded by finely dispersed chromatin particles, giving the nucleus a clear appearance. The remainder of the cell body contains a small amount of Golgi apparatus, rough endoplasmic reticulum, mitochondria, and polyribosomes. The cells, which are long and thin, are widely dispersed and the adjacent extracellular matrix is populated by a few reticular fibrils but is devoid of the other types of collagen fibrils.[13][14]

The International Society for Cellular Therapy (ISCT) has proposed a set of standards to define MSCs. A cell can be classified as an MSC if it shows plastic adherent properties under normal culture conditions and has a fibroblast-like morphology. In fact, some argue that MSCs and fibroblasts are functionally identical.[15] Furthermore, MSCs can undergo osteogenic, adipogenic and chondrogenic differentiation ex-vivo. The cultured MSCs also express on their surface CD73, CD90 and CD105, while lacking the expression of CD11b, CD14, CD19, CD34, CD45, CD79a and HLA-DR surface markers.[16]

MSCs have a great capacity for self-renewal while maintaining their multipotency. Beyond that, there is little that can be definitively said. The standard test to confirm multipotency is differentiation of the cells into osteoblasts, adipocytes, and chondrocytes as well as myocytes and neurons. MSCs have been seen to even differentiate into neuron-like cells,[17][18] but there is lingering doubt whether the MSC-derived neurons are functional.[19] The degree to which the culture will differentiate varies among individuals and how differentiation is induced, e.g., chemical vs. mechanical;[20] and it is not clear whether this variation is due to a different amount of "true" progenitor cells in the culture or variable differentiation capacities of individuals' progenitors. The capacity of cells to proliferate and differentiate is known to decrease with the age of the donor, as well as the time in culture. Likewise, whether this is due to a decrease in the number of MSCs or a change to the existing MSCs is not known.[citation needed]

See the rest here:
Mesenchymal stem cell - Wikipedia, the free encyclopedia

JCI – Bone marrow mesenchymal stem cells and TGF- …

High levels of active TGF- in the bone marrow and abnormalities in bone remodeling are associated with multiple skeletal disorders. Genetic mutations in the TGF- signaling pathway cause premature activation of matrix latent TGF- and may manifest with various skeletal defects. There are additional diseases that result in high levels of active TGF-, which may contribute to the pathology. Here, we discuss how abnormal TGF- signaling results in uncoupled bone remodeling, mainly by loss of site-directed recruitment of MSCs that causes aberrant bone formation. Direct or indirect inhibition of TGF- signaling may provide potential therapeutic options for these disorders.

Genetic disorders. The critical role of TGF-1 in the reversal phase of bone remodeling is demonstrated by the range of skeletal disorders resulting from mutations in genes involved in TGF-1 signaling. Camurati-Engelmann disease (CED), characterized by a fusiform thickening of the diaphysis of the long bones and skull, is caused by mutations in TGFB1 that result in premature activation of TGF-1 (7174). Approximately 11 different TGFB1 mutations have been identified from families affected by CED (75, 76). All of the mutations are located in the region encoding LAP, either destabilizing LAP disulfide bridging or affecting secretion of the protein, both of which increase TGF-1 signaling, as confirmed by in vitro cell cultures and mouse models. Bone histology sections from patients with CED show decreased trabecular connectivity despite normal bone histomorphometric parameters with respect to osteoblast and osteoclast numbers (76, 77), suggestive of uncoupled bone remodeling. In vitro, the ratio of active to total TGF-1 in conditioned medium from cells expressing the CED mutant TGF-1 is significantly higher and enhances MSC migration (18). Targeted recruitment of MSCs to the bone-remodeling site is likely disrupted, secondary to loss of a TGF- gradient.

Elevations in TGF- signaling have also been observed in many genetic connective tissue disorders with craniofacial, skeletal, skin, and cardiovascular manifestations, including Marfan syndrome (MFS), Loeys-Dietz syndrome (LDS), and Shprintzen-Goldberg syndrome (SGS). MFS is caused by mutations in fibrillin and often results in aortic dilation, myopia, bone overgrowth, and joint laxity. Fibrillin is deposited in the ECM and normally binds TGF-, rendering it inactive. In MFS, the decreased level of fibrillin enhances TGF- activity (78). LDS is caused by inactivating mutations in genes encoding TRI and TRII (79). Physical manifestations include arterial aneurysms, hypertelorism, bifid uvula/cleft palate, and bone overgrowth resulting in arachnodactyly, joint laxity, and scoliosis. Pathologic analyses of affected tissue suggest chronically elevated TGF- signaling, despite the inactivating mutation (79). The mechanism of enhanced TGF- signaling remains under investigation. SGS is caused by mutations in the v-ski avian sarcoma viral oncogene homolog (SKI; refs. 80, 81) and causes physical features similar to those of MFS plus craniosynostosis. SKI negatively regulates SMAD-dependent TGF- signaling by impeding SMAD2 and SMAD3 activation, preventing nuclear translocation of the SMAD4 complex, and inhibiting TGF- target gene output by competing with p300/CBP for SMAD binding and recruiting transcriptional repressor proteins, such as mSin3A and HDACs (8284).

