Page 11234..1020..»

Archive for the ‘Cardiac Stem Cells’ Category

Toward drug-induced heart regeneration – Nature.com

Toward drug-induced heart regeneration  Nature.com

See more here:
Toward drug-induced heart regeneration - Nature.com

Scientists are using AI to help detect heart disease, could help everyone live longer – WKRC TV Cincinnati

Scientists are using AI to help detect heart disease, could help everyone live longer  WKRC TV Cincinnati

Go here to read the rest:
Scientists are using AI to help detect heart disease, could help everyone live longer - WKRC TV Cincinnati

Unsupervised analysis of whole transcriptome data from human pluripotent stem cells cardiac differentiation | Scientific … – Nature.com

Unsupervised analysis of whole transcriptome data from human pluripotent stem cells cardiac differentiation | Scientific ...  Nature.com

Read more here:
Unsupervised analysis of whole transcriptome data from human pluripotent stem cells cardiac differentiation | Scientific ... - Nature.com

Treating and preventing abnormal heart beats with stem cell muscle grafts – Medical Xpress

Treating and preventing abnormal heart beats with stem cell muscle grafts  Medical Xpress

Read the rest here:
Treating and preventing abnormal heart beats with stem cell muscle grafts - Medical Xpress

‘Heart-on-a-chip’ to test chemotherapies and other cancer drugs for heart toxicity – Medical Xpress

'Heart-on-a-chip' to test chemotherapies and other cancer drugs for heart toxicity  Medical Xpress

View post:
'Heart-on-a-chip' to test chemotherapies and other cancer drugs for heart toxicity - Medical Xpress

Stem cell therapy trial shows promise for regenerative treatment of heart failure – News-Medical.Net

Stem cell therapy trial shows promise for regenerative treatment of heart failure  News-Medical.Net

Original post:
Stem cell therapy trial shows promise for regenerative treatment of heart failure - News-Medical.Net

Stem Cell Therapy Boosts Quality of Life for People With Advanced Heart Failure – MedicineNet

Stem Cell Therapy Boosts Quality of Life for People With Advanced Heart Failure  MedicineNet

Visit link:
Stem Cell Therapy Boosts Quality of Life for People With Advanced Heart Failure - MedicineNet

Revolutionary Potential of Cell Therapy in Heart Failure Treatment – Medriva

Revolutionary Potential of Cell Therapy in Heart Failure Treatment  Medriva

Read more:
Revolutionary Potential of Cell Therapy in Heart Failure Treatment - Medriva

Cell Therapy Market is estimated to be US$ 19.6 billion by 2030 with a CAGR of 13.1% during the forecast – EIN News

Cell Therapy Market is estimated to be US$ 19.6 billion by 2030 with a CAGR of 13.1% during the forecast  EIN News

Excerpt from:
Cell Therapy Market is estimated to be US$ 19.6 billion by 2030 with a CAGR of 13.1% during the forecast - EIN News

A Possible Connection between Mild Allergic Airway Responses and Cardiovascular Risk Featured in Toxicological Sciences – Newswise

A Possible Connection between Mild Allergic Airway Responses and Cardiovascular Risk Featured in Toxicological Sciences  Newswise

See the original post here:
A Possible Connection between Mild Allergic Airway Responses and Cardiovascular Risk Featured in Toxicological Sciences - Newswise

Stem Cells- Definition, Properties, Types, Uses, Challenges – Microbe Notes

Stem Cells Definition

Stem cells are unique cells present in the body that have the potential to differentiate into various cell types or divide indefinitely to produce other stem cells.

Figure: Stem Cell Renewal and Differentiation. Image Source: Maharaj Institute of Immune Regenerative Medicine.

All the stem cells found throughout all living systems have three important properties. These properties can be visualized in vitro by a process called clonogenic assays, where a single cell is assessed for its ability to differentiate.

The following are some properties of stem cells:

Figure: Techniques for generating embryonic stem cell cultures. Image Source: John Wiley & Sons, Inc. (Nico Heins et al.)

Depending on the source of the stem cells or where they are present, stem cells are divided into various types;

Figure: Human Embryonic Stem Cells Differentiation. Image created with biorender.com

Figure: Preliminary Evidence of Plasticity Among Nonhuman Adult Stem Cells. Image Source: NIH Stem Cell Information.

Figure: Progress in therapies based on iPSCs. Image Source: Nature Reviews Genetics (R. Grant Rowe & George Q. Daley).

Figure: Mesenchymal stem cells (MSCs). Image Source: PromoCell GmbH.

Some of the common and well-known examples of stem cell research are:

Stem cell research has been used in various areas because of their properties. Some of the common applications of stem cells research include;

Because of different ethical and other issues related to stem cell research, there are some limitations or challenges of stem cell research. Some of these are:

Read Also:

Read more here:
Stem Cells- Definition, Properties, Types, Uses, Challenges - Microbe Notes

Induced Pluripotent Stem Cell – an overview – ScienceDirect

13.2.1 Induced pluripotent stem cells

Induced pluripotent stem cells are differentiated cells that have been reprogrammed into an embryonic stem cell like state by the ectopic overexpression of four stem cell specific transcription factors, Oct3/4, Klf4, Sox2, and c-Myc, collectively referred to as OKSM. Induced pluripotent stem cells were first derived in a groundbreaking experiment by Yamanaka and Takashi in 2006 [77]. The team assessed the ability of 24 pluripotency associated candidate genes to covert primarily differentiated mouse tail tip fibroblasts into an embryonic stem cell state. Candidate genes were packaged into individual retroviruses and transduced into Fbx15geo/geo cells, which were grown in G418 containing media, an aminoglycoside antibiotic with conferred resistance to the neomycin gene. If the cells converted to an ESC like fate the embryonic stem cell specific locus Fbx15 containing a -galactosidase and neomycin fused reporter cassette would become activated, thereby inoculating the cells against neomycin. Transduction with all 24 factors proved to be successful in converting the fibroblasts into ESCs. Through the process of elimination, the team narrowed the list of factors down to just four factors needed to reprogram fibroblast cells to an ESC state [77]. Yamanaka and Takashi expanded their groundbreaking discovery to human cells a year later [78].

The discovery of induced pluripotent stem cells ignited the field with possibility. It was a new research tool that could be used to analyze development and cell specialization. Additionally, the possibility of deriving pluripotent stem cells was also a new therapeutic research tool that if harnessed and understood could be used for personalized cell therapy and disease modeling. Researchers quickly began differentiating iPSCs into different cell lineages.

Induced pluripotent stem cell derived-cardiomyocytes (iPSC-CMs) were generated similarly to established methods for differentiating embryonic stem cells into cardiomyocytes [7981] (Fig.13.1C). The cells were first differentiated into embryoid bodies and then exposed to serum-containing medium, which fostered a propensity to differentiate into cardiomyocytes. After 50 or more days in culture, cells derived under these conditions stained positive for sarcomeric myosin light and heavy chains, cardiac troponin T, and alpha-actinin. Additionally, the embryoid bodies demonstrated action potentials akin to atrial, ventricular, and nodal cells, and underwent rapid adaptive response to electrical stimulation and were cable of visible contractions. Despite well-established protocols the purity of cardiomyocytes derived using this technique are often times lower than 1% [8284]. However, the efficiency and purity of cardiomyocytes generated from embryoid body differentiation could be enhanced by following a step wise induction process similar to the naturally occurring cardiac differentiation process in the developing embryo [85].

To increase purity, and the usability for downstream applications monolayer culture methods were developed to facilitate a more controllable and reproducible environment to generate iPSC-CMs [86]. Monoculture conditions consist of growth on Matrigel-coated plates with mouse embryonic fibroblast conditioned media and gradual supplementation with activin A and BMP-4 growth factors. The combination of these conditions have been shown to yield greater than 50% beating iPSC-CMs [87,88]. A variation of this method, called the matrix sandwich method exists and boasts yields of up to 98% beating iPSC-CMs [89]. However, it should be noted that this method only works for some cell lines and requires growth factor batch optimizations to maintain high yields [90]. Alternatively, modifying Wnt/-catenin signaling using shRNA and small molecules has also been shown to increase iPSC-CM yield to approximately 85% [91,92].

The need for complex culture conditions to yield high iPSC-CM outputs makes identifying the biological underpinnings of iPSC-CM differentiation difficult to elucidate. One study claims to have reduced the complexity of iPSC-CM derivation to just three components, referred to as CDM3 [93]. When used in combination with lactate selection the study authors claim to achieve a yield of 80%90% troponin T positive iPSC-CMs [94]. The simplicity of the culture conditions used in this study allowed for the first time the identification of key signaling pathways implicated in iPSC-CM carcinogenesis.

The first and only case thus far of an autologous iPSC derived cell treatment making it to the clinic was reported in 2014. In a trial lead by Takahashi and colleagues, human iPSC derived retinal pigment epithelium cell sheets were transplanted into a human patient to resolve age related macular regeneration [95]. There have been no clinical trials testing iPSC-CM safety or efficacy in repairing the injured heart. However, iPSC-CMs derived using the previously mentioned matrix sandwich technique were transplanted in a non-human primate model, where they were shown to improve cardiac function after induced myocardial infraction. However, the transplanted iPSC-CMs also induced high rates of ventricular arrhythmia [96].

Despite the great hope for patient specific treatments, it is uncertain if autologous iPSC-CM treatments for myocardial infractions will make it to the clinic within the next few years. The production of patient-specific stem cells is expensive and variable. Specifically, iPSC-CM derivation efficiency still remains low and variable without the use of complex culture systems. Streamlining human iPSC cardiomyocyte differentiation to an effective simple differentiation process is key. Large numbers of iPSC-CM cells would be needed for human clinical trials, which would be impractical to accomplish using current culture systems and methods. Currently macaque trials require about 108109 reprogrammed iPSC-CM cells. The number of cells required for a human trial is projected to be a least a magnitude higher [5,97]. Additionally, like all iPSC derived cell therapies, and even embryonic stem cell therapies there is the concern that the transplanted stem cells could develop into tumor and/or cancer cells because of the possible carryover of few highly multi- or pluripotent cells in the transplanted pool [98]. Safety assessment is key before any iPSC-CM trial can make it to the clinical setting.

However, iPSC-CMs do have the potential to be somewhat useful for in vitro screening assays and drug development. iPSC-CMs have been used to improve the identification of false positive and negative data in electrophysiological assays [99]. They have also been shown to be responsive for research purposes to several cardiac and non-cardiac drugs, a prospect that might be of interest for drug screening purposes [100103]. Furthermore, disease-specific iPSC-CMs derived from people with pre-existing heart conditions have been shown to be more responsive to cardiotoxic drugs as measured by action potential duration and drug-induced arrhythmia, consistent with what would be expected naturally in the patient [104].

While iPSC-CMs might have some usefulness for drug screening, the results should be considered in light of the fact that iPSC-CMs are not equivalent to true CMs found in the adult heart. iPSC-CMs have lower conduction velocities and shorter action potential duration. They are altogether functionally immature, disorganized, fetal-like, and are not molecularly equivalent to true cardiomyocytes found in the adult heart [90,105107]. There is a need to understand cardiomyocyte maturation to facilitate regeneration and differentiation into cardiomyocytes capable of maintaining the functions of an adult heart.

View original post here:
Induced Pluripotent Stem Cell - an overview - ScienceDirect

‘Ghost heart’: Built from the scaffolding of a pig and the … – CNN

CNN

The first time molecular biologist Doris Taylor saw heart stem cells beat in unison in a petri dish, she was spellbound.

It actually changed my life, said Taylor, who directed regenerative medicine research at Texas Heart Institute in Houston until 2020. I said to myself, Oh my gosh, thats life. I wanted to figure out the how and why, and re-create that to save lives.

