Page 1,379«..1020..1,3781,3791,3801,381..1,3901,400..»

Massachusetts Stem Cells | Stem Cell TV

Posted: September 10, 2019 at 7:43 pm

James F. Battey, Jr., MD, PhD; Laura K. Cole, PhD; and Charles A. Goldthwaite, Jr., PhD.

Stem cells are distinguished from other cells by two characteristics: (1) they can divide to produce copies of themselves (self-renewal) under appropriate conditions and (2) they are pluripotent, or able to differentiate into any of the three germ layers: the endoderm (which forms the lungs, gastrointestinal tract, and interior lining of the stomach), mesoderm (which forms the bones, muscles, blood, and urogenital tract), and ectoderm (which forms the epidermal tissues and nervous system). Pluripotent cells, which can differentiate into any mature cell type, are distinct from multipotent cells (such as hematopoietic, or blood-forming, cells) that can differ into a limited number of mature cell types. Because of their pluripotency and capacity for self-renewal, stem cells hold great potential to renew tissues that have been damaged by conditions such as type 1 diabetes, Parkinson's disease, heart attacks, and spinal cord injury. Although techniques to transplant multipotent or pluripotent cells are being developed for many specific applications, some procedures are sufficiently mature to be established options for care. For example, human hematopoietic cells from the umbilical cord and bone marrow are currently being used to treat patients with disorders that require replacement of cells made by the bone marrow, including Fanconi's anemia and chemotherapy-induced bone marrow failure after cancer treatment.

However, differentiation is influenced by numerous factors, and investigators are just beginning to understand the fundamental properties of human pluripotent cells. Researchers are gradually learning how to direct these cells to differentiate into specialized cell types and to use them for research, drug discovery, and transplantation therapy (see Figure 8.1). However, before stem cell derivatives are suitable for clinical application, scientists require a more complete understanding of the molecular mechanisms that drive pluripotent cells into differentiated cells. Scientists will need to pilot experimental transplantation therapies in animal model systems to assess the safety and long-term stable functioning of transplanted cells. In particular, they must be certain that any transplanted cells do not continue to self-renew in an unregulated fashion after transplantation, which may result in a teratoma, or stem cell tumor. In addition, scientists must ascertain that cells transplanted into a patient are not recognized as foreign by the patient's immune system and rejected.

Figure 8.1. The Scientific Challenge of Human Stem CellsThe state of the science currently lies in the development of fundamental knowledge of the properties of human pluripotent cells. The scientific capacity needs to be built, an understanding of the molecular mechanisms that drive cell specialization needs to be advanced, the nature and regulation of interaction between host and transplanted cells needs to be explored and understood, cell division needs to be understood and regulated, and the long-term stability of the function in transplanted cells needs to be established.

Stem cells derived from an early-stage human blastocyst (an embryo fertilized in vitro and grown approximately five days in culture) have the capacity to renew indefinitely, and can theoretically provide an unlimited supply of cells. It is also possible to derive stem cells from non-embryonic tissues, including amniotic fluid, placenta, umbilical cord, brain, gut, bone marrow, and liver. These stem cells are sometimes called "adult" stem cells, and they are typically rare in the tissue of origin. For example, blood-forming (hematopoietic) stem cell experts estimate that only 1 in 2000 to fewer than 1 in 10,000 cells found in the bone marrow is actually a stem cell.1 Because so-called "adult" stem cells include cells from the placenta and other early stages of development, they are more correctly termed "non-embryonic stem cells." Non-embryonic stem cells are more limited in their capacity to self renew in the laboratory, making it more difficult to generate a large number of stem cells for a specific experimental or therapeutic application. Under normal conditions, non-embryonic stem cells serve as a repair pool for the body, so they typically differentiate only into the cell types found in the organ of origin. Moreover, there is little compelling evidence for trans-differentiation, whereby a stem cell from one organ differentiates into a mature cell type of a different organ. New discoveries may overcome these limitations of stem cells derived from non-embryonic sources, and research directed toward this goal is currently underway in a number of laboratories.

Cultures of human pluripotent, self-renewing cells enable researchers to understand the molecular mechanisms that regulate differentiation (see Figure 8.2), including epigenetic changes (traits that may be inherited that do not arise from changes in the DNA sequence) in the chromatin structure, developmental changes in gene expression, exposure to growth factors, and interactions between adjacent cells. Understanding these basic mechanisms may enable future scientists to mobilize and differentiate endogenous populations of pluripotent cells to replace a cell type ravaged by injury or disease. Alternatively, scientists may some day be able to coax human pluripotent cells grown in the laboratory to become a specific type of specialized cell, which physicians could subsequently transplant into a patient to replace cells damaged by these same disease processes.

Scientists are gradually learning to direct the differentiation of pluripotent cell cultures into a specific type of cell, which can then be used as cellular models of human disease for drug discovery or toxicity studies. While it is not possible to predict the myriad ways that a basic understanding of stem cell differentiation may lead to new approaches for treating patients with cellular degenerative diseases, some avenues can be theorized. For example, in the case of Huntington's disease, a fatal neurodegenerative disorder, one could imagine that pluripotent cells derived from an embryo that carries Huntington's disease and differentiated into neurons in culture could be used to test drugs to delay or prevent degeneration.

Despite the incredible growth in knowledge that has occurred in stem cell research within the last couple of decades, investigators are just beginning to unravel the process of differentiation. Human pluripotent cell lines are an essential tool to understand this process and to facilitate the ultimate use of these cells in the clinic. To provide background on this fundamental topic, this article reviews the various potential sources and approaches that have been used to generate human pluripotent and multipotent cell lines, both of embryonic and non-embryonic origin.

Currently, at least six embryonic sources have been used to establish human pluripotent stem cell lines. All approaches involve isolation of viable cells during an early phase of development, followed by growth of these cells in appropriate culture medium. The various sources of these initial cell populations are discussed in brief below. It should be noted that the manipulation and use of embryonic tissues has raised a number of ethical issues.2,3 This article focuses on the scientific and technical issues associated with creating pluripotent cells, with the understanding that some of these techniques are currently subject to debates that extend beyond discussions of their scientific merits.

Figure 8.2. The Promise of Stem Cell ResearchStem cell research provides a useful tool for unraveling the molecular mechanisms that determine the differentiation fate of a pluripotent cell and for understanding the gene expression properties and epigenetic modifications essential to maintain the pluripotent state. In the future, this knowledge may be used to generate cells for transplantation therapies, whereby a specific cell population compromised by disease is replaced with new, functional cells. Differentiated derivatives of human pluripotent cells may also prove to be useful as models for understanding the biology of disease and developing new drugs, particularly when there is no animal model for the disease being studied. The greatest promise of stem cell research may lie in an area not yet imagined.

2008 Terese Winslow

Drawing upon twenty years of communal expertise with mouse ES cells,4 and on human inner cell mass culture conditions developed by Ariff Bongso and colleagues,5 James Thomson and colleagues at the University of Wisconsin generated the first hESC lines in 1998 using tissue from embryos fertilized in vitro.6 This method uses embryos generated for in vitro fertilization (IVF) that are no longer needed for reproductive purposes. During IVF, medical professionals usually produce more embryos than a couple attempting to start a family may need. Spare embryos are typically stored in a freezer to support possible future attempts for additional children if desired. It is estimated that there are approximately 400,000 such spare embryos worldwide.6 If these embryos are never used by the couple, they either remain in storage or are discarded as medical waste. Alternatively, these embryos can potentially be used to generate a hESC line.

To generate a hESC line, scientists begin with a donated blastocyst-stage embryo, at approximately five days after IVF (see Figure 8.3a). The blastocyst consists of approximately 150200 cells that form a hollow sphere of cells, the outer layer of which is called the trophectoderm. During normal development, the trophoblast becomes the placenta and umbilical cord. At one pole of this hollow sphere, 3050 cells form a cluster that is called the inner cell mass (ICM), which would give rise to the developing fetus. ICM cells are pluripotent, possessing the capacity to become any of the several hundred specialized cell types found in a developed human, with the exception of the placenta and umbilical cord.

Scientists remove the ICM from the donated blastocyst and place these cells into a specialized culture medium. In approximately one in five attempts, a hESC line begins to grow. Stem cells grown in such a manner can then be directed to differentiate into various lineages, including neural precursor cells,8 cardiomyocytes,9 and hematopoietic (blood forming) precursor cells.10

However, hESC lines are extremely difficult to grow in culture; the cells require highly specialized growth media that contain essential ingredients that are difficult to standardize. Yet the culture conditions are critical to maintain the cells' self-renewing and pluripotent properties. Culture requires the support of mouse or human cells, either directly as a "feeder" cell layer6,11,12 or indirectly as a source of conditioned medium in feeder-free culture systems.13 The feeder cells secrete important nutrients and otherwise support stem cell growth, but are treated so they cannot divide. Although the complete role of these feeder cells is not known, they promote stem cell growth, including detoxifying the culture medium and secreting proteins that participate in cell growth.14 hESC lines used to produce human cells for transplantation therapies may need to be propagated on a human feeder cell layer to reduce the risk of contamination by murine viruses or other proteins that may cause rejection. Thus, hESC lines often grow only under highly specific culture conditions, and the identification of ideal growth conditions presents a challenge regardless of the source of the hESCs.

Furthermore, human ES cell cultures must be expanded using an exacting protocol to avoid cell death and to control spontaneous differentiation. Since a limited number of laboratories in the United States are growing these cells, there is a shortage of people well-versed in the art and science of successful hESC culture. In the short term, challenges of working with these cells include developing robust culture conditions and protocols, understanding the molecular mechanisms that direct differentiation into specific cell types, and developing the infrastructure to advance this scientific opportunity. Once these challenges have been met, scientists will need to conduct transplantation studies in animal models (rodent and non-human primates) to demonstrate safety, effectiveness, and long-term benefit before stem cell therapies may enter clinical trials.

A second method for generating human pluripotent stem cell lines was published in 1998 by John Gearhart and coworkers at The Johns Hopkins Medical School.15

These researchers isolated specialized cells known as primordial germ cells (PGCs) from a 57-week-old embryo and placed these cells into culture (see Figure 8.3b). PGCs are destined to become either oocytes or sperm cells, depending on the sex of the developing embryo. The resulting cell lines are called embryonic germ cell lines, and they share many properties with ES cells. As with ES cells, however, PGCs present challenges with sustained growth in culture.16,17 Spontaneous differentiation, which hinders the isolation of pure clonal lines, is a particular issue. Therefore, the clinical application of these cells requires a more complete understanding of their derivation and maintenance in vitro.

Embryos that stop dividing after being fertilized in vitro are not preferentially selected for implantation in a woman undergoing fertility treatment. These embryos are typically either frozen for future use or discarded as medical waste. In 2006, scientists at the University of Newcastle, United Kingdom, generated hESC lines from IVF embryos that had stopped dividing.18 These scientists used similar methods as described under "Traditional hESC Line Generation" except that their source material was so-called "dead" IVF embryos (see Figure 8.3c). The human stem cells created using this technique behaved like pluripotent stem cells, including producing proteins critical for "stemness" and being able to produce cells from all three germ layers. It has been proposed that an IVF embryo can be considered dead when it ceases to divide.19 If one accepts this definition, such an embryo that "dies" from natural causes presumably cannot develop into a human being, thereby providing a source to derive human ES cells without destroying a living embryo.

Figure 8.3. Alternative Methods for Preparing Pluripotent Stem Cells

2008 Terese Winslow

Couples who have learned that they carry a genetic disorder sometimes use pre-implantation genetic diagnosis (PGD) and IVF to have a child that does not carry the disorder. PGD requires scientists to remove one cell from a very early IVF human embryo and test it for diseases known to be carried by the hopeful couple. Normally, embryos identified with genetic disorders are discarded as medical waste. However, Dr.Yuri Verlinsky and colleagues have capitalized on these embryos as a way to further our understanding of the diseases they carry (see Figure 8.3d) by deriving hESC lines from them.20 These stem cell lines can then be used to help scientists understand genetically-based disorders such as muscular dystrophy, Huntington's disease, thalessemia, Fanconi's anemia, Marfan syndrome, adrenoleukodystrophy, and neurofibromatosis.

