Page 11234..1020..»

Category Archives: Regenerative Medicine

Regenerative medicine UKRI – UK Research and Innovation

Posted: December 10, 2023 at 2:36 am

Funding routes

We are keen to support high quality research into stem cells and regenerative medicine that:

Advanced therapeutics (including cell and gene therapy, regenerative medicine and innovative medicines) is one of three MRC-wide opportunity areas that apply to all boards and panels, used to help prioritise applications for funding.

Our four research boards fund science that enhances our knowledge of the biology of health and disease and new approaches to treatment, including fundamental or investigative research, for example hypothesis-led research seeking to:

The choice of which board to submit to will depend upon the nature of the work and the disease being investigated. Basic stem or progenitor cell research of a more generic nature should be directed to the Molecular and Cellular Medicine Board. Regenerative medicine research that moves beyond underpinning biological mechanisms and focuses on particular organs or tissues and associated dysfunction or disease, other than haematology, are more likely to align with one of the other research boards:

The Developmental Pathway Funding Scheme is a key part of our Translational Research Strategy and supports the translation of fundamental discoveries toward benefits to human health. It funds the pre-clinical development and early clinical testing of novel therapeutics, devices and diagnostics, including repurposing existing therapies.

Regenerative medicine research projects are eligible for all our training investments. More information about training grants is available, specifically for fellowships and studentships.

The Steering Committee for the UK Stem Cell Bank and use of Stem Cell Lines publishes the UK stem cell line registry, which identifies all human embryonic stem cell lines approved for use in the UK. We will not support research using human embryonic stem cell lines that the steering committee has not approved.

Read the UK stem cell line registry.

The UK Stem Cell Bank provides ethically-sourced and quality controlled human embryonic stem cell (hESC) lines and other associated materials to researchers worldwide, and aims to facilitate high quality and standardised research in this area. The bank publishes a catalogue of currently available hESC lines.

The Human Induced Pluripotent Stem Cell Initiative (HipSci) generated a large, high-quality reference panel of genotypically and phenotypically characterised human induced pluripotent stem cells (iPSC). HipSci cell lines are available through the European Collection of Authenticated Cell Cultures (ECACC)

MRC has issued guiding principles on expectations regarding requests for support for establishing new iPSC resources.

The International Stem Cell Banking Initiative (ISCBI) provides information for scientists interested in developing or using a stem cell bank. The initiatives aim is to create a global network of stem cell banks through support for existing banks, and by encouraging the development of new banks.

The ISCBI encourages good practice in stem cell banking scientifically and ethically and provides information on resources and meetings for those involved in stem cell banking.

The MRC-Wellcome Trust human developmental biology resource is a collection of human embryonic and foetal material available for the international scientific community to research.

The NHS Blood and Transplant special health authority includes:

The Innovate UK-funded Cell and Gene Therapy Catapult is an independent centre of excellence, designed to advance the growth of the UK cell and gene therapy industry by bridging the gap between scientific research and full-scale commercialisation. It works with partners in academia and industry to ensure that life-changing therapies can be developed for use in health services throughout the world.

Read the original:
Regenerative medicine UKRI - UK Research and Innovation

Posted in Regenerative Medicine | Comments Off on Regenerative medicine UKRI – UK Research and Innovation

What Is Regenerative Medicine? | Goals and Applications | ISCRM

Posted: December 2, 2023 at 2:34 am

For many centuries, we have looked to medicine to heal us when we are sick or injured. Major breakthroughs, like vaccines and antibiotics, have improved quality of life, and, in some cases, led to the effective eradication of infectious diseases.

While modern medicine has certainly changed the human experience for the better, we remain at the mercy of disease. There are no vaccines for malaria or HIV, for example. And chronic diseases, like heart disease, Alzheimers, diabetes, and osteoporosis, although treatable, are relentless causes of suffering. There are no silver bullet for these conditions. Often the best we can do is manage the symptoms.

One key to changing that may be regenerative medicine, a field of research with its sights set on the root causes of diseases, including many being studied now at the Institute for Stem Cell and Regenerative Medicine (ISCRM).

As a discipline, regenerative medicine combines principles of biology and engineering to develop therapies for diseases characterized by cell depletion, lost tissue, or damaged organs. The broad aim of regenerative medicine is to engineer, regenerate, or replace tissue using natural growth and repair mechanisms, such as stem cells. Organoids, 3D organ printing, and tissue engineering are examples of biopowered technologies used in regenerative medicine.

Many common chronic diseases begin with harmful cell depletion. For example, Alzheimers disease is associated with a loss of brain cells, heart disease is often marked by a loss of healthy heart muscle, and type 1 diabetes occurs when cells in the pancreas fail to produce insulin. In the case of cancer, the problem is that cells grow too quickly. (Click here to read more about diseases being researched at ISCRM.)

For scientists, regenerative medicine is a way to fix the root causes of disease by harnessing the bodys natural capacity to repair itself in other words, to regenerate lost cells and tissue and restore normal functioning. At the Institute for Stem Cell and Regenerative Medicine, researchers are studying how to jump start the growth of cells in the brain, heart, pancreas, liver, kidney, eyes, ears, and muscles.

Ultimately, the goal of regenerative medicine is to improve the daily wellbeing of patients with debilitating chronic diseases by developing a new generation of therapies that go beyond treating symptoms.

Stem cells are powerful tools of discovery used by researchers hoping to understand how regenerative medicine could be used to treat patients. Right now, ISCRM researchers are using stem cells to study how heart diseases develop, testing stem cell-based therapies that could regenerate damaged or lost heart tissue, and even launching heart tissue into space to study the effects of microgravity on cardiovascular health. Many ISCRM scientists use stem cells to create 3D organ models, known as organoids, that allow them to study diseases and test regenerative treatments without involving animals or human subjects.

Heart Regeneration Researchers in multiple ISCRM labs are pursuing novel approaches that can potentially cure rather than manage heart disease. In 2018, a study led by ISCRM Director Dr. Charles Murry demonstrated that stem cell-derived cardiomyocytes have the potential to regenerate heart tissue in large non-human primates, a major step toward human clinical trials. In another investigation, ISCRM faculty members Jen Davis, PhD and Farid Moussavi-Harami, MD are developing new tools to help cardiologists design personalized treatments for certain heart diseases.

Diabetes ISCRM researchers are studying the mechanisms that regulate the development and function of beta cells in the pancreas that produce insulin a key to future treatments for any type of diabetes. Vincenzo Cirulli MD, PhD, is screening for biological factors that could promote the growth of beta cells necessary for insulin production. Dr. Cirullis ISCRM colleague Laura Crisa MD, PhD is using a disease-in-a-dish model to study how islet cells falter and whether they can be regenerated, and eventually transplanted, into patients.

Vision Disorders Researchers at the Institute for Stem Cell and Regenerative Medicine (ISCRM) are using stem cell-derived retinal organoids to study how diseases of the retina form and how they can be treated. Organoids closely approximate human tissue without many of the ethical questions and supply limitations that complicate the use of fetal tissue. Read more about recent efforts to validate stem cell-derived organoids as disease models here.

In an approach could someday be used to help repair the retinas in patients who have lost vision due to macular degeneration, glaucoma and diabetes, the Reh Lab has successfully induced non-neuronal cells to become retinal neurons. In an October 2021 study published in the journal Cell Reports, Reh and his team using proteins (known as transcription factors) that regulate the activity of genes to induce glial cells in the retina to produce neurons. The effort demonstrates that gene therapy could someday be used in clinics to help repair damaged retinas and restore vision.

See the rest here:
What Is Regenerative Medicine? | Goals and Applications | ISCRM

Posted in Regenerative Medicine | Comments Off on What Is Regenerative Medicine? | Goals and Applications | ISCRM

What Is Regenerative Medicine? | Mass General Brigham

Posted: December 2, 2023 at 2:34 am

Prolotherapy injections

Sports medicine specialists use prolotherapy injections to address connective tissue injuries. Research on prolotherapy highlights its potential to reduce pain from early to moderate osteoarthritis, sacroiliac (SI) joint ligaments, and tendinopathy.

Prolotherapy injects a solution made of specific concentrations of dextrose (sugar water), saline (salt and water), and a numbing medication into damaged tissue. Studies have shown the dextrose concentrate itself can stimulate the release of growth factors, explains Dr. Borg Stein. It can also cause some temporary inflammation in the area, which is intended to help kickstart a healing process that may have stalled."

Like all injection therapies, prolotherapy uses a special needling technique during the injection with the goal of increasing your bodys healing response.

Both prolotherapy and extracorporeal shockwave therapy are attractive treatment options when taking time off isnt in the playbook. We often do these treatments with athletes who are in season, sometimes in combination with each other, says Dr. Borg Stein. In most cases, theres really little to no downtime.

Originally posted here:
What Is Regenerative Medicine? | Mass General Brigham

Posted in Regenerative Medicine | Comments Off on What Is Regenerative Medicine? | Mass General Brigham

Advancing Safe and Effective Regenerative Medicine Products

Posted: March 20, 2023 at 12:07 am

By: Peter Marks, M.D., Ph.D., Director, Center for Biologics Evaluation and Research

The U.S. Food and Drug Administration plays a vital role in facilitating the development and availability of innovative medical products. Products such as cellular-derived therapies, including stem cell-based products, offer the potential to treat or even cure diseases or conditions for which few effective treatment options exist.

The FDAs November 2017 regenerative medicine policy framework was developed to help facilitate and support innovation in the area of regenerative medicine therapies. As part of this framework, we encourage sponsors to take advantage of ongoing expedited programs that might be available to them, including Regenerative Medicine Advanced Therapy, breakthrough therapy, and fast track designations, to support product development and licensure.

The framework also outlines the agencys intent to exercise enforcement discretion with respect to the FDAs investigational new drug (IND) and premarket approval requirements for certain regenerative medicine products until November 2020, which was later extended through May 2021. This compliance and enforcement discretion policy gives manufacturers time to determine if certain requirements apply to their products, and if an application is needed, to prepare and submit the appropriate application to the FDA.

We are now reaffirming the timing of the end of the compliance and enforcement discretion policy for certain human cell, tissue, and cellular and tissue-based products (HCT/Ps), including regenerative medicine therapies. The period during which the FDA intends to exercise enforcement discretion with respect to the IND and premarket approval requirements for certain HCT/Ps ends on May 31, 2021, and will not be extended further.

Since November 2017, the FDA has worked with product developers to help them determine if they need to submit an IND or marketing application and, if so, how they should submit their application to the FDA. The FDA developed programs that provide opportunities for engagement between HCT/P manufacturers and the agency, including the Tissue Reference Group (TRG) Rapid Inquiry Program (TRIP). TRIP helped manufacturers of HCT/Ps, including stakeholders that market HCT/Ps to physicians or patients, obtain a rapid, preliminary, informal, non-binding assessment from the FDA regarding how specific HCT/Ps are regulated. TRIP was a temporary program of the TRG. The TRIP began in June 2019 and was extended twice. It recently ended on March 31, 2021.

Despite all of the FDAs efforts to engage industry, there continues to be broad marketing of these unapproved products for the treatment or cure of a wide range of diseases or medical conditions. Many of these unapproved products appear to be HCT/Ps that are regulated as drugs, devices and/or biological products subject to premarket approval requirements. The wide extent of the marketing of such unapproved products is evidenced by their inappropriate advertisement in various media and by the number of consumer complaints about them submitted to the FDA.

These regenerative medicine products are not without risk and are often marketed by clinics as being safe and effective for the treatment of a wide range of diseases or conditions, even though they havent been adequately studied in clinical trials. Weve said previously and want to reiterate here there is no room for manufacturers, clinics, or health care practitioners to place patients at risk through products that violate the law, including by not having an IND in effect or an approved biologics license. We will continue to take action regarding unlawfully marketed products. Our oversight of cellular and related products has included taking compliance actions, including numerous warning and untitled letters, and pursuing enforcement action for serious violations of the law.

Since December 2019, the agency has issued more than 350 letters to manufacturers, clinics, and health care providers, noting that it has come to our attention that they may be offering unapproved regenerative medicine products and reiterating the FDAs compliance and enforcement policy.

We encourage the public and patients who are considering treatment with regenerative medicine products to work with their health care providers to learn about the treatment being offered. Ask questions and understand the potential risks of treatment with unapproved products. It is critical to only seek treatment using legally marketed products, or, for unapproved products, to enroll in clinical trials under FDA oversight. The public can visit the FDAs website to find out if a particular regenerative medicine product is approved.

The FDA remains committed to helping advance the development of safe and effective regenerative medicine products, including stem cell-based products, to benefit individuals in need. We look forward to working with those who share this goal.