That goal has become reality. On Wednesday at the Life Itself conference, a health and wellness event presented in partnership with CNN, Taylor showed the audience the scaffolding of a pigs heart infused with human stem cells creating a viable, beating human heart the body will not reject. Why? Because its made from that persons own tissues.

Now we can truly imagine building a personalized human heart, taking heart transplants from an emergency procedure where youre so sick, to a planned procedure, Taylor told the audience.

That reduces your risk by eliminating the need for (antirejection) drugs, by using your own cells to build that heart it reduces the cost and you arent in the hospital as often so it improves your quality of life, she said.

Debuting on stage with her was BAB, a robot Taylor painstakingly taught to inject stem cells into the chambers of ghost hearts inside a sterile environment. As the audience at Life Itself watched BAB functioning in a sterile environment, Taylor showed videos of the pearly white mass called a ghost heart begin to pinken.

Can we grow a personalized human heart?

Its the first shot at truly curing the number one killer of men, women and children worldwide heart disease. And then I want to make it available to everyone, said Taylor to audience applause.

She never gave up, said Michael Golway, lead inventor of BAB and president and CEO of Advanced Solutions, which designs and creates platforms for building human tissues.

At any point, Dr. Taylor could have easily said Im done, this just isnt going to work. But she persisted for years, fighting setbacks to find the right type of cells in the right quantities and right conditions to enable those cells to be happy and grow.

Taylors fascination with growing hearts began in 1998, when she was part of a team at Duke University that injected cells into a rabbits failed heart, creating new heart muscle. As trials began in humans, however, the process was hit or miss.

We were putting cells into damaged or scarred regions of the heart and hoping that would overcome the existing damage, she told CNN. I started thinking: What if we could get rid of that bad environment and rebuild the house?

Taylors first success came in 2008 when she and a team at the University of Minnesota washed the cells out of a rats heart and began to work with the translucent skeleton left behind.

Soon, she graduated to using pigs hearts, due to their anatomical similarity to human hearts.

We took a pigs heart, and we washed out all the cells with a gentle baby shampoo, she said. What was left was an extracellular matrix, a transparent framework we called the ghost heart.

Then we infused blood vessel cells and let them grow on the matrix for a couple of weeks, Taylor said. That built a way to feed the cells we were going to add because wed reestablished the blood vessels to the heart.

The next step was to begin injecting the immature stem cells into the different regions of the scaffold, and then we had to teach the cells how to grow up.

We must electrically stimulate them, like a pacemaker, but very gently at first, until they get stronger and stronger. First, cells in one spot will twitch, then cells in another spot twitch, but they arent together, Taylor said. Over time they start connecting to each other in the matrix and by about a month, they start beating together as a heart. And let me tell you, its a wow moment!

But thats not the end of the mothering Taylor and her team had to do. Now she must nurture the emerging heart by giving it a blood pressure and teaching it to pump.

We fill the heart chambers with artificial blood and let the heart cells squeeze against it. But we must help them with electrical pumps, or they will die, she explained.

The cells are also fed oxygen from artificial lungs. In the early days all of these steps had to be monitored and coordinated by hand 24 hours a day, 7 days a week, Taylor said.

The heart has to eat every day, and until we built the pieces that made it possible to electronically monitor the hearts someone had to do it person and it didnt matter if it was Christmas or New Years Day or your birthday, she said. Its taken extraordinary groups of people who have worked with me over the years to make this happen.

But once Taylor and her team saw the results of their parenting, any sacrifices they made became insignificant, because then the beauty happens, the magic, she said.

Weve injected the same type of cells everywhere in the heart, so they all started off alike, Taylor said. But now when we look in the left ventricle, we find left ventricle heart cells. If we look in the atrium, they look like atrial heart cells, and if we look in the right ventricle, they are right ventricle heart cells, she said.

So over time theyve developed based on where they find themselves and grown up to work together and become a heart. Nature is amazing, isnt she?

As her creation came to life, Taylor began to dream about a day when her prototypical hearts could be mass produced for the thousands of people on transplant lists, many of whom die while waiting. But how do you scale a heart?

I realized that for every gram of heart tissue we built, we needed a billion heart cells, Taylor said. That meant for an adult-sized human heart we would need up to 400 billion individual cells. Now, most labs work with a million or so cells, and heart cells dont divide, which left us with the dilemma: Where will these cells come from?

The answer arrived when Japanese biomedical researcher Dr. Shinya Yamanaka discovered human adult skin cells could be reprogrammed to behave like embryonic or pluripotent stem cells, capable of developing into any cell in the body. The 2007 discovery won the scientist a Nobel Prize, and his induced pluripotent stem cells (iPS), soon became known as Yamanaka factors.

Now for the first time we could take blood, bone marrow or skin from a person and grow cells from that individual that could turn into heart cells, Taylor said. But the scale was still huge: We needed tens of billions of cells. It took us another 10 years to develop the techniques to do that.

The solution? A bee-like honeycomb of fiber, with thousands of microscopic holes where the cells could attach and be nourished.

The fiber soaks up the nutrients just like a coffee filter, the cells have access to food all around them and that lets them grow in much larger numbers. We can go from about 50 million cells to a billion cells in a week, Taylor said. But we need 40 billion or 50 billion or 100 billion, so part of our science over the last few years has been scaling up the number of cells we can grow.

Another issue: Each heart needed a pristine environment free of contaminants for each step of the process. Every time an intervention had to be done, she and her team ran the risk of opening the heart up to infection and death.

Do you know how long it takes to inject 350 billion cells by hand? Taylor asked the Life Itself audience. What if you touch something? You just contaminated the whole heart.

Once her lab suffered an electrical malfunction and all of the hearts died. Taylor and her team were nearly inconsolable.

When something happens to one of these hearts, its devastating to all of us, Taylor said. And this is going to sound weird coming from a scientist, but I had to learn to bolster my own heart emotionally, mentally, spiritually and physically to get through this process.

Enter BAB, short for BioAssemblyBot, and an uber-sterile cradle created by Advance Solutions that could hold the heart and transport it between each step of the process while preserving a germ-free environment. Taylor has now taught BAB the specific process of injecting the cells she has painstakingly developed over the last decade.

When Dr. Taylor is injecting cells, it has taken her years to figure out where to inject, how much pressure to put on the syringe, and the best speed and pace to add the cells, said BABs creator Golway.

A robot can do that quickly and precisely. And as we know, no two hearts are the same, so BAB can use ultrasound to see inside the vascular pathway of that specific heart, where Dr. Taylor is working blind, so to speak, Golway added. Its exhilarating to watch there are times where the hair on the back of my neck literally stands up.

Taylor left academia in 2020 and is currently working with private investors to bring her creation to the masses. If transplants into humans in upcoming clinical trials are successful, Taylors personalized hybrid hearts could be used to save thousands of lives around the world.

In the US alone, some 3,500 people were on the heart transplant waiting list in 2021.

Thats not counting the people who never make it on the list, due to their age or heath, Taylor said. If youre a small woman, if youre an underrepresented minority, if youre a child, the chances of getting an organ that matches your body are low.

If you do get a heart, many people get sick or otherwise lose their new heart within a decade. We can reduce cost, we can increase access, and we can decrease side effects. Its a win-win-win.

Taylor can even envision a day when people bank their own stem cells at a young age, taking them out of storage when needed to grow a heart and one day even a lung, liver or kidney.

Say they have heart disease in their family, she said. We can plan ahead: Grow their cells to the numbers we need and freeze them, then when they are diagnosed with heart failure pull a scaffold off the shelf and build the heart within two months.

Im just humbled and privileged to do this work, and proud of where we are, she added. The technology is ready. I hope everyone is going to be along with us for the ride because this is game-changing.

See more here:
'Ghost heart': Built from the scaffolding of a pig and the ... - CNN

Stem Cells International | Hindawi

Research Article

09 Nov 2022

Aucubin Impeded Preosteoclast Fusion and Enhanced CD31hi EMCNhi Vessel Angiogenesis in Ovariectomized Mice

Ziyi Li|Chang Liu|...|Peng Xue

Osteogenesis is tightly correlated with angiogenesis during the process of bone development, regeneration, and remodeling. In addition to providing nutrients and oxygen for bone tissue, blood vessels around bone tissue also secrete some factors to regulate bone formation. Type H vessels which were regulated by platelet-derived growth factor-BB (PDGF-BB) were confirmed to couple angiogenesis and osteogenesis. Recently, preosteoclasts have been identified as the most important source of PDGF-BB. Therefore, inhibiting osteoclast maturation, improving PDGF-BB secretion, stimulating type H angiogenesis, and subsequently accelerating bone regeneration may be potent treatments for bone loss disease. In the present study, aucubin, an iridoid glycoside extracted from Aucuba japonica and Eucommia ulmoides, was found to inhibit bone loss in ovariectomized mice. We further confirmed that aucubin could inhibit the fusion of tartrate-resistant acid phosphatase (TRAP)+ preosteoclasts into mature osteoclasts and indirectly increasing angiogenesis of type H vessel. The underlying mechanism is the aucubin-induced inhibition of MAPK/NF-B signaling, which increases the preosteoclast number and subsequently promotes angiogenesis via PDGF-BB. These results prompted that aucubin could be an antiosteoporosis drug candidate, which needs further research.

Review Article

07 Nov 2022

The Influence of Intervertebral Disc Microenvironment on the Biological Behavior of Engrafted Mesenchymal Stem Cells

Jing Zhang|Wentao Zhang|...|Zhonghai Li

Intervertebral disc degeneration is the main cause of low back pain. Traditional treatment methods cannot repair degenerated intervertebral disc tissue. The emergence of stem cell therapy makes it possible to regenerate and repair degenerated intervertebral disc tissue. At present, mesenchymal stem cells are the most studied, and different types of mesenchymal stem cells have their own characteristics. However, due to the harsh and complex internal microenvironment of the intervertebral disc, it will affect the biological behaviors of the implanted mesenchymal stem cells, such as viability, proliferation, migration, and chondrogenic differentiation, thereby affecting the therapeutic effect. This review is aimed at summarizing the influence of each intervertebral disc microenvironmental factor on the biological behavior of mesenchymal stem cells, so as to provide new ideas for using tissue engineering technology to assist stem cells to overcome the influence of the microenvironment in the future.

Research Article

07 Nov 2022

CD44v6+ Hepatocellular Carcinoma Cells Maintain Stemness Properties through Met/cJun/Nanog Signaling

Wei Chen|Ronghua Wang|...|Bin Cheng

Cancer stem cells (CSCs) are characterized by their self-renewal and differentiation abilities. CD44v6 is a novel CSC marker that can activate various signaling pathways. Here, we hypothesized that the HGF/Met signaling pathway promotes stemness properties in CD44v6+ hepatocellular carcinoma (HCC) cells via overexpression of the transcription factor, cJun, thus representing a valuable target for HCC therapy. Magnetic activated cell sorting was used to separate the CD44v6+ from CD44v6- cells, and Met levels were regulated using lentiviral particles and the selective Met inhibitor, PHA665752. An orthotopic liver xenograft tumor model was used to assess the self-renewal ability of CD44v6+ cells in immunodeficient NOD/SCID mice. Luciferase reporter and chromatin immunoprecipitation assays were also conducted using cJun-overexpressing 293 T cells to identify the exact binding site of cJun in the Nanog promoter. Our data demonstrate that CD44v6 is an ideal surface marker of liver CSCs. CD44v6+ HCC cells express higher levels of Met and possess self-renewal and tumor growth abilities. Xenograft liver tumors were smaller in nude mice injected with shMet HCC cells. Immunohistochemical analysis of liver tissue specimens revealed that high Met levels in HCC cells were associated with poor patient prognosis. Further, a cJun binding site was identified 1700 bp upstream of the Nanog transcription start site and mutation of the cJun binding site reduced Nanog expression. In conclusion, the HGF/Met signaling pathway is important for maintenance of stemness in CD44v6+ HCC cells by enhancing expression of cJun, which binds 1700 bp upstream of the Nanog transcription start site.