In 2006, Dr. Robert Lanza and colleagues demonstrated that it is possible to remove a single cell from a pre-implantation mouse embryo and generate a mouse ES cell line.21 This work was based upon their experience with cleavage-stage mouse embryos. Later that same year, Dr. Lanza's laboratory reported that it had successfully established hESC lines (see Figure 8.3e) from single cells taken from pre-implantation human embryos.22 The human stem cells created using this technique behaved like pluripotent stem cells, including making proteins critical for "stemness" and producing cells from all three germ layers. Proponents of this technique suggest that since it requires only one embryonic cell, the remaining cells may yet be implanted in the womb and develop into a human being. Therefore, scientists could potentially derive human embryonic stem cells without having to destroy an embryo. However, ethical considerations make it uncertain whether scientists will ever test if the cells remaining after removal of a single cell can develop into a human being, at least in embryos that are not at risk for carrying a genetic disorder. Moreover, it is unclear whether the single cell used to generate a pluripotent stem cell line has the capacity to become a human being.

Parthenogenesis is the creation of an embryo without fertilizing the egg with a sperm, thus omitting the sperm's genetic contributions. To achieve this feat, scientists "trick" the egg into believing it is fertilized, so that it will begin to divide and form a blastocyst (see Figure 8.3f). In 2007, Dr. E.S. Revazova and colleagues reported that they successfully used parthenogenesis to derive hESCs.23 These stem cell lines, derived and grown using a human feeder cell layer, retained the genetic information of the egg donor and demonstrated characteristics of pluripotency. This technique may lead to the ability to generate tissue-matched cells for transplantation to treat women who are willing to provide their own egg cells.24 It also offers an alternate method for deriving tissue-matched hESCs that does not require destruction of a fertilized embryo.

Amniotic fluid surrounding the developing fetus contains cells shed by the fetus and is regularly collected from pregnant women during amniocentesis. In 2003, researchers identified a subset of cells in amniotic fluid that express Oct-4, a marker for pluripotent human stem cells that is expressed in ES cells and embryonic germ cells.25 Since then, investigators have shown that amniotic fluid stem cells can differentiate into cells of all three embryonic germ layers and that these cells do not form tumors in vivo.26,27

For example, Anthony Atala and colleagues at the Wake Forest University have recently generated non-embryonic stem cell lines from cells found in human and rat amniotic fluid.27 They named these cells amniotic fluid-derived stem cells (AFS). Experiments demonstrate that AFS can produce cells that originate from each of the three embryonic germ layers, and the self-renewing cells maintained the normal number of chromosomes after a prolonged period in culture. However, undifferentiated AFS did not produce all of the proteins expected of pluripotent cells, and they were not capable of forming a teratoma. The scientists developed in vitro conditions that enabled AFS to produce nerve cells, liver cells, and bone-forming cells. AFS-derived human nerve cells could make proteins typical of specialized nerve cells and were able to integrate into a mouse brain and survive for at least two months. Cultured AFS-derived human liver cells secreted urea and made proteins characteristic of normal human liver cells. Cultured AFS-derived human bone cells made proteins expected of human bone cells and formed bone in mice when seeded onto scaffolds and implanted under the mouse's skin. Although scientists do not yet know how many different cell types AFS can generate, AFS may one day allow researchers to establish a bank of cells for transplantation into humans.

An alternative to searching for an existing population of stem cells is to create a new one from a population of non-pluripotent cells. This strategy, which may or may not involve the creation of an embryo, is known as "reprogramming." This section will summarize reprogramming approaches, including several recent breakthroughs in the field..

In SCNT (see Figure 8.3g), human oocytes (eggs) are collected from a volunteer donor who has taken drugs that stimulate the production of more than one oocyte during the menstrual cycle. Scientists then remove the nucleus from the donated oocyte and replace it with the nucleus from a somatic cell, a differentiated adult cell from elsewhere in the body. The oocyte with the newly-transferred nucleus is then stimulated to develop. The oocyte may develop only if the transplanted nucleus is returned to the pluripotent state by factors present in the oocyte cytoplasm. This alteration in the state of the mature nucleus is called nuclear reprogramming. When development progresses to the blastocyst stage, the ICM is removed and placed into culture in an attempt to establish a pluripotent stem cell line. To date, the technique has been successfully demonstrated in two primates: macaque monkeys28 and humans.29

However, successful SCNT creates an embryo-like entity, thereby raising the ethical issues that confront the use of spare IVF embryos. However, pluripotent cell lines created by embryos generated by SCNT offer several advantages over ES cells. First, the nuclear genes of such a pluripotent cell line will be identical to the genes in the donor nucleus. If the nucleus comes from a cell that carries a mutation underlying a human genetic disease such as Huntington's disease, then all cells derived from the pluripotent cell line will carry this mutation. In this case, the SCNT procedure would enable the development of cellular models of human genetic disease that can inform our understanding of the biology of disease and facilitate development of drugs to slow or halt disease progression. Alternatively, if the cell providing the donor nucleus comes from a specific patient, all cells derived from the resulting pluripotent cell line will be genetically matched to the patient with respect to the nuclear genome. If these cells were used in transplantation therapy, the likelihood that the patient's immune system would recognize the transplanted cells as foreign and initiate tissue rejection would be reduced. However, because mitochondria also contain DNA, the donor oocyte will be the source of the mitochondrial genome, which is likely to carry mitochondrial gene differences from the patient which may still lead to tissue rejection.

A technique reported in 2007 by Dr. Kevin Eggan and colleagues at Harvard University may expand scientists' options when trying to "reprogram" an adult cell's DNA30. Previously, successful SCNT relied upon the use of an unfertilized egg. Now, the Harvard scientists have demonstrated that by using a drug to stop cell division in a fertilized mouse egg (zygote) during mitosis, they can successfully reprogram an adult mouse skin cell by taking advantage of the "reprogramming factors" that are active in the zygote at mitosis. They removed the chromosomes from the single-celled zygote's nucleus and replaced them with the adult donor cell's chromosomes (see Figure 8.3h). The active reprogramming factors present in the zygote turned genes on and off in the adult donor chromosomes, to make them behave like the chromosomes of a normally fertilized zygote. After the zygote was stimulated to divide, the cloned mouse embryo developed to the blastocyst stage, and the scientists were able to harvest embryonic stem cells from the resulting blastocyst. When the scientists applied their new method to abnormal mouse zygotes, they succeeded at reprogramming adult mouse skin cells and harvesting stem cells. If this technique can be repeated with abnormal human zygotes created in excess after IVF procedures, scientists could use them for research instead of discarding them as medical waste.

Altered nuclear transfer is a variation on standard SCNT that proposes to create patient-specific stem cells without destroying an embryo. In ANT, scientists turn off a gene needed for implantation in the uterus (Cdx2) in the patient cell nucleus before it is transferred into the donor egg (see Figure 8.3i). In 2006, Dr. Rudolph Jaenisch and colleagues at MIT demonstrated that ANT can be carried out in mice.31 Mouse ANT entities whose Cdx2 gene is switched off are unable to implant in the uterus and do not survive to birth. Although ANT has been used to create viable stem cell lines capable of producing almost all cell types, the authors point out that this technique must still be tested with monkey and human embryos. Moreover, the manipulation needed to control Cdx2 expression introduces another logistical hurdle that may complicate the use of ANT to derive embryonic stem cells. Proponents of ANT, such as William Hurlbut of the Stanford University Medical Center, suggest that the entity created by ANT is not a true embryo because it cannot implant in the uterus.32, 33 However, the technique is highly controversial, and its ethical implications remain a source of current debate.4,32

In 2005, Kevin Eggan and colleagues at Harvard University reported that they had fused cultured adult human skin cells with hESCs (see Figure 8.3j).36 The resulting "hybrid" cells featured many characteristics of hESCs, including a similar manner of growth and division and the manufacture of proteins typically produced by hESCs. Some factor(s) within the hESCs enabled them to "reprogram" the adult skin cells to behave as hESCs. However, these cells raised a significant technical barrier to clinical use. Because fused cells are tetraploid (they contain four copies of the cellular DNA rather than the normal two copies), scientists would need to develop a method to remove the extra DNA without eliminating their hESC-like properties. The fusion method serves as a useful model system for studying how stem cells "reprogram" adult cells to have properties of pluripotent cells. However, if the reprogramming technique could be carried out without the fusion strategy, a powerful avenue for creating patient-specific stem cells without using human eggs could be developed.

In 2007, two independent research groups published manuscripts that described successful genetic reprogramming of human adult somatic cells into pluripotent human stem cells.34,35 Although some technical limitations remain, this strategy suggests a promising new avenue for generating pluripotent cell lines that can inform drug development, models of disease, and ultimately, transplantation medicine. These experiments, which are discussed below, were breakthroughs because they used adult somatic cells to create pluripotent stem cells that featured hallmarks of ES cells.

In 2006, Shinya Yamanaka and colleagues at Kyoto University reported that they could use a retroviral expression vector to introduce four important stem cell factors into adult mouse cells and reprogram them to behave like ES cells (see Figure 8.3k).37 They called the reprogrammed cells "iPSCs," for induced pluripotent stem cells. However, iPSCs produced using the original technique failed to produce sperm and egg cells when injected into an early mouse blastocyst and did not make certain critical DNA changes. These researchers then modified the technique to select for iPSCs that can produce sperm and eggs,38 results that have since been reproduced by Rudolph Jaenisch and colleagues at the Massachusetts Institute of Technology (MIT).39

In addition, the MIT scientists determined that iPSCs DNA is modified in a manner similar to ES cells, and important stem cell genes are expressed at similar levels. They also demonstrated that iPSCs injected into an early mouse blastocyst can produce all cell types within the developing embryo, and such embryos can complete gestation and are born alive.

Once these research advances were made in mice, they suggested that similar techniques might be used to reprogram adult human cells. In 2007, Yamanaka and coworkers reported that introducing the same four genetic factors that reprogrammed the mouse cells into adult human dermal fibroblasts reprogrammed the cells into human iPSCs.35 These iPSCs were similar to human ES cells in numerous ways, including morphology, proliferative capacity, expression of cell surface antigens, and gene expression. Moreover, the cells could differentiate into cell types from the three embryonic germ layers both in vitro and in teratoma assays. Concurrent with the Yamanaka report, James Thomson and coworkers at the University of Wisconsin published a separate manuscript that detailed the creation of human iPSCs through somatic cell reprogramming using four genetic factors (two of which were in common with the Yamanaka report).34 The cells generated by the Thomson group met all defining criteria for ES cells, with the exception that they were not derived from embryos.

These breakthroughs have spurred interest in the field of iPSCs research. In early 2008, investigators at the Massachusetts General Hospital40 and the University of California, Los Angeles41 reported generating reprogrammed cells. As scientists explore the mechanisms that govern reprogramming, it is anticipated that more reports will be forthcoming in this emerging area. Although these reprogramming methods require the use of a virus, non-viral strategies may also be possible in the future. In any case, these approaches have created powerful new tools to enable the "dedifferentation" of cells that scientists had previously believed to be terminally differentiated.42,43

Although further study is warranted to determine if iPS and ES cells differ in clinically significant ways, these breakthrough reports suggest that reprogramming is a promising strategy for future clinical applications. Induced pluripotent cells offer the obvious advantage that they are not derived from embryonic tissues, thereby circumventing the ethical issues that surround use of these materials. Successful reprogramming of adult somatic cells could also lead to the development of stem cell lines from patients who suffer from genetically-based diseases, such as Huntington's Disease, spinal muscular atrophy, muscular dystrophy, and thalessemia. These lines would be invaluable research tools to understand the mechanisms of these diseases and to test potential drug treatments. Additionally, reprogrammed cells could potentially be used to repair damaged tissues; patient-specific cell lines could greatly reduce the concerns of immune rejection that are prevalent with many transplantation strategies.

However, several technical hurdles must be overcome before iPSCs can be used in humans. For example, in preliminary experiments with mice, the virus used to introduce the stem cell factors sometimes caused cancers.37 The viral vectors used in these experiments will have to be selected carefully and tested fully to verify that they do not integrate into the genome, thereby harboring the potential to introduce genetic mutations at their site of insertion. This represents a significant concern that must be addressed before the technique can lead to useful treatments for humans. However, this strategy identifies a method for creating pluripotent stem cells that, together with studies of other types of pluripotent stem cells, will help researchers learn how to reprogram cells to repair damaged tissues in the human body.