Originally posted here:
Advancing Safe and Effective Regenerative Medicine Products

Posted in Regenerative Medicine | Comments Off on Advancing Safe and Effective Regenerative Medicine Products

Can This Companys Research Help Transform Regenerative Medicine As Its …

Posted: December 18, 2022 at 12:49 am

Learn More about Organicell Regenerative Medicine, Inc. by gaining access to the latest research report

In the medical community, the popularity of regenerative medicine is reportedly soaring primarily because research around these therapies shows promise in treating a wide range of musculoskeletal pain conditions.

Armed with the belief that the human body can heal itself, regenerative medicine treatments like exosome therapy and stem cell therapy are advancing.

But regenerative medicine has sometimes been embroiled in controversy beginning in 1981 when scientistsdiscoveredways to derive embryonic stem cells from early mouse embryos.

That discovery was followed by a method to derive stem cells from human embryos and grow the cells in the laboratory in 1998.

Massive players likeAmgen Inc.AMGN,Sanofi SASNY, andGilead Sciences Inc.GILD have advanced stem cell research and therapies.

Unlike stem cell therapy, Organicells OCEL exosome therapy doesn'tinvolveusing donor cells or umbilical cords. Instead, the exosomes are extracted from full term, planned C Section, Amniotic fluid. The extracted exosome solution is derived from amniotic fluid and contains over 300 soluble proteins as growth factors, cytokines and chemokines, extracellular matrix proteins as fibronectin and collagen, structural molecules as hyaluronic acid, Nanoparticles as extracellular vesicles, exosomes and surfactants proteins, valuable lipids, micro-RNA, messenger-RNA, and cytokines.

Exosome therapy, according toexperts, is a highly targeted, flexible treatment believed to help inflammatory conditions like COPD, Long Covid, musculoskeletal injuries, osteoarthritis, and chronic pain.

Try to think of Stem Cells as microchips in a computer. Exosomes are like the software.

South Florida-basedOrganicell Regenerative Medicine Inc.OCEL is a company that is a pioneer in the exosome space. Their products are designed to operate like software for the computers of our body known as cells. However, the software Organicell uses is naturally occurring and unaltered. Organicell extracts its exosomes from what is believed to be one of the most naturally healing fluids on the planet, amniotic fluid. The software/exosomes in this fluid reminds the body what it used to do when it was young.

To put things plainly, the software (exosomes) is a set of instructions, data or programs used to operate the computers (cells) and execute specific tasks, while a computer-chip (stem cells) are used more as building blocks in regenerative medicine.

The potential benefits of Organicells products could be enormous. For example, the company's investigational productZofinis an acellular, non-HCT/P biologic, currently being studied in clinical trials. The company accomplished the difficult task of an approved investigational new drug (IND)from the U.S. Food and Drug Administration (FDA) which lead to approved clinical trials..

Zofin is derived from perinatal sources and is manufactured to retain the naturally occurring nanoparticles andmicroRNAswithout manipulation or adding or combining any other substance.

Zofin is currently being studied in FDAPhase I/II clinical trials for COVID-19, Long Covid, Chronic Obstructive Pulmonary Disease (COPD), and ready to begin a trial for Osteoarthritis of the knee.

Below are the results that were published on the COVID patients treated with Zofin under eIND approval. The important items in the chart are the key biomarkers that measure inflammation: IL-6 and CRP as well as the chest X Rays that show the structural difference in the lungs over time.

Organicellreports that its mission is to be the first company to prove the efficacy ofextracellular vesicles (EV's) a.k.a exosomes on inflammatory ailments. Organicell has two active FDA approved clinical trials for Long Covid and COPD.

The company says through its ground-breaking research in the field of nanotechnology, specifically perinatal-derived EVs, it has created the drug candidate, Zofin, that could be the next frontier of regenerative biologic therapeutics.

Organicells proprietary products biological representation is allogenic amniotic fluid (secreted body fluid, non-HCT/Ps), which is an acellular product derived from human amniotic fluid (HAF). The product contains no addition or combination of any other substance or diluent during production.

Moreover, the product quality assurance is seemingly unmatched in the regenerative medicine space. the donor from whom this product was derived has been tested and found negative for the following: HBsAg (Hepatitis B Surface Antigen), HBcAb (hepatitis B core antibody), HCV (hepatitis C antibody), HIV I/II-Ab (Antibody to Human Immunodeficiency Virus Types 1 and 2), Syphilis detection test, HIV NAT (HIV Nucleic Acid Test), and HCV NAT (HCV Nucleic Acid Test).

Additional donor screening tests may have been performed on the donor. If additional tests for Human Immunodeficiency Virus, Hepatitis B, Hepatitis C or Syphilis were performed, the results were reviewed and found to be NEGATIVE.

Additional tests for other communicable diseases, such as West Nile Virus, T. Cruzi, Cytomegalovirus and Epstein Barr Virus may have been performed. The results of all additional communicable disease tests have been evaluated by the Medical Director and have been found acceptable according to regulations, standards and Standard Operating Procedures (SOPs).

Donor screening tests are performed by laboratories certified under the Clinical Laboratory Improvement Amendments of 1988 (CLIA) and American Association of Blood Banks (AABB) using FDA licensed tests when available performed by VRL Laboratories.

The donor is selected based on medical and social history which meets the Standards of the American Association of Tissue Banks (AATB) United States Public Health Service (USPHS), and the Federal Food and Drug Administration (FDA).

Donor suitability was determined by the Manufacturing Facilities Medical Directors (Organicell Regenerative Medicine, 1951 NW 7th Ave, Suite 300, Miami, FL 33136). Organicell Factor X is manufactured for clinical use under cGMP-compliant manufacturing facility, to control potency and purity of the product.

This post contains sponsored advertising content. This content is for informational purposes only and is not intended to be investing advice.

Featured Photo by Shutterstock

Read the original post:
Can This Companys Research Help Transform Regenerative Medicine As Its ...

Posted in Regenerative Medicine | Comments Off on Can This Companys Research Help Transform Regenerative Medicine As Its …

Regenerative Medicine in Pharma Market Size, Share and Trends Analysis by Region, Drug Class, Route of Administration, and Segment Forecast, 2022-2027…

Posted: December 10, 2022 at 12:15 am

Regenerative Medicine in Pharma Market Size, Share and Trends Analysis by Region, Drug Class, Route of Administration, and Segment Forecast, 2022-2027  Longview News-JournalView Full Coverage on Google News

Excerpt from:
Regenerative Medicine in Pharma Market Size, Share and Trends Analysis by Region, Drug Class, Route of Administration, and Segment Forecast, 2022-2027...

Posted in Regenerative Medicine | Comments Off on Regenerative Medicine in Pharma Market Size, Share and Trends Analysis by Region, Drug Class, Route of Administration, and Segment Forecast, 2022-2027…

Stem Cells Applications in Regenerative Medicine and Disease …

Posted: November 16, 2022 at 2:34 am

Int J Cell Biol. 2016; 2016: 6940283.

Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, Madhya Pradesh 462066, India

Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, Madhya Pradesh 462066, India

Academic Editor: Paul J. Higgins

Received 2016 Mar 13; Accepted 2016 Jun 5.

This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Regenerative medicine, the most recent and emerging branch of medical science, deals with functional restoration of tissues or organs for the patient suffering from severe injuries or chronic disease. The spectacular progress in the field of stem cell research has laid the foundation for cell based therapies of disease which cannot be cured by conventional medicines. The indefinite self-renewal and potential to differentiate into other types of cells represent stem cells as frontiers of regenerative medicine. The transdifferentiating potential of stem cells varies with source and according to that regenerative applications also change. Advancements in gene editing and tissue engineering technology have endorsed the ex vivo remodelling of stem cells grown into 3D organoids and tissue structures for personalized applications. This review outlines the most recent advancement in transplantation and tissue engineering technologies of ESCs, TSPSCs, MSCs, UCSCs, BMSCs, and iPSCs in regenerative medicine. Additionally, this review also discusses stem cells regenerative application in wildlife conservation.

Regenerative medicine, the most recent and emerging branch of medical science, deals with functional restoration of specific tissue and/or organ of the patients suffering with severe injuries or chronic disease conditions, in the state where bodies own regenerative responses do not suffice [1]. In the present scenario donated tissues and organs cannot meet the transplantation demands of aged and diseased populations that have driven the thrust for search for the alternatives. Stem cells are endorsed with indefinite cell division potential, can transdifferentiate into other types of cells, and have emerged as frontline regenerative medicine source in recent time, for reparation of tissues and organs anomalies occurring due to congenital defects, disease, and age associated effects [1]. Stem cells pave foundation for all tissue and organ system of the body and mediates diverse role in disease progression, development, and tissue repair processes in host. On the basis of transdifferentiation potential, stem cells are of four types, that is, (1) unipotent, (2) multipotent, (3) pluripotent, and (4) totipotent [2]. Zygote, the only totipotent stem cell in human body, can give rise to whole organism through the process of transdifferentiation, while cells from inner cells mass (ICM) of embryo are pluripotent in their nature and can differentiate into cells representing three germ layers but do not differentiate into cells of extraembryonic tissue [2]. Stemness and transdifferentiation potential of the embryonic, extraembryonic, fetal, or adult stem cells depend on functional status of pluripotency factors like OCT4, cMYC, KLF44, NANOG, SOX2, and so forth [35]. Ectopic expression or functional restoration of endogenous pluripotency factors epigenetically transforms terminally differentiated cells into ESCs-like cells [3], known as induced pluripotent stem cells (iPSCs) [3, 4]. On the basis of regenerative applications, stem cells can be categorized as embryonic stem cells (ESCs), tissue specific progenitor stem cells (TSPSCs), mesenchymal stem cells (MSCs), umbilical cord stem cells (UCSCs), bone marrow stem cells (BMSCs), and iPSCs (; ). The transplantation of stem cells can be autologous, allogenic, and syngeneic for induction of tissue regeneration and immunolysis of pathogen or malignant cells. For avoiding the consequences of host-versus-graft rejections, tissue typing of human leucocyte antigens (HLA) for tissue and organ transplant as well as use of immune suppressant is recommended [6]. Stem cells express major histocompatibility complex (MHC) receptor in low and secret chemokine that recruitment of endothelial and immune cells is enabling tissue tolerance at graft site [6]. The current stem cell regenerative medicine approaches are founded onto tissue engineering technologies that combine the principles of cell transplantation, material science, and microengineering for development of organoid; those can be used for physiological restoration of damaged tissue and organs. The tissue engineering technology generates nascent tissue on biodegradable 3D-scaffolds [7, 8]. The ideal scaffolds support cell adhesion and ingrowths, mimic mechanics of target tissue, support angiogenesis and neovascularisation for appropriate tissue perfusion, and, being nonimmunogenic to host, do not require systemic immune suppressant [9]. Stem cells number in tissue transplant impacts upon regenerative outcome [10]; in that case prior ex vivo expansion of transplantable stem cells is required [11]. For successful regenerative outcomes, transplanted stem cells must survive, proliferate, and differentiate in site specific manner and integrate into host circulatory system [12]. This review provides framework of most recent (; Figures ) advancement in transplantation and tissue engineering technologies of ESCs, TSPSCs, MSCs, UCSCs, BMSCs, and iPSCs in regenerative medicine. Additionally, this review also discusses stem cells as the tool of regenerative applications in wildlife conservation.

Promises of stem cells in regenerative medicine: the six classes of stem cells, that is, embryonic stem cells (ESCs), tissue specific progenitor stem cells (TSPSCs), mesenchymal stem cells (MSCs), umbilical cord stem cells (UCSCs), bone marrow stem cells (BMSCs), and induced pluripotent stem cells (iPSCs), have many promises in regenerative medicine and disease therapeutics.

ESCs in regenerative medicine: ESCs, sourced from ICM of gastrula, have tremendous promises in regenerative medicine. These cells can differentiate into more than 200 types of cells representing three germ layers. With defined culture conditions, ESCs can be transformed into hepatocytes, retinal ganglion cells, chondrocytes, pancreatic progenitor cells, cone cells, cardiomyocytes, pacemaker cells, eggs, and sperms which can be used in regeneration of tissue and treatment of disease in tissue specific manner.

TSPSCs in regenerative medicine: tissue specific stem and progenitor cells have potential to differentiate into other cells of the tissue. Characteristically inner ear stem cells can be transformed into auditory hair cells, skin progenitors into vascular smooth muscle cells, mesoangioblasts into tibialis anterior muscles, and dental pulp stem cells into serotonin cells. The 3D-culture of TSPSCs in complex biomaterial gives rise to tissue organoids, such as pancreatic organoid from pancreatic progenitor, intestinal tissue organoids from intestinal progenitor cells, and fallopian tube organoids from fallopian tube epithelial cells. Transplantation of TSPSCs regenerates targets tissue such as regeneration of tibialis muscles from mesoangioblasts, cardiac tissue from AdSCs, and corneal tissue from limbal stem cells. Cell growth and transformation factors secreted by TSPSCs can change cells fate to become other types of cell, such that SSCs coculture with skin, prostate, and intestine mesenchyme transforms these cells from MSCs into epithelial cells fate.