The neurocutaneous syndrome neurofibromatosis type 1 (NF1) has been noted to have skeletal features similar to those of CED, MFS, and LDS, including kyphoscoliosis, osteoporosis, and tibial pseudoarthrosis. Hyperactive TGF-1 signaling has been implicated as the primary factor underlying the pathophysiology of the osseous defects in Nf1fl/Col2.3Cre mice, a model of NF1 that closely recapitulates the skeletal abnormalities found in human disease (85). The exact mechanisms mediating mutant neurofibrominassociated enhancement of TGF- production and signaling remain unknown.

Osteoarthritis. While genetic disorders are rare, they have provided critical insight into the pathophysiology of more common disorders. Uncoupled bone remodeling accompanies the onset of osteoarthritis. TGF-1 is activated in subchondral bone in response to altered mechanical loading in an anterior cruciate ligament transection (ACLT) mouse model of osteoarthritis (86). High levels of active TGF-1 induced formation of nestin+ MSC clusters via activation of ALK5-SMAD2/3. MSCs underwent osteoblast differentiation in these clusters, leading to formation of marrow osteoid islets. Transgenic expression of active TGF-1 in osteoblastic cells alone was sufficient to induce osteoarthritis, whereas direct inhibition of TGF- activity in subchondral bone attenuated the degeneration of articular cartilage. Knockout of Tgfbr2 in nestin+ MSCs reduced osteoarthritis development after ACLT compared with wild-type mice, which confirmed that MSCs are the target cell population of TGF- signaling. High levels of active TGF-1 in subchondral bone likely disrupt the TGF- gradient and interfere with targeted migration of MSCs. Furthermore, mutations of ECM proteins that bind to latent TGF-s, such as small leucine-rich proteoglycans (87) and fibrillin (88), or mutations in genes involved in activation of TGF-, such as in CED (76) and LDS (89), are associated with high osteoarthritis incidence. Osteoblast differentiation of MSCs in aberrant locations appears histologically as subchondral bone osteoid islets and alters the thickness of the subchondral plate and calcified cartilage zone, changes known to be associated with osteoarthritis (90, 91). A computer-simulated model found that a minor increase in the size of the subchondral bone (1%2%) causes significant changes in the mechanical load properties on articular cartilage, which likely leads to degeneration (86). Importantly, inhibition of the TGF- signaling pathway delayed the development of osteoarthritis in both mouse and rat models (86).

MSCs in bone loss. Aging leads to deterioration of tissue and organ function. Skeletal aging is especially dramatic: bone loss in both women and men begins as early as the third decade, immediately after peak bone mass. Aging bone loss occurs when bone formation does not adequately compensate for osteoclast bone resorption during remodeling. Age-associated osteoporosis was previously believed to be due to a decline in survival and function of osteoblasts and osteoprogenitors; however, recent work by Park and colleagues found that mature osteoblasts and osteoprogenitors are actually nonreplicative cells and require constant replenishment from bone marrow MSCs (92). When MSCs fail to migrate to bone-resorptive sites or are unable to commit and differentiate into osteoblasts, new bone formation is impaired. Therefore, insufficient recruitment of MSCs, or their differentiation to osteoblasts, at the bone remodeling surface may contribute to the decline in bone formation in the elderly.

There are multiple hypotheses regarding the decreased osteogenic potential of MSCs during aging. For example, during aging, the bone marrow environment has an increased concentration of ROS and lipid oxidation that may decrease osteoblast differentiation, yet increase osteoclast activity (93, 94). MSCs also undergo senescence, which decreases proliferative capacity and contributes to decreased bone formation (95, 96). Cellular senescence involves the secretion of a plethora of factors, including TGF-, which induces expression of cyclin-dependent kinase inhibitors 2A and 2B (p16INK4A and p15INK4B, respectively; refs. 97).

