Page 120«..1020..119120121122..130140..»

A short history of gene therapy – Boston Children’s Answers

Posted: December 27, 2022 at 12:44 am

As early as the 1960s, scientists speculated that DNA sequences could be introduced into patients cells to cure genetic disorders. In the early 1980s, David Williams, MD, and David Nathan, MD, at Boston Childrens Hospital published the first paper showing one could use a virus to insert genes into blood-forming stem cells. In 2003, the Human Genome Project wrapped up, giving us a complete blueprint of our DNA. In the past decade, gene therapy has become a reality for multiple diseases, especially those caused by mutations in a single gene.

Gene therapy falls into two main categories. Ex vivo gene therapy removes cells from the patient, introduces new genetic material, packaged in a delivery vehicle called a vector, then returns the cells to the patient. Boston Childrens is using this method for such disorders as sickle cell disease, adrenoleukodystrophy, chronic granulomatous disease and others. In vivo gene therapy involves direct IV infusion of the vector into the bloodstream or injection into a target organ like the eye. Boston Childrens uses in vivo gene therapy for several disorders, including hemophilia and ornithine transcarbamylase deficiency.

After a rocky start, gene therapy is on fire, drawing keen interest from the biopharmaceutical industry. And its still evolving and improving.

In 1990, 4-year-old Ashanthi de Silva became the first gene therapy success story. She was born with a severe combined immunodeficiency (SCID) due to lack of the enzyme adenosine deaminase (ADA). Without ADA, her T cells died off, leaving her unable to fight infections. Injections of a synthetic ADA enzyme helped, but only temporarily.

Doctors decided to deliver a healthy ADA gene into her blood cells, using a disabled virus that cannot spread in the body. Their success spurred more trials in the 1990s for the same form of SCID. Now in her 30s, de Silva is active in the rare disease community.

European researchers in the 1990s focused on SCID-X1, another form of SCID linked to the X chromosome. They reported the first cures in 2000, but within several years, five of the 20 treated children developed cancer.The viral vector that delivered the gene to their T cells had also activated an oncogene, triggering leukemia.

The U.S. saw another early setback: the 1999 death of 18-year-old Jesse Gelsinger, after receiving gene therapy for a rare metabolic disorder. In his case, the viral vector caused a fatal immune response.

Gene therapy came to a halt.

In the early 2010s, gene therapy experienced a renaissance. Scientists developed better viral vectors to deliver genetic therapies. They added regulatory elements called promoters and enhancers to direct the genes activity. These elements specified where and when the gene should turn on, and at what level. Investigators at Boston Childrens, in a global collaborative effort, led work that addressed the problem of leukemia, allowing gene therapy to resume for SCID-X1.

A REBIRTH IN BOSTON: GENE THERAPY TURNS 10

Born in 2010 with X-linkedsevere combined immunodeficiency(SCID-X1), Agustn spent the first few months of his life in isolation. He became the first patient to receive gene therapy at Boston Childrens and today is an active fifth-grade soccer and tennis player.

The new, modified vectors can more precisely target expression of genes in specific cell types, dont go astray in the body, and dont trigger the immune system. Some deliver genes meant to work for a short while and then inactivate themselves. Others carry genes that remain active long-term and pass to daughter cells as the cells divide. Popular viruses for gene therapy include adenoviruses, adeno-associated virus, and lentiviruses.

An example of an improved vector is the lentivirus vector used for sickle cell gene therapy at Boston Childrens. The vector silences a gene calledBCL11A, leading to production of fetal hemoglobin that is not affected by the sickle cell mutation. It was precision engineered to silence the gene only in precursors of red blood cells, a tweak that enabled the treated blood stem cells to live long-term in patients bone marrow. Williams led the vectors development, based on seminal research by Vijay Sankaran, MD, PhD, and Stuart Orkin, MD, in the Hematology/Oncology Program at Boston Childrens.

Traditional gene therapyuses viruses to carry healthy genes into cells, compensating for a faulty or missing gene. But the past decade has seen an explosion of other methods for delivering or fixing genes.

Gene editing uses various molecular tools that precisely target problematic genes and create a cut or break in their DNA. It can knock out a faulty gene, insert a new DNA sequence, or both in a cut and paste operation. The best-known gene editing systems are CRISPR/Cas 9, zinc-finger nucleases (ZFNs), and transcription activator-like effector nucleases (TALENs). The next generation of gene therapy for sickle cell disease is utilizing CRISPR to edit the BCL11A gene, based on work by Dan Bauer, MD, PhD, at Boston Childrens, then in Orkins laboratory.

Base editingis even more fine-tuned. It leverages the targeting ability of CRISPR, but relies on enzymes to chemically change one letter of a genes code at a time changing, say, C to T or A to G. These small changes can correct a spelling error mutation, silence a disease-causing gene, or help activate a specific gene. Unlike gene editing, base editing hasnt yet been tested in clinical trials, but it offers the promise of more precision, efficiency, and safety.Boston Childrens has several base editing projects on deck.

Other new approaches blur the line between gene therapy and drug treatment. For example, antisense oligonucleotides (ASOs) are drugs made up of short, synthetic pieces of DNA or RNA that target the messenger RNA made by the faulty gene. They prevent the gene from being translated into a bad protein or, in some cases, trick the cells machinery into making a good protein. Researchers can even customize ASOs to single patients. Tim Yu, MD, PhD, in the Division of Genetics and Genomics at Boston Childrens, has developed this approach to treat several very rare genetic conditions.

Another approach, RNA interference, uses small RNAs to silence a targeted gene by neutralizing the genes mRNA. (The lentivirus described above uses RNA interference to silence the BCL11A gene.)

Even the messenger RNAs used for some COVID-19 vaccines represent a form of gene therapy. The mRNAs introduce genetic code that cells then use to make the coronavirus spike protein, encouraging people to develop antibodies to the virus.

Today, ClinicalTrials.gov lists nearly 400 active gene therapy studies all over the world, and more than a dozen gene therapy drugs are on the market. At Boston Childrens, the Gene Therapy Program has more than 20 human trials completed or underway, with more in the pipeline. While gene therapies are currently expensive, its expected that prices will come down over time. And as a one-time treatment, gene therapy promises to save money in the long run by preventing a lifetime of illness a true revolution in medicine.