Stem cell research is a rapidly evolving field, and researchers continue to isolate new pluripotent cells and create additional cell lines. This section briefly reviews other sources of pluripotent cells and the implications that their discovery may have on future research.

Epiblast Cells. While rodent and human ES cells are pluripotent, they maintain their respective pluripotencies through different molecular signaling pathways. It is not known why these differences exist. Recently, several research groups have reported the generation of stable, pluripotent cell lines from mouse and rat epiblast, a tissue of the post-implantation embryo that ultimately generates the embryo proper.44,45 These cells are distinct from mouse ES cells in terms of the signals that control their differentiation. However, the cells share patterns of gene expression and signaling responses with human ES cells. The establishment of epiblast cell lines can therefore provide insight into the distinctions between pluripotent cells from different species and illuminate ways that pluripotent cells pursue distinct fates during early development.

Existing Adult Stem Cells. As has been discussed in other chapters, numerous types of precursor cells have been isolated in adult tissues.46 Although these cells tend to be relatively rare and are dispersed throughout the tissues, they hold great potential for clinical application and tissue engineering. For example, tissues created using stem cells harvested from an adult patient could theoretically be used clinically in that patient without engendering an immune response. Moreover, the use of adult stem cells avoids the ethical concerns associated with the use of ES cells. In addition, adult-derived stem cells do not spontaneously differentiate as do ES cells, thus eliminating the formation of teratomas often seen with implantation of ES cells. The potential of adult stem cells for regenerative medicine is great; it is likely that these various cells will find clinical application in the upcoming decades.

Although the recent advances in reprogramming of adult somatic cells has generated a wave of interest in the scientific community, these cell lines will not likely replace hESC lines as tools for research and discovery. Rather, both categories of cells will find unique uses in the study of stem cell biology and the development and evaluation of therapeutic strategies. Pluripotent cells offer a number of potential clinical applications, especially for diseases with a genetic basis. However, researchers are just beginning to unlock the many factors that govern the cells' growth and differentiation. As scientists make strides toward understanding how these cells can be manipulated, additional applications, approaches, and techniques will likely emerge. As such, pluripotent cells will play a pivotal role in future research into the biology of development and the treatment of disease.

Chapter7|Table of Contents|Chapter9

Excerpt from:Alternate Methods for Preparing Pluripotent Stem Cells ...

See more here:
Massachusetts Stem Cells | Stem Cell TV

Posted in Massachusetts Stem Cells | Comments Off on Massachusetts Stem Cells | Stem Cell TV

Stem Cell Therapy | Las Vegas, NV

Posted: September 10, 2019 at 7:43 pm

As an interventional pain management specialist with over 25 years of experience, Dr. Crispino Santos is a leader of medical innovations that provide longer lasting solutions for chronic pain and specialized treatments to help reduce the problems associated with injuries and degenerative diseases through Stem Cell Therapy in Las Vegas, NV.

Dr. Santos is the President and medical director of both the Regenerative Cell Institute (RCI) and Interventional Pain Medicine (IPM). He attended medical school at the Far Eastern University in Manila, Philippines, and did his post-doctoral residency training in general surgery and anesthesiology at King Drew Medical Center in Los Angeles, California. He also completed one year of advanced anesthesiology fellowship at University of South Florida-Tampa General Hospital in Tampa, Florida. He graduated and is a recipient of Winthrop Stearns Scholar and Rolando Solis Cardiology Award in 1979.

Dr. Santos first practiced medicine in Mason City, Iowa where he worked for nine years and started the cardiac anesthesia program and established the Pain Management Center at St. Joseph Mercy Hospital.In February 1998, Dr. Santos moved to Las Vegas, Nevada and joined Nevada Orthopedic and Spine Center. There, he began his full-time practice as an interventional pain physician. He founded Regenerative Cell Institute of Las Vegas, Nevada in 2013 and opened the Institutes second location in Mason City, Iowa in 2014. In February 2017, he extended his practice to his native home, Philippines, where he performs procedures at the Panay Healthcare Hospital in Kalibo, Aklan Province, Philippines.

Dr. Santos is board-certified in both anesthesiology and pain medicine by the American Board of Anesthesiology, the American Board of Pain Medicine, and the American Board of Pain Management. He is also board certified by the American Board of Interventional Pain Physicians and a Fellow of Interventional Pain Practice. Additionally, he is an active member of the World Institute of Pain, the International Spine Intervention Society, the American Society of Interventional Pain Physicians, the International Neuromodulation Society, the American Academy of Pain Medicine, the American Association of Pain Management in Ultrasound, the American Society of Cosmetic Physicians, the American Academy of Anti-Aging Medicine, the International Society of Plastic and Regenerative Surgeons, and the American Association of Orthopedic Medicine.

He specializes in minimally invasive interventional procedures for the treatment of spinal pain and other complex chronic pain conditions. Dr. Santos unparalleled expertise in minimally invasive injection techniques, utilizing live image guidance, allows him to administer the biologic solutions are placed precisely in the areas of tissue damage. Using the latest stem cell research and cutting-edge regenerative medical techniques, he performs the most advanced regenerative cell treatments for an array of medical conditions including arthritis, joint injuries, spinal pain, chronic pain conditions, neurological conditions, auto-immune diseases, and aesthetics. He has performed several thousands of interventional pain procedures, have trained other doctors, given lectures, and treated patients locally and internationally.

He is always researching the most advanced, safe, and effective therapies to improve and prolong patients lives using the most natural way of healing.

About Regenerative Cell Institute

With locations in Las Vegas, Nevada, and Mason City, Iowa, the Regenerative Cell Institute (RCI) has changed the practice of traditional medicine by revolutionizing pain management and regenerative medical treatments to reduce symptoms associated with the diseases of aging. Combining cutting-edge regenerative medical techniques utilizing stem cells and high-density activated platelet-rich plasma (PRP) with several stem cell procedures, Dr. Santos and his team offer the most advanced regenerative cell treatments for an array of medical conditions including arthritis, joint injuries, spinal pain, chronic pain conditions, neurological conditions, auto-immune diseases, sexual wellness, and aesthetics.

Dr. Santos splits his time between both offices, performs all procedures, and offers customized treatment options and care plans. He carefully evaluates each patients condition and body composition to determine the best stem cell extraction technique, harvesting process, and treatment plan to ensure patients receive the highest concentrations of stem cells to spark the bodys natural healing and rejuvenation processes. While other doctors often require multiple appointments and procedures which drives up patient costs, Dr. Santos patients enjoy a 90% success rate after one treatment*; 10-20% of patients require a booster dose** to achieve optimal results.

Why Choose RCI?

We provide the highest quality patient care available with over 90% satisfaction rate.

View post:
Stem Cell Therapy | Las Vegas, NV

Posted in Nevada Stem Cells | Comments Off on Stem Cell Therapy | Las Vegas, NV

Regenerative Therapy in Northeast Ohio | Ohio Therapy Centers

Posted: September 9, 2019 at 4:48 am

The video above is Dr. Nick Fabian from our Elyria location talking about Regenerative Therapyduring a Facebook live segment with Jenny from the Block on Fox 8 Cleveland.

Unlike other cells, stem cells are unspecialized or undifferentiated in our bodies that have the capacity to change into any healthy cell in our body. Meaning they can change into skin, bone, heart, and muscle cells to name a few. They have the unique ability to divide or differentiate into many types of cells with specific functions such as muscle, skin or bone cells.

Stem cells can also give rise to new generations of undifferentiated stem cells, thus renewing themselves. Stem cells are located throughout our body in almost every organ and tissue such as bone marrow, fat, teeth, muscles, etc.

While cortisone and other drugs only provide temporary pain relief, Regenerative Therapy actually restores degenerated tissue while providing pain relief. Additionally, the injections contain collagen, proteins and hyaluronic acid, which acts as a lubricant on worn and damaged joints while encouraging new, healthy cartilage tissue growth.

Some people will feel immediate relief from their pain and will notice continued improvements in pain reduction, mobility, and range of motion following the treatment. Most results are seen within one to three months after injection.

The wonderful thing about Regenerative Therapy is that its being found to be a safer and more effective pain relief treatment than addicting prescription medications and surgeries that require weeks and sometimes months downtime from your active life.

However, this therapy doesnt just put a Band-Aid on the problem and walk away; it encourages your own body to start healing. The end result is reduced or eliminatedpain, healthier joint tissue, increased mobility, and the ability to once again engage in all of your favorite activities, allowing you to live a vibrant, healthy, and pain-free life!

View original post here:
Regenerative Therapy in Northeast Ohio | Ohio Therapy Centers

Posted in Ohio Stem Cells | Comments Off on Regenerative Therapy in Northeast Ohio | Ohio Therapy Centers

Stem Cell Procedures | Motus Biologics of Kansas City

Posted: September 9, 2019 at 4:47 am

key benefits ofstem cell treatment

NOBEL PRIZE IN PHYSIOLOGY & MEDICINE

The Nobel Prize in physiology and medicine was awarded to Drs. Gurdon and Yamanaka in 2012 for achieving the regress mature cells of living human beings into these pluripotent cells. These cells are known as induced pluripotent stem cells (iPSC) and are in their infancy as part of the landscape of orthopedic biologics.

Combining Treatment Elements

In addition to the mesenchymal and hematopoietic stem cells, the concentrate includes similar growth factors found in PRP as well as substances known as cytokines, which recruit more healing cells to the site. The concentrate is injected into the affected site under ultrasound-guidance to ensure accurate placement.

Common Uses

Bone marrow and fat aspirate are commonly used in degenerative conditions such as osteoarthritis and cartilage defects. A retrospective study performed in 2014 by Centeno et al found significant improvements in pain and function at all follow-up intervals after treatment of 840 knees with osteoarthritis using bone marrow aspirate with fat grafting.

Same Day In-Office Procedure

Eliminating the risks or costs associated with anesthesia. Procedures are conveniently performed in a comfortable office setting. PRP procedures take less time in this setting.

Less Cost & Risk

Total knee replacement surgery costs between $56,000 and $58,000. Not including pre-operative costs for office visits,xrays, laboratory work, or the post-surgical costs for physical therapy which can average $2,500 to $4,500. This treatment is conservative, minimally invasive, able to be performed in patients with comorbid conditions.

See more here:
Stem Cell Procedures | Motus Biologics of Kansas City

Posted in Kansas Stem Cells | Comments Off on Stem Cell Procedures | Motus Biologics of Kansas City

"M*A*S*H" Preventative Medicine (TV Episode 1979) – IMDb

Posted: September 7, 2019 at 4:35 pm

Edit Storyline

BJ is appalled when his patient has entry wounds entering his body from 3 different directions. Col. Potter reminds him of Lt. Col. Lacy, 163rd Combat Infantry, the CO with the highest casualty rate of any single battalion in the sector. Apparently, Lacy refused to obey an order to retreat and subjected his men to hell. Poor Klinger: he has tried chicanery, malingering and endless flim-flammery, but now, Klinger is pulling out the heavy artillery, voodoo, to get his Section 8. Lacy visits the Post Op and one of his own men, Corporal North, turns away. Margaret is intrigued with the virile Lacy until she lunches with him and Lacy tells Margaret of his latest plan to take Hill 403. His plan is based on a plan used in the WWII Battle of Monte Casino...and it has a 20-30% casualty rate. Margaret understands this translates to 100 men and she leaves the table, sick. BJ and Hawkeye despise Lacy and his hypocracy; he thrives on his war games. Potter writes an unprecedented letter to I Corp ... Written byLA-Lawyer

Certificate: TV-PG

Runtime: 24 min

Aspect Ratio: 1.33 : 1

See the rest here:
"M*A*S*H" Preventative Medicine (TV Episode 1979) - IMDb

Posted in Preventative Medicine | Comments Off on "M*A*S*H" Preventative Medicine (TV Episode 1979) – IMDb

University of Pennsylvania || Cell and Molecular Biology …

Posted: September 7, 2019 at 4:35 pm

Overview|Leadership|Research |Communication & Outreach| Academic Requirements

Program OverviewStudents within DSRB address key questions in Developmental biology, Stem cell/niche interactions, and Regenerative Biology. The thread that unites these areas is the compelling drive to understand how tissues are first formed, how they are maintained, and how they can be repaired. The primary mission of DSRB faculty is to train students in hypothesis-driven research at the forefront of these areas.