Microgravity experienced by astronauts during spaceflight causes severe physiological alterations in the human body, including a 1%2% loss of bone mass every month during spaceflight (98). Several studies have shown decreases in osteoblastic markers of bone formation and increases in bone resorption (99101). The underlying molecular mechanisms responsible for the apparent concurrent decrease in bone formation and increase in bone resorption remain under investigation. Work by the McDonald group suggests that bone remodeling may become uncoupled under zero-gravity conditions secondary to decreased RhoA activity and resultant changes in actin stress fiber formation (102). In modeled microgravity, cultured human MSCs exhibit disruption of F-actin stress fibers within three hours of initiation of microgravity; the fibers are completely absent after seven days. RhoA activity is significantly reduced, and introduction of an adenoviral construct expressing constitutively active RhoA can reverse the elimination of stress fibers, significantly increasing markers of osteoblast differentiation (102). Under zero-gravity conditions, RhoA is unable to bind to its receptor, and a sufficient number of MSCs may not be able to migrate correctly to the bone-resorptive site for osteoblast differentiation, ultimately leading to bone loss with every cycle of remodeling.

Bone metastases are a frequent complication of cancer and often have both osteolytic and osteoblastic features, indicative of dysregulated bone remodeling. The importance of the bone marrow microenvironment contributing to the spread of cancer was first described in 1889 (103), postulating that tumor cells can grow only if they are in a conducive environment. Activation of matrix TGF- during bone remodeling plays a central role in the initiation of bone metastases and tumor expansion by regulating osteolytic and prometastatic factors (reviewed in refs. 104110). For example, TGF- can induce osteoclastic bone destruction by upregulating tumor cell expression of PTHrP and IL-11. Additionally, upregulation of CXCR4 by TGF- may home cancer cells to bones.

Link:
JCI - Bone marrow mesenchymal stem cells and TGF- ...