MSCs in regenerative medicine: mesenchymal stem cells are CD73+, CD90+, CD105+, CD34, CD45, CD11b, CD14, CD19, and CD79a cells, also known as stromal cells. These bodily MSCs represented here do not account for MSCs of bone marrow and umbilical cord. Upon transplantation and transdifferentiation these bodily MSCs regenerate into cartilage, bones, and muscles tissue. Heart scar formed after heart attack and liver cirrhosis can be treated from MSCs. ECM coating provides the niche environment for MSCs to regenerate into hair follicle, stimulating hair growth.

UCSCs in regenerative medicine: umbilical cord, the readily available source of stem cells, has emerged as futuristic source for personalized stem cell therapy. Transplantation of UCSCs to Krabbe's disease patients regenerates myelin tissue and recovers neuroblastoma patients through restoring tissue homeostasis. The UCSCs organoids are readily available tissue source for treatment of neurodegenerative disease. Peritoneal fibrosis caused by long term dialysis, tendon tissue degeneration, and defective hyaline cartilage can be regenerated by UCSCs. Intravenous injection of UCSCs enables treatment of diabetes, spinal myelitis, systemic lupus erythematosus, Hodgkin's lymphoma, and congenital neuropathies. Cord blood stem cells banking avails long lasting source of stem cells for personalized therapy and regenerative medicine.

BMSCs in regenerative medicine: bone marrow, the soft sponge bone tissue that consisted of stromal, hematopoietic, and mesenchymal and progenitor stem cells, is responsible for blood formation. Even halo-HLA matched BMSCs can cure from disease and regenerate tissue. BMSCs can regenerate craniofacial tissue, brain tissue, diaphragm tissue, and liver tissue and restore erectile function and transdifferentiation monocytes. These multipotent stem cells can cure host from cancer and infection of HIV and HCV.

iPSCs in regenerative medicine: using the edge of iPSCs technology, skin fibroblasts and other adult tissues derived, terminally differentiated cells can be transformed into ESCs-like cells. It is possible that adult cells can be transformed into cells of distinct lineages bypassing the phase of pluripotency. The tissue specific defined culture can transform skin cells to become trophoblast, heart valve cells, photoreceptor cells, immune cells, melanocytes, and so forth. ECM complexation with iPSCs enables generation of tissue organoids for lung, kidney, brain, and other organs of the body. Similar to ESCs, iPSCs also can be transformed into cells representing three germ layers such as pacemaker cells and serotonin cells.

Stem cells in wildlife conservation: tissue biopsies obtained from dead and live wild animals can be either cryopreserved or transdifferentiated to other types of cells, through culture in defined culture medium or in vivo maturation. Stem cells and adult tissue derived iPSCs have great potential of regenerative medicine and disease therapeutics. Gonadal tissue procured from dead wild animals can be matured, ex vivo and in vivo for generation of sperm and egg, which can be used for assistive reproductive technology oriented captive breeding of wild animals or even for resurrection of wildlife.

Application of stem cells in regenerative medicine: stem cells (ESCs, TSPSCs, MSCs, UCSCs, BMSCs, and iPSCs) have diverse applications in tissue regeneration and disease therapeutics.

For the first time in 1998, Thomson isolated human ESCs (hESCs) [13]. ESCs are pluripotent in their nature and can give rise to more than 200 types of cells and promises for the treatment of any kinds of disease [13]. The pluripotency fate of ESCs is governed by functional dynamics of transcription factors OCT4, SOX2, NANOG, and so forth, which are termed as pluripotency factors. The two alleles of the OCT4 are held apart in pluripotency state in ESCs; phase through homologues pairing during embryogenesis and transdifferentiation processes [14] has been considered as critical regulatory switch for lineage commitment of ESCs. The diverse lineage commitment potential represents ESCs as ideal model for regenerative therapeutics of disease and tissue anomalies. This section of review on ESCs discusses transplantation and transdifferentiation of ESCs into retinal ganglion, hepatocytes, cardiomyocytes, pancreatic progenitors, chondrocytes, cones, egg sperm, and pacemaker cells (; ). Infection, cancer treatment, and accidents can cause spinal cord injuries (SCIs). The transplantation of hESCs to paraplegic or quadriplegic SCI patients improves body control, balance, sensation, and limbal movements [15], where transplanted stem cells do homing to injury sites. By birth, humans have fixed numbers of cone cells; degeneration of retinal pigment epithelium (RPE) of macula in central retina causes age-related macular degeneration (ARMD). The genomic incorporation of COCO gene (expressed during embryogenesis) in the developing embryo leads lineage commitment of ESCs into cone cells, through suppression of TGF, BMP, and Wnt signalling pathways. Transplantation of these cone cells to eye recovers individual from ARMD phenomenon, where transplanted cone cells migrate and form sheet-like structure in host retina [16]. However, establishment of missing neuronal connection of retinal ganglion cells (RGCs), cones, and PRE is the most challenging aspect of ARMD therapeutics. Recently, Donald Z Jacks group at John Hopkins University School of Medicine has generated RGCs from CRISPER-Cas9-m-Cherry reporter ESCs [17]. During ESCs transdifferentiation process, CRIPER-Cas9 directs the knock-in of m-Cherry reporter into 3UTR of BRN3B gene, which is specifically expressed in RGCs and can be used for purification of generated RGCs from other cells [17]. Furthermore, incorporation of forskolin in transdifferentiation regime boosts generation of RGCs. Coaxing of these RGCs into biomaterial scaffolds directs axonal differentiation of RGCs. Further modification in RGCs generation regime and composition of biomaterial scaffolds might enable restoration of vision for ARMD and glaucoma patients [17]. Globally, especially in India, cardiovascular problems are a more common cause of human death, where biomedical therapeutics require immediate restoration of heart functions for the very survival of the patient. Regeneration of cardiac tissue can be achieved by transplantation of cardiomyocytes, ESCs-derived cardiovascular progenitors, and bone marrow derived mononuclear cells (BMDMNCs); however healing by cardiomyocytes and progenitor cells is superior to BMDMNCs but mature cardiomyocytes have higher tissue healing potential, suppress heart arrhythmias, couple electromagnetically into hearts functions, and provide mechanical and electrical repair without any associated tumorigenic effects [18, 19]. Like CM differentiation, ESCs derived liver stem cells can be transformed into Cytp450-hepatocytes, mediating chemical modification and catabolism of toxic xenobiotic drugs [20]. Even today, availability and variability of functional hepatocytes are a major a challenge for testing drug toxicity [20]. Stimulation of ESCs and ex vivo VitK12 and lithocholic acid (a by-product of intestinal flora regulating drug metabolism during infancy) activates pregnane X receptor (PXR), CYP3A4, and CYP2C9, which leads to differentiation of ESCs into hepatocytes; those are functionally similar to primary hepatocytes, for their ability to produce albumin and apolipoprotein B100 [20]. These hepatocytes are excellent source for the endpoint screening of drugs for accurate prediction of clinical outcomes [20]. Generation of hepatic cells from ESCs can be achieved in multiple ways, as serum-free differentiation [21], chemical approaches [20, 22], and genetic transformation [23, 24]. These ESCs-derived hepatocytes are long lasting source for treatment of liver injuries and high throughput screening of drugs [20, 23, 24]. Transplantation of the inert biomaterial encapsulated hESCs-derived pancreatic progenitors (CD24+, CD49+, and CD133+) differentiates into -cells, minimizing high fat diet induced glycemic and obesity effects in mice [25] (). Addition of antidiabetic drugs into transdifferentiation regime can boost ESCs conservation into -cells [25], which theoretically can cure T2DM permanently [25]. ESCs can be differentiated directly into insulin secreting -cells (marked with GLUT2, INS1, GCK, and PDX1) which can be achieved through PDX1 mediated epigenetic reprogramming [26]. Globally, osteoarthritis affects millions of people and occurs when cartilage at joints wears away, causing stiffness of the joints. The available therapeutics for arthritis relieve symptoms but do not initiate reverse generation of cartilage. For young individuals and athletes replacement of joints is not feasible like old populations; in that case transplantation of stem cells represents an alternative for healing cartilage injuries [27]. Chondrocytes, the cartilage forming cells derived from hESC, embedded in fibrin gel effectively heal defective cartilage within 12 weeks, when transplanted to focal cartilage defects of knee joints in mice without any negative effect [27]. Transplanted chondrocytes form cell aggregates, positive for SOX9 and collagen II, and defined chondrocytes are active for more than 12wks at transplantation site, advocating clinical suitability of chondrocytes for treatment of cartilage lesions [27]. The integrity of ESCs to integrate and differentiate into electrophysiologically active cells provides a means for natural regulation of heart rhythm as biological pacemaker. Coaxing of ESCs into inert biomaterial as well as propagation in defined culture conditions leads to transdifferentiation of ESCs to become sinoatrial node (SAN) pacemaker cells (PCs) [28]. Genomic incorporation TBox3 into ESCs ex vivo leads to generation of PCs-like cells; those express activated leukocyte cells adhesion molecules (ALCAM) and exhibit similarity to PCs for gene expression and immune functions [28]. Transplantation of PCs can restore pacemaker functions of the ailing heart [28]. In summary, ESCs can be transdifferentiated into any kinds of cells representing three germ layers of the body, being most promising source of regenerative medicine for tissue regeneration and disease therapy (). Ethical concerns limit the applications of ESCs, where set guidelines need to be followed; in that case TSPSCs, MSCs, UCSCs, BMSCs, and iPSCs can be explored as alternatives.