Learn more about the Gene Therapy Program at Boston Childrens Hospital

See the original post:
A short history of gene therapy - Boston Children's Answers

Posted in Gene therapy | Comments Off on A short history of gene therapy – Boston Children’s Answers

Gene Therapy Analytical Development Summit 2022 | Home

Posted: December 27, 2022 at 12:44 am

The past twelve months have culminated in an unprecedented level of excitement, investment, and clinical progress within the gene therapy field. As the field strives to strike a delicate balance between safety and efficacy, in the context of increased regulatory scrutiny and safety challenges, attending the 4th Annual Gene Therapy Analytical Development as an analytical scientist has never been so important.

This years summit returns in-person to Boston to reunite 300+ analytical experts in innovative biotech, pharma and academia to continue to develop resilient, long-lasting and robust analytical tools to enhance the safety, quality and efficacy of gene therapy products.

Whether you are focusing on specific characterization methods, enhancing your genome sequencing, advancing your understanding of full and partial particles, or advancing your early-stage bioassays, with 4 tracks, 8 pre-conference workshops and a post-conference focus day, the 4th Gene Therapy Analytical Development Summit will encompass all aspects of analytical development, giving you the chance to address and overcome challenges.

If you work in quality control, quality assurance, or process development - weve listened and weve answered. This years agenda includes a novel track designed for quality control and process development groups working in gene therapy. Talks include enhancing the knowledge transfer between departments, bridging between analytical methods with regards to QC/PD, and enhancing in-process development support.

Whether you're working with AAV, non-viral vectors or lentiviral vectors, this is your opportunity to enhance your existing analytical methods and explore innovative new tools to support safe and effective gene therapy development.

Read more:
Gene Therapy Analytical Development Summit 2022 | Home

Posted in Gene therapy | Comments Off on Gene Therapy Analytical Development Summit 2022 | Home

Stem cell transplant – NHS

Posted: December 27, 2022 at 12:40 am

A stem cell or bone marrow transplant replaces damaged blood cells with healthy ones. It can be used to treat conditions affecting the blood cells, such as leukaemia and lymphoma.

Stem cells arespecial cells produced bybone marrow (aspongytissue found in the centre of some bones) that can turn into different types of blood cells.

The 3 maintypes of blood cellthey can become are:

A stem cell transplant involves destroying any unhealthy blood cells and replacing them with stem cells removed from the blood or bone marrow.

Stem cell transplants are used to treat conditions in which the bone marrow is damaged and is no longer able to produce healthy blood cells.

Transplants can also be carried out to replace blood cells that are damaged or destroyed as a result of intensive cancer treatment.

Conditions that stem cell transplants can be used to treat include:

A stem cell transplant will usually only be carried out if other treatments have not helped, the potential benefits of a transplant outweigh the risks and you're in relatively good health, despite your underlying condition.

A stem cell transplant can involve taking healthy stem cells from the blood or bone marrow of one person ideally a close family member with the same or similar tissue type and transferring them to another person. This is called an allogeneic transplant.

It's also possible to remove stem cells from your own body and transplant them later, after any damaged or diseased cells have been removed. This is called an autologous transplant.

Astem celltransplant has 5 main stages. These are:

Having a stem cell transplant can be an intensive and challenging experience. You'll usually need to stay in hospital forat least a few weeks until the transplant starts to take effect and itcan take up toa year or longer to fully recover.

Read more about what happens during a stem cell transplant.

Stem celltransplants arecomplicated procedures with significant risks. It's important that you're aware of both the risks and possible benefits before treatment begins.

Possible problems you can have during or after the transplant process include:

Read more about the risks of having a stem cell transplant.

Ifit is not possible to use your own stem cells for the transplant, stem cells will need to come from a donor.

To improve the chances ofthetransplant being successful, donated stem cells need tocarry a special genetic marker known as a human leukocyte antigen (HLA) that'sidentical or very similar to that of the person receiving the transplant.

The best chance of getting a match is from a brother or sister, or sometimes another close family member. If there are no matches in your close family,a search of theBritish Bone Marrow Registry will be carried out.

Most peoplewill eventually find a donor in the registry,although a small number of people may find it very hard or impossibleto find a suitable match.

The NHS Blood and Transplant website and the Anthony Nolan website have more information about stem cell and bone marrow donation.

Page last reviewed: 07 September 2022Next review due: 07 September 2025

Here is the original post:
Stem cell transplant - NHS

Posted in Stem Cells | Comments Off on Stem cell transplant – NHS

Stem cells: a brief history and outlook – Science in the News

Posted: December 27, 2022 at 12:40 am

Stem cells have been the object of much excitement and controversy amongst both scientists and the general population. Surprisingly, though, not everybody understands the basic properties of stem cells, let alone the fact that there is more than one type of cell that falls within the stem cell category. Here, Ill lay out the basic concepts of stem cell biology as a background for understanding the stem cell research field, where it is headed, and the enormous promise it offers for regenerative medicine.

Fertilization of an egg cell by a sperm cell results in the generation of a zygote, the single cell that, upon a myriad of divisions, gives rise to our whole body. Because of this amazing developmental potential, the zygote is said to be totipotent. Along the way, the zygote develops into the blastocyst, which implants into the mothers uterus. The blastocyst is a structure comprising about 300 cells that contains two main regions: the inner cell mass (ICM) and the trophoblast. The ICM is made of embryonic stem cells (ES cells), which are referred to as pluripotent. They are able to give rise to all the cells in an embryo proper, but not to extra-embryonic tissues, such as the placenta. The latter originate from the trophoblast [].

Even though it is hard to pinpoint exactly when or by whom what we now call stem cells were first discovered, the consensus is that the first scientists to rigorously define the key properties of a stem cell were Ernest McCulloch and James Till. In their pioneering work in mice in the 1960s, they discovered the blood-forming stem cell, the hematopoietic stem cell (HSC) [2, 3]. By definition, a stem cell must be capable of both self-renewal (undergoing cell division to make more stem cells) and differentiation into mature cell types. HSCs are said to be multipotent, as they can still give rise to multiple cell types, but only to other types of blood cells (see Figure 1, left column). They are one of many examples of adult stem cells, which are tissue-specific stem cells that are essential for organ maintenance and repair in the adult body. Muscle, for instance, also possesses a population of adult stem cells. Called satellite cells, these muscle cells are unipotent, as they can give rise to just one cell type, muscle cells.