Haematopoietic stem cells and early lymphoid progenitors …

UMS#7=U/U'K 6U,leTC(HG~}fllvFp0LN*i0`U~1Y:,WxX*3h"jIDfU!`8) ty@$7;hi6 vCvZLEM{$DfCxh&dE#]}Q |Yk{=$ &}SyX((n&'weuQ:ir^fN?<4X/iJ`fdfRP2Yn}4:kqoGSP9%%!^]?#9+A^Qg Q2:?putgZW&w*8glqcVO~VYpKtBPbP$ra ;wA&r:r$|K!,_Q:xtr[Wmo6 _WI$ui`Z:[$J#);Ie;2Cr Ew bv@~[.LZB[DHCvkav-E! Kh$Y`X:PS=2kXpm@7E!LFFpnZ9&n0:64Y4ZS Md:K`Mc JOj {WybC8"UO_}v;noMTHc080C g7G Mbsu 8_bY} !ie-j"w)7=Xm_3!_cLI%[s}x5wC/F!QfG" V>UKlKkkst&e9~AAI.)cE;pFOx@n >gmA&C.]2~<=yQZ(^$x(X/'UB/CT_PyR -.hQ &5v :s5N !S%(VYQt>1Z*4Z-D' Q;(%zcLHaQ3_1>~W0{En6x&1GyX:^=6+{lJ2pr0dD `jV{ndd[T)x1#$ HxEpTxo+sEL!SfRn2/|V7zBi6yL{7@KG?mq>_&BVZRah>4#A{%;7!Kh837*qR ^8[}u&D5 s/n8Wb%,"mg5HhZcu1ZV<]kw)UQmn!EQUrK(OYV%KPdebCnJ._>2&jbg%|6z/dl% B28XTD6?4qixs6%*{ }k8:-TE,H/mWd%+9f /Y8OyV@*SsAnIHV`CbL#54K`u5VsQfyy)iF>j18<'VsCW631Zg80/Zn*L8?wIs;3tBX6x M|]2R.I(@]px)Myl ["rDTsXhNU!?7Shrt8]J!E.#Hp2xk~GS 7c2!uk'QrQJp;$[> s]7_z}hed1+Q2ld;|Yo]WT-y)~&/U C[UwZUMi,Lx?`/iUf%6::c$")+IHSM5#zS-rkR>G7:Pe=A(4NZK<=qZ0R1zb8SEU5Yy3t*#jkr a::2qx haqn[bd;ZC~k!2bCdC3AZ3CV1{k|:VgB-s)44XhytxPcd=c,8Q86KjNH$(~N5TbI{fA^% INEO(%IjEhcH[GeMC{8;/o=vKE3RSKoUeof[26tO0yhs+[+:/+W*uNLvLz 2!cW[B= "?z8YhZ>q7[ aS{[!I8-Z^N/{o: 6pEZrB#= 3G"t:sos|7A7P:1pr3X /i`Rzt2v-@M_dIS nS[Ahx48q|x ?^u$kMv W??_[V |}8(5z$iO/MOW~mg `l:U@t :`+#Y()[}w"%*udO{axwx#Y *QQ!hN fd ,9Ap*7$?6Y`ZJwTx7.TV(f A#sC<1e%d[;ZBb? AJFhm!>=P'M !L=Nw%R6Q$8 _?n+G}_HJ8b'E_k#.2Yy'8Y'2Y Y1#*N++Vbq1y,.h>Rro!W8q"k"0} iYCciD?HVc=eGM}'?]9)P|[c T;DT!`3_;2VI1lESwC9k:VpbyyT x5roxwyeJV)v<}SZngxGALAhlenoj%t41 ;iKz'-nMNlJiRHr&'v/?2RY(&zu)IItPEtX0! [.`']xo# XG7tO;45l( '6JI3byE"j(XYCX#GpM]1F#L63^QY7LKs?6)AL ^`ler'Xtt0gNHoIQ!`F7N hxB l^,2?WtMSLij-'i0xxm* SuHOv2+2ca1m yRH_&uglME`tDD"5Rr=GJ^Gs {'3U +7~NUY*$5BUBu.-tDQt=>L7kd2zInYJmEK**f-*|fKIhcS&|<)Z#$9 vHtRQs!qp+cf(?l]}1#~-?$=-yFcs_|O |0#~gCl^>v2eW 44ZqqLXXx][O4HD6kxzr;?/VY_/Nug hyrW3,DeJ$9,yw{EJJMQoH DUCVr]D&AJz06&cC>+"G^^~1-z4IuQZ1&4Khqw$a-^ DE S; P!PH-t*bY](W+ehhD{>pNFZ| GHUYrdV* F[jLw:fax@GD82%)SPNyK$,}k3QWx7e*ILXss8ur:~K]Mb#sj /p~*QGFhrD]c:P*K"iT/QJ}6w58RW#F6kmQLFz'|ej)v$UARS 3hz'[C-S&78iN6BZQ8?W[o6}N (,b']T0kAQRCR%u0d^w96zi]9P*/3_?9vxI.W?mdW9t~ zKo-X3KRVHtiK-5fJ1blt:|;'RkH9a@ C~)mlu%PBG/E PTWA]XM^|os~]>c'7Aw %V Q%tkU zW(P!r4Z_m[W)4VMIho;qPc86ht5Be8JqhBowv3GCLP *Pd=. NY-q`_|ptqC'p]w[kr!~$MpqO~O!82#.)-J.qEqZUD0sy+^CH@U{%#b>^~GBP24 *FYSy~/rYxu/HJi`Y VDm{t<j-ylstO+L1naByt8bs"vr&xp;CK_SY-~CTCKt2<5Gwdn@bO+wD.00(. dq!,|=h4frY7OD]u{`Sqw,uk'nqxjLF(!0hr^"C=Q_De=?M3gBef16-ScDw}2/%r};2eDH v, N>s}=1wK6^NKS6z1$*Ti6y$Xa;9d'Y/e%/K/#Nx;DB6{]g"+$2ZmNB-pm=HD,4*y$cy,Q<:mK2.;oro,!U bBu(aZV#Vi_5s|} 5=A2Mg}$te^r}bg"2O2dCx}[Z^sj%5wN0I>;P$"gd^~Brtk}af#zKz;l="-sj/ }MfdhDJ:9akc T(HrRuScQTtG?`@;S0(/ T2B]_bg4mri5 :>jMIA?clL]=cA n#N Y.E'y:;mn};j' Dzn zx>U5ux`hQl>9^UoV1:ps1.'N&O.uu^<{;Ie& oY~pl-"Tn DE2bs)/p@avA,1T@G{wc&k@DJCRmXkCMC,/*4+?"y_mm@Ej(Y7M3f< Qh@@ {9R28k?8P ?Q"*=C57jK1H>dv?'