TSPSCs maintain tissue homeostasis through continuous cell division, but, unlike ESCs, TSPSCs retain stem cells plasticity and differentiation in tissue specific manner, giving rise to few types of cells (). The number of TSPSCs population to total cells population is too low; in that case their harvesting as well as in vitro manipulation is really a tricky task [29], to explore them for therapeutic scale. Human body has foundation from various types of TSPSCs; discussing the therapeutic application for all types is not feasible. This section of review discusses therapeutic application of pancreatic progenitor cells (PPCs), dental pulp stem cells (DPSCs), inner ear stem cells (IESCs), intestinal progenitor cells (IPCs), limbal progenitor stem cells (LPSCs), epithelial progenitor stem cells (EPSCs), mesoangioblasts (MABs), spermatogonial stem cells (SSCs), the skin derived precursors (SKPs), and adipose derived stem cells (AdSCs) (; ). During embryogenesis PPCs give rise to insulin-producing -cells. The differentiation of PPCs to become -cells is negatively regulated by insulin [30]. PPCs require active FGF and Notch signalling; growing more rapidly in community than in single cell populations advocates the functional importance of niche effect in self-renewal and transdifferentiation processes. In 3D-scaffold culture system, mice embryo derived PPCs grow into hollow organoid spheres; those finally differentiate into insulin-producing -cell clusters [29]. The DSPSCs, responsible for maintenance of teeth health status, can be sourced from apical papilla, deciduous teeth, dental follicle, and periodontal ligaments, have emerged as regenerative medicine candidate, and might be explored for treatment of various kinds of disease including restoration neurogenic functions in teeth [31, 32]. Expansion of DSPSCs in chemically defined neuronal culture medium transforms them into a mixed population of cholinergic, GABAergic, and glutaminergic neurons; those are known to respond towards acetylcholine, GABA, and glutamine stimulations in vivo. These transformed neuronal cells express nestin, glial fibrillary acidic protein (GFAP), III-tubulin, and voltage gated L-type Ca2+ channels [32]. However, absence of Na+ and K+ channels does not support spontaneous action potential generation, necessary for response generation against environmental stimulus. All together, these primordial neuronal stem cells have possible therapeutic potential for treatment of neurodental problems [32]. Sometimes, brain tumor chemotherapy can cause neurodegeneration mediated cognitive impairment, a condition known as chemobrain [33]. The intrahippocampal transplantation of human derived neuronal stem cells to cyclophosphamide behavioural decremented mice restores cognitive functions in a month time. Here the transplanted stem cells differentiate into neuronal and astroglial lineage, reduce neuroinflammation, and restore microglial functions [33]. Furthermore, transplantation of stem cells, followed by chemotherapy, directs pyramidal and granule-cell neurons of the gyrus and CA1 subfields of hippocampus which leads to reduction in spine and dendritic cell density in the brain. These findings suggest that transplantation of stem cells to cranium restores cognitive functions of the chemobrain [33]. The hair cells of the auditory system produced during development are not postmitotic; loss of hair cells cannot be replaced by inner ear stem cells, due to active state of the Notch signalling [34]. Stimulation of inner ear progenitors with -secretase inhibitor ({"type":"entrez-nucleotide","attrs":{"text":"LY411575","term_id":"1257853995","term_text":"LY411575"}}LY411575) abrogates Notch signalling through activation of transcription factor atonal homologue 1 (Atoh1) and directs transdifferentiation of progenitors into cochlear hair cells [34]. Transplantation of in vitro generated hair cells restores acoustic functions in mice, which can be the potential regenerative medicine candidates for the treatment of deafness [34]. Generation of the hair cells also can be achieved through overexpression of -catenin and Atoh1 in Lrg5+ cells in vivo [35]. Similar to ear progenitors, intestine of the digestive tract also has its own tissue specific progenitor stem cells, mediating regeneration of the intestinal tissue [34, 36]. Dysregulation of the common stem cells signalling pathways, Notch/BMP/TGF-/Wnt, in the intestinal tissue leads to disease. Information on these signalling pathways [37] is critically important in designing therapeutics. Coaxing of the intestinal tissue specific progenitors with immune cells (macrophages), connective tissue cells (myofibroblasts), and probiotic bacteria into 3D-scaffolds of inert biomaterial, crafting biological environment, is suitable for differentiation of progenitors to occupy the crypt-villi structures into these scaffolds [36]. Omental implementation of these crypt-villi structures to dogs enhances intestinal mucosa through regeneration of goblet cells containing intestinal tissue [36]. These intestinal scaffolds are close approach for generation of implantable intestinal tissue, divested by infection, trauma, cancer, necrotizing enterocolitis (NEC), and so forth [36]. In vitro culture conditions cause differentiation of intestinal stem cells to become other types of cells, whereas incorporation of valproic acid and CHIR-99021 in culture conditions avoids differentiation of intestinal stem cells, enabling generation of indefinite pool of stem cells to be used for regenerative applications [38]. The limbal stem cells of the basal limbal epithelium, marked with ABCB5, are essential for regeneration and maintenance of corneal tissue [39]. Functional status of ABCB5 is critical for survival and functional integrity of limbal stem cells, protecting them from apoptotic cell death [39]. Limbal stem cells deficiency leads to replacement of corneal epithelium with visually dead conjunctival tissue, which can be contributed by burns, inflammation, and genetic factors [40]. Transplanted human cornea stem cells to mice regrown into fully functional human cornea, possibly supported by blood eye barrier phenomena, can be used for treatment of eye diseases, where regeneration of corneal tissue is critically required for vision restoration [39]. Muscle degenerative disease like duchenne muscular dystrophy (DMD) can cause extensive thrashing of muscle tissue, where tissue engineering technology can be deployed for functional restoration of tissue through regeneration [41]. Encapsulation of mouse or human derived MABs (engineered to express placental derived growth factor (PDGF)) into polyethylene glycol (PEG) fibrinogen hydrogel and their transplantation beneath the skin at ablated tibialis anterior form artificial muscles, which are functionally similar to those of normal tibialis anterior muscles [41]. The PDGF attracts various cell types of vasculogenic and neurogenic potential to the site of transplantation, supporting transdifferentiation of mesoangioblasts to become muscle fibrils [41]. The therapeutic application of MABs in skeletal muscle regeneration and other therapeutic outcomes has been reviewed by others [42]. One of the most important tissue specific stem cells, the male germline stem cells or spermatogonial stem cells (SSCs), produces spermatogenic lineage through mesenchymal and epithets cells [43] which itself creates niche effect on other cells. In vivo transplantation of SSCs with prostate, skin, and uterine mesenchyme leads to differentiation of these cells to become epithelia of the tissue of origin [43]. These newly formed tissues exhibit all physical and physiological characteristics of prostate and skin and the physical characteristics of prostate, skin, and uterus, express tissue specific markers, and suggest that factors secreted from SSCs lead to lineage conservation which defines the importance of niche effect in regenerative medicine [43]. According to an estimate, more than 100 million people are suffering from the condition of diabetic retinopathy, a progressive dropout of vascularisation in retina that leads to loss of vision [44]. The intravitreal injection of adipose derived stem cells (AdSCs) to the eye restores microvascular capillary bed in mice. The AdSCs from healthy donor produce higher amounts of vasoprotective factors compared to glycemic mice, enabling superior vascularisation [44]. However use of AdSCs for disease therapeutics needs further standardization for cell counts in dose of transplant and monitoring of therapeutic outcomes at population scale [44]. Apart from AdSCs, other kinds of stem cells also have therapeutic potential in regenerative medicine for treatment of eye defects, which has been reviewed by others [45]. Fallopian tubes, connecting ovaries to uterus, are the sites where fertilization of the egg takes place. Infection in fallopian tubes can lead to inflammation, tissue scarring, and closure of the fallopian tube which often leads to infertility and ectopic pregnancies. Fallopian is also the site where onset of ovarian cancer takes place. The studies on origin and etiology of ovarian cancer are restricted due to lack of technical advancement for culture of epithelial cells. The in vitro 3D organoid culture of clinically obtained fallopian tube epithelial cells retains their tissue specificity, keeps cells alive, which differentiate into typical ciliated and secretory cells of fallopian tube, and advocates that ectopic examination of fallopian tube in organoid culture settings might be the ideal approach for screening of cancer [46]. The sustained growth and differentiation of fallopian TSPSCs into fallopian tube organoid depend both on the active state of the Wnt and on paracrine Notch signalling [46]. Similar to fallopian tube stem cells, subcutaneous visceral tissue specific cardiac adipose (CA) derived stem cells (AdSCs) have the potential of differentiation into cardiovascular tissue [47]. Systemic infusion of CA-AdSCs into ischemic myocardium of mice regenerates heart tissue and improves cardiac function through differentiation to endothelial cells, vascular smooth cells, and cardiomyocytes and vascular smooth cells. The differentiation and heart regeneration potential of CA-AdSCs are higher than AdSCs [48], representing CA-AdSCs as potent regenerative medicine candidates for myocardial ischemic therapy [47]. The skin derived precursors (SKPs), the progenitors of dermal papilla/hair/hair sheath, give rise to multiple tissues of mesodermal and/or ectodermal origin such as neurons, Schwann cells, adipocytes, chondrocytes, and vascular smooth muscle cells (VSMCs). VSMCs mediate wound healing and angiogenesis process can be derived from human foreskin progenitor SKPs, suggesting that SKPs derived VSMCs are potential regenerative medicine candidates for wound healing and vasculature injuries treatments [49]. In summary, TSPSCs are potentiated with tissue regeneration, where advancement in organoid culture (; ) technologies defines the importance of niche effect in tissue regeneration and therapeutic outcomes of ex vivo expanded stem cells.

MSCs, the multilineage stem cells, differentiate only to tissue of mesodermal origin, which includes tendons, bone, cartilage, ligaments, muscles, and neurons [50]. MSCs are the cells which express combination of markers: CD73+, CD90+, CD105+, CD11b, CD14, CD19, CD34, CD45, CD79a, and HLA-DR, reviewed elsewhere [50]. The application of MSCs in regenerative medicine can be generalized from ongoing clinical trials, phasing through different state of completions, reviewed elsewhere [90]. This section of review outlines the most recent representative applications of MSCs (; ). The anatomical and physiological characteristics of both donor and receiver have equal impact on therapeutic outcomes. The bone marrow derived MSCs (BMDMSCs) from baboon are morphologically and phenotypically similar to those of bladder stem cells and can be used in regeneration of bladder tissue. The BMDMSCs (CD105+, CD73+, CD34, and CD45), expressing GFP reporter, coaxed with small intestinal submucosa (SIS) scaffolds, augment healing of degenerated bladder tissue within 10wks of the transplantation [51]. The combinatorial CD characterized MACs are functionally active at transplantation site, which suggests that CD characterization of donor MSCs yields superior regenerative outcomes [51]. MSCs also have potential to regenerate liver tissue and treat liver cirrhosis, reviewed elsewhere [91]. The regenerative medicinal application of MSCs utilizes cells in two formats as direct transplantation or first transdifferentiation and then transplantation; ex vivo transdifferentiation of MSCs deploys retroviral delivery system that can cause oncogenic effect on cells. Nonviral, NanoScript technology, comprising utility of transcription factors (TFs) functionalized gold nanoparticles, can target specific regulatory site in the genome effectively and direct differentiation of MSCs into another cell fate, depending on regime of TFs. For example, myogenic regulatory factor containing NanoScript-MRF differentiates the adipose tissue derived MSCs into muscle cells [92]. The multipotency characteristics represent MSCs as promising candidate for obtaining stable tissue constructs through coaxed 3D organoid culture; however heterogeneous distribution of MSCs slows down cell proliferation, rendering therapeutic applications of MSCs. Adopting two-step culture system for MSCs can yield homogeneous distribution of MSCs in biomaterial scaffolds. For example, fetal-MSCs coaxed in biomaterial when cultured first in rotating bioreactor followed with static culture lead to homogeneous distribution of MSCs in ECM components [7]. Occurrence of dental carries, periodontal disease, and tooth injury can impact individual's health, where bioengineering of teeth can be the alternative option. Coaxing of epithelial-MSCs with dental stem cells into synthetic polymer gives rise to mature teeth unit, which consisted of mature teeth and oral tissue, offering multiple regenerative therapeutics, reviewed elsewhere [52]. Like the tooth decay, both human and animals are prone to orthopedic injuries, affecting bones, joint, tendon, muscles, cartilage, and so forth. Although natural healing potential of bone is sufficient to heal the common injuries, severe trauma and tumor-recession can abrogate germinal potential of bone-forming stem cells. In vitro chondrogenic, osteogenic, and adipogenic potential of MSCs advocates therapeutic applications of MSCs in orthopedic injuries [53]. Seeding of MSCs, coaxed into biomaterial scaffolds, at defective bone tissue, regenerates defective bone tissues, within fourwks of transplantation; by the end of 32wks newly formed tissues integrate into old bone [54]. Osteoblasts, the bone-forming cells, have lesser actin cytoskeleton compared to adipocytes and MSCs. Treatment of MSCs with cytochalasin-D causes rapid transportation of G-actin, leading to osteogenic transformation of MSCs. Furthermore, injection of cytochalasin-D to mice tibia also promotes bone formation within a wk time frame [55]. The bone formation processes in mice, dog, and human are fundamentally similar, so outcomes of research on mice and dogs can be directional for regenerative application to human. Injection of MSCs to femur head of Legg-Calve-Perthes suffering dog heals the bone very fast and reduces the injury associated pain [55]. Degeneration of skeletal muscle and muscle cramps are very common to sledge dogs, animals, and individuals involved in adventurous athletics activities. Direct injection of adipose tissue derived MSCs to tear-site of semitendinosus muscle in dogs heals injuries much faster than traditional therapies [56]. Damage effect treatment for heart muscle regeneration is much more complex than regeneration of skeletal muscles, which needs high grade fine-tuned coordination of neurons with muscles. Coaxing of MSCs into alginate gel increases cell retention time that leads to releasing of tissue repairing factors in controlled manner. Transplantation of alginate encapsulated cells to mice heart reduces scar size and increases vascularisation, which leads to restoration of heart functions. Furthermore, transplanted MSCs face host inhospitable inflammatory immune responses and other mechanical forces at transplantation site, where encapsulation of cells keeps them away from all sorts of mechanical forces and enables sensing of host tissue microenvironment, and respond accordingly [57]. Ageing, disease, and medicine consumption can cause hair loss, known as alopecia. Although alopecia has no life threatening effects, emotional catchments can lead to psychological disturbance. The available treatments for alopecia include hair transplantation and use of drugs, where drugs are expensive to afford and generation of new hair follicle is challenging. Dermal papillary cells (DPCs), the specialized MSCs localized in hair follicle, are responsible for morphogenesis of hair follicle and hair cycling. The layer-by-layer coating of DPCs, called GAG coating, consists of coating of geletin as outer layer, middle layer of fibroblast growth factor 2 (FGF2) loaded alginate, and innermost layer of geletin. GAG coating creates tissue microenvironment for DPCs that can sustain immunological and mechanical obstacles, supporting generation of hair follicle. Transplantation of GAG-coated DPCs leads to abundant hair growth and maturation of hair follicle, where GAG coating serves as ECM, enhancing intrinsic therapeutic potential of DPCs [58]. During infection, the inflammatory cytokines secreted from host immune cells attract MSCs to the site of inflammation, which modulates inflammatory responses, representing MSCs as key candidate of regenerative medicine for infectious disease therapeutics. Coculture of macrophages (M) and adipose derived MSCs from Leishmania major (LM) susceptible and resistant mice demonstrates that AD-MSCs educate M against LM infection, differentially inducing M1 and M2 phenotype that represents AD-MSC as therapeutic agent for leishmanial therapy [93]. In summary, the multilineage differentiation potential of MSCs, as well as adoption of next-generation organoid culture system, avails MSCs as ideal regenerative medicine candidate.