Individualized Training:The curriculum in DSRB is set up to provide students with a strong foundation in fundamental developmental biology, as well as in stem cell and regenerative biology (see Academics). The curriculum is complemented by an Individualized Training Plan tailored to your background, to your interests, and to your career plans. You will develop this plan in consultation with faculty advisors, and take advantage of unique resources provided by the program, such as the extensive network of alumnae distributed among various post-PhD career paths.

Stunning Research Accomplishments:Evidence of the quality of research and the strength of the training plan is shown by the success rate among DSRB students who have applied for individual research training grants. Fully two-thirds of our students have been awarded NIH grants for their work (20/30; an unheard of 67% success rate). Those successes are spread across diverse areas of research within DSRB labs .

A Cohesive Program:We celebrate the diverse research within our community in our renowned Research-in-Progress Lunchtime series, where students present current work, with opportunity for discussion over good food. The series serves as one mechanism to polish communication skills and receive timely feedback, both on the science and on its presentation. Connections among our students are also reinforced by participation in the Developmental Biology Journal Club and the Student-invited Distinguished Lecturer Series. Our students support each other through a peer-mentoring program, and by running mock critique sessions to prepare for the oral Preliminary Exam and for fellowship proposals. Finally, students appointed to the Developmental Biology Training Grant (https://www.med.upenn.edu/dbtraininggrant/) are fully integrated into our program.

Continued Success as Your Career Progresses:Once students have received their PhD, their success continues as they leave our program and step onto their chosen path. Of the almost 50 DSRB students who received their PhD over the last 10 years, 90% went directly into academic research, medicine, the pharmaceutical or biotech industries.

PhiladelphiaMany other major Academic and Medical Institutions are right here, across Philadelphia, including the Childrens Hospital of Philadelphia, Fox Chase Cancer Center, The Wistar Institute, Thomas Jefferson University and several others. The Greater Philadelphia area has always been home to various Biotech and Pharma institutions. But, over the last five years it is becoming a hub for Next Generation therapeutics:http://www.phillymag.com/business/2017/06/17/cellacon-valley-philadelphia-medical-technology/ All of this converges to provide wonderful opportunities as you navigate your career choices.

This all can happen for you while experiencing an historical and vibrant city, alive with the Performing Arts, Culture, Sports and rich in world-class Museums and unique Collections. Both the University and the City of Philadelphia are incredibly diverse and welcoming communities, with opportunities for all: http://www.upenn.edu/life-at-penn.

DSRB Faculty Leadership & Committee Structure

DSRB Executive Committee:Jonathan Raper, Professor of NeuroscienceMichael Granato, Professor of Cell & Developmental BiologySarah Millar, Professor of DermatologyMontserrat Anguera, Assistant Professor of Animal Biology, the Vet SchoolCamille Syrett, DSRB Student Representative

DSRB Admissions Committee:Greg Bashaw, Professor of NeurosciencePaul Gadue, Associate Professor, Pathology & Lab Medicine

DSRB First year Advisors:Mary Mullins, Professor of Cell & Developmental BiologyMeera Sundaram, Professor of Genetics

DSRB Second (Prelim) year advisors:Paul Gadue, Associate Professor, Pathology & Lab Medicine Judith Grinspan, Professor of NeurologyWenqin Luo, Assistant Professor of NeurosciencePatrick Seale, Associate Professor of Cell & Developmental Biology

DSRB Curriculum Committee:Dan Kessler, Professor of Cell & Developmental BiologyChris Lengner, Associate Professor of Animal BiologyMary Mullins, Professor of Cell & Developmental Biology

ResearchBreadth of Research in DSRBDSRB Students are exposed to interdisciplinary training in gametogenesis; embryonic and fetal development; nervous system development and its wiring; the genesis of tissues and organ systems as well as their homeostasis, metabolism and repair. Exciting work is unraveling the basic biology of these processes, as well as understanding the consequences of and working toward the amelioration of diseases affecting their function. Complementing these areas is work on Embryonic Stem Cells (ESCs) and Induced Pluripotent Stem Cells (iPSCs; patient-derived iPSCs), using genome-scale approaches to understand disease processes in vitro in order to develop cell replacement strategies for therapy. The proximity of the University of Pennsylvania Health System, a world-renowned academic medical center, provides unparalleled opportunity for translational research.

State-of-the-Art Tools at your disposalDSRB Students have at their disposal all the tools of the modern genomic era. For example, some students have been developing high-throughput and genomic-scale screens, using deep sequencing, ChIP-SEQ, RNAi tools or small molecule libraries, while others are capitalizing on classical genetic analyses or CRISPR/Cas9-based screens. Some have been utilizing high-resolution microscopy techniques, including live-imaging of cells, organs and tissues in vivo, while others have adapted novel biochemical, epigenetic and cell biological approaches to their questions. Some students are actively isolating and manipulating ESCs and iPSCs, as well as characterizing the nuclear reprogramming events required to coax these cells into distinct neuroepithelial, mesodermal or endodermal lineages, an essential step in taking these powerful, biomedically promising entities into the clinic. Facilitating this work is access to world-class cores, including those for Next Generation Sequencing, Proteomics, the Zebrafish facility, Stem cell & Xenografting, and Microscopy (http://www.med.upenn.edu/cores/).

Expansive Choice of Systems in which to workYou have the opportunity to choose among a wide variety of experimental model systems. This facilitates comparative and collaborative work, and can significantly speed your work. For example, you might begin attacking a question in a system that is highly adapted to a particular approach, and then can shift your work to another system more amenable to a different approach, or for addressing the follow-up part of the question. The systems used in DSRB labs include plants, such as Arabidopsis; the yeasts; several invertebrate organisms, such as the nematode worm, fruitfly and ant; various vertebrate models, such as zebra fish, the frog, chick, rat and mouse; and, of course, various pluripotent types of stem cells, both normal and patient-derived.

Links to the Faculty areas of research in DSRB:

GametogenesisEmbryo & Fetal developmentCell Migration, Polarity and Morphogenesis Neurogenesis and Axonogenesis Tissue & Organ FormationStem Cell/Niche Interactions Regenerative Biology Epigenetics, gene regulatory networks & signal integration

Communication & Outreach

Communicating your science:An important facet of your training is developing the ability to communicate effectively and interact productively with scientific peers. For this reason, the DSRB program uses several mechanisms to promote communication of your research. This includes the popular student research-in-progress lunches -- a very successful forum, fostering cohesion among the students in DSRB. Communication skills are also sharpened by opportunities to host student-invited seminar speakers; to give presentations at various symposia, such as the CAMB Graduate Symposium, various Departmental or Institute symposia, and, of course, at National and International meetings.

Outreach to the Next GenerationStudents trained in DSRB have an important responsibility to enhance scientific and technological understanding within our communities. We want our students to convey to diverse audiences the general excitement about DSRB research as well as of their own specific accomplishments. For these reasons, we support and encourage the participation in any of several acclaimed outreach activities. Opportunities range from a semester as a teaching assistant to participation in a nationally renowned outreach program, or mentoring an intern in a research project for a summer. An appropriate time for our graduate students to participate in outreach would be in the middle-to-out years of their work, when coursework is complete and a thesis project is solidly underway.

-- BioEYES: with over 12 years in area public schools, the program is run by Dr. Jamie Shuda, an experienced educator who develops university-community partnerships in science. She will provide training, help establish relationships with students and teachers, and serve as a mentor to help assess impact and effectiveness. Students are encouraged to adapt the existing programs to best communicate their research focus to the most appropriate audience. http://bioeyes.org/index.html

-- The Summer Undergraduate Internship Program (SUIP):http://www.med.upenn.edu/bgs/applicants_suip.shtmlRun successfully by Biomedical Graduate Studies for over 15 years, the program serves some 30 college interns each summer, and particularly so those from underrepresented minority groups or disadvantaged backgrounds, those with disabilities, or who attend small colleges. In this program you will directly oversee the interns research in collaboration with your PI, and coach them in their summers end presentation to the Leadership Alliance National Symposium.

-- Mentoring local high school students in STEM science fair projects (Science, Technology, Engineering & Math); Running our DSRB booth at the Philadelphia Science on the parkway fair.

Academics

Program course requirementsTypical course & research schedule for DSRB students:

Fall of first year:- Cell Biology (BIOM 600)- First year seminar (literature readings + discussions; CAMB 605)- Lab Rotation #1 (11 weeks)

Spring of first year: - Regulation of the Genome (BIOM 555)- Biological Data Analysis (BIOM 611) - Principles of Development (required for DSRB; CAMB 511) - Lab Rotations #2 & #3 (each lasting 11 weeks)

Summer between first & second year:- Research in prospective thesis lab

Fall of second year:- Required for DSRB: one of two concept-based, seminar offerings:

- Neurodevelopment, Regeneration & Repair Seminar (Bashaw & Luo; CAMB 597).The course integrates neural specification & development, with essential functions of neurons in guidance, synapse formation and function, along with neural regeneration & repair.

- Stem Cells (Gadue & Rompolas; CAMB 697) This course covers the challenges in stem cell & regeneration biology including reprogramming, embryonic and tissue-specific stem cells, tissue regeneration, and tissue engineering.

- One Elective: any graduate-level course selected based on research interest (can also be whichever of the above was not chosen to satisfy the DSRB requirement)

- Thesis lab Research

Spring of second year:

- Two Electives: selected based on research interest- Prelim prep writing course (CAMB 695)- Thesis lab Research

Elective choices:Students have tremendous flexibility, and in consultation with faculty advisors will select electives that fit their research interests and prospective directions. Suggested electives:

CAMB 534:Seminar on Current Genetic ResearchCAMB 542:Topics in Molecular MedicineCAMB 550:Genetic PrinciplesCAMB 597:Neural Development, Regeneration and RepairCAMB 608:Regulation of Eukaryotic Gene ExpressionCAMB 632:Cell Control by Signal Transduction PathwaysCAMB 691/CAMB 692:Advanced Topics in Cell Biology and PhysiologyCAMB 697:The Biology of Stem CellsCAMB 709: Quantitative Imaging and Analysis for Biologists (QIAB)GCB 535: Introduction to BioInformaticsGCB 536: Computational BiologyBIOL 446: Statistics for BiologistsEPID 575: Introduction to Genetic Epidemiology

[top of page]

See the rest here:
University of Pennsylvania || Cell and Molecular Biology ...

Posted in Pennsylvania Stem Cells | Comments Off on University of Pennsylvania || Cell and Molecular Biology …

Georgia Stem Cells | Stem Cell TV

Posted: September 7, 2019 at 4:35 pm

Theranostics 2017; 7(7):2067-2077. doi:10.7150/thno.19427

Review

Kiwon Ban1, Seongho Bae2, Young-sup Yoon2, 3

1. Department of Biomedical Sciences, City University of Hong Kong, Hong Kong;2. Department of Medicine, Division of Cardiology, Emory University, Atlanta, Georgia, USA;3. Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea.

This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions.

Cardiomyocytes (CMs) derived from human pluripotent stem cells (hPSCs) are considered a most promising option for cell-based cardiac repair. Hence, various protocols have been developed for differentiating hPSCs into CMs. Despite remarkable improvement in the generation of hPSC-CMs, without purification, these protocols can only generate mixed cell populations including undifferentiated hPSCs or non-CMs, which may elicit adverse outcomes. Therefore, one of the major challenges for clinical use of hPSC-CMs is the development of efficient isolation techniques that allow enrichment of hPSC-CMs. In this review, we will discuss diverse strategies that have been developed to enrich hPSC-CMs. We will describe major characteristics of individual hPSC-CM purification methods including their scientific principles, advantages, limitations, and needed improvements. Development of a comprehensive system which can enrich hPSC-CMs will be ultimately useful for cell therapy for diseased hearts, human cardiac disease modeling, cardiac toxicity screening, and cardiac tissue engineering.

Keywords: Cardiomyocytes, hPSCs

Heart failure is the leading cause of death worldwide [1]. Approximately 6 million people suffer from heart failure in the United States every year [1]. Despite this high incidence, existing surgical and pharmacological interventions for treating heart failure are limited because these approaches only delay the progression of the disease; they cannot directly repair the damaged hearts [2]. In the case of large myocardial infarction (MI), patients progress to heart failure and die within short time from the onset of symptoms [3].