}h3'oTcy<@ ?/~q(k;u)9;Sk$@HRKi"5`BI51`"&RV%Fz 6Q;ZN`ut#_y*y|^B~uIJ1n%asXE5m(Fx$moy,3>zt>9w9 @VE8$nUX`O2Lxl6K|t?5(71A}-^?`8P'6BdF(P.kPsL@].czqa- e F(+27QS:l?6A) W"&4t{})]CvR);s e7ETjf|*.3mmM8zN{3e c/Qh-3b'dd%gu:#",1lR2*igpM}N0+8kSmV`l/ Mk_U}#jWb?|5~K(w1Sze< NM`4xa]0`Ya23}ikK2"oP6{:nHWmB=Gn`cq?Ab ]zKD)2R|AX)L[xq!21`DB9_U41FIg^9wHFPAy5W0A@`k8b0[Ehk`24TKp~|B ]AJiNB*(kxRLDn{,% lXxhO 8(lRJ}8."@; 8zldQxu@/ GTBDlav+|7j0zL5Xu S !H#L*OpP]Ebk.!tCBlHar%&R'Cn{??nl|LfZzvJE*qqD> #GLNe|Tm|2 mE!9k,p6w:]j7LK.~6| MMHrIR?@#@Wb0egu5NCfAg?^x[.lnReJSf}26"PTF5% UR`0u5dZ}[^%|DEb ,R}tR4wrnK0h3eP.0fPy.f"eX4QZb<~j@4HrP{jOo}0g/qB$ Eq_GcO|7q ;lOD_6q(EGLp8Ta&Vn$'Ns0we}SZYaYTl4eNU^au3i/K2ZJ`q#t *AzsYjR$b9zlU:g2D%|aUyh>ZF~2Xh>{assRyf=68sq/"Nk.X=XLF{Ckmj]58l3v3 M +~W#?R)ra,%,{1vJx,I3Ml7|. 3f1IACqX0M9F@3`hrof,adUluXj]gu5gOxp4m-XpKVs=,9j7xCl0@ {m8'8vh4*oSmTJkZ/d0w;C_^_^>+{M0Y3a%YsJ~lW&v%F7k*q:HP[{pHA(F]P(C49W4G/Lc[) QGmHSCMa^dcb[m5Ys%O<:7Jl]a+,rrFWzY94&5!K_w4qDU;xV`-4f./x6Gn;/4$TK%?uU)/#?HEeBM?AYF-.#Ir}Q(fiV $%Sw`s$WRM*kQ-;)0j&LX, B"uN*k]UZv9_7Ns={190t= ^/- `@pH_e}]n/^JW?]K_ex?rb+!~a):b{0yQ?8N4ITh4,0V.5q_Nbe $~l)wC%WGpd"RLU!I8(b)H&`?os&h"/4]_:>GH|iX^wh:8xj^#r-,lK(BrDw{:mZV_/ LOW5?P%w)LMu6DZ.sR5yIaKq ]|geX+My8@+Fm1>Tm%`x|> $KFqE)Xe< ]9,4WAW;2]|8awCtQs0&!cs ?jS.B321:7;2B#XjOGR4' m1R0vj1v;]Zc|Dg2CIX)]@]l~^*:;@:t|w$_JJBCG Z36gaf-8 d .)+j6%bCxu=`[[5dz&m,b|E5oCw=lkOY~B}H -#N3i~/n-h9AEgUn^5!$2HjYww l|!R%;.),thH1MMYf- $,XJ_?=? 59I>)wMyNcl8BN3#~N |F(LI&u$3Z+vI.p?'Rr:fpmk`tUi? `)[q@0mJt(*K;osI]imSk0s[NT.T~8-hz / xkT5'IN#E568pZDD"Q4 #@L*pE,!Z3>PY($2Ia"I%/m ^?! $eK,Nvi.X7%R2;(/Dk<,p(&>+2=&@T_p Fn =czBN'H:N;pOu$I::?d~1lj|(n4W?KK2!`p!h79x4c; hGoQ=wyZTt<hxDmV~E>*m l vKeC2`$b!*Ie~K]] l"ER{oE`Z[`]U+#jn= YRWM+*&! h5IO=pM;!9}]qM Ir4K _ ^UM_btECOynxFw{}u>{J#v[VMM jK*@gr[^FA4R~|vFi+X&U xLP;[~t3zA<$|g1NEV7m6DeRtTBQ5 `WnF}vbLD$[, nyC"G:+.{7)A/3S0_mtzC)fKoEZZO%yCd(#/gG'GzvLJWF4k~.Md9^Eh 4 wy@ W Q+mZhWU)iW@oD):5dBPdQ]+n'g;C/a6`#JdE0XkY"3WEu>+M&s&VZ %oE@B((UMy(hvk*E 3SM^5{U66M5?OywZ<rWVVwDw.cuJK=]E/RiI37^Jn8/LmbdmsRR^/|~NqGZtP CE#TSN~.CYW^M: xI*xREd0fNDx{ NDkr$!Xox:GfrNG~2Zf^_bCQDA<[=|Hw<_Pd)foctg+skcIDDY!&5W@:Jc-3_`9_@UH &>lVlGJAxH;w$pp4TvstbqvH1#-Ti -QJ3 J"ZL5e&<@ NY>{C?,Xhia[^x;hnz:8}[%7D7T]mDU=A*MML4!7MZp5RWyDE.Q&* ~WSUST[x2eBo(wTsVB>{wPqSSi kAc12/h|c{w`eB|L9;8!&k 6}%4$bI9[&K]4ZC RcL q'c](^.'U`dxT5]~dW@6e, 1]zL9S +U"))$VYg[:AfKXjI9T{tDZe~h603B0 @l'> m%`. /1iP^*Fe6&dr$lYD da"6IEKTfYN zyZ-pACv[/c=hp{7L b+H< m L+B0f|',qd$[[Sf=ZWR8+Sn FkQ&-9LI.E%Dk/&gSyZUECw;4+yQ'L*VX~BEA|6brDpb m$hNlLs#| Wz$@g|q1B10#8Q8l>8{;nl.m?G "Q2ed)*zxl|v/m>zq M^- J$:';%m6oI1lE=]fTvLI=c|>#2r fKfC um"%Tj!b`+NeaB53GhL 5koC.HR%(*K yR^k@s.)u_u"4"XaK$ao$-ewBXZptjAykHFJHU B@BuneR=IQ,,"*iK*"9qI9;q7p_E4xu98mh~hL|G4l3Iy|