Therefore, the foundations of stem cell research lie not with the famous (or infamous) human embryonic stem cells, but with HSCs, which have been used in human therapy (such as bone marrow transplants) for decades. Still, what ultimately fueled the enormous impact that the stem cell research field has today is undoubtedly the isolation and generation of pluripotent stem cells, which will be the main focus of the remainder of the text.

Figure 1: Varying degrees of stem cell potency. Left: The fertilized egg (totipotent) develops into a 300-cell structure, the blastocyst, which contains embryonic stem cells (ES cells) at the inner cell mass (ICM). ES cells are pluripotent and can thus give rise to all cell types in our body, including adult stem cells, which range from multipotent to unipotent. Right: An alternative route to obtain pluripotent stem cells is the generation of induced pluripotent stem cells (iPS cells) from patients. Cell types obtained by differentiation of either ES cell (Left) or iPS cells (Right) can then be studied in the dish or used for transplantation into patients. Figure drawn by Hannah Somhegyi.

Martin Evans (Nobel Prize, 2007) and Matt Kauffman were the first to identify, isolate and successfully culture ES cells using mouse blastocysts in 1981 []. This discovery opened the doors to the creation of murine genetic models, which are mice that have had one or several of their genes deleted or otherwise modified to study their function in disease []. This is possible because scientists can modify the genome of a mouse in its ES cells and then inject those modified cells into mouse blastocysts. This means that when the blastocyst develops into an adult mouse, every cell its body will have the modification of interest.

The desire to use stem cells unique properties in medicine was greatly intensified when James Thomson and collaborators first isolated ES cells from human blastocysts []. For the first time, scientists could, in theory, generate all the building blocks of our body in unlimited amounts. It was possible to have cell types for testing new therapeutics and perhaps even new transplantation methods that were previously not possible. Yet, destroying human embryos to isolate cells presented ethical and technical hurdles. How could one circumvent that procedure? Sir John Gurdon showed in the early 1960s that, contrary to the prevalent belief back then, cells are not locked in their differentiation state and can be reverted to a more primitive state with a higher developmental potential. He demonstrated this principle by injecting the nucleus of a differentiated frog cell into an egg cell from which the nucleus had been removed. (This is commonly known as reproductive cloning, which was used to generate Dolly the Sheep.) When allowed to develop, this egg gave rise to a fertile adult frog, proving that differentiated cells retain the information required to give rise to all cell types in the body. More than forty years later, Shinya Yamanaka and colleagues shocked the world when they were able to convert skin cells called fibroblasts into pluripotent stem cells by altering the expression of just four genes []. This represented the birth of induced pluripotent stem cells, or iPS cells (see Figure 1, right column). The enormous importance of these findings is hard to overstate, and is perhaps best illustrated by the fact that, merely six years later, Gurdon and Yamanaka shared the Nobel Prize in Physiology or Medicine 2012 [].

Since the generation of iPS cells was first reported, the stem cell eld has expanded at an unparalleled pace. Today, these cells are the hope of personalized medicine, as they allow one to capture the unique genome of each individual in a cell type that can be used to generate, in principle, all cell types in our body, as illustrated on the right panel of Figure 1. The replacement of diseased tissues or organs without facing the barrier of immune rejection due to donor incompatibility thus becomes approachable in this era of iPS cells and is the object of intense research [].

The first proof-of-principle study showing that iPS cells can potentially be used to correct genetic diseases was carried out in the laboratory of Rudolf Jaenisch. In brief, tail tip cells from mice with a mutation causing sickle cell anemia were harvested and reprogrammed into iPS cells. The mutation was then corrected in these iPS cells, which were then differentiated into blood progenitor cells and transplanted back into the original mice, curing them []. Even though iPS cells have been found not to completely match ES cells in some instances, detailed studies have failed to find consistent differences between iPS and ES cells []. This similarity, together with the constant improvements in the efficiency and robustness of generating iPS cells, provides bright prospects for the future of stem cell research and stem cell-based treatments for degenerative diseases unapproachable with more conventional methods.

Leonardo M. R. Ferreira is a graduate student in Harvard Universitys Department of Molecular and Cellular Biology

[] Stem Cell Basics: http://stemcells.nih.gov/info/basics/Pages/Default.aspx

[] Becker, A. J., McCulloch, E.A., Till, J.E. Cytological demonstration of the clonal nature of spleen colonies derived from transplanted mouse marrow cells. Nature 1963. 197: 452-4

[] Siminovitch, L., McCulloch, E.A., Till, J.E. The distribution of colony-forming cells among spleen colonies. J Cell Comp Physiol 1963, 62(3): 327-336

[] Evans, M. J. and Kaufman, M. Establishment in culture of pluripotential stem cells from mouse embryos. Nature 1981, 292: 151156

[] Simmons, D. The Use of Animal Models in Studying Genetic Disease: Transgenesis and Induced Mutation. Nature Education 2008,1(1):70

[] Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 1998, 282(5391): 1145-1147

[] Takahashi, K. and Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006. 126(4): 663-76

[] The Nobel Prize in Physiology or Medicine 2012:

[] Ferreira, L.M.R. and Mostajo-Radji, M.A. How induced pluripotent stem cells are redefining personalized medicine. Gene 2013. 520(1): 1-6 [] Hanna J. et al. Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science 2007. 318: 1920-1923

[] Yee,J.Turning Somatic Cells into Pluripotent Stem Cells.Nature Education 2010.3(9):25

Excerpt from:
Stem cells: a brief history and outlook - Science in the News

Posted in Stem Cells | Comments Off on Stem cells: a brief history and outlook – Science in the News

RUDN Physician And Russian Scientists Investigate Long-term Effects Of Treating Diabetic Ulcers With Stem Cells – India Education Diary