Research Article

26 Oct 2022

Stage-Dependent Regulation of Dental Pulp Stem Cell Odontogenic Differentiation by Transforming Growth Factor-1

Yu Bai|Xin Liu|...|Wenxi He

Transforming growth factor-1 (TGF-1) is an important multifunctional cytokine with dual effects on stem cell differentiation. However, the role of TGF-1 on odontogenic differentiation of dental pulp stem cells (DPSCs) remains to be entirely elucidated. In the present study, we initially investigated the effect of TGF-1 at a range of concentrations (0.1-5ng/mL) on the proliferation, cell cycle, and apoptosis of DPSCs. Subsequently, to determine the effect of TGF-1 on odontogenic differentiation, alkaline phosphatase (ALP) activity and Alizarin Red S (ARS) staining assays at different concentrations and time points were performed. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis were used to determine the levels of odonto-/osteo-genic differentiation-related gene and protein expression, respectively. For in vivo studies, newly formed tissue was assessed by Massons trichrome and von Kossa staining. Data indicated that TGF-1 inhibited DPSCs proliferation in a concentration-and time-dependent manner () and induced cell cycle arrest but did not affect apoptosis. ALP activity was enhanced, while ARS reduced gradually with increasing TGF-1 concentrations, accompanied by increased expression of early marker genes of odonto-/osteo-genic differentiation and decreased expression of late-stage mineralization marker genes (). ALP expression was elevated in the TGF-1-treatment group until 14 days, and the intensity of ARS staining was attenuated at days 14 and 21 (). Compared with the control group, abundant collagen but no mineralized tissues were observed in the TGF-1-treatment group in vivo. Overall, these findings indicate that TGF-1 promotes odontogenic differentiation of DPSCs at early-stage while inhibiting later-stage mineralization processes.

Research Article

20 Oct 2022

miR-31 from Mesenchymal Stem Cell-Derived Extracellular Vesicles Alleviates Intervertebral Disc Degeneration by Inhibiting NFAT5 and Upregulating the Wnt/-Catenin Pathway

Baodong Wang|Na Xu|...|Yang Cao

In this study, we explored the regulatory mechanism of intervertebral disc degeneration (IDD) that involves miR-31 shuttled by bone marrow mesenchymal stem cell-derived extracellular vesicles (BMSC-EVs) and its downstream signaling molecules. Nucleus pulposus cells (NPCs) were isolated and treated with TNF- to simulate IDD in vitro. The TNF--exposed NPCs were then cocultured with hBMSCs or hBMSC-EVs in vitro to detect the effects of hBMSC-EVs on NPC viability, apoptosis, and ECM degradation. Binding between miR-31 and NFAT5 was determined. A mouse model of IDD was prepared by vertebral disc puncture and injected with EVs from hBMSCs with miR-31 knockdown to discern the function of miR-31 in vivo. The results demonstrated that hBMSC-EVs delivered miR-31 into NPCs. hBMSC-EVs enhanced NPC proliferation and suppressed cell apoptosis and ECM degradation, which was associated with the transfer of miR-31 into NPCs. In NPCs, miR-31 bound to the 3UTR of NFAT5 and inhibited NFAT5 expression, leading to activation of the Wnt/-catenin pathway and thus promoting NPC proliferation and reducing cell apoptosis and ECM degradation. In addition, miR-31 in hBMSC-EVs alleviated the IDD in mouse models. Taken together, miR-31 in hBMSC-EVs can alleviate IDD by targeting NFAT5 and activating the Wnt/-catenin pathway.

Review Article

20 Oct 2022

Variability in Platelet-Rich Plasma Preparations Used in Regenerative Medicine: A Comparative Analysis

Raghvendra Vikram Tey|Pallavi Haldankar|...|Ravindra Maradi

Background. Platelet-rich plasma (PRP) and its derivatives are used in several aesthetic, dental, and musculoskeletal procedures. Their efficacy is primarily due to the release of various growth factors (GF), interleukins, cytokines, and white blood cells. However, the PRP preparation methods are highly variable, and studies lack consistency in reporting complete procedures to prepare PRP and characterize PRP and its derivatives. Also, all the tissue-specific (in vivo and in vitro) interactions and functional properties of the various derivatives/factors of the PRP have not been taken into consideration by any study so far. This creates a potential space for further standardization of the PRP preparation methods and customization of PRP/PRP derivatives targeted at tissue-specific/pathology specific requirements that would enable efficacious and widely acceptable usage of PRP as main therapy, rather than being used as adjuvant therapy. The main objective of our study was to investigate the variability in PRP preparation methods and to analyze their efficacy and reliability. Method. This study considered articles published in the last 5 years, highlighting the variability in their PRP preparation methods and characterization of PRP. Following the PRISMA protocol, we selected 13 articles for the study. The selected articles were assessed using NHLBI quality assessment tool. Results. We noted differences in (1) approaches to producing PRP, (2) extent of characterization of PRP, (3) small scale and large-scale preparation methods, (4) in vitro and in vivo studies. Conclusion. We identified two studies describing the procedures which are simple, reproducible, economical, provide a good yield of platelets, and therefore can be considered methods for further tissue-specific and pathology-specific standardizations of PRP and its derivatives. We recommend further randomized studies to understand the full therapeutic potential of the constituents of PRP and its derivatives.

See the rest here:
Stem Cells International | Hindawi

Stem Cell Differentiation | Stem Cells | Tocris Bioscience

Stem Cell Differentiation Target Files

Stem cell differentiation involves the changing of a cell to a more specialized cell type, involving a switch from proliferation to specialization. This involves a succession of alterations in cell morphology, membrane potential, metabolic activity and responsiveness to certain signals. Differentiation leads to the commitment of a cell to developmental lineages and the acquisition of specific functions of committed cells depending upon the tissue in which they will finally reside. Stem cell differentiation is tightly regulated by signaling pathways and modifications in gene expression.

Stem cells can be categorized into groups depending on their ability to differentiate.

Embryonic stem cells (ESCs) are pluripotent cells that differentiate as a result of signaling mechanisms. These are tightly controlled by most growth factors, cytokines and epigenetic processes such as DNA methylation and chromatin remodeling. ESCs divide into two cells: one is a duplicate stem cell (the process of self-renewal) and the other daughter cell is one which will differentiate. The daughter cells divides and after each division it becomes more specialized. When it reaches a mature cell type downstream (for example, becomes a red blood cell) it will no longer divide. The ability of ESCs to differentiate is currently being researched for the treatment of many diseases including Parkinson's disease and cancer.

Adult or 'somatic' stem cells are thought to be undifferentiated. Their primary role is to self-renew and maintain or repair the tissue in which they reside.

View all pluripotent stem cell resources available from Bio-Techne.

Regenerative medicine is the repair or replacement of damaged or diseased tissue to restore normal tissue function. This blog post discusses the development of a new cell therapy product derived from PSCs for regenerative medicine use in Parkinson's disease.

Neurons derived from pluripotent stem cells (PSCs) are a source of considerable therapeutic potential for neurodegenerative diseases. This blog post outlines the development of a small molecule-based protocol for the differentiation of human induced PSCs into functional cortical neurons.

Tocris offers the following scientific literature for Stem Cell Differentiation to showcase our products. We invite you to request* your copy today!

*Please note that Tocris will only send literature to established scientific business / institute addresses.

This product guide provides a background to the use of small molecules in stem cell research and lists over 200 products for use in:

Written by Kirsty E. Clarke, Victoria B. Christie, Andy Whiting and Stefan A. Przyborski, this review provides an overview of the use of small molecules in the control of stem cell growth and differentiation. Key signaling pathways are highlighted, and the regulation of ES cell self-renewal and somatic cell reprogramming is discussed. Compounds available from Tocris are listed.

Stem cells have potential as a source of cells and tissues for research and treatment of disease. This poster summarizes some key protocols demonstrating the use of small molecules across the stem cell workflow, from reprogramming, through self-renewal, storage and differentiation to verification. Advantages of using small molecules are also highlighted.

Written by Rebecca Quelch and Stefan Przyborski from Durham University (UK), this poster describes the isolation of pluripotent stem cells, their maintenance in culture, differentiation, and the generation and potential uses of organoids.

Link:
Stem Cell Differentiation | Stem Cells | Tocris Bioscience

Adult Stem Cells // Center for Stem Cells and Regenerative Medicine …

Adult stem cells, also called somatic stem cells, are undifferentiated cells that are found in many different tissues throughout the body of nearly all organisms, including humans. Unlike embryonic stem cells, which can become any cell in the body (called pluripotent), adult stem cells, which have been found in a wide range of tissues including skin, heart, brain, liver, and bone marrow are usually restricted to become any type of cell in the tissue or organ that they reside (called multipotent). These adult stem cells, which exist in the tissue for decades, serve to replace cells that are lost in the tissue as needed, such as the growth of new skin every day in humans.

Scientists discovered adult stem cells in bone marrow more than 50 years ago. These blood-forming stem cells have been used in transplants for patients with leukemia and several other diseases for decades. By the 1990s, researchers confirmed that nerve cells in the brain can also be regenerated from endogenous stem cells. It is thought that adult stem cells in a variety of different tissues could lead to treatments for numerous conditions that range from type 1 diabetes (providing insulin-producing cells) to heart attack (repairing cardiac muscle) to neurological disease (regenerating lost neurons in the brain or spinal cord).

Efforts are underway to stimulate these adult stem cells to regenerate missing cells within damaged tissues. This approach will utilize the existing tissue organization and molecules to stimulate and guide the adult stem cells to correctly regenerate only the necessary cell types. Alternatively, the adult stem cells could be isolated from the tissue and grown outside of the body, in cultures. This would allow the cells to be easily manipulated, although they are often relatively rare and difficult to grow in culture.

Because the isolation of adult stem cells does not result in the destruction of human life, research involving adult stem cells does not raise any of the ethical issues associated with research utilizing human embryonic stem cells. Thus, research involving adult stem cells has the potential for therapies that will heal disease and ease suffering, a major focus of Notre Dames stem cell research. Combined with our efforts with induced pluripotent stem (iPS) cells, the Center for Stem Cells and Regenerative Medicine will advance the Universitys mission to ease suffering and heal disease.

Original post:
Adult Stem Cells // Center for Stem Cells and Regenerative Medicine ...

What are Stem Cells? – Types, Applications and Sources – BYJUS

Stem cells are special human cells that can develop into many different types of cells, from muscle cells to brain cells.

Stem cells also have the ability to repair damaged cells. These cells have strong healing power. They can evolve into any type of cell.

Research on stem cells is going on, and it is believed that stem cell therapies can cure ailments like paralysis and Alzheimers as well. Let us have a detailed look at stem cells, their types and their functions.

Also Read: Gene Therapy

Stem cells are of the following different types:

The fertilized egg begins to divide immediately. All the cells in the young embryo are totipotent cells. These cells form a hollow structure within a few days. Cells in one region group together to form the inner cell mass. This contains pluripotent cells that make up the developing foetus.

The embryonic stem cells can be further classified as:

These stem cells are obtained from developed organs and tissues. They can repair and replace the damaged tissues in the region where they are located. For eg., hematopoietic stem cells are found in the bone marrow. These stem cells are used in bone marrow transplants to treat specific types of cancers.

These cells have been tested and arranged by converting tissue-specific cells into embryonic cells in the lab. These cells are accepted as an important tool to learn about the normal development, onset and progression of the disease and are also helpful in testing various drugs. These stem cells share the same characteristics as embryonic cells do. They also have the potential to give rise to all the different types of cells in the human body.