Go here to read the rest:
Haematopoietic stem cells and early lymphoid progenitors ...

Side effects of bone marrow and stem cell transplants …

You will have a low white blood cell count after your treatment. This means you are more at risk of getting an infection. You are likely to get an infection from the normally harmless bacteria we all have in our digestive systems and on our skin.

To stop this from happening your nurse may give you tablets called gut sterilisers (antibiotics) and mouthwashes. And they will encourage you to have a shower each day.

You are also at risk of infection from food. The nurses on the ward will tell you and your relatives about the food you can and can't eat. The rules vary from hospital to hospital but you may be told that

Your room will be thoroughly cleaned every day. Your visitors will be asked to wash their hands before they come into your room. They may also have to wear disposable gloves and aprons. Visitors with coughs and colds are not allowed. Some hospitals don't allow you to have plants or flowers in your room because bacteria and fungi can grow in the soil or water, and may cause infection.

Even with all these precautions, most people do get an infection at some point and need to have antibiotics. You can help yourself by trying to do your mouth care properly and getting up to shower and have your bed changed even on the days you don't feel too good.

After a transplant you will have lost immunity to diseases you were vaccinated against as a child. The team caring for you will advise you about the immunisations you need and when. You should only have inactivated immunisations and not live ones. To lower the risk of you getting any of these infections it is important that all your family have the flu vaccine and any children have all their immunisations.

Continue reading here:
Side effects of bone marrow and stem cell transplants ...

Stem Cell vs. Bone Marrow Transplant: Whats the …

With Brigham and Womens Hospital and Boston Childrens Hospital, Dana-Farber has performed thousands of stem cell/bone marrow transplants for adult and pediatric patients with blood cancers and other serious illnesses.

Whats the difference between these two terms? As it turns out, the only real distinction is in the method of collecting the stem cells.

Lets start with the basics.

Stem cells are versatile cells with the ability to divide and develop into many other kinds of cells.

Hematopoietic stem cells produce red blood cells, which deliver oxygen throughout the body; white blood cells, which help ward off infections; and platelets, which allow blood to clot and wounds to heal.

While chemotherapy and/or radiation therapy are essential treatments for the majority of cancer patients, high doses can severely weakenand even wipe outhealthy stem cells. Thats where stem cell transplantation comes in.

Stem cell transplantation is a general term that describes the procedures performed by the Adult Stem Cell Transplantation Program at Dana-Farber/Brigham and Womens Cancer Center and the Pediatric Stem Cell Transplantation Program at Dana-Farber/Boston Childrens Cancer and Blood Disorders Center.

Stem cells for transplant can come from bone marrow or blood.

When stem cells are collected from bone marrow and transplanted into a patient, the procedure is known as a bone marrow transplant. If the transplanted stem cells came from the bloodstream, the procedure is called a peripheral blood stem cell transplantsometimes shortened to stem cell transplant.

Whether you hear someone talking about a stem cell transplant or a bone marrow transplant, they are still referring to stem cell transplantation. The only difference is where in the body the transplanted stem cells came from. The transplants themselves are the same.

Read more here:
Stem Cell vs. Bone Marrow Transplant: Whats the ...

Stem Cell Basics: Introduction [Stem Cell Information]

Introduction: What are stem cells, and why are they important? What are the unique properties of all stem cells? What are embryonic stem cells? What are adult stem cells? What are the similarities and differences between embryonic and adult stem cells? What are induced pluripotent stem cells? What are the potential uses of human stem cells and the obstacles that must be overcome before these potential uses will be realized? Where can I get more information?