Posted: December 27, 2022 at 12:40 am

RUDN Physician And Russian Scientists Investigate Long-term Effects Of Treating Diabetic Ulcers With Stem Cells  India Education Diary

See the article here:
RUDN Physician And Russian Scientists Investigate Long-term Effects Of Treating Diabetic Ulcers With Stem Cells - India Education Diary

Posted in Stem Cells | Comments Off on RUDN Physician And Russian Scientists Investigate Long-term Effects Of Treating Diabetic Ulcers With Stem Cells – India Education Diary

Adoptive Cell Therapy – Cancer Research Institute (CRI)

Posted: December 27, 2022 at 12:36 am

How Cellular Immunotherapies Are Changing the Outlook for Cancer Patients

Reviewed By:

Philip D. Greenberg, MD.Fred Hutchinson Cancer Research Center

Some of these approaches involve directly isolating our own immune cells and simply expanding their numbers, whereas others involve genetically engineering our immune cells (via gene therapy) to enhance their cancer-fighting capabilities.

Our immune system is capable of recognizing and eliminating cells that have become infected or damaged as well as those that have become cancerous. In the case of cancer, immune cells known as killer T cells are particularly powerful against cancer, due to their ability to bind to markers known as antigens on the surface of cancer cells. Cellular immunotherapies take advantage of this natural ability and can be deployed in different ways:

Today, cell therapies are constantly evolving and improving and providing new options to cancer patients. Cell therapies are currently being evaluated, both alone and in combination with other treatments, in a variety of cancer types in clinical trials.

Cancer patients have naturally occurring T cells that are often capable of targeting their cancer cells. These T cells are some of the most powerful immune cells in our body, and come in several types. The killer T cells, especially, are capable of recognizing and eliminating cancer cells in a very precise way.

The existence of these T cells alone, however, isnt always enough to guarantee that they will be able to carry out their mission to eliminate tumors. One potential roadblock is that these T cells must first become activated before they can effectively kill cancer cells, and then they must be able to maintain that activity for a sufficiently long time to sustain an effective anti-tumor response. Another is that these T cells might not exist in sufficient numbers.

One form of adoptive cell therapy that attempts to address these issues is called tumor-infiltrating lymphocyte (TIL) therapy. This approach harvests naturally occurring T cells that have already infiltrated patients tumors, and then activates and expands them. Then, large numbers of these activated T cells are re-infused into patients, where they can then seek out and destroy tumors.

Unfortunately, not all patients have T cells that have already recognized their tumors. Others patients might, but for a number of reasons, these T cells may not be capable of being activated and expanded to sufficient numbers to enable rejection of their tumors. For these patients, doctors may employ an approach known as engineered T cell receptor (TCR) therapy.

This approach also involves taking T cells from patients, but instead of just activating and expanding the available anti-tumor T cells, the T cells can also be equipped with a new T cell receptor that enables them to target specific cancer antigens. By allowing doctors to choose an optimal target for each patients tumor and distinct types of T cell to engineer, the treatment can be further personalized to individuals and, ideally, provide patients with greater hope for relief.

The previously mentioned TIL and TCR therapies can only target and eliminate cancer cells that present their antigens in a certain context (when the antigens are bound by the major histocompatibility complex, or MHC).

Recent advances in cell-based immunotherapy have enabled doctors to overcome this limitation. Scientists equip a patients T cells with a synthetic receptor known as a CAR, which stands for chimeric antigen receptor.

A key advantage of CARs is their ability to bind to cancer cells even if their antigens arent presented on the surface via MHC, which can render more cancer cells vulnerable to their attacks. However, CAR T cells can only recognize antigens that themselves are naturally expressed on the cell surface, so the range of potential antigen targets is smaller than with TCRs. In October 2017, the U.S. Food and Drug Administration (FDA) approved the first CAR T cell therapy to treat adults with certain types of large B-cell lymphoma.

Given their power, CARs are being explored in a variety of strategies for many cancer types. One approach currently in clinical trials is using stem cells to create a limitless source of off-the-shelf CAR T cells. This may have application to only selected settings, but could allow doctors to treat patients in a timelier fashion.

More recently, adoptive cell therapy strategies have begun to incorporate other immune cells, such as Natural Killer (NK) cells. One application being explored in the clinic involves equipping these NK cells with cancer-targeting CARs.

There are currently two adoptive cell therapies that are approved by the FDA for the treatment of cancer.

Side effects may vary according to the type of adoptive cell immunotherapyand what exactly it targetsand may also be influenced by the location and type of cancer as well as a patients overall health. Potential cell therapy-related side effects often take the form of an overactive immune response and may lead to excessive inflammation via cytokine release syndrome (also known as cytokine storm), and also to neurotoxicity from inflammation in the brain. Side effects can range from mild to moderate and may become potentially life-threatening under certain circumstances.

Fortunately, in most cases, potential immunotherapy-related side effects can be managed safely as long as the potential side effects are recognized and addressed early. Therefore, its extremely important that patients inform their medical care team as soon as possible if they experience any unusual symptoms during or after treatment with cancer immunotherapy. In addition, patients should always consult their doctors and the rest of their care team to gain a better and fuller understanding of the potential risks and side effects associated with specific adoptive cell immunotherapies.

Common side effects associated with currently approved adoptive cell therapies may include but are not limited to: acute kidney injury, bleeding episodes, heart arrhythmias, chills, constipation, cough, cytokine release syndrome (cytokine storm), decreased appetite, delirium, diarrhea, dizziness, edema, encephalopathy, fatigue, febrile neutropenia, fever, headache, hypogammaglobulinemia, hypotension, hypoxia, infections, nausea, neurotoxicity, pyrexia, tachycardia, tremors, and vomiting.

Throughout its history, CRI has supported a variety of basic research projects aimed at improving our understanding of the identity and functions of our many immune cells as well as translational and clinical efforts that seek to use these insights in the development of cellular immunotherapies for cancer patients in the clinic.