These cells are mainly formed from the connective tissues surrounding other tissues and organs, known as the stroma. These mesenchymal stem cells are accurately called stromal cells. The first mesenchymal stem cells were found in the bone marrow that is capable of developing bones, fat cells, and cartilage.

There are different mesenchymal stem cells that are used to treat various diseases as they have been developed from different tissues of the human body. The characteristics of mesenchymal stem cells depend on the organ from where they originate.

Following are the important applications of stem cells:

This is the most important application of stem cells. The stem cells can be used to grow a specific type of tissue or organ. This can be helpful in kidney and liver transplants. The doctors have already used the stem cells from beneath the epidermis to develop skin tissue that can repair severe burns or other injuries by tissue grafting.

A team of researchers have developed blood vessels in mice using human stem cells. Within two weeks of implantation, the blood vessels formed their network and were as efficient as the natural vessels.

Stem cells can also treat diseases such as Parkinsons disease and Alzheimers. These can help to replenish the damaged brain cells. Researchers have tried to differentiate embryonic stem cells into these types of cells and make it possible to treat diseases.

The adult hematopoietic stem cells are used to treat cancers, sickle cell anaemia, and other immunodeficiency diseases. These stem cells can be used to produce red blood cells and white blood cells in the body.

Stem Cells originate from different parts of the body. Adult stem cells can be found in specific tissues in the human body. Matured cells are specialized to conduct various functions. Generally, these cells can develop the kind of cells found in tissues where they reside.

Embryonic Stem Cells are derived from 5-day-old blastocysts that develop into embryos and are pluripotent in nature. These cells can develop any type of cell and tissue in the body. These cells have the potential to regenerate all the cells and tissues that have been lost because of any kind of injury or disease.

To know more about stem cells, their types, applications and sources, keep visiting BYJUS website.

Stem-cell therapy is the use of stem cells to cure or prevent a disease or condition. The damaged cells are repaired by the generated stem cells, which can also hasten the healing process in the injured tissue. These cells are essential for the regeneration and transplanting of tissue.

Stem cells have the capacity to self-renew and differentiate into specialized cell types. Totipotent stem cells come from an early embryo and can differentiate into all possible types of stem cells.

The four types of stem cells are the embryonic stem cells, adult stem cells, induced pluripotent stem cells and mesenchymal stem cells

Adult stem cells are undifferentiated cells taken from tissues and developing organs. They can replace and restore damaged tissues. Example hematopoietic stem cells in the bone marrow.

See original here:
What are Stem Cells? - Types, Applications and Sources - BYJUS

Global Induced Pluripotent Stem Cells Market (2022 to 2027) – Growth, Trends, Covid-19 Impact and Forecasts – ResearchAndMarkets.com – Business Wire

DUBLIN--(BUSINESS WIRE)--The "Induced Pluripotent Stem Cells Market - Growth, Trends, Covid-19 Impact, and Forecasts (2022 - 2027)" report has been added to ResearchAndMarkets.com's offering.

The Induced Pluripotent Stem Cells Market is projected to register a CAGR of 8.4% during the forecast period (2022 to 2027).

Companies Mentioned

Key Market Trends

The Drug Development Segment is Expected to Hold a Major Market Share in the Induced Pluripotent Stem Cells Market.

By application, the drug development segment holds the major segment in the induced pluripotent stem cell market. Various research studies focusing on drug development studies with induced pluripotent stem cells have been on the rise in recent years.

For instance, an article titled "Drug Development and the Use of Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Disease Modeling and Drug Toxicity Screening" published in the International Journal of Molecular Science in October 2020 discussed the broad use of iPSC derived cardiomyocytes for drug development in terms of adverse drug reactions, mechanisms of cardiotoxicity, and the need for efficient drug screening protocols.

Another article published in the Journal of Cells in December 2021 titled "Human Induced Pluripotent Stem Cell as a Disease Modeling and Drug Development Platform-A Cardiac Perspective" focused on methods to reprogram somatic cells into human induced pluripotent stem cells and the solutions to overcome the immaturity of the human induced pluripotent stem cells derived cardiomyocytes to mimic the structure and physiological properties of adult human cardiomyocytes to accurately model disease and test drug safety. Thus, this increase in the research of induced pluripotent stem cells for drug development and drug modeling is likely to propel the segment's growth over the study period.

Furthermore, as per an article titled "Advancements in Disease Modeling and Drug Discovery Using iPSC-Derived Hepatocyte-like Cells" published in the Multi-Disciplinary Publishing Institute journal of Cells in March 2022, preserved differentiation and physiological function, amenability to genetic manipulation via tools such as CRISPR/Cas9, and availability for high-throughput screening, make induced pluripotent stem cell systems increasingly attractive for both mechanistic studies of disease and the identification of novel therapeutics.

North America is Expected to Hold a Significant Share in the Market and Expected to do Same in the Forecast Period

The rise in the adoption of highly advanced technologies and systems in drug development, toxicity testing, and disease modeling coupled with the growing acceptance of stem cell therapies in the region are some of the major factors driving the market growth in North America.

The United States Food and Drug Administration in March 2022 discussed the development of strategies to improve cell therapy product characterization. The agency focused on the development of improved methods for testing stem cell products to ensure the safety and efficacy of such treatments when used as therapies.

Likewise, in March 2020, the Food and Drug Administration announced that ImStem drug IMS001, which uses AgeX's pluripotent stem cell technology, would be available for the treatment of multiple sclerosis. Similarly, REPROCELL introduced a customized iPSC generation service in December 2020, as well as a new B2C website to promote the "Personal iPS" service. This service prepares and stores an individual's iPSCs for future injury or disease regeneration treatment.

Thus, the increasing necessity for induced pluripotent stem cells coupled with increasing investment in the health care department is known to propel the growth of the market in this region.

Key Topics Covered:

1 INTRODUCTION

2 RESEARCH METHODOLOGY

3 EXECUTIVE SUMMARY

4 MARKET DYNAMICS

4.1 Market Overview

4.2 Market Drivers

4.2.1 Increase in Research and Development Activities in Stem Cells Therapies

4.2.2 Surge in Adoption of Personalized Medicine

4.3 Market Restraints

4.3.1 Lack of Awareness Regarding Stem Cell Therapies

4.3.2 High Cost of Treatment

4.4 Porter's Five Force Analysis

5 MARKET SEGMENTATION

5.1 By Derived Cell Type

5.2 Application

5.3 End User

5.4 Geography

6 COMPETITIVE LANDSCAPE

6.1 Company Profiles

7 MARKET OPPORTUNITIES AND FUTURE TRENDS

For more information about this report visit https://www.researchandmarkets.com/r/ylzwhr

See the original post here:
Global Induced Pluripotent Stem Cells Market (2022 to 2027) - Growth, Trends, Covid-19 Impact and Forecasts - ResearchAndMarkets.com - Business Wire

Bone Marrow market estimated to reach US$13899.60 Million during the forecast period – Digital Journal

ThisBone Marrow MarketReport provides details on Recent New Developments, Trade Regulations, Import-Export Analysis, Production Analysis, Value Chain Optimization, Market Share, Impact of Domestic and Localized Market Players, Analyzes opportunities in terms of emerging revenue pockets, changing market regulations, strategic market growth analysis, market size, market category growth, niche and application dominance, product endorsements, product launches, geographic expansions , technological innovations in the market.For more information on the bone marrow market, please contact Data Bridge Market Research for a summary of theanalyst, our team will help you make an informed market decision to achieve market growth.

Bone Marrow Market is expected to experience market growth during the forecast period of 2021 to 2028. Data Bridge Market Research analyzes that the market is growing with a CAGR of 5.22% during the forecast period of 2021 to 2028 and it is projected to reach USD 13,899.60 Million by 2028. The increasing number of bone marrow diseases will help accelerate the growth of the bone marrow market.Bone marrow transplant also called hematopoietic stem cell.It is a soft vascular tissue present inside the long bones.It includes two types of stem cells, namely hematopoietic and mesenchymal stem cells.The bone marrow is primarily responsible for hematopoiesis (blood cell formation), lymphocyte production, and fat storage.

Get Report Sample PDF: https://www.databridgemarketresearch.com/request-a-sample/?dbmr=global-bone-marrow-market

The main factors driving the growth of the bone marrow market during the forecast period are the growth in the incidence of non-Hodgkins and Hodgkins lymphoma, thalassemia, and leukemia, as well as common bone marrow diseases worldwide, developments in technology and improvements.in health infrastructure.In addition, advanced signs of bone marrow transplantation for cardiac and neural disorders, increased funding for logistics services, and rising health care spending per capita are some of the other factors expected to further drive growth. growth of the bone marrow market in the coming years.years.However, the high costs of treatment,

Key Players Covered in the Bone Marrow Market Report are AGendia, Agilent Technologies, Inc., Ambrilia Biopharma Inc., Astellas Pharma Inc., diaDexus, Illumina, Inc., QIAGEN, F Hoffmann-La Roche Ltd, Sanofi, Stryker Corporation, PromoCell GmbH, STEMCELL Technologies Inc., Lonza, ReachBio LLC, AllCells, ATCC, Lifeline Cell Technology, Conversant bio, HemaCare, Mesoblast Ltd., Merck KGaA, Discovery Life Sciences, ReeLabs Pvt. Ltd., Gamida Cell, among others national and global players.Market share data is available separately for Global, North America, Europe, Asia-Pacific (APAC), Middle East and Africa (MEA), and South America.DBMR analysts understand competitive strengths and provide competitive analysis for each competitor separately.

For More Information On Market Analysis, View Research Report Summary At :-https://www.databridgemarketresearch.com/reports/global-bone-marrow-market

Bone MarrowMarket Scope and Market Size

The bone marrow market is segmented based on transplant type, disease indication, and end user.Growth between these segments will help you analyze weak growth segments in industries and provide users with valuable market overview and market insights to help them make strategic decisions to identify leading market applications.

Country-level analysis of thebone marrow market

The bone marrow market is analyzed and information is provided on market size and trends by country, transplant type, disease indication, and end user, as mentioned above.Countries Covered in Bone Marrow Market Report are USA, Canada, and Mexico, North America, Germany, France, UK, Netherlands, Switzerland, Belgium, Russia, Italy, Spain, Turkey, Rest of Europe in Europe, China, Japan, India, South Korea, Singapore, Malaysia, Australia, Thailand, Indonesia, the Philippines, Rest of Asia-Pacific (APAC) in the Asia-Pacific region (APAC), Saudi Arabia, United Arab Emirates , South Africa, Egypt, Israel, Rest of the Middle East and Africa (MEA) under Middle East and Africa (MEA), Brazil,

Europe dominates the bone marrow market due to the proliferation of innovative health centers.Furthermore, the health systems have introduced bone marrow transplantation in their contributions and state-of-the-art public facilities that will further drive the growth of the bone marrow market in the region during the forecast period.North America is expected to witness significant growth in the bone marrow market due to increasing cases of chronic diseases such as blood cancer.In addition, the increase in the geriatric population is one of the factors that is expected to drive the growth of the bone marrow market in the region in the coming years.