Stem cells have the remarkable potential to develop into many different cell types in the body during early life and growth. In addition, in many tissues they serve as a sort of internal repair system, dividing essentially without limit to replenish other cells as long as the person or animal is still alive. When a stem cell divides, each new cell has the potential either to remain a stem cell or become another type of cell with a more specialized function, such as a muscle cell, a red blood cell, or a brain cell.

Stem cells are distinguished from other cell types by two important characteristics. First, they are unspecialized cells capable of renewing themselves through cell division, sometimes after long periods of inactivity. Second, under certain physiologic or experimental conditions, they can be induced to become tissue- or organ-specific cells with special functions. In some organs, such as the gut and bone marrow, stem cells regularly divide to repair and replace worn out or damaged tissues. In other organs, however, such as the pancreas and the heart, stem cells only divide under special conditions.

Until recently, scientists primarily worked with two kinds of stem cells from animals and humans: embryonic stem cells and non-embryonic "somatic" or "adult" stem cells. The functions and characteristics of these cells will be explained in this document. Scientists discovered ways to derive embryonic stem cells from early mouse embryos more than 30 years ago, in 1981. The detailed study of the biology of mouse stem cells led to the discovery, in 1998, of a method to derive stem cells from human embryos and grow the cells in the laboratory. These cells are called human embryonic stem cells. The embryos used in these studies were created for reproductive purposes through in vitro fertilization procedures. When they were no longer needed for that purpose, they were donated for research with the informed consent of the donor. In 2006, researchers made another breakthrough by identifying conditions that would allow some specialized adult cells to be "reprogrammed" genetically to assume a stem cell-like state. This new type of stem cell, called induced pluripotent stem cells (iPSCs), will be discussed in a later section of this document.

Stem cells are important for living organisms for many reasons. In the 3- to 5-day-old embryo, called a blastocyst, the inner cells give rise to the entire body of the organism, including all of the many specialized cell types and organs such as the heart, lungs, skin, sperm, eggs and other tissues. In some adult tissues, such as bone marrow, muscle, and brain, discrete populations of adult stem cells generate replacements for cells that are lost through normal wear and tear, injury, or disease.

Given their unique regenerative abilities, stem cells offer new potentials for treating diseases such as diabetes, and heart disease. However, much work remains to be done in the laboratory and the clinic to understand how to use these cells for cell-based therapies to treat disease, which is also referred to as regenerative or reparative medicine.

Laboratory studies of stem cells enable scientists to learn about the cells essential properties and what makes them different from specialized cell types. Scientists are already using stem cells in the laboratory to screen new drugs and to develop model systems to study normal growth and identify the causes of birth defects.

Research on stem cells continues to advance knowledge about how an organism develops from a single cell and how healthy cells replace damaged cells in adult organisms. Stem cell research is one of the most fascinating areas of contemporary biology, but, as with many expanding fields of scientific inquiry, research on stem cells raises scientific questions as rapidly as it generates new discoveries.

I.Introduction|Next

See the original post:
Stem Cell Basics: Introduction [Stem Cell Information]

Bone marrow or stem cell transplants for AML | Cancer …

Having someone elses marrow or stem cells is called a donor transplant, or an allogeneic transplant. This is pronounced al-lo-jen-ay-ik.

The donors bone marrow cells must match your own as closely as possible. The most suitable donor is usually a close relative, such as a brother or sister. It is sometimes possible to find a match in an unrelated donor. Doctors call this a matched unrelated donor (MUD). To find out if there is a suitable donor for you, your doctor will contact The Anthony Nolan Bone Marrow Register and other UK based and international bone marrow registers.

To make sure that your donors cells match, you and the donor will have blood tests. These are to see how many of the proteins on the surface of their blood cells match yours. This is called tissue typing or HLA matching. HLA stands for human leucocyte antigen.

Once you have a donor and are in remission, you have high dose chemotherapy either on its own or with radiotherapy. A week later the donor goes into hospital and their stem cells or marrow are collected. You then have the stem cells or bone marrow as a drip through your central line.

If you've had a transplant from a donor, there is a risk of graft versus host disease (GVHD). This happens because the transplanted stem cells or bone marrow contain cells from your donor's immune system. These cells can sometimes recognise your own tissues as being foreign and attack them. This can be an advantage because the immune cells may also attack any leukaemia cells left after your treatment.

Acute GVHD starts within 100 days of the transplant and can cause

If you develop GVHD after your transplant, your doctor will prescribe medicines to damp down this immune reaction. These are called immunosuppressants.

Chronic GVHD starts more than 100 days after the transplant and you may have

Your doctor is likely to suggest that you stay out of the sun because GVHD skin rashes can often get worse in the sun.