Umbilical cord, generally thrown at the time of child birth, is the best known source for stem cells, procured in noninvasive manner, having lesser ethical constraints than ESCs. Umbilical cord is rich source of hematopoietic stem cells (HSCs) and MSCs, which possess enormous regeneration potential [94] (; ). The HSCs of cord blood are responsible for constant renewal of all types of blood cells and protective immune cells. The proliferation of HSCs is regulated by Musashi-2 protein mediated attenuation of Aryl hydrocarbon receptor (AHR) signalling in stem cells [95]. UCSCs can be cryopreserved at stem cells banks (; ), in operation by both private and public sector organization. Public stem cells banks operate on donation formats and perform rigorous screening for HLA typing and donated UCSCs remain available to anyone in need, whereas private stem cell banks operation is more personalized, availing cells according to donor consent. Stem cell banking is not so common, even in developed countries. Survey studies find that educated women are more eager to donate UCSCs, but willingness for donation decreases with subsequent deliveries, due to associated cost and safety concerns for preservation [96]. FDA has approved five HSCs for treatment of blood and other immunological complications [97]. The amniotic fluid, drawn during pregnancy for standard diagnostic purposes, is generally discarded without considering its vasculogenic potential. UCSCs are the best alternatives for those patients who lack donors with fully matched HLA typing for peripheral blood and PBMCs and bone marrow [98]. One major issue with UCSCs is number of cells in transplant, fewer cells in transplant require more time for engraftment to mature, and there are also risks of infection and mortality; in that case ex vivo propagation of UCSCs can meet the demand of desired outcomes. There are diverse protocols, available for ex vivo expansion of UCSCs, reviewed elsewhere [99]. Amniotic fluid stem cells (AFSCs), coaxed to fibrin (required for blood clotting, ECM interactions, wound healing, and angiogenesis) hydrogel and PEG supplemented with vascular endothelial growth factor (VEGF), give rise to vascularised tissue, when grafted to mice, suggesting that organoid cultures of UCSCs have promise for generation of biocompatible tissue patches, for treating infants born with congenital heart defects [59]. Retroviral integration of OCT4, KLF4, cMYC, and SOX2 transforms AFSCs into pluripotency stem cells known as AFiPSCs which can be directed to differentiate into extraembryonic trophoblast by BMP2 and BMP4 stimulation, which can be used for regeneration of placental tissues [60]. Wharton's jelly (WJ), the gelatinous substance inside umbilical cord, is rich in mucopolysaccharides, fibroblast, macrophages, and stem cells. The stem cells from UCB and WJ can be transdifferentiated into -cells. Homogeneous nature of WJ-SCs enables better differentiation into -cells; transplantation of these cells to streptozotocin induced diabetic mice efficiently brings glucose level to normal [7]. Easy access and expansion potential and plasticity to differentiate into multiple cell lineages represent WJ as an ideal candidate for regenerative medicine but cells viability changes with passages with maximum viable population at 5th-6th passages. So it is suggested to perform controlled expansion of WJ-MSCS for desired regenerative outcomes [9]. Study suggests that CD34+ expression leads to the best regenerative outcomes, with less chance of host-versus-graft rejection. In vitro expansion of UCSCs, in presence of StemRegenin-1 (SR-1), conditionally expands CD34+ cells [61]. In type I diabetic mellitus (T1DM), T-cell mediated autoimmune destruction of pancreatic -cells occurs, which has been considered as tough to treat. Transplantation of WJ-SCs to recent onset-T1DM patients restores pancreatic function, suggesting that WJ-MSCs are effective in regeneration of pancreatic tissue anomalies [62]. WJ-MSCs also have therapeutic importance for treatment of T2DM. A non-placebo controlled phase I/II clinical trial demonstrates that intravenous and intrapancreatic endovascular injection of WJ-MSCs to T2DM patients controls fasting glucose and glycated haemoglobin through improvement of -cells functions, evidenced by enhanced c-peptides and reduced inflammatory cytokines (IL-1 and IL-6) and T-cells counts [63]. Like diabetes, systematic lupus erythematosus (SLE) also can be treated with WJ-MSCs transplantation. During progression of SLE host immune system targets its own tissue leading to degeneration of renal, cardiovascular, neuronal, and musculoskeletal tissues. A non-placebo controlled follow-up study on 40 SLE patients demonstrates that intravenous infusion of WJ-MSC improves renal functions and decreases systematic lupus erythematosus disease activity index (SLEDAI) and British Isles Lupus Assessment Group (BILAG), and repeated infusion of WJ-MSCs protects the patient from relapse of the disease [64]. Sometimes, host inflammatory immune responses can be detrimental for HSCs transplantation and blood transfusion procedures. Infusion of WJ-MSC to patients, who had allogenic HSCs transplantation, reduces haemorrhage inflammation (HI) of bladder, suggesting that WJ-MSCs are potential stem cells adjuvant in HSCs transplantation and blood transfusion based therapies [100]. Apart from WJ, umbilical cord perivascular space and cord vein are also rich source for obtaining MSCs. The perivascular MSCs of umbilical cord are more primitive than WJ-MSCs and other MSCs from cord suggest that perivascular MSCs might be used as alternatives for WJ-MSCs for regenerative therapeutics outcome [101]. Based on origin, MSCs exhibit differential in vitro and in vivo properties and advocate functional characterization of MSCs, prior to regenerative applications. Emerging evidence suggests that UCSCs can heal brain injuries, caused by neurodegenerative diseases like Alzheimer's, Krabbe's disease, and so forth. Krabbe's disease, the infantile lysosomal storage disease, occurs due to deficiency of myelin synthesizing enzyme (MSE), affecting brain development and cognitive functions. Progression of neurodegeneration finally leads to death of babies aged two. Investigation shows that healing of peripheral nervous system (PNS) and central nervous system (CNS) tissues with Krabbe's disease can be achieved by allogenic UCSCs. UCSCs transplantation to asymptomatic infants with subsequent monitoring for 46 years reveals that UCSCs recover babies from MSE deficiency, improving myelination and cognitive functions, compared to those of symptomatic babies. The survival rate of transplanted UCSCs in asymptomatic and symptomatic infants was 100% and 43%, respectively, suggesting that early diagnosis and timely treatment are critical for UCSCs acceptance for desired therapeutic outcomes. UCSCs are more primitive than BMSCs, so perfect HLA typing is not critically required, representing UCSCs as an excellent source for treatment of all the diseases involving lysosomal defects, like Krabbe's disease, hurler syndrome, adrenoleukodystrophy (ALD), metachromatic leukodystrophy (MLD), Tay-Sachs disease (TSD), and Sandhoff disease [65]. Brain injuries often lead to cavities formation, which can be treated from neuronal parenchyma, generated ex vivo from UCSCs. Coaxing of UCSCs into human originated biodegradable matrix scaffold and in vitro expansion of cells in defined culture conditions lead to formation of neuronal organoids, within threewks' time frame. These organoids structurally resemble brain tissue and consisted of neuroblasts (GFAP+, Nestin+, and Ki67+) and immature stem cells (OCT4+ and SOX2+). The neuroblasts of these organoids further can be differentiated into mature neurons (MAP2+ and TUJ1+) [66]. Administration of high dose of drugs in divesting neuroblastoma therapeutics requires immediate restoration of hematopoiesis. Although BMSCs had been promising in restoration of hematopoiesis UCSCs are sparely used in clinical settings. A case study demonstrates that neuroblastoma patients who received autologous UCSCs survive without any associated side effects [12]. During radiation therapy of neoplasm, spinal cord myelitis can occur, although occurrence of myelitis is a rare event and usually such neurodegenerative complication of spinal cord occurs 624 years after exposure to radiations. Transplantation of allogenic UC-MSCs in laryngeal patients undergoing radiation therapy restores myelination [102]. For treatment of neurodegenerative disease like Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), traumatic brain injuries (TBI), Parkinson's, SCI, stroke, and so forth, distribution of transplanted UCSCs is critical for therapeutic outcomes. In mice and rat, injection of UCSCs and subsequent MRI scanning show that transplanted UCSCs migrate to CNS and multiple peripheral organs [67]. For immunomodulation of tumor cells disease recovery, transplantation of allogenic DCs is required. The CD11c+DCs, derived from UCB, are morphologically and phenotypically similar to those of peripheral blood derived CTLs-DCs, suggesting that UCB-DCs can be used for personalized medicine of cancer patient, in need for DCs transplantation [103]. Coculture of UCSCs with radiation exposed human lung fibroblast stops their transdifferentiation, which suggests that factors secreted from UCSCs may restore niche identity of fibroblast, if they are transplanted to lung after radiation therapy [104]. Tearing of shoulder cuff tendon can cause severe pain and functional disability, whereas ultrasound guided transplantation of UCB-MSCs in rabbit regenerates subscapularis tendon in fourwks' time frame, suggesting that UCB-MSCs are effective enough to treat tendons injuries when injected to focal points of tear-site [68]. Furthermore, transplantation of UCB-MSCs to chondral cartilage injuries site in pig knee along with HA hydrogel composite regenerates hyaline cartilage [69], suggesting that UCB-MSCs are effective regenerative medicine candidate for treating cartilage and ligament injuries. Physiologically circulatory systems of brain, placenta, and lungs are similar. Infusion of UCB-MSCs to preeclampsia (PE) induced hypertension mice reduces the endotoxic effect, suggesting that UC-MSCs are potential source for treatment of endotoxin induced hypertension during pregnancy, drug abuse, and other kinds of inflammatory shocks [105]. Transplantation of UCSCs to severe congenital neutropenia (SCN) patients restores neutrophils count from donor cells without any side effect, representing UCSCs as potential alternative for SCN therapy, when HLA matched bone marrow donors are not accessible [106]. In clinical settings, the success of myocardial infarction (MI) treatment depends on ageing, systemic inflammation in host, and processing of cells for infusion. Infusion of human hyaluronan hydrogel coaxed UCSCs in pigs induces angiogenesis, decreases scar area, improves cardiac function at preclinical level, and suggests that the same strategy might be effective for human [107]. In stem cells therapeutics, UCSCs transplantation can be either autologous or allogenic. Sometimes, the autologous UCSCs transplants cannot combat over tumor relapse, observed in Hodgkin's lymphoma (HL), which might require second dose transplantation of allogenic stem cells, but efficacy and tolerance of stem cells transplant need to be addressed, where tumor replace occurs. A case study demonstrates that second dose allogenic transplants of UCSCs effective for HL patients, who had heavy dose in prior transplant, increase the long term survival chances by 30% [10]. Patients undergoing long term peritoneal renal dialysis are prone to peritoneal fibrosis and can change peritoneal structure and failure of ultrafiltration processes. The intraperitoneal (IP) injection of WJ-MSCs prevents methylglyoxal induced programmed cell death and peritoneal wall thickening and fibrosis, suggesting that WJ-MSCs are effective in therapeutics of encapsulating peritoneal fibrosis [70]. In summary, UCB-HSCs, WJ-MSCs, perivascular MSCs, and UCB-MSCs have tissue regeneration potential.