Some of the most important contributions made by CRI scientists in the area of adoptive cell therapy include:

Go here to see the original:
Adoptive Cell Therapy - Cancer Research Institute (CRI)

Posted in Cell Therapy | Comments Off on Adoptive Cell Therapy – Cancer Research Institute (CRI)

Stem Cell Therapy for Treatment of Ocular Disorders – Hindawi

Posted: December 27, 2022 at 12:25 am

Sustenance of visual function is the ultimate focus of ophthalmologists. Failure of complete recovery of visual function and complications that follow conventional treatments have shifted search to a new form of therapy using stem cells. Stem cell progenitors play a major role in replenishing degenerated cells despite being present in low quantity and quiescence in our body. Unlike other tissues and cells, regeneration of new optic cells responsible for visual function is rarely observed. Understanding the transcription factors and genes responsible for optic cells development will assist scientists in formulating a strategy to activate and direct stem cells renewal and differentiation. We review the processes of human eye development and address the strategies that have been exploited in an effort to regain visual function in the preclinical and clinical state. The update of clinical findings of patients receiving stem cell treatment is also presented.

Blindness or loss of visual function can be caused by failure of the light path to reach the retina or failure of the retina to capture and convert light to an electrochemical signal before transmission to the brain via optic nerve [1]. The major causes contributing to blindness include age-related macular degeneration (ARMD), diabetic retinopathy, cataracts, and glaucoma [24], which are genetically linked [5] and associated with multiple risk factors including diet [6], hypertension [7], pregnancy [8], and smoking [9]. The occurrences of these pathologies increase with the age of the patient and are thus widely spread among aging populations. Blindness is an extensive disease that not only affects the quality of life of the patients themselves but may have a negative impact on the socioeconomic status of their immediate families [10, 11].

Current treatments have aimed at protecting vision and preventing visual impairment by early diagnosis using various methods of intervention such as surgery, ionizing radiation, laser, or drug treatments [1214]. Despite the efficiencies of these treatment modalities, they do not provide a complete solution to stop the progression to blindness.

Many recent findings from preclinical data have supported the notion that stem cells have the capacity to revive degenerated cells or replace cells in many major diseases including ocular disorders [1518]. Stem cells are present in all tissues in our body and are self-renewable and capable of maintaining a certain level of differentiation in response to injury for tissue repair [1921]. We mainly aimed this review at both clinicians and academicians, so we presented the localization of stem cell progenitors with eye development in different regions in the eye, the functions of these progenitors, and the current clinical trials and their exploitation of nontissue specific stem cells as alternative sources for regaining lost vision.

Eye development involves indispensable participation of the neural ectoderm (NE), surface ectoderm (SE), ectomesenchymal/cranial neural crest cell (CNCC), and modicum of mesenchymal tissues [22]. During the fourth week of intrauterine life, the forebrain gives rise to two bulges called optic vesicles that extend like a stalk and a cup to trigger the surface ectoderm on both sides [22]. The retinal pigmented epithelium (RPE) and neural retina (NR) are developed from outer and inner layer of optic cup, while the optic nerve is developed from optic stalk [22]. The cup tip becomes the ciliary body and iris by integrating with the CNCC [23]. The surface ectoderm is repressible for the lens, cornea, and conjunctiva [24]. The sclera, corneal endothelium, corneal stroma, iridial stroma, and iridial muscles are contributed by the CNCC [25]. The neural ectodermal derivatives of eye are permanent cells and lack the self-renewal, as like other nervous tissues. But unlike other surface ectodermal derivatives, the ocular ectodermal derivatives do lack the self-renewal in the eye during aging which collectively results in various degenerative disorders.

The well-organized time-dependent interactions and gene expression of all these layers for initiation, pattern determination, and organogenesis are significant for eye development [22, 2427]. Eye development in an embryonic mouse at 9.5 days is shown in Figure 1 [26]. The neural ectoderm bulges as the optic vesicle to reach the surface ectoderm. The surface ectoderm becomes thicker on contact with the neural ectoderm to become the lens placode. Except in the lens placode region, the neural ectoderm and the surface ectoderm are separated by the extraocular mesenchyme. In the NE, the presumptive RPE, NR, and optic tract are colored red, green, and yellow, respectively, in Figure 1. The lens placode is colored blue in Figure 1. The transcription factors described in Figure 1 are involved in the regulation of eye development.

Pax6 is a crucial and evolutionarily conserved homeobox gene of eye development [28, 29]. Along with Pax6, the other associated genes reported for eye development are Rx/Rax, Pax2, Lhx2, Mitf, Otx2, Sox2, Six3, Pitx, Vsx2, Crx, Optx2, and FaxL2 [2839]. The expression of Pax6 is upregulated by Six3 and downregulated by Shh (Sonic hedgehog) [35] to help eye formation on both sides [39]. The transcription factor Pax2 is important for the formation of the optic stalk (which becomes the optic nerve). Retinal axons from both the eyes selectively decussate at the midline named optic chiasma (crucial for vision) which is failed when the Pax2 mutates (optic chiasma) [23].

Initiation of optic vesicle formation from the neural ectoderm by Rx/Rax involves extensive cell movements and proliferation [36]. In addition, Rx is essential for expression of Lhx2, Pax6, Mab2112, and Six3, which specifies the retinal progenitor cells in the optic cup [30, 31, 36, 37]. Lhx2, a patterning gene expressed in the neural ectoderm, is important for expression of Mitf [32]. Mitf is a governing gene for RPE that specifies the neural retina, and in the neural ectoderm, RPE regulates the vesicle to cup transformation and activates the retinoid acid receptor, which is another important factor for eye development [33, 34]. In Lhx2 mutant mouse, Mitf and Vsx2 are never initiated and Pax2, Vax2, and Rx are initiated but not maintained, resulting in arrest of eye development in the optic vesicle stage [40]. The optic vesicle is important for lens formation and the lens is important for the vesicle to cup formation. The surface ectoderm will not form the lens if the optic vesicle is removed. In contrast, when provided with an optic vesicle, any primitive ectoderm will develop into the lens [40].