Explore Full TOC At:- https://www.databridgemarketresearch.com/toc/?dbmr=global-bone-marrow-market

The country section of the Bone Marrow market report also provides individual market impact factors and regulatory changes in the country market that affect current and future market trends.Data points such as consumption volumes, production sites and volumes, import and export analysis, price trend analysis, raw material cost, Downstream and Upstream value chain analysis are some of the main indicators used to forecast the scenario. of the market for each country.Additionally, the presence and availability of global brands and the challenges they face due to significant or rare competition from local and national brands,

Top Healthcare Report Links:

https://www.digitaljournal.com/pr/osteomyelitis-drugs-market-global-key-players-market-dynamics-future-demand-analysis-development-business-industry-technology-opportunity-and-forecasts-to-2029

https://www.digitaljournal.com/pr/glioblastoma-multiforme-treatment-market-global-industry-analysis-trend-development-supply-chain-key-players-share-scope-size-growthsegmentation-forecasts-to-2029

https://www.digitaljournal.com/pr/digital-behavioral-health-services-market-overview-share-trend-demand-supply-analysis-size-segmentation-and-forecast-to-2029

https://www.digitaljournal.com/pr/asia-pacific-biopreservation-market-development-trend-channel-vendors-key-players-analysis-supply-research-and-forecast-to-2029

https://www.digitaljournal.com/pr/u-s-integrated-distribution-network-market-will-reach-usd-40-91-billion-with-a-cagr-of-9-67during-the-forecast-period-from-2022-to-2029

About Data Bridge Market Research:

An absolute way to forecast what future holds is to comprehend the trend today! Data Bridge Market Research set forth itself as an unconventional and neoteric Market research and consulting firm with unparalleled level of resilience and integrated approaches. We are determined to unearth the best market opportunities and foster efficient information for your business to thrive in the market. Data Bridge endeavours to provide appropriate solutions to the complex business challenges and initiates an effortless decision-making process. Data Bridge is an aftermath of sheer wisdom and experience which was formulated and framed in the year 2015 in Pune.

Data Bridge Market Research has over 500 analysts working in different industries. We have catered more than 40% of the fortune 500 companies globally and have a network of more than 5000+ clientele around the globe. Data Bridge adepts in creating satisfied clients who reckon upon our services and rely on our hard work with certitude. We are content with our glorious 99.9 % client satisfying rate.

Contact Us:-

Data Bridge Market Research

US: +1 888 387 2818

UK: +44 208 089 1725

Hong Kong: +852 8192 7475

Email:-[emailprotected]

Go here to see the original:
Bone Marrow market estimated to reach US$13899.60 Million during the forecast period - Digital Journal

3D Printers in Zero-G Flights? There Have Been a Few of Those – 3DPrint.com

In 2011, Made In Space created the first 3D printer for microgravity; what sounded like science fiction suddenly became a reality. Since then, at least 15 experimental 3D printers have been tested aboard Zero-G flights worldwide. Powered by companies, academic institutions, and space agencies, this type of 3D printing research has been successful, from a few printers occasionally tested between 2011 and 2018 to half a dozen in 2022 alone.

Looking back at 2011, we might remember it as a year of transition for the space industry, chiefly because it was the beginning of the end for NASAs Space Shuttle program, which took its final flight in July of that year. With a budget trim to go with it, NASA would soon turn to private industry for many of its space needs. One company, in particular, was keen to leave its mark. Known today as the firm that creates 3D printers for the International Space Station (ISS), Made In Space came out of Singularity University looking to fill a space manufacturing gap.

At its core, Made In Space founders believed that 3D printing and in-space manufacturing would dramatically change the way we look at space exploration, commercialization, and mission design today.

Like Made In Space (now part of Redwire), other companies also decided to test their 3D printing technology in parabolic flights. For example, in 2016, engineering firm and regular NASA contractor Techshot (also acquired by Redwire) partnered with manufacturer nScrypt to create the first microgravity bioprinter and tested it in an aircraft flown by the Zero Gravity Corporation, which operates weightless flights from U.S. airports.

Flying at 30,000 ft (roughly 9,144 meters) over the Gulf of Mexico, the plane simulated weightlessness while the bioprinter created cardiac and vascular structures using human stem cells. Like Made In Space, Techshot and nScrypt later sent the bioprinter to the ISS U.S. National Laboratory, where astronauts are using it for manufacturing human knee cartilage test prints and other human tissue.

The idea of manufacturing in space has long posed several obvious challenges, primarily gravity issues, quality controls, and raw material sourcing. However, once in place, in-situ manufacturing has the potential to relax the dependence on resource resupply from Earth, making survival in space a little bit easier.

For decades, in-space manufacturing has been investigated as a method for producing parts and components in orbit that would otherwise be almost impossible to obtain immediately or at all. In the late 1960s, Soviet cosmonauts conducted the first welding experiments in space as part of their space manufacturing research. In the next decade, the United States began experimenting with space manufacturing in Skylab, the first space station launched by NASA.

But the gateway to space manufacturing lies in the investigations of parabolic flights that can reproduce gravity-free conditions in an aircraft right here on Earth. By alternating upward and downward arcs, they provide the necessary microgravity environment for scientists to conduct research without actually traveling to space. This simulated weightlessness may have started in the 1960s with the first flying space labs aboard U.S. military planes. Still, it has expanded to incorporate several private businesses, like US-based company Zero-G and French-based Novespace.

With more options to recreate the unique weightlessness of space, we have witnessed a series of printers that have been successfully tested in parabolic flights. For example, in late 2016, Luke Carter of the Advanced Materials and Processing Laboratory (AMP Lab) at the University of Birmingham demonstrated metal 3D printing in microgravity aboard three separate parabolic flights. By creating a printing process much like directed energy deposition (DED), and using aluminum wire as feedstock, Carter and his team made a near-net shape part.

Then in 2017, the Canadian Reduced Gravity Experiment Design Challenge (CAN-RGX), supported by the National Research Council and the Canadian Space Agency, chose two teams to test their 3D printing experiments in parabolic flights. Team AVAIL (Analyzing Viscosity and Inertia in Liquids) from the University of Toronto built a system that controls the flow of a viscous liquid (corn syrup, in this case) through 15 different nozzles, and Team iSSELab (Interfacial Science and Surface Engineering Lab), hailing from the University of Alberta, collected data from 3D printing materials in a reduced gravity environment.

The following year, a European parabolic flight aircraft in New Zealand took scientists from the Technology and Engineering Center for Space Utilization of the Chinese Academy of Sciences (CAS) to test the first ceramic DLP 3D printer in microgravity. Following this successful event, NASA chose Associate Professor Gregory Whiting and his research group to test and model how 3D printing functional materials would work in lunar gravity. Whitings research group, the Boulder Experimental Electronics and Manufacturing Lab, geared up for two parabolic flights in 2021.

Around that time, engineering students of the Munich University of Applied Sciences built a 3D printer with an extruder to dispense a liquid photopolymer that took off on the European Space Agency (ESA)s 74th parabolic flight campaign from Paderborn-Lippstadt Airport in Germany.

A few memorable 3D printing experiments in zero gravity in 2022 include Space Foundrys testing of space-based electronic printing, supported by NASAs Flight Opportunities and Small Business Innovation Research (SBIR) programs. In addition, UC Berkeley research teams tested the replicator, a light-based 3D printer, on May 10, printing more than 100 objects. Also, a German consortium tested out its patented 3D printing process and, for the first time, used metallic powders to 3D print in zero gravity.

This is just a taste of what is possible here on Earth, thanks to gravity-free flights. These and other experiments that took place in the last few years can be found below.

Stay up-to-date on all the latest news from the 3D printing industry and recieve information and offers from thrid party vendors.

Read the original:
3D Printers in Zero-G Flights? There Have Been a Few of Those - 3DPrint.com

Stem cell-based therapy for human diseases | Signal Transduction and Targeted Therapy – Nature.com

Mellman, I., Coukos, G. & Dranoff, G. Cancer immunotherapy comes of age. Nature 480, 480489 (2011).

PubMed PubMed Central Article CAS Google Scholar

Ancans, J. Cell therapy medicinal product regulatory framework in Europe and its application for MSC-based therapy development. Front. Immunol. 3, 253 (2012).

PubMed PubMed Central Article Google Scholar

Yin, J. Q., Zhu, J. & Ankrum, J. A. Manufacturing of primed mesenchymal stromal cells for therapy. Nat. Biomed. Eng. 3, 90104 (2019).

PubMed Article CAS Google Scholar

OBrien, T. & Barry, F. P. Stem cell therapy and regenerative medicine. Mayo Clin. Proc. 84, 859861 (2009).

PubMed PubMed Central Article Google Scholar

Mousaei Ghasroldasht, M., Seok, J., Park, H. S., Liakath Ali, F. B. & Al-Hendy, A. Stem cell therapy: from idea to clinical practice. Int. J. Mol. Sci. 23, 2850 (2022).

Kuriyan, A. E. et al. Vision loss after intravitreal injection of autologous stem cells for AMD. N. Engl. J. Med. 376, 10471053 (2017).

PubMed PubMed Central Article Google Scholar

Biehl, J. K. & Russell, B. Introduction to stem cell therapy. J. Cardiovasc. Nurs. 24, 98103 (2009). quiz 104-105.

PubMed PubMed Central Article Google Scholar

Srijaya, T. C., Ramasamy, T. S. & Kasim, N. H. Advancing stem cell therapy from bench to bedside: lessons from drug therapies. J. Transl. Med. 12, 243 (2014).

PubMed PubMed Central Article Google Scholar

Ramalho-Santos, M. & Willenbring, H. On the origin of the term stem cell. Cell Stem Cell 1, 3538 (2007).

PubMed Article CAS Google Scholar

Konstantinov, I. E. In search of Alexander A. Maximow: the man behind the unitarian theory of hematopoiesis. Perspect. Biol. Med. 43, 269276 (2000).

PubMed Article CAS Google Scholar

Droscher, A. Images of cell trees, cell lines, and cell fates: the legacy of Ernst Haeckel and August Weismann in stem cell research. Hist. Philos. Life Sci. 36, 157186 (2014).

PubMed Article Google Scholar

Jansen, J. The first successful allogeneic bone-marrow transplant: Georges Mathe. Transfus. Med. Rev. 19, 246248 (2005).

PubMed Article Google Scholar

Blume, K. G. & Weissman, I. L. E. Donnall Thomas (1920-2012). Proc. Natl Acad. Sci. USA 109, 2077720778 (2012).

PubMed PubMed Central Article CAS Google Scholar

Cheng, M. Hartmann Stahelin (1925-2011) and the contested history of cyclosporin A. Clin. Transpl. 27, 326329 (2013).

Article CAS Google Scholar

Thomas, E. D. et al. Aplastic anaemia treated by marrow transplantation. Lancet 1, 284289 (1972).

PubMed Article CAS Google Scholar

Friedenstein, A. J., Chailakhyan, R. K. & Gerasimov, U. V. Bone marrow osteogenic stem cells: in vitro cultivation and transplantation in diffusion chambers. Cell Tissue Kinet. 20, 263272 (1987).

PubMed CAS Google Scholar

Friedenstein, A. J., Chailakhjan, R. K. & Lalykina, K. S. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 3, 393403 (1970).

PubMed CAS Google Scholar

Caplan, A. I. Mesenchymal stem cells. J. Orthop. Res. 9, 641650 (1991).

PubMed Article CAS Google Scholar

Bolli, R., Tang, X. L., Guo, Y. & Li, Q. After the storm: an objective appraisal of the efficacy of c-kit+ cardiac progenitor cells in preclinical models of heart disease. Can. J. Physiol. Pharm. 99, 129139 (2021).

Article CAS Google Scholar

Liu, C., Han, D., Liang, P., Li, Y. & Cao, F. The current dilemma and breakthrough of stem cell therapy in ischemic heart disease. Front. Cell Dev. Biol. 9, 636136 (2021).

PubMed PubMed Central Article Google Scholar

Zhang, J. et al. Basic and translational research in cardiac repair and regeneration: JACC state-of-the-art review. J. Am. Coll. Cardiol. 78, 20922105 (2021).