The adult human heart has minimal regenerative capacity, because during mammalian development, the proliferative capacity of cardiomyocytes (CMs) progressively diminishes and becomes terminally differentiated shortly after birth [4].Therefore, once CMs are damaged, they are rarely restored [5]. When MI occurs, the infarcted area is easily converted to non-contractile scar tissue due to loss of CMs and replacement by fibrosis [6]. Development of a fibroblastic scar initiates a series of events that lead to adverse remodeling, hypertrophy, and eventual heart failure [2, 3, 7].

While heart transplantation is considered the most viable option for treating advanced heart failure, the number of available donor hearts is always less than needed [6]. Therefore, more realistic therapeutic options have been required [2]. Accordingly, over the past two decades, cell-based cardiac repair has been intensively pursued [2, 7]. Several different cell types have been tested and varied outcomes were obtained. Indeed, the key factor for successful cell-based cardiac repair is to find the optimal cell type that can restore normal heart function. Naturally, CMs have been considered the best cell type to repair a damaged heart [8]. In fact, many scientists hypothesized that implanted CMs would survive in damaged hearts and form junctions with host CMs and synchronously contract with the host myocardium [9]. In fact, animal studies with primary fetal or neonatal CMs demonstrated that transplanted CMs could survive in infarcted hearts [9-11]. These primary CMs reduced scar size, increased wall thickness, and improved cardiac contractile function with signs of electro-mechanical integration [9-11]. These studies strongly suggest that CMs can be a promising source to repair the heart. However, the short supply and ethical concerns disallow using primary human CMs. In a patient with ischemic cardiomyopathy, about 40-50% of the CMs are lost in 40 to 60 grams of heart tissue [7]. Even if we seek to regenerate a fairly small portion of the damaged myocardium, a large number of human primary CMs would be required, which is impossible.

Accordingly, CMs differentiated from human pluripotent stem cells (hPSCs) including both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have emerged as a promising option for candidate CMs for cell therapy [12, 13]. hPSCs have many advantages as a source for CMs. First, hPSCs have obvious cardiomyogenic potential. hPSC derived-CMs (hPSC-CMs) possess a clear cardiac phenotype, displaying spontaneous contraction, cardiac excitation-contraction (EC) coupling, and expression of cardiac transcription factors, cardiac ion channels, and cardiac structural proteins [14, 15]. Second, undifferentiated hPSCs and their differentiated cardiac progeny display significant proliferation capacity, allowing generation of a large number of hPSC-CMs. Lastly, many pre-clinical studies demonstrated that implantation of hPSC-CMs can repair injured hearts and improve cardiac function [16-19]. Histologically, implanted hPSC-CMs are engrafted, aligned and coupled with the host CMs in a synchronized manner [16-19].

In the last two decades, various protocols for differentiating hPSCs into CMs have been developed to improve the efficiency, purity and clinical compatibility [20] [18]. The reported differentiation methods include, but are not limited to: differentiation via embryoid body (EB) formation [20], co-culture with END-2 cells [18], and monolayer culture [15, 21, 22]. The EB-mediated CM differentiation protocol is one of the most widely employed methods due to its simple procedure and low cost. However, it often becomes labor-intensive to produce scalable EBs for further differentiation, which makes it difficult for therapeutic applications. EB-mediated differentiation also produces inconsistent results, showing beating CMs from 5% to 70% of EBs. Recently, researchers developed monolayer methods to complement the problems of EB-based methods [15, 21, 22]. In one representative protocol, hPSCs are cultured at a high density (up to 80%) and treated with a high concentration of Activin A (100 ng/ml) for 1 day and BMP4 (10 ng/ml) for 4 days followed by continuous culture on regular RPMI media with B27 [15]. This protocol induces spontaneous beating at approximately 12 days and produces approximately 40% CMs after 3 weeks. These hPSC-CMs can be further cultured in RPMI-B27 medium for another 2-3 weeks without significant cell damage [15]. However, these protocols use media with proprietary formulations, which complicates clinical application. As shown, most monolayer-based methods employ B27, which is a complex mix of 21 components. Some of the components of B27, including bovine serum albumin (BSA), are animal-derived products, and the effects of B27 components on differentiation, maturation or subtype specification processes are poorly defined. In 2014, Burridge and his colleagues developed an advanced protocol that is defined, cost-effective and efficient [22]. By subtracting one component from B27 at a time and proceeding with cardiac differentiation, the researchers reported that BSA and L-ascorbic acid 2-phosphate are essential components in cardiac differentiation. Subsequently, by replacing BSA with rice-derived recombinant human albumin, the chemically defined medium with 3 components (CDM3) was produced. The application of a GSK-inhibitor, CHIR99021, for the first 2 days followed by 2 days of the Wnt-inhibitor Wnt-59 to cells is an optimal culture condition in CDM3 resulting in similar levels of live-cell yields and CM differentiation [22].

Despite remarkable improvement in the generation of hPSC-CMs, obtaining pure populations of hPSC-CMs still remains challenging. Currently available methods can only generate a mixture of cells which include not only CMs but other cell types. This is one of the most critical barriers for applications of hPSC-CMs in regenerative therapy, drug discovery, and disease investigation. For Instance, cardiac transplantation of non-pure hPSC-CMs mixed with undifferentiated hPSCs or other cell types may produce tumors or unwanted cell types in hearts [23-28]. Accordingly, a pure or enriched population of hPSC-CMs would be required, particularly for cardiac cell therapy. Enriched hPSC-CMs would also be more beneficial for myocardial repair due to improved electric and mechanical properties [29]. A pure, homogeneous population of hPSC-CMs would pose less arrhythmic risk and have enhanced contractile performance, and would be more useful in disease modeling as they better reflect native CM physiology. Finally, purified hPSC-CMs would better serve for testing drug efficacy and toxicity. Therefore, many researchers have tried to develop methods to purify CMs from cardiomyogenically differentiated hPSCs.

There are three important topics that are not addressed in this review. First is the beneficial role of other cell types such as endothelial cells and fibroblasts in the integration, survival, and function of CMs [30-32]. We did not discuss this issue because it would need a separate review due to the volume of material. While the roles of such cells are important, the value of having purified hPSC-CMs is not diminished. Although cell mixtures or tissue engineered products can be used, unless purified CMs are employed, they would form tumors or other cells/tissues when implanted in vivo. Our point here is that even if cardiomyocytes are mixed with non-CMs, all cells should be clearly defined and purified as well. If the mixture is made in a non-purified or non-defined manner (for example, an unsophisticated top-down approach), there would be undefined cells that are neither CMs, ECs, nor fibroblasts and these unidentified cells will make aberrant tissues or tumors. Second, we did not deal with maturation of hPSC-CMs because of its broad scope and depth [33, 34]. Third is direct reprogramming or conversion of somatic cells into CMs. There has been another advancement in the generation of CMs by directly reprogramming or converting somatic cells into CM-like cells by introducing a combination of cardiac transcription factors (TFs) or muscle-specific microRNAs (miRNAs) both in vitro and in vivo [35-41]. These cells are referred to as induced CMs (iCMs) or cardiac-like myocytes (iCLMs). While this is an important advancement, we did not cover this topic either due to its size. Accordingly, this review will focus on the various strategies for purifying or enriching hPSC-CMs reported to date (Figure 1).

Early on, researchers isolated hPSC-CMs manually under microscopy by mechanically separating out the beating areas from myogenically differentiating hPSC cultures [18, 20, 42]. This method usually generates 5-70% hPSC-CMs. Although generally crude, it can enrich even higher percentages of CMs with further culture. This manual isolation method has the advantage of being easy, but while it can be useful for small-scale research, it is very labor intensive and not scalable, precluding large scale research or clinical application.

Currently available strategies for enriching cardiomyocytes derived from human pluripotent stem cells.

Xu et el. reported that hPSC-CMs, due to their physical and structural properties, can be enriched by Percoll density gradient centrifugation [43]. Percoll was first formulated by Pertoft et al [44] and it was originally developed for the isolation of cells, organelles, or viruses by density centrifugation. The Percoll-based method has several advantages. The procedure for Percoll-based separation is very simple and easy, it is inexpensive, and its low viscosity allows more rapid sedimentation and lower centrifugal forces compared to a sucrose density gradient. Lastly, it can be prepared and kept for a long time in an isotonic solution to maintain osmolarity. Although Percoll separation has resulted in major improvements in hPSC-CM isolation procedures, it has clear limitations with regard to purity and scalability. Previous studies found that Percoll separation is only able to enrich 40 -70% of hPSC-CMs. It is also not compatible with large-scale enrichment of hPSC-CMs.

Another traditional method for purifying hPSC-CMs is based on the expression of a drug resistant gene or a fluorescent reporter gene such as eGFP or DsRed, which is driven by a cardiac specific promoter in genetically modified hPSC lines [45, 46]. Here, enrichment of hPSC-CMs can be achieved by either drug treatment to eliminate cells that do not express the drug resistant gene or with FACS to isolate fluorescent cells [47, 48].

Briefly, enrichment of PSC-CMs by genetically based selection was first reported by Klug et al [49]. The authors generated murine ES cell lines via permanent gene transfection of the aminoglycoside phosphotransferase gene driven by the MHC (MYH7) promoter. With this approach, highly purified murine ESC-CMs up to 99% were achieved. Next, several studies reported the use of various CM-specific promoters to enrich ESC-CMs such as Mhc (Myh6), Myh7, Ncx (Sodium Calcium exchanger) and Mlc2v (Myl2) [46, 50, 51]. In the case of hESCs, MHC/EGFP hESCs were generated by permanent transfection of the EGFP-tagged MHC promoter [52]. Similarly, an NKX2.5/eGFP hESC line was generated to enrich GFP positive CMs [53]. However, since MHC and NKX2.5 are expressed in general CMs, the resulting CMs contain a mixture of the three subtypes of CMs, nodal-, atrial-, and ventricular-like CMs. To enrich only ventricular-like CMs, Huber et al. generated MLC2v/GFP ESCs to be able to isolate MLC2v/GFP positive ventricular-like cells by FACS [52] [54-57]. In addition, the cGATA6 gene was used to purify nodal-like hESC-CMs [58]. Future studies should focus on testing new types of cardiac specific promoters and devising advanced selection procedures to improve this strategy.

While fluorescence-based cell sorting is more widely used, the drug selection method may be a better approach to enrich high purity of hPSC-CMs during differentiation/culture as it does not require FACS. The advantage is its capability for high-purity cell enrichment due to specific gene-based cell sorting. These highly pure cells can allow more precise mechanistic studies and disease modeling. Despite its many advantages, the primary weakness of genetic selection is genetic manipulation, which disallows its use for therapeutic application. Insertion of reporter genes into the host genome requires viral or nonviral transfection/transduction methods, which can induce mutagenesis and tumor formation [50, 59-61].

Practically, antibody-based cell enrichment is the best method for cell purification to date. When cell type-specific surface proteins or marker proteins are known, one can tag cells with antibodies against the proteins and sort the target cells by FACS or magnetic-activated cell sorting (MACS). The main advantage is its specificity and sensitivity, and its utility is well demonstrated in research and even in clinical therapy with hematopoietic cells [62]. Another advantage is that multiple surface markers can be used at the same time to isolate target cells when one marker is not sufficient. However, no studies have reported surface markers that are specific for CMs, even after many years. Recently, though, several researchers demonstrated that certain proteins can be useful for isolating hPSC-CMs.