The neural retina, the brain of eye with nine distinct layers, transmits color signals in and out as vision [22]. During development, the neural retina depends on the expression of Vsx2, an important homeobox gene for early patterning and maintenance of cell proliferation and fate [41]. MAPK/FGF signaling is important for neural retina and upregulates Vsx2 [42] and Vsx2 downregulates Mitf [4244]. This regulation helps control the distinct neural retina and RPE specification in the optic cup. FGF9, normally expressed in the distal optic vesicle, is important for the boundary between the neural retina and the RPE [4446]. FGF receptor activation is crucial in chicks but not in mice [46]. This suggests that there is species-specific neurogenesis. Interestingly, specific activation of MAPK/FGF can induce neural retina formation from presumptive RPE with distinct layers [43, 45, 4749]. BMP is important for Vsx2 regulation [50]. Figure 2 shows a schematic of the adult eye of different vertebrates (frogs and fish, birds, and human) [27]. The ciliary marginal zone (CMZ, yellow color in Figure 2) is progressively reduced in higher vertebrates. Unlike the earlier vertebrates, the neural retina in mammals (blue color in Figure 2) is not renewed continuously because of the absence of the CMZ [51]. The neural progenitor marker Nestin is expressed in the junction of the ciliary body with the neural retina, suggesting the remaining of a CMZ even though the relationship is not clear [52]. The regeneration studies reported with RPE to neural retina are akin to transdifferentiation under suitable conditions [53].

Mller glial cells are a progenitor glial component of the neural retina, which arise from activation of Notch, Rax, and Jak signaling pathway [54]. The RPE is an array of uniformly arranged cells in a single layer between the retina and choroid [22]. MITF governs the RPE, the bHLH transcription factor that is the first and critical gene expressed in presumptive RPE and is specific for patterning and cell proliferation [38]. Mitf, the regulator of the pigmented cells (both in the RPE and in the CNCC) is expressed even before the pigments are formed in the RPE [24]. Mitf is initially expressed throughout the optic vesicle but is later downregulated in neural retina for layer specification. Otx2 is important for Mitf expression [33]. Pax regulates both the Mitf and Otx2 [55]. Pax2 and Pax6 bind and activate the Mitf A enhancer [35]. Retinoid acid signaling regulates the optic cup morphogenesis and induces apoptosis in extraocular mesenchyme [33]. Retinoid acid receptors (RAR-,,) are important for signal transduction of retinoic acid, which is important for the maintenance of the RPE [56]. The enzymes, retinaldehyde dehydrogenases (Raldh) 1, 2, and 3, are vital for retinoid acid synthesis. Raldh 3 originates from the RPE, and Raldh 2 originates from the surrounding mesenchyme [57]. Pitx2 is also important for RPE differentiation [58]. The fate of RPE is influenced by Shh [59]. Growth arrest specific 1 (Gas1) is a (GPI) protein that binds and coregulates with Shh [60]. Gas1 downregulates the proliferation of the RPE to maintain a single cell-layered structure [60]. There are reports of the distinct control mechanisms by BMP in the ventral and dorsal aspects of the RPE [61]. The Wnt/-catenin pathway also controls the optic cup differentiation by activating Mitf and Otx2 [62].

The ciliary body and the iris are developed from the optic cup tip with the incorporated connective tissue stroma derived from the migrated CNCC. The smooth muscles of the iris, namely, the sphincter and dilator pupillae, are derived by transdifferentiation of the pigmented epithelial cells of neural origin [43, 63]. The iris and ciliary body regulate the light reaching the retina and maintain the intraocular pressure by maintaining the aqueous humor secretion [61, 64]. The pigmented cells of the iris possess the ability to differentiate into RPE, neural retina, and lens, and a potential source of stem cells in mammals [65]. FGF, BMP, and Wnt/-catenin participate in the differentiation of progenitor cells into ciliary and iris epithelium [45, 66].

The lens is derived from the surface ectoderm upon receiving instruction by Pax6 to respond to FGF, BMP, and Sox2. Fox-3 helps in the separation of the lens from the surface ectoderm and formation of lens fibers. Lens fibers are epithelial cells that undergo clever modification to become transparent fibers by losing their organelles and accumulating crystalline protein; Pax6, Pitx3, c-Maf, HSF4, RAR, Six, Sox, and Prox are the transcription factors related to crystalline genes [66, 67]. The CNCC is crucial for eye development and restricts the lens formation area in the surface ectoderm by inhibiting cells other than those for the lens. The lens is under the control of the retina throughout life. The retinal secretion of FGF accumulates in the vitreous humor and stimulates the lens part facing the retina to form lens fibers. If the developing lens is rotated, the cell type changes to form lens fibers from the surface facing the retina (Figure 3) [23]. Attempt can be made to turn the defective lens, front to back to find the results, because the side of the lens which faces the retina is influenced with better survival.

The optic cup is surrounded by mesenchymal cells predominantly of CNCC origin that help in the formation of the vascular coat called the choroid and fibrous coat, namely, the sclera. Transcription factors involved with the scleral development are Foxc1, Foxc2, Lmx1b, Pax6, Pitx2, RARb, RARg, RXRa, Six3, and Smad2 [30, 31, 36, 39].

The corneal epithelium is continuous with the conjunctiva covering the visible part of the sclera. The junction between the corneal and conjunctiva is named the limbus, which holds stem cells for the renewal of the epithelium throughout life [22]. The corneal epithelium is constantly renewed every 7 to 10 days. Corneal epithelium expresses Np63, ABCG2, integrin 9, Bmi-1, EGFR, TGF, and PDGF growth factor indicators for their stemness. The stromal interaction is important for the cell renewal achieved by paracrine factors, hepatocyte growth factor (HGF), and keratinocyte growth factor (KGF). These factors are fibroblast-derived epithelial mitogens of the FGF7 family. In the corneal endothelium the morphology, collagen expression, and cell proliferation are maintained by TGF-1 and TGF-2 [46, 49]. Altogether, the corneal endothelial integrity is preserved by Pax6, Lmx1b, and Pitx2 [37].

The tissues of eye which are commonly associated with diseases are the surface ectoderm derivatives cornea and lens and the neural ectoderm derivatives RPE and retina. Since the lens and cornea do lack the renewal capacity during aging, stem cells from other surface ectoderm derivatives which are relatively easy to collect can be reprogrammed by manipulating target genes and proteins with the help of gained knowledge from molecular biology for regenerative therapy. Regenerated lenses from stem cells can be more exciting personalized regenerative treatment.