Bone marrow found in soft spongy bones is responsible for formation of all peripheral blood and comprises hematopoietic stem cells (producing blood cells) and stromal cells (producing fat, cartilage, and bones) [108] (; ). Visually bone marrow has two types, red marrow (myeloid tissue; producing RBC, platelets, and most of WBC) and yellow marrow (producing fat cells and some WBC) [108]. Imbalance in marrow composition can culminate to the diseased condition. Since 1980, bone marrow transplantation is widely accepted for cancer therapeutics [109]. In order to avoid graft rejection, HLA typing of donors is a must, but completely matched donors are limited to family members, which hampers allogenic transplantation applications. Since matching of all HLA antigens is not critically required, in that case defining the critical antigens for haploidentical allogenic donor for patients, who cannot find fully matched donor, might relieve from donor constraints. Two-step administration of lymphoid and myeloid BMSCs from haploidentical donor to the patients of aplastic anaemia and haematological malignancies reconstructs host immune system and the outcomes are almost similar to fully matched transplants, which recommends that profiling of critically important HLA is sufficient for successful outcomes of BMSCs transplantation. Haploidentical HLA matching protocol is the major process for minorities and others who do not have access to matched donor [71]. Furthermore, antigen profiling is not the sole concern for BMSCs based therapeutics. For example, restriction of HIV1 (human immune deficiency virus) infection is not feasible through BMSCs transplantation because HIV1 infection is mediated through CD4+ receptors, chemokine CXC motif receptor 4 (CXCR4), and chemokine receptor 5 (CCR5) for infecting and propagating into T helper (Th), monocytes, macrophages, and dendritic cells (DCs). Genetic variation in CCR2 and CCR5 receptors is also a contributory factor; mediating protection against infection has been reviewed elsewhere [110]. Engineering of hematopoietic stem and progenitor cells (HSPCs) derived CD4+ cells to express HIV1 antagonistic RNA, specifically designed for targeting HIV1 genome, can restrict HIV1 infection, through immune elimination of latently infected CD4+ cells. A single dose infusion of genetically modified (GM), HIV1 resistant HSPCs can be the alternative of HIV1 retroviral therapy. In the present scenario stem cells source, patient selection, transplantation-conditioning regimen, and postinfusion follow-up studies are the major factors, which can limit application of HIV1 resistant GM-HSPCs (CD4+) cells application in AIDS therapy [72, 73]. Platelets, essential for blood clotting, are formed from megakaryocytes inside the bone marrow [74]. Due to infection, trauma, and cancer, there are chances of bone marrow failure. To an extent, spongy bone marrow microenvironment responsible for lineage commitment can be reconstructed ex vivo [75]. The ex vivo constructed 3D-scaffolds consisted of microtubule and silk sponge, flooded with chemically defined organ culture medium, which mimics bone marrow environment. The coculture of megakaryocytes and embryonic stem cells (ESCs) in this microenvironment leads to generation of functional platelets from megakaryocytes [75]. The ex vivo 3D-scaffolds of bone microenvironment can stride the path for generation of platelets in therapeutic quantities for regenerative medication of burns [75] and blood clotting associated defects. Accidents, traumatic injuries, and brain stroke can deplete neuronal stem cells (NSCs), responsible for generation of neurons, astrocytes, and oligodendrocytes. Brain does not repopulate NSCs and heal traumatic injuries itself and transplantation of BMSCs also can heal neurodegeneration alone. Lipoic acid (LA), a known pharmacological antioxidant compound used in treatment of diabetic and multiple sclerosis neuropathy when combined with BMSCs, induces neovascularisation at focal cerebral injuries, within 8wks of transplantation. Vascularisation further attracts microglia and induces their colonization into scaffold, which leads to differentiation of BMSCs to become brain tissue, within 16wks of transplantation. In this approach, healing of tissue directly depends on number of BMSCs in transplantation dose [76]. Dental caries and periodontal disease are common craniofacial disease, often requiring jaw bone reconstruction after removal of the teeth. Traditional therapy focuses on functional and structural restoration of oral tissue, bone, and teeth rather than biological restoration, but BMSCs based therapies promise for regeneration of craniofacial bone defects, enabling replacement of missing teeth in restored bones with dental implants. Bone marrow derived CD14+ and CD90+ stem and progenitor cells, termed as tissue repair cells (TRC), accelerate alveolar bone regeneration and reconstruction of jaw bone when transplanted in damaged craniofacial tissue, earlier to oral implants. Hence, TRC therapy reduces the need of secondary bone grafts, best suited for severe defects in oral bone, skin, and gum, resulting from trauma, disease, or birth defects [77]. Overall, HSCs have great value in regenerative medicine, where stem cells transplantation strategies explore importance of niche in tissue regeneration. Prior to transplantation of BMSCs, clearance of original niche from target tissue is necessary for generation of organoid and organs without host-versus-graft rejection events. Some genetic defects can lead to disorganization of niche, leading to developmental errors. Complementation with human blastocyst derived primary cells can restore niche function of pancreas in pigs and rats, which defines the concept for generation of clinical grade human pancreas in mice and pigs [111]. Similar to other organs, diaphragm also has its own niche. Congenital defects in diaphragm can affect diaphragm functions. In the present scenario functional restoration of congenital diaphragm defects by surgical repair has risk of reoccurrence of defects or incomplete restoration [8]. Decellularization of donor derived diaphragm offers a way for reconstruction of new and functionally compatible diaphragm through niche modulation. Tissue engineering technology based decellularization of diaphragm and simultaneous perfusion of bone marrow mesenchymal stem cells (BM-MSCs) facilitates regeneration of functional scaffolds of diaphragm tissues [8]. In vivo replacement of hemidiaphragm in rats with reseeded scaffolds possesses similar myography and spirometry as it has in vivo in donor rats. These scaffolds retaining natural architecture are devoid of immune cells, retaining intact extracellular matrix that supports adhesion, proliferation, and differentiation of seeded cells [8]. These findings suggest that cadaver obtained diaphragm, seeded with BM-MSCs, can be used for curing patients in need for restoration of diaphragm functions (; ). However, BMSCs are heterogeneous population, which might result in differential outcomes in clinical settings; however clonal expansion of BMSCs yields homogenous cells population for therapeutic application [8]. One study also finds that intracavernous delivery of single clone BMSCs can restore erectile function in diabetic mice [112] and the same strategy might be explored for adult human individuals. The infection of hepatitis C virus (HCV) can cause liver cirrhosis and degeneration of hepatic tissue. The intraparenchymal transplantation of bone marrow mononuclear cells (BMMNCs) into liver tissue decreases aspartate aminotransferase (AST), alanine transaminase (ALT), bilirubin, CD34, and -SMA, suggesting that transplanted BMSCs restore hepatic functions through regeneration of hepatic tissues [113]. In order to meet the growing demand for stem cells transplantation therapy, donor encouragement is always required [8]. The stem cells donation procedure is very simple; with consent donor gets an injection of granulocyte-colony stimulating factor (G-CSF) that increases BMSCs population. Bone marrow collection is done from hip bone using syringe in 4-5hrs, requiring local anaesthesia and within a wk time frame donor gets recovered donation associated weakness.

The field of iPSCs technology and research is new to all other stem cells research, emerging in 2006 when, for the first time, Takahashi and Yamanaka generated ESCs-like cells through genetic incorporation of four factors, Sox2, Oct3/4, Klf4, and c-Myc, into skin fibroblast [3]. Due to extensive nuclear reprogramming, generated iPSCs are indistinguishable from ESCs, for their transcriptome profiling, epigenetic markings, and functional competence [3], but use of retrovirus in transdifferentiation approach has questioned iPSCs technology. Technological advancement has enabled generation of iPSCs from various kinds of adult cells phasing through ESCs or direct transdifferentiation. This section of review outlines most recent advancement in iPSC technology and regenerative applications (; ). Using the new edge of iPSCs technology, terminally differentiated skin cells directly can be transformed into kidney organoids [114], which are functionally and structurally similar to those of kidney tissue in vivo. Up to certain extent kidneys heal themselves; however natural regeneration potential cannot meet healing for severe injuries. During kidneys healing process, a progenitor stem cell needs to become 20 types of cells, required for waste excretion, pH regulation, and restoration of water and electrolytic ions. The procedure for generation of kidney organoids ex vivo, containing functional nephrons, has been identified for human. These ex vivo kidney organoids are similar to fetal first-trimester kidneys for their structure and physiology. Such kidney organoids can serve as model for nephrotoxicity screening of drugs, disease modelling, and organ transplantation. However generation of fully functional kidneys is a far seen event with today's scientific technologies [114]. Loss of neurons in age-related macular degeneration (ARMD) is the common cause of blindness. At preclinical level, transplantation of iPSCs derived neuronal progenitor cells (NPCs) in rat limits progression of disease through generation of 5-6 layers of photoreceptor nuclei, restoring visual acuity [78]. The various approaches of iPSCs mediated retinal regeneration including ARMD have been reviewed elsewhere [79]. Placenta, the cordial connection between mother and developing fetus, gets degenerated in certain pathophysiological conditions. Nuclear programming of OCT4 knock-out (KO) and wild type (WT) mice fibroblast through transient expression of GATA3, EOMES, TFAP2C, and +/ cMYC generates transgene independent trophoblast stem-like cells (iTSCs), which are highly similar to blastocyst derived TSCs for DNA methylation, H3K7ac, nucleosome deposition of H2A.X, and other epigenetic markings. Chimeric differentiation of iTSCs specifically gives rise to haemorrhagic lineages and placental tissue, bypassing pluripotency phase, opening an avenue for generation of fully functional placenta for human [115]. Neurodegenerative disease like Alzheimer's and obstinate epilepsies can degenerate cerebrum, controlling excitatory and inhibitory signals of the brain. The inhibitory tones in cerebral cortex and hippocampus are accounted by -amino butyric acid secreting (GABAergic) interneurons (INs). Loss of these neurons often leads to progressive neurodegeneration. Genomic integration of Ascl1, Dlx5, Foxg1, and Lhx6 to mice and human fibroblast transforms these adult cells into GABAergic-INs (iGABA-INs). These cells have molecular signature of telencephalic INs, release GABA, and show inhibition to host granule neuronal activity [81]. Transplantation of these INs in developing embryo cures from genetic and acquired seizures, where transplanted cells disperse and mature into functional neuronal circuits as local INs [82]. Dorsomorphin and SB-431542 mediated inhibition of TGF- and BMP signalling direct transformation of human iPSCs into cortical spheroids. These cortical spheroids consisted of both peripheral and cortical neurons, surrounded by astrocytes, displaying transcription profiling and electrophysiology similarity with developing fetal brain and mature neurons, respectively [83]. The underlying complex biology and lack of clear etiology and genetic reprogramming and difficulty in recapitulation of brain development have barred understanding of pathophysiology of autism spectrum disorder (ASD) and schizophrenia. 3D organoid cultures of ASD patient derived iPSC generate miniature brain organoid, resembling fetal brain few months after gestation. The idiopathic conditions of these organoids are similar with brain of ASD patients; both possess higher inhibitory GABAergic neurons with imbalanced neuronal connection. Furthermore these organoids express forkhead Box G1 (FOXG1) much higher than normal brain tissue, which explains that FOXG1 might be the leading cause of ASD [84]. Degeneration of other organs and tissues also has been reported, like degeneration of lungs which might occur due to tuberculosis infection, fibrosis, and cancer. The underlying etiology for lung degeneration can be explained through organoid culture. Coaxing of iPSC into inert biomaterial and defined culture leads to formation of lung organoids that consisted of epithelial and mesenchymal cells, which can survive in culture for months. These organoids are miniature lung, resemble tissues of large airways and alveoli, and can be used for lung developmental studies and screening of antituberculosis and anticancer drugs [87]. The conventional multistep reprogramming for iPSCs consumes months of time, while CRISPER-Cas9 system based episomal reprogramming system that combines two steps together enables generation of ESCs-like cells in less than twowks, reducing the chances of culture associated genetic abrasions and unwanted epigenetic [80]. This approach can yield single step ESCs-like cells in more personalized way from adults with retinal degradation and infants with severe immunodeficiency, involving correction for genetic mutation of OCT4 and DNMT3B [80]. The iPSCs expressing anti-CCR5-RNA, which can be differentiated into HIV1 resistant macrophages, have applications in AIDS therapeutics [88]. The diversified immunotherapeutic application of iPSCs has been reviewed elsewhere [89]. The -1 antitrypsin deficiency (A1AD) encoded by serpin peptidase inhibitor clade A member 1 (SERPINA1) protein synthesized in liver protects lungs from neutrophils elastase, the enzyme causing disruption of lungs connective tissue. A1AD deficiency is common cause of both lung and liver disease like chronic obstructive pulmonary disease (COPD) and liver cirrhosis. Patient specific iPSCs from lung and liver cells might explain pathophysiology of A1AD deficiency. COPD patient derived iPSCs show sensitivity to toxic drugs which explains that actual patient might be sensitive in similar fashion. It is known that A1AD deficiency is caused by single base pair mutation and correction of this mutation fixes the A1AD deficiency in hepatic-iPSCs [85]. The high order brain functions, like emotions, anxiety, sleep, depression, appetite, breathing heartbeats, and so forth, are regulated by serotonin neurons. Generation of serotonin neurons occurs prior to birth, which are postmitotic in their nature. Any sort of developmental defect and degeneration of serotonin neurons might lead to neuronal disorders like bipolar disorder, depression, and schizophrenia-like psychiatric conditions. Manipulation of Wnt signalling in human iPSCs in defined culture conditions leads to an in vitro differentiation of iPSCs to serotonin-like neurons. These iPSCs-neurons primarily localize to rhombomere 2-3 segment of rostral raphe nucleus, exhibit electrophysiological properties similar to serotonin neurons, express hydroxylase 2, the developmental marker, and release serotonin in dose and time dependent manner. Transplantation of these neurons might cure from schizophrenia, bipolar disorder, and other neuropathological conditions [116]. The iPSCs technology mediated somatic cell reprogramming of ventricular monocytes results in generation of cells, similar in morphology and functionality with PCs. SA note transplantation of PCs to large animals improves rhythmic heart functions. Pacemaker needs very reliable and robust performance so understanding of transformation process and site of transplantation are the critical aspect for therapeutic validation of iPSCs derived PCs [28]. Diabetes is a major health concern in modern world, and generation of -cells from adult tissue is challenging. Direct reprogramming of skin cells into pancreatic cells, bypassing pluripotency phase, can yield clinical grade -cells. This reprogramming strategy involves transformation of skin cells into definitive endodermal progenitors (cDE) and foregut like progenitor cells (cPF) intermediates and subsequent in vitro expansion of these intermediates to become pancreatic -cells (cPB). The first step is chemically complex and can be understood as nonepisomal reprogramming on day one with pluripotency factors (OCT4, SOX2, KLF4, and hair pin RNA against p53), then supplementation with GFs and chemical supplements on day seven (EGF, bFGF, CHIR, NECA, NaB, Par, and RG), and two weeks later (Activin-A, CHIR, NECA, NaB, and RG) yielding DE and cPF [86]. Transplantation of cPB yields into glucose stimulated secretion of insulin in diabetic mice defines that such cells can be explored for treatment of T1DM and T2DM in more personalized manner [86]. iPSCs represent underrated opportunities for drug industries and clinical research laboratories for development of therapeutics, but safety concerns might limit transplantation applications (; ) [117]. Transplantation of human iPSCs into mice gastrula leads to colonization and differentiation of cells into three germ layers, evidenced with clinical developmental fat measurements. The acceptance of human iPSCs by mice gastrula suggests that correct timing and appropriate reprogramming regime might delimit human mice species barrier. Using this fact of species barrier, generation of human organs in closely associated primates might be possible, which can be used for treatment of genetic factors governed disease at embryo level itself [118]. In summary, iPSCs are safe and effective for treatment of regenerative medicine.