In earlier studies, KDR (FLK1 or VEGFR2) and PDGFR- were used to isolate cardiac progenitor cells [63]. However, since these markers are also expressed on hematopoietic cells, endothelial cells, and smooth muscle cells, they could not enrich only hPSC-CMs. Next, two independent studies reported two surface proteins, SIRPA [64] and VCAM-1 [65], which it was claimed could specifically identify hPSC-CMs. Dubois et al. screened a panel of 370 known antibodies against CMs differentiated from hESCs and identified SIRPA as a specific surface protein expressed on hPSC-CMs [64]. FACS with anti-SIRPA antibody enabled the purification of CMs and cardiac precursors from cardiomyogenically differentiating hPSC cultures, producing cardiac troponin T (TNNT2, also known as cTNT)-positive cells, which are generally considered hPSC-CMs, with up to 98% purity. In addition, a study performed by Elliot and colleagues identified another cell surface marker, VCAM1 [53]. In this study, the authors used NKX2.5/eGFP hESCs to generate hPSC-CMs, allowing the cells to be sorted by their NKX2.5 expression. NKX2.5 is a well-known cardiac transcription factor and a specific marker for cardiac progenitor cells [66, 67]. To identify CM-specific surface proteins, the authors performed expression profiling analyses and found that expression levels of both VCAM1 and SIRPA were significantly upregulated in NKX2.5/eGFP+ cells. Flow cytometry results showed that both proteins were expressed on the cell surface of NKX2.5/eGFP+ cells. Differentiation day 14 NKX2.5/eGFP+ cells expressed VCAM1 (71 %) or SIRPA (85%) or both VCAM1 and SIRPA (37%). When the FACS-sorted SIRPA-VCAM1-, SIRPA+ or SIRPA+VCAM1+ cells were further cultured, only SIRPA+ or SIRPA+VCAM1+ cells showed NKX2.5/eGFP+ contracting portion. Of note, NKX2.5/eGFP and SIRPA positive cells showed higher expression of smooth muscle cell and endothelial cell markers indicating that cells sorted solely based on SIRPA expression may not be of pure cardiac lineage. Hence, the authors concluded that a more purified population of hPSC-CMs could be isolated by sorting with both cell surface markers. Despite significant improvements, it appears that these surface markers are not exclusively specific for CMs as these antibodies also mark other cell types including smooth muscle cells and endothelial cells. Furthermore, they are also known to be expressed in the brain and the lung, which raises concerns whether these surface proteins can be used as sole markers for the purification of hPSC-CMs compatible for clinical applications.

More recently, Protze et al. reported successful differentiation and enrichment of sinoatrial node-like pacemaker cells (SANLPCs) from differentiating hPSCs by using cell surface markers and an NKX2-5-reporter hPSC line [68]. They found that BMP signaling specified cardiac mesoderm toward the SANLPC fate and retinoic acid signaling enhanced the pacemaker phenotype. Furthermore, they showed that later inhibition of the FGF pathway, the TFG pathway, and the WNT pathway shifted cell fate into SANLPCs, and final cell sorting for SIRPA-positive and CD90-negative cells resulted in enrichment of SANLPCs up to ~83%. These SIRPA+CD90- cells showed the molecular, cellular and electrophysiological characteristics of SANLPCs [68]. While this study makes important progress in enriching SANLPCs by modulating signaling pathways, no specific surface markers for SANLPCs were identified and the yield was still short of what is usually expected for cells purified via FACS.

Hattori et al. developed a highly efficient non-genetic method for purifying hPSC-derived CMs, in which they employed a red fluorescent dye, tetramethylrhodamine methyl ester perchlorate (TMRM), that can label active mitochondria. Since CMs contain a large number of mitochondria, CMs from mice and marmosets (monkey) could be strongly stained with TMRM [69]. They further found that primary CMs from several different types of animals and CMs derived from both mESCs and hESCs were successfully purified by FACS up to 99% based on the TMRM signals. In addition to its efficiency for CM enrichment, TMRM did not affect cell viability and disappeared completely from the cells within 24 hrs. Importantly, injected hPSC-CMs purified in this way did not form teratoma in the heart tissues. However, since TMRM only functions in CMs with high mitochondrial density, this method cannot purify entire populations of hPSC-CMs [64]. While originally TMRM was claimed to be able to isolate mature hPSC-CMs, mounting evidence indicates that hPSC-CMs are similar to immature human CMs at embryonic or fetal stages. Therefore, both the exact phenotype of the cells isolated by TMRM and its utility are rather questionable [33, 34]. Two subsequent studies demonstrated that TMRM failed to accurately distinguish hPSC-CMs due to the insufficient amounts of mitochondria [64].

Employing the unique metabolic properties of CMs, Tohyama et al. developed an elegant purification method to enrich PSC-CMs [70]. This approach is based on the remarkable biochemical differences in lactate and glucose metabolism between CMs and non-CMs, including undifferentiated cells. Mammalian cells use glucose as their main energy source [71]. However, CMs are capable of energy production from different sources such as lactate or fatty acids [71]. A comparative transcriptome analysis was performed to detect metabolism-related genes which have different expression patterns between newborn mouse CMs and undifferentiated mouse ESCs. These results showed that CMs expressed genes encoding tricarboxylic acid (TCA) cycle enzymes more than genes related to lipid and amino acid synthesis and the pentose phosphate cycle compared to undifferentiated ESCs. To further prove this observation, they compared the metabolites of these pathways using fluxome analysis between CMs and other cell types such as ESCs, hepatocytes and skeletal muscle cells, and found that CMs have lower levels of metabolites related to lipid and amino acid synthesis and pentose phosphate. Subsequently, authors cultured newborn rat CMs and mouse ESCs in media with lactate, forcing the cells to use the TCA cycle instead of glucose, and they observed that CMs were the only cells to survive this condition for even 96 hrs. They further found that when PSC derivatives were cultured in lactate-supplemented and glucose-depleted culture medium, only CMs survived. Their yield of CM population was up to 99% and no tumors were formed when these CMs were transplanted into hearts. This lactate-based method has many advantages: its simple procedures, ease of application, no use of FACS for cell sorting, and relatively low cost. More recently, this method was applied to large-scale CM aggregates to ensure scalability. As a follow-up study, the same group recently reported a more refined lactate-based enrichment method which further depletes glutamine in addition to glucose [72]. The authors found that glutamine is essential for the survival of hPSCs since hPSCs are highly dependent on glycolysis for energy production rather than oxidative phosphorylation. The use of glutamine- and glucose-depleted lactate-containing media resulted in more highly purified hPSC-CMs with less than 0.001% of residual PSCs [72]. One concern of this lactate-based enrichment method is the health of the purified hPSC-CMs, because physiological and functional characteristics of hPSC-CMs cultured in glucose- and glutamine-depleted media for a long time may have functional impairment since CMs with mature mitochondria were not able to survive without glucose and glutamine, although they were able to use lactate to synthesize pyruvate and glutamate [72]. In addition, this lactate-based strategy can only be applied to hPSC- CMs, but not other hPSC derived cells such as neuron or -cells.

Our group also recently reported a new method to isolate hPSC-CMs by directly labelling cardiac specific mRNAs using nano-sized probes called molecular beacons (MBs) [29, 73, 74]. Designed to detect intracellular mRNA targets, MBs are dual-labeled antisense oligonucleotide (ODN) nano-scale probes with a DNA or RNA backbone, a Cy3 fluorophore at the 5' end, and a Black Hole quencher 2 (BHQ2) at the 3' end [75, 76]. They form a stem-loop (hairpin) structure in the absence of a complementary target, quenching the fluorescence of the reporter. Hybridization with the target mRNA opens the hairpin and physically separates the reporter from the quencher, allowing a fluorescence signal to be emitted upon excitation. The MB-based method can be applied to the purification of any cell type that has known specific gene(s) [77].

In one study [29], we designed five MBs targeting unique sites in TNNT2 or MYH6/7 mRNA in both mouse and human. To determine the most efficient transfection method to deliver MBs into living cells, various methods were tested and nucleofection was found to have the highest efficiency. Next, we tested the sensitivity and specificity of MBs using an immortalized mouse CM cell line, HL-1, and other cell types. Finally, we narrowed it down to one MB, MHC-MB, which showed >98% sensitivity and > 95% specificity. This MHC-MB was applied to cardiomyogenically differentiated mouse and human PSCs and FACS sorting was performed. The resultant MHC-MB-positive cells expressed cardiac proteins at ~97% when measured by flow cytometry. These sorted cells also demonstrated spontaneous contraction and all the molecular and electrophysiological signatures of human CMs. Importantly, when these purified CMs were injected into the mouse infarcted myocardium, they were well integrated into the myocardium without forming any tumors, and they improved cardiac function.

In a subsequent study [74], we refined a method to enrich ventricular CMs from differentiating PSCs (vCMs) by targeting a transcription factor which is not robustly expressed in cells. Since vCMs are the main source for generating cardiac contractile forces and the most frequently damaged in the heart, there has been great demand to develop a method that can obtain a pure population of vCMs for cardiac repair. Despite this critical unmet need, no studies have demonstrated the feasibility of isolating ventricular CMs without permanently altering their genome. Accordingly, we first designed MBs targeting the Iroquois homeobox protein 4 (Irx4) mRNA, a vCM specific transcription factor [78, 79]. After testing sensitivity and specificity, one IRX4-MB was selected and applied to myogenically differentiated mPSCs. The FACS-sorted IRX4-MB-positive cells exhibited vCM-like action potentials in more than 98% of cells when measured by several electrophysiological analyses including patch clamp and Ca2+ transient analyses. Furthermore, these cells maintained spontaneous contraction and expression of vCM-specific proteins.

The MB-based cell purification method is theoretically the most broadly applicable technology among the purification methods because it can isolate any target cells expressing any specific gene. Thus, the MB-based sorting technique can be applied to the isolation of other cell types such as neural-lineage cells or islet cells, which are critical elements in regenerative medicine but do not have specific surface proteins identified to date. In addition, theoretically, this technology may have the highest efficiency when MBs are designed to have the maximum sensitivity and specificity for the cells of interest, but not others. These characteristics are particularly important for cell therapy. Despite these advantages, the delivery method of MB into the cells needs to be improved. So far, nucleofection is the best delivery method, but caused some cell damage with

Recently, Miki and colleagues reported a novel method for purifying cells of interest based on endogenous miRNA activity [80]. Miki et al. employed several synthetic mRNA switches (= miRNA switch), which consist of synthetic mRNA sequences that include a recognition sequence for miRNA and an open reading frame that codes a desired gene, such as a regulatory protein that emits fluorescence or promotes cell death. If the miRNA recognition sequence binds to miRNA expressed in the desired cells, the expression of the regulatory protein is suppressed, thus distinguishing the cell type from others that do not contain the miRNA and express the protein.

Briefly, the authors first identified 109 miRNA candidates differentially expressed in distinct stages of hPSC-CMs (differentiation day 8 and 20). Next, they found that 14 miRNAs were co-expressed in hPSC-CMs at day 8 and day 20 and generated synthetic mRNAs that recognize these 14 miRNA, called miRNA switches. Among those miRNA switches, miR-1-, miR-208a-, and miR-499a-5p-switches successfully enriched hPSC-CMs with purity of sorted cells up to 96% determined by TNNT2 intracellular flow cytometry. Particularly, hPSC-CMs enriched by the miR-1-switch showed substantially higher expression of several cardiac specific genes/proteins and lower expression of non-CM genes/proteins compared with control cells. Patch clamp confirmed that these purified hPSC-CMs possessed both ventricular-like and atrial-like action potentials.

One of the major advantages of this technology is its wider applicability to other cell types. miRNA switches have the flexibility to design the open reading frame in the mRNA sequence such that any desired transgene can be incorporated into the miRNA switches to regulate the cell phenotype based on miRNA activity. The authors tested this possibility by incorporating BIM sequence, an apoptosis inducer, into the cardiac specific miR-1- and miR-208a switches and tested whether they could selectively induce apoptosis in non-CMs. They found that miR-1- and miR-208a-Bim-switches successfully enriched cTNT-positive hPSC-CMs without cell sorting. Enriched hPSC-CMs by 208a-Bim-switch were injected into the hearts of mice with acute MI and they engrafted, survived, expressed both cTNT and CX43, and formed gap junctions with the host myocardium. No teratoma was detected. In addition, other miRNA switches such as miR-126-, miR-122-5p-, and miR-375-switches targeting endothelial cells, hepatocytes, and -cells, respectively, successfully enriched these cell types differentiated from hPSCs. However, identification of specific miRNAs expressed only in the specific cell type of interest and verification of their specificity in target cells will be key issues for continuing to use this miRNA-based cell enrichment method.

Recent advances in biomedical engineering have contributed to developing systems that can isolate target cells using physicochemical properties of the cells. Microfluidic systems have been intensively applied for cell separation due to recent improvements in miniaturizing a cell culture system [81-83]. These advances made possible the design of automated microfluidic devices with cellular microenvironments and controlled fluid flows that save time and cost in experiments. Thus, there have been an increasing number of studies seeking to apply the microfluidic system for cell separation. Among the first, Singh et al. tested the possibility of using a microfluidic system for the separation of hPSC [84] by preparative detachment of hPSCs from differentiating cultures based on differences in the adhesion properties of different cell types. Distinct streams of buffer that generated varying levels of shear stress further allowed selective enrichment of hPSC colonies from mixed populations of adherent non-hPSCs, achieving up to 95% purity. Of note, this strategy produced hPSC survival rates almost two times higher than FACS, reaching 80%.