The aim of understanding the sequential events during embryogenesis at molecular level cell to cell communication is to understand the pathogenesis and to design the regenerative or genetic therapy to restore normal. The commonest degenerative eye disorders can be tactfully managed by delivering target proteins to prevent and to repair several types of ocular diseases. Stem cells of eye are closely associated with maxillofacial tissues including dental stem cells (a derivative of CNCC) during embryogenesis which retain the stem cells till life can be traced and reprogrammed for stem cell therapy. Researchers differentiated retina [68] from dental pulp stem cells. Epigenetic memory explains that the differentiated cells retain the memory of their original tissue and on reprograming they spontaneously dedifferentiate to its original tissue [69, 70]. If the suggested RPE differentiation from dental stem cells [71] is succeeded, it will be more acceptable than the controversial embryonic stem cells, which has proven its success after 2 years of follow-up of clinical trial [72, 73]. Autologous oral mucosal epithelial cells have been successfully reconstructed to fabricate cornea to restore vision [74].

To dispense a suitable intervention, the mechanism that regulates cell renewal, differentiation, and maturation change in a diseased microenvironment needs to be understood. One of the major inherited ocular disorders, Retinitis Pigmentosa (RP), is characterized by progressive degeneration of photoreceptors in the retina [7578]. Complete blindness in most cases proves that humans lack a homeostatic mechanism to replace lost photoreceptors [79].

The earliest interventions used autologous tissue resident stem cells such as RPE cell suspensions or RPE-choroid sheets to improve vision of patients affected by age-related macular degeneration via subretinal translocation [80]. Other sources of stem or progenitors cells from extraocular tissues such as hematopoietic stem cells (HSCs) [8183], dental pulp stem cells (DPSCs) [68], hair follicle stem cells (HFSCs) [84], mesenchymal stem cells (MSCs) [76, 8589], and induced pluripotent stem cells (iPSCs) [9092] have been explored for regenerating retinal neurons, corneal or conjunctival epithelial cells, and the RPE. The reason for using these stem cells is their capability to form neural progenitor cells or mature optic cells and the release of trophic factors important for reparative mechanism (Table 1 [76, 82, 83, 8588, 90, 91, 9399]). The manipulation of these cells raises less debate over moral and ethical issues than the use of ESCs [93] and fetal stem cells [100, 101]. Moreover, the eye is a suitable target organ for stem cell transplantation because it is immune-privileged, and strict containment by the blood-retinal barrier will disable the emigration of possibly maltransformed injected cells to extraocular tissues [102].

Figure 4 shows microcomputed tomography images to track the injected human Whartons jelly-derived MSCs (hWJ-MSCs) in a Retinitis Pigmentosa rat model [103]. The gold-loaded hWJ-MSC remained in the eye with no systemic migration to other organs detected on day 70 after injection. This study indicated that the injected MSCs were confined to the subretinal layer of experimented eyes and that no systemic migration occurred in the rat model [103]. Figure 5 shows rat retinal cell phenotypes exhibiting modest level of human MSCs marker, as observed by confocal microscopy. Colocalization of stem 121 (mesenchymal stem cell marker, red color in Figure 5) with rhodopsin (green), GFAP (Mller glial cells, green color in Figure 5), and PKC- (bipolar cells, green color in Figure 5) [103] was found, implying that MSCs could have differentiated into specific retinal cell phenotypes upon activation by cytokines released by the dying cells or fused with the degenerating cells to rescue tissue death [104, 105]. It is noteworthy that other studies have also demonstrated differentiation of human Whartons jelly-derived MSCs into neurons [106], glia [107], and retinal progenitor cells [108]. Hence, introduction of hWJ-MSCs might be beneficial in inducing certain level of cell repair or regeneration in retinal degeneration.

However, the most significant barrier for successful visual restoration has been the failure of these neuron-derived stem cells to integrate into the retinal circuitry. In central nervous system, stem cells and its neuron derivatives were reported to successfully integrate into the host neural circuitry [109112]. On the contrary, the integration of transplanted cells might be influenced by the molecular predisposition in the damaged eye tissues, which could vary even between different regions [111] and the ontogenetic stage of transplanted neurons [104]. MacLaren et al. first demonstrated that physiologically older retinal tissues showed predilection and tissue acceptance to later ontogenetic stage of transplanted retinal cells, that is, immature postmitotic photoreceptors over neural progenitor cells [104]. Human ESCs-directed differentiated retinal cells could migrate and integrate into the retinal layer and form synapses in transgenic mice following intravitreal injection at birth or postnatal day 1 [113]. Conversely, there is also a report that ESCs-derived neural stem cells showed lesser migration and integration in the retina. To prove that ontogenetic stage of transplanted neurons would also determine the level of integration, West et al. used three-dimensional culture of mouse ESCs with overexpression of Rax genes to direct generation of retinal neuron cells at different time points to establish an equivalent retinal developmental stage for a retinal cell integration study [114]. Unfortunately, their results were not able to prove that the transplantation of photoreceptors at the late ontogenetic stage has better integration into the retinal layer. However, a significant reduction of tumorigenic formation in the retina was observed when photoreceptors were used than when ESCs were used. The difference in the gene expression profile of the different ontogenetic stage of stem cells or progenitors may not mimic the native characteristics of retinal neurons, hence, an incomplete integration into the retinal circuitry. The characteristics of transplanted cells can be significantly affected by the choice of culture methods [115]. Generally, future studies should widen focus on the determination of geographical protein expression in different ocular disorders and identification of similarities in gene expression, rather than mere dependence on morphological observation or in vitro functional studies. It is hoped that these efforts would provide clue on tissue predilection over specific stem cells or its neuron derivatives for maximum therapeutic efficacy.

There is also a suggestion that concomitant transplantation of stem cells with telocytes may help restore the microenvironment. Telocytes are interstitial cells that reside in close contact with stem cells (Figure 6) and may be responsible for the transfer of bioactive molecules (nutrients and paracrine factors) among neighboring cells such as nerve cells and blood vessels [116]. The presence of telocytes has been reported in skeletal muscle [116], uterus [117], skin [118], heart [119], digestive tract [120], lung [121], and iris and uvea of mouse eyes [122].