The unstable growth of human population threatens the existence of wildlife, through overexploitation of natural habitats and illegal killing of wild animals, leading many species to face the fate of being endangered and go for extinction. For wildlife conservation, the concept of creation of frozen zoo involves preservation of gene pool and germ plasm from threatened and endangered species (). The frozen zoo tissue samples collection from dead or live animal can be DNA, sperms, eggs, embryos, gonads, skin, or any other tissue of the body [119]. Preserved tissue can be reprogrammed or transdifferentiated to become other types of tissues and cells, which opens an avenue for conservation of endangered species and resurrection of life (). The gonadal tissue from young individuals harbouring immature tissue can be matured in vivo and ex vivo for generation of functional gametes. Transplantation of SSCs to testis of male from the same different species can give rise to spermatozoa of donor cells [120], which might be used for IVF based captive breeding of wild animals. The most dangerous fact in wildlife conservation is low genetic diversity, too few reproductively capable animals which cannot maintain adequate genetic diversity in wild or captivity. Using the edge of iPSC technology, pluripotent stem cells can be generated from skin cells. For endangered drill, Mandrillus leucophaeus, and nearly extinct white rhinoceros, Ceratotherium simum cottoni, iPSC has been generated in 2011 [121]. The endangered animal drill (Mandrillus leucophaeus) is genetically very close to human and often suffers from diabetes, while rhinos are genetically far removed from other primates. The progress in iPSCs, from the human point of view, might be transformed for animal research for recapturing reproductive potential and health in wild animals. However, stem cells based interventions in wild animals are much more complex than classical conservation planning and biomedical research has to face. Conversion of iPSC into egg or sperm can open the door for generation of IVF based embryo; those might be transplanted in womb of live counterparts for propagation of population. Recently, iPSCs have been generated for snow leopard (Panthera uncia), native to mountain ranges of central Asia, which belongs to cat family; this breakthrough has raised the possibilities for cryopreservation of genetic material for future cloning and other assisted reproductive technology (ART) applications, for the conservation of cat species and biodiversity. Generation of leopard iPSCs has been achieved through retroviral-system based genomic integration of OCT4, SOX2, KLF4, cMYC, and NANOG. These iPSCs from snow leopard also open an avenue for further transformation of iPSCs into gametes [122]. The in vivo maturation of grafted tissue depends both on age and on hormonal status of donor tissue. These facts are equally applicable to accepting host. Ectopic xenografts of cryopreserved testis tissue from Indian spotted deer (Moschiola indica) to nude mice yielded generation of spermatocytes [123], suggesting that one-day procurement of functional sperm from premature tissue might become a general technique in wildlife conservation. In summary, tissue biopsies from dead or live animals can be used for generation of iPSCs and functional gametes; those can be used in assisted reproductive technology (ART) for wildlife conservation.

The spectacular progress in the field of stem cells research represents great scope of stem cells regenerative therapeutics. It can be estimated that by 2020 or so we will be able to produce wide array of tissue, organoid, and organs from adult stem cells. Inductions of pluripotency phenotypes in terminally differentiated adult cells have better therapeutic future than ESCs, due to least ethical constraints with adult cells. In the coming future, there might be new pharmaceutical compounds; those can activate tissue specific stem cells, promote stem cells to migrate to the side of tissue injury, and promote their differentiation to tissue specific cells. Except few countries, the ongoing financial and ethical hindrance on ESCs application in regenerative medicine have more chance for funding agencies to distribute funding for the least risky projects on UCSCs, BMSCs, and TSPSCs from biopsies. The existing stem cells therapeutics advancements are more experimental and high in cost; due to that application on broad scale is not feasible in current scenario. In the near future, the advancements of medical science presume using stem cells to treat cancer, muscles damage, autoimmune disease, and spinal cord injuries among a number of impairments and diseases. It is expected that stem cells therapies will bring considerable benefits to the patients suffering from wide range of injuries and disease. There is high optimism for use of BMSCs, TSPSCs, and iPSCs for treatment of various diseases to overcome the contradictions associated with ESCs. For advancement of translational application of stem cells, there is a need of clinical trials, which needs funding rejoinder from both public and private organizations. The critical evaluation of regulatory guidelines at each phase of clinical trial is a must to comprehend the success and efficacy in time frame.

Dr. Anuradha Reddy from Centre for Cellular and Molecular Biology Hyderabad and Mrs. Sarita Kumari from Department of Yoga Science, BU, Bhopal, India, are acknowledged for their critical suggestions and comments on paper.

There are no competing interests associated with this paper.

See the original post here:
Stem Cells Applications in Regenerative Medicine and Disease ...

Posted in Regenerative Medicine | Comments Off on Stem Cells Applications in Regenerative Medicine and Disease …

Neuroregeneration – Center for Regenerative Biotherapeutics – Mayo …

Posted: November 16, 2022 at 2:34 am

Spinal neuron growth cones

Fluorescent spinal neurons in the developing Xenopus embryo

Hippocampal neuron immunostained to reveal green microtubule cytoskeleton

Nerve muscle co-culture

Contact adhesions in the nerve growth cone (paxillin in red, microtubules in green)

Substrate adhesions in the growth cone induced by brain derived neurotrophic factor

The complex, delicate structures that make up the nervous system the brain, spinal cord and peripheral nerves are susceptible to various types of injury ranging from trauma to neurodegenerative diseases that cause progressive deterioration: Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis (ALS, also known as Lou Gehrig's disease), multiple sclerosis and multiple system atrophy.

Unfortunately, because of the complexity of the brain and spinal cord, little spontaneous regeneration, repair or healing occurs. Therefore, brain damage, paralysis from spinal cord injury and peripheral nerve damage are often permanent and incapacitating.

Patients with serious nervous system injuries or strokes often require lifelong assistance, which puts a tremendous burden on patients, their families and society. Innovative, paradigm-shifting strategies are required to advance treatment of neurological injury. The neuroregeneration research at Mayo Clinic is at the forefront of healing the nervous system.

For an in-depth look at neuroregeneration, see the neuroregenerative medicine at Mayo Clinic booklet.

Mayo Clinic clinicians, scientists, engineers and other specialists in the Center for Regenerative Biotherapeutics are taking a multidisciplinary integrative approach to neuroregeneration for a number of devastating neurological conditions. The research is multifaceted, ranging from basic science discovery to clinical applications.

Alzheimer's disease. Alzheimer's disease is the major cause of dementia in older adults, with progressive loss of neurons in areas of the brain responsible for learning and memory. Efforts in Alzheimer's disease research focus on understanding why neurons degenerate in brains with Alzheimer's disease and how to either slow down the process or replace lost neurons.

Mayo researchers are investigating the effects of restoring cerebrovascular function, through transplantation of induced pluripotent stem (iPS) cell-derived vascular progenitor cells, on amyloid pathology and cognitive function in amyloid Alzheimer's disease model mice. The iPS cells converted from skin fibroblasts by transducing four transcription factors (Oct3/4, SOX2, Klf4, c-Myc) have the potential to generate all tissues in the body, including vascular cells.

This innovative approach will likely allow for rational designs of vascular regenerative therapy against Alzheimer's disease.

Anthony J. Windebank, M.D., and Nathan P. Staff, M.D., Ph.D., both neurologists and researchers at Mayo Clinic, discuss the latest research into ALS treatments.

Multiple sclerosis. While scientists understand much about the damage that happens to nerves and their insulating sheath (myelin) during multiple sclerosis (MS)and how the immune system causes this damage, the exact reasons for the immune system attack are very poorly understood. The lack of understanding of the exact cause of MS is a challenge for the development of effective therapies, and Mayo Clinic laboratories are working to better understand this disease.

Protecting nerves and myelin from damage, or repairing myelin after it's been damaged, also holds potential for the treatment of MS. Injury to nerves and myelin can be severe in MS and is the major cause of functional impairments. However, spontaneous repair of this damage is sometimes observed in people with MS. Researchers in the Center for Regenerative Biotherapeutics are actively engaged in developing therapies designed to stimulate this repair and thereby promote recovery of lost function.

Antibodies that bind to myelin and nerve cells and protect nerves from damage and stimulate myelin regeneration have been identified. A recent study also has found that regeneration of the myelin sheath can be stimulated by small, folded DNA molecules (aptamers).

Multiple system atrophy.Multiple system atrophy (MSA) is a progressive, fatal neurodegenerative disorder. The hallmark of the disease is glial cytoplasmic inclusions. The main component of glial cytoplasmic inclusions is alpha-synuclein. Aggregation of alpha-synuclein microfibrils leads to a chain of events, including microglial activation, inflammation, and glial and neuronal degeneration. The likely mechanisms involved include growth factor (BDNF, GDNF) deficiency, toxic cytokines and oxidative injury.

Research focuses on the prevention of alpha-synuclein aggregation by drugs such as rifampicin or paroxetine; the use of mesenchymal stem cells to provide and deliver growth factors; and attacking microglial activation and the inflammatory response by agents such as intravenous immunoglobulin.

Immune response and neuroregeneration. Researchers in the Mayo Clinic Center for Regenerative Biotherapeutics are developing numerous approaches to attenuate specific immune cell types in central nervous system (CNS) inflammation and applying strategies to a variety of diseases, including inflammation developing in the course of stem cell transplant, gene therapy or factor-driven regeneration of CNS tissues.

Studies have demonstrated a therapeutic effect in reducing motor dysfunction and blood-brain barrier disruption in model systems of multiple sclerosis through the removal of antigen-specific CD8 T cell responses. By optimizing the imaging of neuroinflammation with high-resolution confocal microscopy, small animal MRI and the profiling of CNS-infiltrating immune cells using flow cytometry, it's possible to isolate and phenotype CNS-infiltrating immune cells in vivo and visualize in real time the events leading to inflammatory destruction of nervous tissue.

Spinal cord repair. Regrowth of nerve fibers (axons) is essential to repair and functional recovery of the spinal cord. Tissue destruction with cysts and gliosis at the site of injury forms a barrier to regeneration.

Ongoing research is using tissue engineering with biodegradable polymer scaffolds (PLGA, PCLF, OPF) loaded with different growth-promoting cells (Schwann cells, neural progenitor cells, mesenchymal stem cells) and different growth factors (GDNF, NT3, BDNF) to bridge the gap, and to promote axonal regeneration and functional restoration in the spinal cords of rats and mice, eventually for future use in patients.

Further, Mayo Clinic researchers are investigating the effects of exercise training and local delivery of steroids on axon regeneration and functional recovery.

Peripheral nerve regeneration and repair. The Center for Regenerative Biotherapeutics is developing strategies to expand the time window of opportunity and improve the functional recovery followingperipheral nerve injury and repair.