Subsequently, for hPSC-CMs purification, Xin et al. developed a microfluidic system with integrated ridge-like flow derivations and fishnet-like microcolumns for the enrichment of hiPSC-CMs [85]. This device is composed of a 250 mm-long microfluidic channel, which has two integrated parallel microcolumns with surfaces functionalized with anti-human TRA-1 antibody for undifferentiated hiPSC trapping. Aided by the ridge-like surface patterns on the upper wall of the channel, micro-streams are generated so that the cell suspension of mixed undifferentiated hiPSCs and hiPSC-CMs are forced to cross the functionalized fishnet-like microcolumns, resulting in trapping of undifferentiated hiPSCs due to the interaction between the hiPSCs and the columns, and the untrapped hiPSC-CMs are eventually separated. By modulating flow and coating with anti-human TRA-1 antibody, they were able to enrich CMs to more than 80% purity with 70% viability. While this study demonstrated that a microfluidic device could be used for purifying hPSC-CMs, it was not realistic because the authors used a mixture of only undifferentiated hiPSCs and hiPSC-CMs. In real cardiomyogenically differentiated hiPSCs, undifferentiated hiPSCs are rare and many intermediate stage cells or other cell types are present, so the idea that this simple device can select only hiPSC-CMs from a complex mixture is uncertain.

Overall, the advantages of microfluidic system based cell isolation include fast speed, improved cell viability and low cost owing to the automated microfluidic devices that can control cellular microenvironments and fluid flows [86-88]. However, microfluidic-based cell purification methods have limitations in terms of low purity and scalability [89-92]. In fact, there have been only a few studies demonstrating the feasibility that microfluidic device-based cell separation could achieve higher than 80% purity of target cells. Furthermore, currently available microfluidic devices allow only separation of a small number of cells ( 95% purity.

Having available a large quantity of a homogeneous population of cells of interest is an important factor in advancing biomedical research and clinical medicine, and is especially true for hPSC-CMs. While remarkable progress has been made in the methods for differentiating hPSCs into CMs, technologies to enrich hPSC-CMs, particularly those which are clinically applicable, have been emerging only over the last few years. Contamination with other cell types and even the heterogeneous nature of hPSC-CMs significantly hinder their use for several future applications such as cardiac drug toxicology screening, human cardiac disease modeling, and cell-based cardiac repair. For instance, cardiac drug-screening assays require pure populations of hPSC-CMs, so that the observed signals can be attributed to effects on human CMs. Studies of human cardiac diseases can also be more adequately interpreted with purified populations of patient derived hiPSC-CMs. Clinical applications with hPSC-CMs will need to be free of other PSC derivatives to minimize the risk of teratoma formation and other adverse outcomes.

Summary of representative methods for hPSC-CM purification

Schematic pictures of microfluidic device for enriching hiPSC-CMs. (A) The part of the device designed for trapping undifferentiated hiPSCs. (B) (Left) Illustration of the overall microfluidic device assembled with peristaltic pump, cell suspension reservoirs, and a serpentine channel. (Right) Magnified image showing a channel combining microcolumns and ridge-like flow derivation structures. Modified from Li et al. On chip purification of hiPSC-derived cardiomyocytes using a fishnet-like microstructure. Biofabrication. 2016 Sep 8;8(3): 035017

Therefore, development of reproducible, effective, non-mutagenic, scalable, and economical technologies for purifying hPSC-CMs, independent of hPSC lines or differentiation protocols, is a fundamental requirement for the success of hPSC-CM applications. Fortunately, new technologies based on the biological specificity of CMs such as MITO-tracker, molecular beacons, lactate-enriched-glucose depleted-media, and microRNA switches have been developed. In addition, technologies based on engineering principles have recently yielded a promising platform using microfluidic technology. While due to the short history of this field, more studies are needed to verify the utility of these technologies, the growing attention toward this research is a welcome move.

Another important question raised recently is how to non-genetically purify chamber-specific subtypes of CMs such as ventricular-like, atrial-like and nodal-like hPSC-CMs. So far, only a few studies have addressed this potential with human PSCs. We also showed that a molecular beacon-based strategy could enrich ventricular CMs differentiated from PSCs [74]. Another study demonstrated generation of SA-node like pacemaker cells by using a stepwise treatment of various morphogens and small molecules followed by cell sorting with several sub-specific surface markers. However, the yield of both studies was relatively low (

In summary, technological advances in the purification of hPSC-CMs have opened an avenue for realistic application of hPSC-CMs. Although initial success was achieved for purification of CMs from differentiating hPSC cultures, questions such as scalability, clinical compatibility, and cellular damage remain to be answered and isolation of human subtype CMs has yet to be demonstrated. While there are other challenges such as maturity, in vivo integration, and arrhythmogenecity, this development of purification technology represents major progress in the field and will provide unprecedented opportunities for cell-based therapy, disease modeling, drug discovery, and precision medicine. Furthermore, the availability of chamber-specific CMs with single cell analyses will facilitate more sophisticated investigation of human cardiac development and cardiac pathophysiology.

This work was supported by the Bio & Medical Technology Development Program of the National Research Foundation (NRF) funded by the Korean government (MSIP) (No 2015M3A9C6031514), the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (HI15C2782, HI16C2211) and grants from NHLBI (R01HL127759, R01HL129511), NIDDK (DP3-DK108245). This work was also supported by a CityU Start-up Grant (No 7200492), a CityU Research Project (No 9610355), and a Georgia Immuno Engineering Consortium through funding from Georgia Institute of Technology, Emory University, and the Georgia Research Alliance.

The authors have declared that no competing interest exists.

1. Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M. et al. Heart disease and stroke statistics2016 update. Circulation. 2016;133:e38-e360

2. Ptaszek LM, Mansour M, Ruskin JN, Chien KR. Towards regenerative therapy for cardiac disease. Lancet. 2012;379:933-42

3. Jessup M, Brozena S. Heart failure. N Engl J Med. 2003;348:2007-18

4. Pasumarthi KBS, Field LJ. Cardiomyocyte cell cycle regulation. Circ Res. 2002;90:1044-54

5. Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnab-Heider F, Walsh S. et al. Evidence for cardiomyocyte renewal in humans. Science. 2009;324:98-102

6. Passier R, van Laake LW, Mummery CL. Stem-cell-based therapy and lessons from the heart. Nature. 2008;453:322-9

7. Laflamme MA, Murry CE. Heart regeneration. Nature. 2011;473:326-35

8. Tulloch NL, Muskheli V, Razumova MV, Korte FS, Regnier M, Hauch KD. et al. Growth of engineered human myocardium with mechanical loading and vascular coculture / Novelty and significance. Circ Res. 2011;109:47-59

9. Reinecke H, Zhang M, Bartosek T, Murry CE. Survival, integration, and differentiation of cardiomyocyte grafts: A study in normal and injured rat hearts. Circulation. 1999;100:193-202

10. Leor J, Patterson M, Quinones MJ, Kedes LH, Kloner RA. Transplantation of fetal myocardial tissue into the infarcted myocardium of rat. A potential method for repair of infarcted myocardium?. Circulation. 1996;94:II332-6

11. Li R-K, Mickle DAG, Weisel RD, Zhang J, Mohabeer MK. In vivo survival and function of transplanted rat cardiomyocytes. Circ Res. 1996;78:283-8

12. Shiba Y, Fernandes S, Zhu WZ, Filice D, Muskheli V, Kim J. et al. Human ES-cell-derived cardiomyocytes electrically couple and suppress arrhythmias in injured hearts. Nature. 2012;489:322-5

13. Shigeru M, Satsuki F, Yukiko I, Takuji K, Noriko Mochizuki O, Shigeo M. et al. Building a new treatment for heart failure-transplantation of induced pluripotent stem cell-derived cells into the heart. Curr Gene Ther. 2016;16:5-13

14. Mignone JL, Kreutziger KL, Paige SL, Murry CE. Cardiogenesis from human embryonic stem cells - Mechanisms and applications. Circulation J. 2010;74:2517-26

15. Laflamme MA, Chen KY, Naumova AV, Muskheli V, Fugate JA, Dupras SK. et al. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat Biotechnol. 2007;25:1015-24

16. Yuasa S, Itabashi Y, Koshimizu U, Tanaka T, Sugimura K, Kinoshita M. et al. Transient inhibition of BMP signaling by Noggin induces cardiomyocyte differentiation of mouse embryonic stem cells. Nat Biotechnol. 2005;23:607-11

17. Nemir M, Croquelois A, Pedrazzini T, Radtke F. Induction of cardiogenesis in embryonic stem cells via downregulation of Notch1 signaling. Circ Res. 2006;98:1471-8

18. Mummery C, Ward-van Oostwaard D, Doevendans P, Spijker R, van den Brink S, Hassink R. et al. Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells. Circulation. 2003;107:2733-40

19. Passier R, Oostwaard DW, Snapper J, Kloots J, Hassink RJ, Kuijk E. et al. Increased cardiomyocyte differentiation from human embryonic stem cells in serum-free cultures. Stem Cells. 2005;23:772-80

20. Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein A. et al. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest. 2001;108:407-14

21. Yang L, Soonpaa MH, Adler ED, Roepke TK, Kattman SJ, Kennedy M. et al. Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature. 2008;453:524-8

22. Burridge PW, Matsa E, Shukla P, Lin ZC, Churko JM, Ebert AD. et al. Chemically defined generation of human cardiomyocytes. Nat Methods. 2014;11:855-60

23. Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein A. et al. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest. 2001;108:407-14

24. Zhang J, Wilson GF, Soerens AG, Koonce CH, Yu J, Palecek SP. et al. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ Res. 2009;104:e30-41

25. Nussbaum J, Minami E, Laflamme MA, Virag JAI, Ware CB, Masino A. et al. Transplantation of undifferentiated murine embryonic stem cells in the heart: teratoma formation and immune response. FASEB J. 2007;21:1345-57

26. Lee AS, Tang C, Rao MS, Weissman IL, Wu JC. Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies. Nat Med. 2013;19:998-1004

27. Masuda S, Miyagawa S, Fukushima S, Sougawa N, Ito E, Takeda M. et al. Emerging innovation towards safety in the clinical application of ESCs and iPSCs. Nat Rev Cardiol. 2014;11:553-4

28. Masuda S, Miyagawa S, Fukushima S, Sougawa N, Okimoto K, Tada C. et al. Eliminating residual iPS cells for safety in clinical application. Protein Cell. 2015;6:469-71

29. Ban K, Wile B, Kim S, Park H-J, Byun J, Cho K-W. et al. Purification of cardiomyocytes from differentiating pluripotent stem cells using molecular beacons that target cardiomyocyte-specific mRNA. Circulation. 2013;128:1897-909

30. Masumoto H, Ikuno T, Takeda M, Fukushima H, Marui A, Katayama S. et al. Human iPS cell-engineered cardiac tissue sheets with cardiomyocytes and vascular cells for cardiac regeneration. Sci Rep. 2014;4:6716

31. Kensah G, Roa Lara A, Dahlmann J, Zweigerdt R, Schwanke K, Hegermann J. et al. Murine and human pluripotent stem cell-derived cardiac bodies form contractile myocardial tissue in vitro. Eur Heart J. 2013;34:1134-46

32. Thavandiran N, Dubois N, Mikryukov A, Mass S, Beca B, Simmons CA. et al. Design and formulation of functional pluripotent stem cell-derived cardiac microtissues. Proc Natl Acad Sci U S A. 2013;110:E4698-E707

33. Yang X, Pabon L, Murry CE. Engineering adolescence: maturation of human pluripotent stem cell-derived cardiomyocytes. Circ Res. 2014;114:511-23

34. Robertson C, Tran DD, George SC. Concise review: maturation phases of human pluripotent stem cell-derived cardiomyocytes. Stem Cells. 2013;31:829-37

35. Ieda M, Fu JD, Delgado-Olguin P, Vedantham V, Hayashi Y, Bruneau BG. et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell. 2010;142:375-86