PubMed PubMed Central Article CAS Google Scholar

Gyongyosi, M., Wojakowski, W., Navarese, E. P., Moye, L. A. & Investigators, A. Meta-analyses of human cell-based cardiac regeneration therapies: controversies in meta-analyses results on cardiac cell-based regenerative studies. Circ. Res. 118, 12541263 (2016).

PubMed PubMed Central Article CAS Google Scholar

Okamoto, R., Matsumoto, T. & Watanabe, M. Regeneration of the intestinal epithelia: regulation of bone marrow-derived epithelial cell differentiation towards secretory lineage cells. Hum. Cell 19, 7175 (2006).

PubMed Article Google Scholar

Gehart, H. & Clevers, H. Tales from the crypt: new insights into intestinal stem cells. Nat. Rev. Gastroenterol. Hepatol. 16, 1934 (2019).

PubMed Article Google Scholar

Santos, A. J. M., Lo, Y. H., Mah, A. T. & Kuo, C. J. The intestinal stem cell niche: homeostasis and adaptations. Trends Cell Biol. 28, 10621078 (2018).

PubMed PubMed Central Article CAS Google Scholar

Roda, G. et al. Crohns disease. Nat. Rev. Dis. Prim. 6, 22 (2020).

PubMed Article Google Scholar

Kobayashi, T. et al. Ulcerative colitis. Nat. Rev. Dis. Prim. 6, 74 (2020).

PubMed Article Google Scholar

Gratwohl, A. et al. Autologous hematopoietic stem cell transplantation for autoimmune diseases. Bone Marrow Transpl. 35, 869879 (2005).

Article CAS Google Scholar

Kashyap, A. & Forman, S. J. Autologous bone marrow transplantation for non-Hodgkins lymphoma resulting in long-term remission of coincidental Crohns disease. Br. J. Haematol. 103, 651652 (1998).

PubMed Article CAS Google Scholar

Hurley, J. M., Lee, S. G., Andrews, R. E. Jr., Klowden, M. J. & Bulla, L. A. Jr. Separation of the cytolytic and mosquitocidal proteins of Bacillus thuringiensis subsp. israelensis. Biochem Biophys. Res. Commun. 126, 961965 (1985).

PubMed Article CAS Google Scholar

Oyama, Y. et al. Autologous hematopoietic stem cell transplantation in patients with refractory Crohns disease. Gastroenterology 128, 552563 (2005).

PubMed Article Google Scholar

Burt, R. K. et al. Autologous nonmyeloablative hematopoietic stem cell transplantation in patients with severe anti-TNF refractory Crohn disease: long-term follow-up. Blood 116, 61236132 (2010).

PubMed Article CAS Google Scholar

Hasselblatt, P. et al. Remission of refractory Crohns disease by high-dose cyclophosphamide and autologous peripheral blood stem cell transplantation. Aliment Pharm. Ther. 36, 725735 (2012).

Article CAS Google Scholar

Hawkey, C. J. et al. Autologous hematopoetic stem cell transplantation for refractory Crohn disease: a randomized clinical trial. J. Am. Med. Assoc. 314, 25242534 (2015).

Article CAS Google Scholar

Lindsay, J. O. et al. Autologous stem-cell transplantation in treatment-refractory Crohns disease: an analysis of pooled data from the ASTIC trial. Lancet Gastroenterol. Hepatol. 2, 399406 (2017).

PubMed Article Google Scholar

Wang, R. et al. Stem cell therapy for Crohns disease: systematic review and meta-analysis of preclinical and clinical studies. Stem Cell Res Ther. 12, 463 (2021).

PubMed PubMed Central Article CAS Google Scholar

Hawkey, C. J. Hematopoietic stem cell transplantation in Crohns disease: state-of-the-art treatment. Dig. Dis. 35, 107114 (2017).

PubMed Article CAS Google Scholar

Si-Tayeb, K., Lemaigre, F. P. & Duncan, S. A. Organogenesis and development of the liver. Dev. Cell 18, 175189 (2010).

PubMed Article CAS Google Scholar

Xue, R. et al. Clinical performance of stem cell therapy in patients with acute-on-chronic liver failure: a systematic review and meta-analysis. J. Transl. Med. 16, 126 (2018).

PubMed PubMed Central Article CAS Google Scholar

Shi, M. et al. Human mesenchymal stem cell transfusion is safe and improves liver function in acute-on-chronic liver failure patients. Stem Cells Transl. Med. 1, 725731 (2012).

PubMed PubMed Central Article CAS Google Scholar

Liu, Y., Dong, Y., Wu, X., Xu, X. & Niu, J. The assessment of mesenchymal stem cells therapy in acute on chronic liver failure and chronic liver disease: a systematic review and meta-analysis of randomized controlled clinical trials. Stem Cell Res. Ther. 13, 204 (2022).

PubMed PubMed Central Article Google Scholar

Lin, B. L. et al. Allogeneic bone marrow-derived mesenchymal stromal cells for hepatitis B virus-related acute-on-chronic liver failure: a randomized controlled trial. Hepatology 66, 209219 (2017).

PubMed Article CAS Google Scholar

Gordon, M. Y. et al. Characterization and clinical application of human CD34+ stem/progenitor cell populations mobilized into the blood by granulocyte colony-stimulating factor. Stem Cells 24, 18221830 (2006).

PubMed Article Google Scholar

Arroyo, V. et al. Acute-on-chronic liver failure in cirrhosis. Nat. Rev. Dis. Prim. 2, 16041 (2016).

PubMed Article Google Scholar

Zhang, Z. et al. Human umbilical cord mesenchymal stem cells improve liver function and ascites in decompensated liver cirrhosis patients. J. Gastroenterol. Hepatol. 27(Suppl 2), 112120 (2012).

PubMed Article CAS Google Scholar

El-Ansary, M. et al. Phase II trial: undifferentiated versus differentiated autologous mesenchymal stem cells transplantation in Egyptian patients with HCV induced liver cirrhosis. Stem Cell Rev. Rep. 8, 972981 (2012).

PubMed Article CAS Google Scholar

Xu, L. et al. Randomized trial of autologous bone marrow mesenchymal stem cells transplantation for hepatitis B virus cirrhosis: regulation of Treg/Th17 cells. J. Gastroenterol. Hepatol. 29, 16201628 (2014).

PubMed Article CAS Google Scholar

Suk, K. T. et al. Transplantation with autologous bone marrow-derived mesenchymal stem cells for alcoholic cirrhosis: Phase 2 trial. Hepatology 64, 21852197 (2016).

PubMed Article CAS Google Scholar

Fang, X. et al. A study about immunomodulatory effect and efficacy and prognosis of human umbilical cord mesenchymal stem cells in patients with chronic hepatitis B-induced decompensated liver cirrhosis. J. Gastroenterol. Hepatol. 33, 774780 (2018).

PubMed Article CAS Google Scholar

Mohamadnejad, M. et al. Randomized placebo-controlled trial of mesenchymal stem cell transplantation in decompensated cirrhosis. Liver Int. 33, 14901496 (2013).

PubMed Article CAS Google Scholar

Nguyen, T. L. et al. Autologous bone marrow mononuclear cell infusion for liver cirrhosis after the Kasai operation in children with biliary atresia. Stem Cell Res. Ther. 13, 108 (2022).

Follow this link:
Stem cell-based therapy for human diseases | Signal Transduction and Targeted Therapy - Nature.com

Protocol for a Nested, Retrospective Study of the Australian Placental Transfusion Study Cohort – Cureus

Immediate cord clamping (ICC), within a few seconds after birth, became routine in the latter half of the 20th century, as part of a tranche of medical birth-related interventions that collectively, undoubtedly improved maternal and neonatal survival and outcomes [1]. The trend to ICC (within 15-20 seconds after birth) was partly driven by some early studies suggesting that the most benefit in terms of blood volume is achieved within this time frame [2], and that deferred cord clamping (DCC) increased rates of polycythemia and jaundice [1]. It may also have been partly driven by increased rates of operative deliveries and consequent pressure to minimize surgical times, as well as the increased availability and effectiveness of neonatal resuscitation. Furthermore, ICC was proposed as a means to reduce the risk of maternal exposure to fetal blood group antigens at a time (before RhD immunoprophylaxis) when hemolytic disease of the fetus and newborn was far more common than it is now.

Formal evidence that ICC was beneficial was never sought, and recent research summarized in systematic reviews [3-6] has suggested that it may be harmful when compared with DCC for various intervals from 30 seconds until when the cord stops pulsating (defined in some studies as physiological cord clamping). ICC before the onset of breathing exposes the newborn baby to a period of significantly restricted cardiac function, whereas DCC until after the onset of breathing (which often does not occur until late in the first minute after birth) may mean that the expanding pulmonary circulation is able to fill with blood from the placenta, rather than by reverse flow across the ductus arteriosus [7]. This may improve left ventricular preload and stabilize pressures and flows in major vessels [7].

In addition, when cord clamping is deferred, babies may receive a transfusion of blood from the umbilical cord and placenta. A recent systematic review demonstrated that DCC in preterm babies improves peak hematocrit in the first week by 2.7% (95% confidence intervals (CI) 1.88-3.52) and reduced the proportion of babies receiving any subsequent blood transfusion (RD: -0.07, 95%CI -0.11 to -0.04) [6]. Some studies have found a weight increase in the first two minutes after birth when the cord is not clamped, supporting the hypothesis of placental transfusion [8]. Yet, recent evidence shows that placental transfusion may not always occur (Conference abstract: Vijayaselvi R, Abraham A, Kumar M, Kuruvilla A, Mathews J, Duley L. Measuring Umbilical Flow and Placental Transfusion for Preterm Births: Weighing Babies at 33-36 Weeks Gestation with Cord Intact. 1st Congress of Joint European Neonatal Societies; 2015).

The relative roles of cardiovascular stabilization at birth versus placental transfusion in improving outcomes have not been established. Understanding the contributions of these two mechanisms has significant implications for research and practice: for example, if the size of placental transfusion is more important, then prescribing a top-up transfusion soon after birth for babies with lower than average hemoglobin (who are known to be at higher risk of various adverse outcomes) [9] may be justified, especially for the babies for whom DCC has been precluded by maternal or fetal conditions. These include significant maternal bleeding, and monochorionic twins, where deferred cord clamping in the first twin could lead to one twin losing blood to the other. However, if it is the effects on improving cardiovascular stability in the first minutes (with consequential benefits for cardiorespiratory function and reducing severity of illness during the subsequent neonatal intensive care unit (NICU) stay), regardless of the magnitude of transfusion, then early top-up transfusion is unlikely to be helpful.

Observational studies suggest that exposure to blood transfusion itself is harmful to preterm babies, increasing the risk of adverse outcomes [10]. However, this suggestion has not been supported by the small number (to date) of randomized controlled trials of blood (red cell) transfusion thresholds [11-14]. It is unlikely to be the means by which DCC reduced deaths in the largest trial to date of deferred cord clamping in preterm babies, the Australian Placental Transfusion Study (APTS), and in the most recent systematic review on this, because neither showed a difference in rates of other adverse outcomes [6,15].

Another possibility is that it is the umbilical cord blood stem cells received by the baby are the main reason for the observed benefits to both survival and reduced requirement for later blood transfusion [16]. Umbilical cord blood has been demonstrated to be such a good contributor to hematopoiesis that it is a recognized stem cell resource for pediatric and adult hematopoietic stem cell transplant [17]. In addition, umbilical cord blood is a potential regenerative and immunomodulatory agent for a variety of clinical conditions [18], so in this case, the extent of placental transfusion would be critical to the improvement of outcomes, and transfusion with adult red cells would not suffice. There are no established methods to quantify the contribution of umbilical cord stem cells to placental transfusion. However, a larger volume of placental transfusion results in the baby receiving more nucleated cells [19], including more umbilical cord stem cells.