One strategy is to apply polymer microspheres to deliver vascular endothelial growth factor (VEGF) to the nerve repair site in a controlled sustainable release manner. VEGF promotes angiogenesis and neurogenesis, and thus leads to a better functional outcome and larger window of opportunity for the nerve to be permissive to prolonged regeneration.

The other strategy is to counteract the lack of healthy Schwann cells at the nerve repair site by supplementing functioning Schwann cells derived from nerves prepared in an in vitro system or Schwann cells induced from stem cells of the adipose tissue.

Novel animal models are being developed to delineate the nature and time course of denervation muscle changes; identify the key indicators of muscle receptivity, including electromyographic changes, muscle fiber type changes and changes of myogenic genes; and evaluate the impact of these changes on nerve regeneration and the potential success of a nerve repair.

Stroke neuroregeneration. After stroke, neurons near the penumbra are vulnerable to delayed but progressive damage as a result of ischemia. There is no effective treatment to rescue such dying neurons. Researchers in the Center for Regenerative Biotherapeutics hypothesized that mesenchymal stem cells (MSC) can rescue damaged neurons after exposure to oxygen-glucose deprivation (OGD) stress.

Studies have demonstrated that the MSC can differentiate into bone, cartilage and fat tissues. Experiments in animal models of hemorrhagic stroke showed MSC therapy improves limb function. Taken together, this data will form the basis for using MSC to treat patients with recent hemorrhagic stroke.

Neuro-oncology and neuroregenerative research. Research currently focuses on invasive brain tumors (gliomas) for which patients receive a very poor prognosis. However, there are other brain tumors oligodendroglioma and astrocytoma that have a much better prognosis. Mayo Clinic researchers are interested in the mutations that are involved in the development of each of these different tumor types and why the tumors behave differently.

A target locus in a gene-poor region initially discovered by genome scanning has been identified. Research efforts are focused on studying the function of this alteration. Using mouse models, murine and human neural stem cells, and human induced pluripotent stem cells, Mayo researchers are investigating how the alteration modifies glial cell development.

Neuroregeneration and inflammation. The limited capacity for repair in the nervous system is a significant medical challenge. The Center for Regenerative Biotherapeutics is developing new tools to effectively control the process of neural injury and degeneration and to create a microenvironment that enhances the capacity for innate repair and the efficacy of other regeneration strategies, including neural cell replacement and neurorehabilitation.

Research efforts focus on how highly druggable proteases (kallikreins) can be targeted to prevent the complex cascade of tissue injury and aberrant reorganization that is a well-recognized component of CNS trauma and which is increasingly recognized as an integral factor underlying the progression of many neurological disorders, including those classified as neurodegenerative or neuroinflammatory as well as those having an oncogenic basis.

Efforts are directed at understanding the physiological and pathophysiological consequences of a family of G protein-coupled receptors (protease-activated receptors, or PARs), and determining whether PARs or the proteases that activate them can be targeted therapeutically to prevent pathogenesis and to promote CNS plasticity and repair to improve patient functional outcomes.

Deep brain stimulation for Alzheimer's disease. Anecdotal and initial trial reports concerning deep brain stimulation (DBS) to the fornix and hypothalamus have been associated with improvement in memory function and reductions in expected cognitive decline in patients with early Alzheimer's disease. The fornix constitutes the major inflow and output pathway from the hippocampus and medial temporal lobe.

Mayo researchers have started an innovative pilot study of dual-hemispheric stimulation of the subthalamic nucleus and fornix and hypothalamus to determine if this approach may have positive effects in attenuating cognitive decline. If this study provides positive data, then the potential of using DBS of the fornix as a treatment for Alzheimer's disease will be considered.

Pediatric anesthesia, apoptosis and safety. Exposure to multiple anesthetics at a young age may be associated with later problems, such as learning disabilities and attention-deficit/hyperactivity disorder. Researchers in the Center for Regenerative Biotherapeutics are working on a large project involving the detailed testing of 1,000 children to try to better define what injury (if any) may be associated with anesthetic exposure. This information will be important to see if this is really a problem in clinical practice, and if so, to change practice to minimize problems.

Researchers are performing detailed neurodevelopmental testing on a sample from a birth cohort of children, including a testing battery previously used in primates shown to be affected by anesthesia exposure. The aim is to confirm (or refute) prior findings and provide for the first time a detailed phenotype of anesthesia-associated injury (if present).

Neurogenesis. By increasing the understanding of the molecular targets involved in regulation of adult hippocampal neurogenesis (neuron generation) and related behavioral responses altered in neuropathological conditions, scientists can study underlying cellular and molecular mechanisms that regulate the production, maturation and integration of new neurons in the circuitry, and how aberrant neurogenesis plays a role in disease pathogenesis. Researchers are employing behavioral neuroscience to quantify cognition such as learning, memory and anxiety.

Recognizing the therapeutic potential of adult neurogenesis, Mayo Clinic researchers are characterizing treatment systems and clinically approved medication that can allow dictation of neuronal development in the correct direction. The long-term goal is to harness the regenerative capacity of adult neurogenesis toward an optimal clinical outcome and improved treatment options for brain disorders.

Neurorehabilitation. This research focuses on improving participation and the quality of life in people whose brain functions have been altered by injury or disease. The focus is regenerative in that improved behavioral performance is possible only when adaptive anatomic and physiological change occurs within and between brain systems in response to therapeutic intervention.

By developing treatment approaches that lead to improved function and independence, researchers in the Center for Regenerative Biotherapeutics promote the adaptive regenerative changes in brain function that make this improved behavioral performance possible.

Transduction mechanisms mediating bidirectional nerve growth. Cues released from the breakdown of myelin after injury in the brain and spinal cord may act as chemorepellents and inhibit axon extension, which limits functional recovery. In contrast, positive cues such as neurotrophins can promote axon extension and elicit chemoattraction.

This research aims to determine how chemotropic cues in the microenvironment guide nerve growth and how dysfunctional guidance mechanisms can cause disease. Understanding these mechanisms and discovering methods to manipulate them are important for developing new therapies to promote neural regeneration after degenerative disease or injury.

Researchers are determining how chemotropic cues in the microenvironment guide nerve growth and how dysfunctional guidance mechanisms can cause disease. This will allow scientists to define the spatiotemporal signal transduction mechanisms by which nerve growth cones detect extracellular guidance cues and dynamically regulate cellular effectors to control the direction of axon extension during normal embryonic development and neural regeneration after injury.

Longer term, the research goal is to define mechanisms for priming and guiding regenerating axons to appropriate synaptic targets to complete functional circuits.

Neuroregenerative Medicine at Mayo Clinic (PDF)

.

See the original post:
Neuroregeneration - Center for Regenerative Biotherapeutics - Mayo ...

Posted in Regenerative Medicine | Comments Off on Neuroregeneration – Center for Regenerative Biotherapeutics – Mayo …

Journal of Immunology and Regenerative Medicine – ScienceDirect

Posted: November 16, 2022 at 2:34 am

As the name implies, the Journal of Immunology and Regenerative Medicine provides a forum for the publication of studies that explore the potential and essential roles of the immune system in tissue and organ development, maintenance, response to environmental stressors, response to injury, and in the processes of tissue repair and regeneration.

The field of tissue engineering and regenerative medicine (TE/RM) has only recently recognized the potential benefits of modulating or instructing the immune system, rather than suppressing the immune response, as a viable strategy for promoting tissue reconstruction and improving functional outcomes. Biomaterials, both synthetic and naturally occurring, play an important role in TE/RM strategies. Therefore, it is only logical that the immune response to biomaterials is a critical consideration with respect to clinical outcomes.

Similarly, the immunology community has only recently recognized the potential benefits of cross-disciplinary interactions between biomaterials and stem cell scientists who target regenerative medicine applications and clinical translation. The immunomodulatory effects of various stem and tissue progenitor cells, signaling molecules derived from extracellular matrix, and the essential contribution of regulatory macrophages, T-cells, and other immune cells in functional tissue restoration and maintenance has also taken center stage.

These newly recognized targets of research and development provide exciting opportunities for immunologists, regenerative medicine scientists, biomaterial and stem cell scientists, and clinicians alike. The Journal of Immunology and Regenerative Medicine provides a venue for publication of this research. The journal will accept basic and applied research manuscripts, selected review articles, clinical study reports, and short communications for peer review and publication. The Journal of Immunology and Regenerative Medicine aims to support conferences and symposia on topics within its remit through the publication of meeting reports. The Editors would, therefore, welcome suggestions from event organisers who would like to submit a meeting report in order to increase the impact of their conference through the dissemination of research findings and discussion to a broader audience.

Scope of Subject Matter: Role of the innate and adaptive immune system in TE/RM strategies for functional tissue repair, regeneration or replacement Cross talk between "immune cells" and "stem cells" The host response to Biomaterials and the effect upon outcome The role of the immune system in tissue and organ development Nanoparticles as immune modulators Animal models that facilitate exploration of the role of the immune system in TE/RM strategies Clinical studies and case reports of the role of the immune system in regenerative vs non-regenerative tissues and organs Manipulation of the innate or adaptive immune system to facilitate success TE/RM strategies The effects of age upon the immune system and its role in TE/RM

See the article here:
Journal of Immunology and Regenerative Medicine - ScienceDirect

Posted in Regenerative Medicine | Comments Off on Journal of Immunology and Regenerative Medicine – ScienceDirect

A Look At Some Of The Companies Innovating In the Cutting-Edge …

Posted: November 16, 2022 at 2:34 am

Biotechnology companies have broken many medical barriers in the past 40 years, harnessing the technological revolution to bring innovative solutions to medical problems.

One flourishing field in biotech is regenerative medicine, a market worth $16.9 billion in 2021. Regenerative medicine harnesses the bodys amazing ability to heal itself, using cutting-edge technology to apply this regenerative power to prompt the body to recover from diseases previously uncured.

An estimated one-third of Americans would benefit from regenerative therapeutic cures. There are wide applications for such regenerative therapy, with categories including stem cell research, gene therapy and tissue engineering. Some difficulties for regenerative medicine companies include arduous Food and Drug Administration (FDA) trial processes and the need for vertical integration of their product development to cut expenses.

Biotech companies involved in regenerative medicine include Mesoblast Ltd. MESO, Brainstorm Cell Therapeutics Inc. BCLI, Lineage Cell Therapeutics LCTX and BioRestorative Therapies Inc. BRTX.

Here is a look at some companies looking to be leaders in the regenerative therapy field:

Mesoblast Ltd. develops novel treatments for back pain and various cardiovascular conditions. This Australia-based company focuses primarily on cell therapy solutions with a mesenchymal lineage stem cell (MSC) technology platform. This develops MSCs, highly multipotent cells taken from healthy bone marrow, and develops treatments for tissue damage, heart disease and more.

Lineage Cell Therapeutics is a company pioneering cell-based therapies to treat serious diseases, including ocular disorders and cancer. It uses its proprietary cell-therapy platform to develop and manufacture self-renewing stem cells into differentiated cells, which can be transplanted to treat problems including cancer or degenerative diseases.

Brainstorm Therapeutics focuses on cell therapies for neurodegenerative diseases. Its autologous cellular therapeutics platform NurOwn treats the disease by differentiating the patients healthy MSCs. Brainstorms work may eventually provide treatments for such neurodegenerative diseases as Alzheimers.

BioRestorative Therapies, which primarily develops products using highly therapeutic adult stem cells, focuses on disc/spine disease and metabolic disorders. BioRestoratives brtxDisc program is developing a treatment for the millions of Americans suffering from either chronic or acute back pain. Its product BRTX-100, which uses autologous stem cells to treat degenerative spinal discs, is in a Phase Two FDA trial.

BioRestorative is also tackling obesity, which currently affects over 40% of Americans. It is developing the product ThermoStem, which harnesses the bodys natural production of healthy brown fat cells to target patient obesity and other metabolic issues associated with obesity.

BioRestorative believes that its treatments will also help condition the body for better future regeneration and responses to medical treatment. A significant advantage for the company is the vertical integration of development and production it has through its clinical-grade cell therapy manufacturing facility. This facility, completed in April, gives BioRestorative control and oversight in the cell manufacturing process, apart from the flexibility to make its own decisions and to correct quality issues in real-time. Owning the facility mitigates the expense normally associated with these activities, which is a great benefit when conducting FDA trials.

Learn more about BioRestorative by visiting its website.

This post contains sponsored advertising content. This content is for informational purposes only and is not intended to be investing advice.

Image sourced from Shutterstock

Read the original post:
A Look At Some Of The Companies Innovating In the Cutting-Edge ...

Posted in Regenerative Medicine | Comments Off on A Look At Some Of The Companies Innovating In the Cutting-Edge …

Page 11234..1020..»