There is detailed information about graft versus host disease in the section about coping physically with cancer.

See the rest here:
Bone marrow or stem cell transplants for AML | Cancer ...

5. Hematopoietic Stem Cells [Stem Cell Information]

With more than 50 years of experience studying blood-forming stem cells called hematopoietic stem cells, scientists have developed sufficient understanding to actually use them as a therapy. Currently, no other type of stem cell, adult, fetal or embryonic, has attained such status. Hematopoietic stem cell transplants are now routinely used to treat patients with cancers and other disorders of the blood and immune systems. Recently, researchers have observed in animal studies that hematopoietic stem cells appear to be able to form other kinds of cells, such as muscle, blood vessels, and bone. If this can be applied to human cells, it may eventually be possible to use hematopoietic stem cells to replace a wider array of cells and tissues than once thought.

Despite the vast experience with hematopoietic stem cells, scientists face major roadblocks in expanding their use beyond the replacement of blood and immune cells. First, hematopoietic stem cells are unable to proliferate (replicate themselves) and differentiate (become specialized to other cell types) in vitro (in the test tube or culture dish). Second, scientists do not yet have an accurate method to distinguish stem cells from other cells recovered from the blood or bone marrow. Until scientists overcome these technical barriers, they believe it is unlikely that hematopoietic stem cells will be applied as cell replacement therapy in diseases such as diabetes, Parkinson's Disease, spinal cord injury, and many others.

Blood cells are responsible for constant maintenance and immune protection of every cell type of the body. This relentless and brutal work requires that blood cells, along with skin cells, have the greatest powers of self-renewal of any adult tissue.

The stem cells that form blood and immune cells are known as hematopoietic stem cells (HSCs). They are ultimately responsible for the constant renewal of bloodthe production of billions of new blood cells each day. Physicians and basic researchers have known and capitalized on this fact for more than 50 years in treating many diseases. The first evidence and definition of blood-forming stem cells came from studies of people exposed to lethal doses of radiation in 1945.

Basic research soon followed. After duplicating radiation sickness in mice, scientists found they could rescue the mice from death with bone marrow transplants from healthy donor animals. In the early 1960s, Till and McCulloch began analyzing the bone marrow to find out which components were responsible for regenerating blood [56]. They defined what remain the two hallmarks of an HSC: it can renew itself and it can produce cells that give rise to all the different types of blood cells (see Chapter 4. The Adult Stem Cell).

A hematopoietic stem cell is a cell isolated from the blood or bone marrow that can renew itself, can differentiate to a variety of specialized cells, can mobilize out of the bone marrow into circulating blood, and can undergo programmed cell death, called apoptosisa process by which cells that are detrimental or unneeded self-destruct.

A major thrust of basic HSC research since the 1960s has been identifying and characterizing these stem cells. Because HSCs look and behave in culture like ordinary white blood cells, this has been a difficult challenge and this makes them difficult to identify by morphology (size and shape). Even today, scientists must rely on cell surface proteins, which serve, only roughly, as markers of white blood cells.

Identifying and characterizing properties of HSCs began with studies in mice, which laid the groundwork for human studies. The challenge is formidable as about 1 in every 10,000 to 15,000 bone marrow cells is thought to be a stem cell. In the blood stream the proportion falls to 1 in 100,000 blood cells. To this end, scientists began to develop tests for proving the self-renewal and the plasticity of HSCs.

The "gold standard" for proving that a cell derived from mouse bone marrow is indeed an HSC is still based on the same proof described above and used in mice many years ago. That is, the cells are injected into a mouse that has received a dose of irradiation sufficient to kill its own blood-producing cells. If the mouse recovers and all types of blood cells reappear (bearing a genetic marker from the donor animal), the transplanted cells are deemed to have included stem cells.

These studies have revealed that there appear to be two kinds of HSCs. If bone marrow cells from the transplanted mouse can, in turn, be transplanted to another lethally irradiated mouse and restore its hematopoietic system over some months, they are considered to be long-term stem cells that are capable of self-renewal. Other cells from bone marrow can immediately regenerate all the different types of blood cells, but under normal circumstances cannot renew themselves over the long term, and these are referred to as short-term progenitor or precursor cells. Progenitor or precursor cells are relatively immature cells that are precursors to a fully differentiated cell of the same tissue type. They are capable of proliferating, but they have a limited capacity to differentiate into more than one cell type as HSCs do. For example, a blood progenitor cell may only be able to make a red blood cell (see Figure 5.1. Hematopoietic and Stromal Stem Cell Differentiation).

Follow this link:
5. Hematopoietic Stem Cells [Stem Cell Information]

Archives