Discerning whether these effects (initial enhanced cardiovascular stability leading to early and sustained reduction in severity of illness or volume of placental transfusion) appear to be the main driver of improved outcomes is likely to contribute to practice change, as well as to informing the design of future research studies into methods to improve outcomes of high-risk newborn babies and reduce their transfusion dependence.

The causal mechanisms of reduced transfusion requirements found in DCC relative to ICC are yet to be resolved. The aim of the study is to address the question; In preterm infants (P) does DCC (I) compared to ICC (C) reduce dependence on red cell transfusion via enhanced cardiovascular stability (mediator 1, M1) or via an increased volume of placental transfusion (M2).

The study is a nested retrospective study, called the Transfusions in the APTS Newborns Study (TITANS) (study registration: ACTRN12620000195954), of the cohort of babies who were enrolled and randomly assigned to ICC or DCC in the Australian and New Zealand (NZ) sites for APTS (study registration: ACTRN12610000633088). This design has been developed to take advantage of the comprehensive dataset already collected for APTS, and because there is currently no suitable prospective study that could address the same research questions in such a large group of participants.

Babies had been considered eligible for APTS if obstetricians or maternal-fetal medicine specialists anticipated that delivery would occur before 30 weeks of gestation. Exclusion criteria included fetal hemolytic disease, hydrops fetalis, twin-twin transfusion, genetic syndromes, and potentially lethal malformations. Further details are available in the original APTS publication [15]. In the present TITANS analysis, we will also exclude any baby with a diagnosis of hemolytic anemia or aplastic/hypoplastic anemia.

There were 1401 babies enrolled for APTS from the 13 Australian and 5 NZ hospital sites [15]. APTS data was provided to the TITANS team on 31 July, 2020. It is planned to collect additional data from Australian and NZ APTS sites using a customised, secure web-based database application (REDCap) [20], which is maintained by the University of Sydney, Sydney, Australia. Data will be obtained from source documents (patient hospital records and laboratory reports) using the electronic data collection application from each study site. The individual participant data collected will correspond to the minimum data required to answer the research questions. Baby identification (ID) and other babies details from APTS will be used to re-identify participants and link them to hospital records. Identified data will be collected, in order to allow linkage between the data newly collected from patient records and hospital laboratories and the existing APTS dataset. The data will be checked with respect to range, internal consistency, consistency with published reports and missing items. After data cleaning and analysis, data will be stored in re-identifiable form, with each participants data being identified with the same study numbering system as used for the APTS study.

We will combine the data already extracted, stored and cleaned from APTS with the additional data obtained from study sites for each participating baby, to determine which factors are most influential in reducing transfusion requirements. The specific objectives are, after adjustment for prior risk factors (listed below), to determine:

1.Whether the effect of the intervention (cord clamping) on the outcome (blood transfusions) is mediated by placental transfusion (measured by hematocrit (Hct)) as seen in Figure 1 (a, c) following the causal path X M1 Y, where X is the intervention, ICC or DCC, Y is the outcome, mediator M1 is placental transfusion, and M2 is initial severity of illness stability

2.Whether the effect of the intervention (cord clamping) on the outcome (blood transfusions) is mediated by initial severity of illness (respiratory support, sampling line yes/no and total duration number, blood pressure, cumulative blood sample volume) as seen in Figure 1 (b, c) following the causal path X M2 Y

3.Whether the effect of cord clamping intervention on the outcome (blood transfusions) is driven by multiple mediators (placental transfusion and initial severity of illness) as seen in Figure 1 (c)

4.Whether cording clamping intervention (ICC or DCC) has a direct effect on the outcome after accounting for the mediators as seen in all panels of Figure 1: X Y.

The protocol was approved by the Northern Sydney Local Health District Human Research Ethics Committee in November 2019 (Version 3.0, Reference 2019/ETH12819), the Mater Misericordiae Ltd Human Research Ethics Committee (Version 1.0, Reference HREC/MML/56247), the Mercy Health Human Research Ethics Committee (Version 2.0, Reference 2020-078), and the Southern Health and Disability Ethics Committee (Version 1.0, Reference 19/STH/195). The ethics committees have granted a waiver of consent. The study is conducted in accordance with the National Health and Medical Research Council Statement on Ethical Conduct in Research Involving Humans.

Intervention

The intervention consisted of either immediate or delayed cord clamping (as assigned in APTS). Immediate clamping was defined as clamping the cord within 10 seconds of delivery. Delayed clamping was defined as clamping the cord at least 60 seconds after delivery, with the infant held as low as possible, below the introitus or placenta, and with no palpation of the cord. Variations in the protocol were allowed if they would aid the mother, baby, or both. If the baby was non-vigorous (heart rate <100 beats per minute, low muscle tone, or lack of breathing, or crying), clinicians were allowed to break protocol using their discretion. Cord milking was not part of the protocol for either intervention. Further details may be sourced from the original APTS publication [15].

Outcomes

The primary outcome is the proportion of babies receiving red cell transfusion (for restoration of hemoglobin or blood volume). The secondary outcomes are number of transfusions per baby, cumulative transfusion volume (mL/kg) per baby, and primary reasons for each transfusion.

Putative Mediators

M1: Indicators of placental transfusion to be assessed will be hematocrit (on admission, highest on the first day, highest in the first week collected before any postnatal transfusion).

M2: Indicators of initial severity of illness to be assessed will be cumulative blood sample volume collected throughout hospital stay (number of blood tests multiplied by hospitals usual sample volume for each type of test), sampling line (umbilical arterial line or peripheral arterial line) - yes/no and total duration, mechanical ventilation or inspired O2, and blood pressure.

Sensitivity Analyses (For the Primary Outcome Analysis Only)

Sensitivity analyses will adjust for the following variables: gender, birth <27 weeks vs. 27 weeks, method of delivery (vaginal versus cesarean), intraventricular hemorrhage (IVH) (yes/no and grade III/IV yes/no), surgery for patent ductus arteriosus (PDA), necrotizing enterocolitis (NEC), and sodium in the first 24 hours of life. We will also test model assumptions relating to sequential ignorability and post-randomization confounding (discussed further in the data analysis plan).

Potential Confounders (Covariates)

The following covariates may be used for adjustment in the analysis: gestational age at randomization before birth and any oral iron supplement pre-transfusion.

Timing of Assessments

Putative mediating variables will only be analyzed if they have been measured before the outcome and will be excluded if there is not adequate time and date information available. If the multiple mediator model is applied, careful consideration of timing information will be evaluated. If there is insufficient empirical information to conclude the causal ordering of mediators (M1 causes M2), we will adjust our analytic approach (as discussed in the analysis plan) and discuss any limitations.

Data Analysis Plan

The analysis will include all babies who were initially randomized in the APTS trial for whom we were able to obtain the relevant data and be based on intention-to-treat. All statistical analyses will be conducted in R version 4.1.3 (2022-03-10; R Foundation for Statistical Computing, Vienna, Austria). Descriptive characteristics for continuous data will be presented as means or medians, as appropriate, and categorical data will be presented as frequencies and percentages.

A model-based inference approach will be applied to estimate the average causal mediation effect (ACME), average direct effect (ADE), and the average total effect as recommended [23-25]. This approach will be applied with the R mediation package [26]. We will initially fit two models, one model with mediation as the dependent variable and intervention as the independent variable (mediator model), and a second model with the outcome as the dependent variable, and both mediation and intervention as independent variables (outcome model). To account for the clustering of multiples, estimates will be calculated with generalized estimating equations with a compound symmetric correlation structure to account for within subject correlations. Depending on the outcome (binary, count, skew) these will be modelled with the appropriate family and link functions.

A counterfactual framework will be applied to the mediator and outcome models to simulate the values of the mediator and outcome to estimate the potential values of the mediator. This process is used to estimate the ACME, ADE, and average total effects; 95%CI will be estimated with 1000 bootstrap simulations.

We will apply single mediator models on both placental transfusion variables and initial severity of illness variables if mediators are statistically independent, as seen in Table 1. Independence will be tested using linear regression and any appropriate link functions. If both mediators are not statistically independent, we will investigate the possibility of multiple mediator models, which require an expanded framework for analysis [21]. Here we assume that initial severity of illness is causally related to placental transfusion. For this process, we will use the method developed by Imai and Yamamoto [21] to estimate the ACME and ADE. Following this, 95%CI will be estimated with 1000 bootstrap simulations. If theoretical and empirical timing data and sensitivity analyses suggest that M1 and M2 have non-causal correlation and may be affected by an unmeasured latent mediator, we will adjust our approach to estimate interventional direct and path-specific indirect effects [27,28].

Sensitivity analyses have been limited to a set of biologically plausible and clinically meaningful groups that will be explored by including them for adjustment with covariates, and with the introduction of interaction terms if appropriate. Missing data will be described, reasons for missing data will be explored, and the impact of missing data on conclusions about the treatment effect on the primary outcome will also be explored where possible (e.g., using sensitivity analyses and multiple imputation techniques).

Methodological Assumptions

The causal mediation approach assumes sequential ignorability: that the treatment effect on the outcome is not confounding and that the mediator effect on the outcome is not confounded. As treatment was randomly allocated to neonates, we will assume that the treatment-mediator relationship is not confounded. However, the mediator itself has not been randomized. Thus, unknown confounders may be driving a spurious effect in the mediator-outcome relationship. We will employ additional sensitivity analyses to estimate whether any mediation effects are sensitive to the violation of the assumption of sequential ignorability. To test the possibility of unmeasured confounders we will examine the correlation between residuals in the mediator model and the outcome model. If there is no correlation this would suggest there is no unmeasured confounding, if there is correlation between the residuals, an unmeasured mediator may be affecting both the measured mediator and the outcome. We will apply the method developed by Imai et al. andTingley et al. [23,26] that uses sensitivity analyses to evaluate if the ACME estimate is sensitive to unmeasured confounding.

Post-randomization confounders are dependent on the treatment allocated, affect both mediator and outcome, and can corrupt the mediation estimate. In the context of the present trial, it is possible that non-adherence to the intervention is a post-randomization confounder. We are analyzing our data based on intention to treat principles; however, a sensitivity analysis based on the actual time of cord clamping to assess the influence of non-adherence with the treatment protocol on our estimates may be performed.

Blood transfusions of neonates have been associated with a number of serious adverse outcomes [29]. Nevertheless, there are few evidence-based methods to reduce transfusion exposure [30]. The APTS study found that DCC was associated with a statistically significant reduced need for red cell transfusions by about 10% compared to ICC [15]. However, the mechanism remains unclear.

The study will, at a minimum, provide further information that should increase clinicians understanding of the pathways by which DCC (or other methods to accomplish placental transfusion) results in beneficial patient outcomes. Since one of the main barriers to implementation is lack of understanding about the mechanisms by which such a simple practice change should have such dramatic effects, this should improve adherence to recommendations to defer cord clamping for most babies, thereby reducing mortality and transfusion incidence.

By elaborating on the mechanisms, it may also provide good evidence for how other routine neonatal intensive care practices and interventions affect likelihood of needing to transfuse. Better understanding of these effects may lead to other testable hypotheses or improvements in other aspects of practice, further reducing transfusion exposure and improving other outcomes.

Potential limitations of the study include the dependence on some routinely collected clinical data, which were not collected at the time by the original study according to predefined research definitions. However, we have no reason to think that potential problems of data quality would have been influenced by study group allocation and so do not anticipate that this will be a source of bias.