36. Qian L, Huang Y, Spencer CI, Foley A, Vedantham V, Liu L. et al. In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature. 2012;485:593-8

37. Jayawardena TM, Egemnazarov B, Finch EA, Zhang L, Payne JA, Pandya K. et al. MicroRNA-mediated in vitro and in vivo direct reprogramming of cardiac fibroblasts to cardiomyocytes. Circ Res. 2012;110:1465-73

38. Song K, Nam YJ, Luo X, Qi X, Tan W, Huang GN. et al. Heart repair by reprogramming non-myocytes with cardiac transcription factors. Nature. 2012;485:599-604

39. Nam YJ, Song K, Luo X, Daniel E, Lambeth K, West K. et al. Reprogramming of human fibroblasts toward a cardiac fate. Proc Natl Acad Sci U S A. 2013;110:5588-93

40. Jayawardena TM, Finch EA, Zhang L, Zhang H, Hodgkinson CP, Pratt RE. et al. MicroRNA induced cardiac reprogramming in vivo: Evidence for mature cardiac myocytes and improved cardiac function. Circ Res. 2015;116:418-24

41. Wada R, Muraoka N, Inagawa K, Yamakawa H, Miyamoto K, Sadahiro T. et al. Induction of human cardiomyocyte-like cells from fibroblasts by defined factors. Proc Natl Acad Sci U S A. 2013;110:12667-72

42. Caspi O, Huber I, Kehat I, Habib M, Arbel G, Gepstein A. et al. Transplantation of human embryonic stem cell-derived cardiomyocytes improves myocardial performance in infarcted rat hearts. J Am Coll Cardiol. 2007;50:1884-93

43. Xu C, Police S, Rao N, Carpenter MK. Characterization and enrichment of cardiomyocytes derived from human embryonic stem cells. Circ Res. 2002;91:501-8

44. Pertoft H, Laurent TC, Ls T, Kgedal L. Density gradients prepared from colloidal silica particles coated by polyvinylpyrrolidone (Percoll). Anal Biochem. 1978;88:271-82

45. Doevendans PA, Becker KD, An RH, Kass RS. The utility of fluorescentin vivoreporter genes in molecular cardiology. Biochem Biophys Res Commun. 1996;222:352-8

46. Ritner C, Wong SSY, King FW, Mihardja SS, Liszewski W, Erle DJ. et al. An engineered cardiac reporter cell line identifies human embryonic stem cell-derived myocardial precursors. PLoS One. 2011;6:e16004

47. Ma J, Guo L, Fiene SJ, Anson BD, Thomson JA, Kamp TJ. et al. High purity human-induced pluripotent stem cell-derived cardiomyocytes: electrophysiological properties of action potentials and ionic currents. Am J Physiol Heart Circ Physiol. 2011;301:H2006-H17

48. Xu XQ, Zweigerdt R, Soo SY, Ngoh ZX, Tham SC, Wang ST. et al. Highly enriched cardiomyocytes from human embryonic stem cells. Cytotherapy. 2008;10:376-89

49. Klug MG, Soonpaa MH, Koh GY, Field LJ. Genetically selected cardiomyocytes from differentiating embronic stem cells form stable intracardiac grafts. J Clin Invest. 1996;98:216-24

50. Anderson D, Self T, Mellor IR, Goh G, Hill SJ, Denning C. Transgenic enrichment of cardiomyocytes from human embryonic stem cells. Mol Ther. 2007;15:2027-36

51. Fu J-D, Jiang P, Rushing S, Liu J, Chiamvimonvat N, Li RA. Na+/Ca2+ exchanger is a determinant of excitation-contraction coupling in human embryonic stem cell-derived ventricular cardiomyocytes. Stem Cells Dev. 2009;19:773-82

52. Huber I, Itzhaki I, Caspi O, Arbel G, Tzukerman M, Gepstein A. et al. Identification and selection of cardiomyocytes during human embryonic stem cell differentiation. FASEB J. 2007;21:2551-63

53. Elliott DA, Braam SR, Koutsis K, Ng ES, Jenny R, Lagerqvist EL. et al. NKX2-5(eGFP/w) hESCs for isolation of human cardiac progenitors and cardiomyocytes. Nat Methods. 2011;8:1037-40

54. Bizy A, Guerrero-Serna G, Hu B, Ponce-Balbuena D, Willis BC, Zarzoso M. et al. Myosin light chain 2-based selection of human iPSC-derived early ventricular cardiac myocytes. Stem Cell Res. 2013;11:1335-47

55. Lee MY, Sun B, Schliffke S, Yue Z, Ye M, Paavola J. et al. Derivation of functional ventricular cardiomyocytes using endogenous promoter sequence from murine embryonic stem cells. Stem Cell Res. 2012;8:49-57

56. MLLER M, FLEISCHMANN BK, SELBERT S, JI GJ, ENDL E, MIDDELER G. et al. Selection of ventricular-like cardiomyocytes from ES cells in vitro. FASEB J. 2000;14:2540-8

57. Zhang Q, Jiang J, Han P, Yuan Q, Zhang J, Zhang X. et al. Direct differentiation of atrial and ventricular myocytes from human embryonic stem cells by alternating retinoid signals. Cell Res. 2011;21:579-87

View post:
Georgia Stem Cells | Stem Cell TV

Posted in Georgia Stem Cells | Comments Off on Georgia Stem Cells | Stem Cell TV

Genetic Medicine | List of High Impact Articles | PPts …

Posted: September 7, 2019 at 4:34 pm

Genetic medicine is the integration and application of genomic technologies allows biomedical researchers and clinicians to collect data from large study population and to understand disease and genetic bases of drug response. It includes genome structure, functional genomics, epigenomics, genome scale population genomics, systems analysis, pharmacogenomics and proteomics. The Division of Genetic Medicine provides an academic environment enabling researchers to explore new relationships between disease susceptibility and human genetics. The Division of Genetic Medicine was established to host both research and clinical research programs focused on the genetic basis of health and disease. Equipped with state-of-the-art research tools and facilities, our faculty members are advancing knowledge of the common genetic determinants of cancer, congenital neuropathies, and heart disease.

Related Journals of Genetic Medicine

Cellular & Molecular Medicine, Translational Biomedicine, Biochemistry & Molecular Biology Journal, Cellular & Molecular Medicine, Electronic Journal of Biology, Molecular Enzymology and Drug Targets, Journal of Applied Genetics, Journal of Medical Genetics, Genetics in Medicine, Journal of Anti-Aging Medicine, Reproductive Medicine and Biology, Romanian journal of internal medicine

Read this article:
Genetic Medicine | List of High Impact Articles | PPts ...

Posted in Genetic medicine | Comments Off on Genetic Medicine | List of High Impact Articles | PPts …

Stem Cell Therapy Salem OR – oregonmedicalcenters.com

Posted: September 7, 2019 at 4:33 pm

Stem cell therapy in Salem, Oregon can help relieve your discomfort and repair damaged tissues in your body without the need for surgery. Led by Dr. Sunita Bhasin, who has a team of medical providers that specialize in regenerative medicine and have advanced training and experience with safe, effective stem cell therapy in Salem, Oregon. Schedule a consultation online or by phone now to learn more.

Stem cell therapy is a form of regenerative medicine that uses stem cells to replace and restore damaged tissue in the body. Its administered through an injection into your joints, muscles, ligaments, and tendons to relieve pain, reduce inflammation, and improve the function of the affected area.

at Oregon Medical Centers in Salem, Oregon, we take stem cells from placental tissue and processes them for safety and effectiveness. Once stem cells are injected into your body, they work in two ways to repair the damage. First, stem cells are able to differentiate into the various types of damaged tissues in the area. Second, stem cells can replicate and multiply to replace damaged or injured tissues.

With tissue damage, chronic inflammation causes pain and prevents your body from healing properly. Stem cells reduce inflammation in the area by repairing and replacing the damaged tissues, stimulating your body to heal naturally. This reduces the associated inflammation and pain of tissue injury.

Stem cell therapy from our Salem clinic is an effective option if you want to avoid more invasive procedures like surgery for your pain. Common uses for stem cell injections include:

Stem cells can also be used in situations when a previous treatment or surgery hasnt worked or youre still experiencing pain. They can also be used in conjunction with other types of therapy to relieve pain, improve your range of motion, and reduce inflammation.

When you visit Oregon Medical Centers for stem cell treatment, youll meet with your doctor and nurse practitioner to discuss your symptoms, treatments youve tried, and the history of your injury. Your doctor then performs a thorough physical exam, assessing your flexibility and strength in the affected area and checking for signs of more complex injuries.

Once your doctor has completed your physical, they may recommend treatment to relieve your discomfort, including stem cell knee, hip, and joint injections. For the stem cell injection, your nurse practitioner cleans your skin and prepares the area for treatment.

The stem cells are injected into the affected area with a sterile syringe and the area is cleaned and covered with a bandage. In most cases, you can return to your normal activities the same day. Depending on your condition, your doctor may recommend a series of injections over several months to help you get the most benefit and pain relief.

To schedule a consultation at Oregon Medical Centers, call the office or use the online tool now.

Read the rest here:
Stem Cell Therapy Salem OR - oregonmedicalcenters.com

Posted in Oregon Stem Cells | Comments Off on Stem Cell Therapy Salem OR – oregonmedicalcenters.com

About | OHSU

Posted: September 7, 2019 at 4:33 pm

The Oregon Stem Cell Center conducts basic and applied research in the field of Stem Cell Biology with the long-term goal to harness the properties of stem cells for regenerative medicine and cell therapy.

Our mission

Cell and transplantation therapy have been part of mainstream medicine for decades, but cell therapy has made only limited advances in several years. Cell therapy is currently used in transfusion medicine, bone marrow transplantation, orthopedics and in dermatology. Although clinical trials have clearly documented the potential for novel cell therapies, cell supply has been severely limiting and is the main obstacle to more wide-spread success. Islet transplants have worked for type 1 diabetes, fetal cells were successful for severe Parkinson's disease and hepatocytes have been useful for the treatment of metabolic disease. Stem cell biology promises to solve the problem of limited cell availability by finding ways to isolate cells from living donors, cadavers or immortal stem cell sources.

Our cores

The Oregon Stem Cell Center has 3 cores:

Philip Streeter, Ph.D. is the director of these core laboratories. The main goal of the cores is to generate novel reagents for the isolation of stem cells and their differentiated offspring by generating monoclonal antibodies directed against cell surface antigens of living cells. To date, antibodies useful for cell sorting of living cells are only available for a very limited number of tissues, chiefly blood tissues. It therefore has not been possible to isolate and purify living liver stem cells, pancreas stem cells, cardiac stem cells etc.

The Oregon stem cell cores are poised to embark on a systematic effort to produce novel cell surface antibodies for all tissues of the mouse, rat, primates and humans. The cell sorting core uses a state-of-the-art Cytopeia high speed InFlux instrument and is capable of sorting large and fragile cells without loss of viability. Pamela Canaday is the FACS operator. The cell isolation core will provide cell isolations services including tissue procurement and protease digestion of these tissues.

Center description

The Oregon Stem Cell Center was created on January 1, 2004 and is directed by Markus Grompe, M.D. The center is housed on the top (7th) floor of the Biomedical Research Building.In 2009, the center administratively became part of the Pap Family Pediatric Research Institute.

The center has both primary and affiliate faculty members representing multiple departments and centers at OHSU. Research topics that are covered include pluripotent stem cells, hematopoietic stem cells, leukemia stem cells, hepatic and pancreatic progenitors, mesenchymal stem cells, neural stem cells, muscle stem cells and intestinal stem cells.

The OSCC has 3 cores, a monoclonal antibody production core, a cell sorting core and a cell isolation core. Philip Streeter, Ph.D. is the director of these core laboratories. The main goal of the cores is to generate novel reagents for the isolation of stem cells and their differentiated offspring by generating monoclonal antibodies directed against cell surface antigens of living cells. The cell sorting core uses a state-of-the-art Cytopeia high speed InFlux instrument and is capable of sorting large and fragile cells without loss of viability. The cell isolation core will provide cell isolations services including tissue procurement and protease digestion of these tissues.

Continue reading here:
About | OHSU

Posted in Oregon Stem Cells | Comments Off on About | OHSU

Page 1,379«..1020..1,3781,3791,3801,381..1,3901,400..»