Go here to read the rest:
Protocol for a Nested, Retrospective Study of the Australian Placental Transfusion Study Cohort - Cureus

Autologous Cell Therapy Market Size to Grow by USD 4.11 billion, Bayer AG and Brainstorm Cell Therapeutics Inc. Among Key Vendors – Technavio – PR…

Get a comprehensive report summary describing the market size and forecast along with research methodology. View our Sample Report

Autologous Cell Therapy Market 2021-2025: Scope

The autologous cell therapy market report covers the following areas:

Autologous Cell Therapy Market 2021-2025: Segmentation

Learn about the contribution of each segment summarized in concise infographics and thorough descriptions. View a PDF Sample Report

Autologous Cell Therapy Market 2021-2025: Vendor Analysis

We provide a detailed analysis of around 25 vendors operating in the autologous cell therapy market, including Bayer AG, Brainstorm Cell Therapeutics Inc., Daiichi Sankyo Co. Ltd., FUJIFILM Holdings Corp., Holostem Terapie Avanzate Srl, Osiris Therapeutics Inc., Takeda Pharmaceutical Co. Ltd., Teva Pharmaceutical Industries Ltd., Sumitomo Chemical Co. Ltd., and Vericel Corp. among others. The key offerings of some of these vendors are listed below:

Get lifetime access to our Technavio Insights! Subscribe to our "Basic Plan" billed annually at USD 5000

Autologous Cell Therapy Market 2021-2025: Key Highlights

Related Reports

Hairy Cell Leukemia Therapeutics Market by Product and Geography - Forecast and Analysis 2022-2026

Cell Therapy Market by Type, Application, and Geography - Forecast and Analysis 2022-2026

Autologous Cell Therapy Market Scope

Report Coverage

Details

Page number

120

Base year

2020

Forecast period

2021-2025

Growth momentum & CAGR

Accelerate at a CAGR of 14.16%

Market growth 2021-2025

USD 4.11 billion

Market structure

Fragmented

YoY growth (%)

13.5

Regional analysis

North America, Europe, APAC, and South America

Performing market contribution

North America at 43%

Key consumer countries

US, UK, Germany, Canada, and Japan

Competitive landscape

Leading companies, competitive strategies, consumer engagement scope

Companies profiled

Bayer AG, Brainstorm Cell Therapeutics Inc., Daiichi Sankyo Co. Ltd., FUJIFILM Holdings Corp., Holostem Terapie Avanzate Srl, Osiris Therapeutics Inc., Takeda Pharmaceutical Co. Ltd., Teva Pharmaceutical Industries Ltd., Sumitomo Chemical Co. Ltd., and Vericel Corp.

Market Dynamics

Parent market analysis, market growth inducers and obstacles, fast-growing and slow-growing segment analysis, COVID-19 impact and future consumer dynamics, market condition analysis for the forecast period

Customization purview

If our report has not included the data that you are looking for, you can reach out to our analysts and get segments customized.

Table Of Contents :

Executive Summary

Market Landscape

Market Sizing

Five Forces Analysis

Market Segmentation by Product

Customer landscape

Geographic Landscape

Vendor Landscape

Vendor Analysis

Appendix

About Us

Technavio is a leading global technology research and advisory company. Their research and analysis focus on emerging market trends and provide actionable insights to help businesses identify market opportunities and develop effective strategies to optimize their market positions. With over 500 specialized analysts, Technavio's report library consists of more than 17,000 reports and counting, covering 800 technologies, spanning across 50 countries. Their client base consists of enterprises of all sizes, including more than 100 Fortune 500 companies. This growing client base relies on Technavio's comprehensive coverage, extensive research, and actionable market insights to identify opportunities in existing and potential markets and assess their competitive positions within changing market scenarios.

Contact

Technavio ResearchJesse MaidaMedia & Marketing ExecutiveUS: +1 844 364 1100UK: +44 203 893 3200Email: [emailprotected]Website: http://www.technavio.com/

SOURCE Technavio

Read the original:
Autologous Cell Therapy Market Size to Grow by USD 4.11 billion, Bayer AG and Brainstorm Cell Therapeutics Inc. Among Key Vendors - Technavio - PR...

UTSW researcher part of team awarded $36 million heart research grant – The Dallas Morning News

The British Heart Foundation announced the winner of its $36 million Big Beat Challenge, one of the largest non-commercial awards ever given for heart research.

The winning team, CureHeart, brings together researchers from the U.K., U.S. and Asia, including Eric Olson, professor and chair of the Department of Molecular Biology at UT Southwestern Medical Center.

Olson is the founding chair of the department and directs the Hamon Center for Regenerative Science and Medicine and the Wellstone Center for Muscular Dystrophy Research. He holds the Robert A. Welch Distinguished Chair in Science and the Annie and Willie Nelson Professorship in Stem Cell Research.

He has spent his career investigating heart and muscle development and disease, leading to his participation on the CureHeart team. The Olson Lab at UTSW has been incredibly successful in muscular research, most recently providing a new way to correct the mutation that causes Duchenne muscular dystrophy through gene editing.

CureHeart made the top of the list with its gene editing therapy aimed at curing inherited heart muscle diseases, known as cardiomyopathies.

A BHF release said the technology will seek to develop the first cures for inherited heart muscle diseases by pioneering revolutionary and ultra-precise gene therapy technologies that could edit or silence the faulty genes that cause these deadly conditions.

The project will use gene-editing technology CRISPR to complete base and prime editing in the heart for the first time.

It works by correcting or silencing a faulty gene in the pumping machinery of the heart, either by re-writing the DNA at a single location or by switching off the entire copy of the faulty gene.

The technique was described as molecules that act like tiny pencils to rewrite the single mutations that are buried within the DNA of heart cells in people with heart conditions.

It can also help the heart produce enough proteins to function normally, again by fixing or stimulating the faulty gene.

With ultra-precise base editing technology, we hope to be able to correct a single letter and larger errors in the genetic code. This would mark a breakthrough for not only genetic cardiomyopathies, but for many heart conditions, said Olson in the release.

The project is the next step toward a real-world application, having already proved successful in animals with cardiomyopathies and in human cells. Members of the team believe therapies could be delivered through an arm injection, slowing or stopping the progression of cardiomyopathies, or even curing the disease entirely.

If successful, the research could have enormous impacts.

Every year in the US, around 2,000 people under the age of 25 die of a sudden cardiac arrest, often caused by one of these inherited muscle diseases, said the release. Current treatments do not prevent the condition from progressing, and around half of all heart transplants are needed because of cardiomyopathy.

The researchers believe it could also be successful in preventing the disease from being expressed if inherited. Children who receive the faulty gene from their parents could receive the injection and never develop cardiomyopathy in the first place.

Over the last 30 years, we have made extraordinary advancements in our understanding of the genetic mistakes that cause cardiomyopathy. CureHeart is a once-in-a-generation opportunity to transform this knowledge into a cure, said Olson in the release.

The technology is still in the research and development phase, but Olson said the funds will be used to optimize the method and expand it to a larger number of genetic diseases of the heart, and could move to clinical trials in the next few years.

Visit link:
UTSW researcher part of team awarded $36 million heart research grant - The Dallas Morning News

Curi Bio Launches Mantarray Platform 3D Engineered Muscle Tissues for Discovery of New Therapeutics – Business Wire

SEATTLE--(BUSINESS WIRE)--Curi Bio, a leading developer of human stem cell-based platforms for drug discovery, today announced the commercial launch of the Mantarray platform for human-relevant 3D heart and skeletal engineered muscle tissue (EMT) contractility analysis. Curis Mantarray platform enables the discovery of new therapeutics by providing parallel analysis of 3D EMTs with adult human-like functional profiles of healthy and disease models. Over the past three years, Curi Bio has developed the Mantarray platform in close beta testing and development partnerships with several leading global pharma companies, biotech companies, and regulatory agencies for use in their drug discovery and development projects. With this commercial launch, the Mantarray instrument and consumables will be available worldwide, on a first-come, first-served basis.

Lack of clinical translatability in current preclinical models, which often rely upon surrogate biomarkers or poorly translational animal models, may be to blame for the unacceptably high clinical trial failure rates plaguing the industry. For cardiac and skeletal muscle diseases, direct assessment of contractile output constitutes the most reliable metric to assess overall tissue function, as other proxy measurements are poor predictors of muscle strength. 3D EMTs derived from human induced pluripotent stem cells (iPSCs) offer a promising route to model the contractile deficiencies seen in the heart and muscles of patients. However, the bioengineering strategies required to generate these predictive models at sufficient scale are oftentimes out of reach for most researchers.

Curis solution to this problem is the Mantarray platform: an easy-to-use, scalable, and flexible biosystem for assaying EMTs. The Mantarray platform leverages a proprietary, label-free, electromagnetic measurement system for parallel contractility assessment of up to 24 3D EMTs simultaneously and can utilize a variety of contractile cell types. The system has built-in electrical stimulation capabilities, enabling users to easily create individual stimulation protocols for each well, for both short and long-term pacing of cardiac and skeletal muscle models. Curi has developed consumable Mantarray plates that are specifically designed for cardiac and skeletal muscle applications. 3D Mantarray tissue models will also be compatible with Curis new Nautilus system (optical mapping system for voltage and calcium analysis).

Additionally, Curi offers services and partnerships leveraging the Mantarray technology for applications in drug discovery, disease modeling, and safety and efficacy screening. Companies interested in fully accessing or licensing Curis proprietary disease models, next-generation iPSC platforms, gene therapy technologies and technical expertise can do so with Curis new Technology Access Partnership program. Further details of this Technology Access Partnership program will be announced at a later date.

High-fidelity models of human diseases can be created and tested on the Mantarray platform using human iPSC-derived cells or patient-derived myoblasts from healthy or affected individuals. For example, Mantarray 3D EMTs can be formed from cells harboring disease-causing mutations (patient-derived or gene-edited; iPSC-derived or primary) to model human diseases such as Duchenne muscular dystrophy, myotonic dystrophy type 1, hypertrophic and dilated cardiomyopathies, and more. Multi-modal data captured with the 3D platform shows enhanced disease stratification for a number of monogenic disorders. Curi Bio is accelerating the path to investigational new drugs, making personalized medicine a reality through clinically-translatable human disease models.

Safety and efficacy testing can also be performed with the Mantarrays novel magnetic detection of drug-induced contractile changes. Acute drug responses can be measured on the order of seconds to minutes with enough sensitivity to detect dose-response-like behavior. Alternatively, chronic experiments can be performed over the course of days to weeks for longer-term studies. Mantarray brings clinically-relevant functional data into the earliest stages of preclinical testing of new therapeutics.

At Curi Bio, our goal is to provide researchers with innovative solutions to accelerate the discovery of new medicines, said Curi CEO Michael Cho. The Mantarray platform provides drug developers with clinically-relevant preclinical models that more closely recapitulate human cardiac and skeletal muscle tissues. With this key milestone, Curi is closing the gap between preclinical results and clinical impact. Our clients are using this data in direct support of IND applications.

To learn more about how the Mantarray system can improve the predictive power of your experiments, or about Curis other human-relevant preclinical platform technologies and services, please reach out at http://www.curibio.com/contact.

About Curi Bio

Curi Bios preclinical discovery platform combines human stem cells, systems, and data to accelerate the discovery of new medicines. The Curi Engine is a comprehensive, bioengineered platform that integrates human iPSC-derived cell models, tissue-specific biosystems, and AI/ML-enabled phenotypic screening data. Curis suite of human stem cell-based products and services enable scientists to build more mature and predictive human iPSC-derived tissueswith a focus on cardiac, musculoskeletal, and neuromuscular modelsfor the discovery, safety testing, and efficacy testing of new drugs in development. By offering drug developers an integrated preclinical platform comprising highly predictive human stem cell models to generate clinically-relevant data, Curi is closing the gap between preclinical data and human results, accelerating the discovery and development of safer, more effective medicines.

For more information, please visit http://www.curibio.com.

Read the original:
Curi Bio Launches Mantarray Platform 3D Engineered Muscle Tissues for Discovery of New Therapeutics - Business Wire

Archives