Advanced techniques have also used a denuded amniotic membrane as a substratum for epithelial cell culture and stratification [123] and used cord blood serum to replace xenobiotic material [124] for conjunctival or corneal transplant. Recently, there is also research effort in developing a new mode of delivery of stem cells through direct application of contact lenses on the ocular surface [125]. Observation of successful stratified epithelization on a corneal wound bed in a rabbit model of limbal stem cell deficiency following application of modified-contact lens (with plasma polymer with high acid functional group) cultured with limbal cells has high clinical indications, suggesting that surgery for corneal transplant may not be needed in the future [125]. Laboratory procedures are getting standardized with simple protocol for culturing limbal cells to adopt with many cell sources [126]. Markers like Keratin 14 is used to map the distribution of precursor cells of cornea and suggested for corneal renewal with stem cells for alternative regenerative therapy [127]. Also to strengthen the universal standard in techniques, good manufacturer practice based on UK facilities on ocular surface reconstruction is suggested for use outside the UK [128].

Retinal degeneration is a medical condition that affects the health and welfare of adults and children in the developed world. It represents a group of blinding diseases that include age-related macular disease, glaucoma, optic neuropathies, and retinal vascular complication. Many clinical trials were performed to develop treatments for these diseases. However, it was reported that those approaches were still unable to entirely cure the disease. Interestingly, a stem cell-based treatment shows an extraordinary potential to rectify some of these diseases. In the past few years, studies strongly propose that stem-cell-based therapy has the ability to correct defective function of retina photoreceptors [114, 126], ganglion cells, retinal pigment epithelium (RPE) [129, 130], and optic nerve [131, 132].

Retinal Pigment Epithelial Cells (RPE) and Age-Related Macular Disease (ARMD). The macula enables people to read, process faces, and drive. Degeneration of the RPE leads to malformation at the macular area of the central vision at the initial phase and eventually progressive loss of central vision. This medical condition, known as age-related macular degeneration (ARMD), contributes to the highest cases of blindness in the elderly population globally [92, 133].

ARMD could be present either in wet or in dry forms (wet and dry ARMD) [134]. Wet ARMD manifests as neovascularization, which can be successfully managed with monthly inoculation of antiangiogenic drugs such as Lucentis [135]. Although effective in treating wet ARMD, the monthly injection into the eye causes discomfort and inconvenience to the patient and is expensive [136]. In contrast, dry ARMD presents as drusenoid aggregates under the basal side of the RPE layer at the early phase. These aggregations will lead to geographic atrophy with pronounced loss of the RPE and photoreceptors at later stage. Most of the ARMD cases (80 to 90% patients) occurred due to the dry form as no effective treatments have been found to date.

Currently, clinical trials using RPE-derived human from ESCs and other stem cell-derived therapy are ongoing and becoming a promising approach for the treatment of ARMD. Several companies and institutions are actively involved in stem cell research to treat various ocular diseases, including institutions in Japan, USA, Europe, South America, China, Iran, Taiwan, and South Korea. To date, stem cell therapies have been administered to over 200 patients globally. Schwartz and his colleagues [72, 130] performed clinical trials on patients affected by dry ARMD (NCT01344993) and Stargardts macular dystrophy (NCT01345006) [72]. In these trials, the researchers injected 50,000 to 200,000 hESC-derived retinal pigment epithelial cells into the worst-affected retina of the patients. Figure 7 shows fundus images taken from the patients following transplantation with hESCs-derived retinal pigment epithelial cells. There were increases in the area size and subretinal pigmentation of patches of transplanted cells in 72% of the treated patients with dry ARMD and Stargardts macular dystrophy at 315 months later [72]. Figures 7(b) and 7(c) showed that the patch of transplanted cells, which were present typically at the boundary of atrophic lesion on the eye of dry ARMD patients, became larger and more pigmented within 6 months. Meanwhile, in a patient with Stargardts macular dystrophy, patches of pigmented cells were found around the boundary of baseline atrophy in retinal pigment epithelium layer (Figure 7(e)) and appeared more prominent after 12 months of transplantation (Figure 7(f)). Figure 7(g) shows preoperative image of another Stargardts macular dystrophic patient with a large central area of atrophy. Six months later after transplantation, the superior half of the atrophic lesion was totally filled in by the transplanted retinal pigment epithelial cells (Figure 7(h)). The filled area became larger in size and more pigmented sites were seen after 15 months of transplantation (Figure 7(i)) [72]. It is important to emphasize that the vision-related quality of life was enhanced in both patients of atrophic ARMD and Stargardts macular dystrophy. None of the patients have reported signs of abnormal tissue formation at either the local or ectopic site of injections or immune rejections even four months after injection [72].

It should be mentioned that Professor Takahashis group [137141] at Kyoto University has been studying the transplantation of retinal pigmented epithelium cells into ARMD patients, which are differentiated from human iPSCs reprogrammed from patient cells. The tissue has maintained its brownish color, which is a sign that it has not been attacked by the immune system [142].

Ocata Therapeutics (formerly known as Advanced Cell Technology) has sponsored the trials at the Jules Stein Eye Institute, Massachusetts Eye & Ear, Wills Eye Institute and Bascom Palmer Eye Institute. Neurotech Pharmaceuticals (NCT00447954) has conducted a trial using encapsulated, modified human RPE cells to express ciliary neurotrophic factor for intraocular implantation into ARMD patients. Another report (NCT01518127) shows that Siqueira has been engaged in wet ARMD treatment using bone marrow-derived stem cells in a prospective phase I/II clinical trial [143]. Unfortunately, the complete outcomes have not yet been posted in the clinical trial registry of US National Institutes of Health (ClinicalTrials.gov) despite the fact that the trial was ended in December 2015. Other institutes, such as CHA Bio & Diostech (NCT01674829), Janssen R&D (NCT01226628), University of California, Davis Eye Center (NCT01736059) [83], University College London, Moorfields Eye Hospital (NCT01691261) [144], Hollywood Eye Institute (NCT02024269), and Stem Cells Inc. (NCT01632527) [145], were also engaged in stem cell therapy for ARMD.

Glaucoma is the most common neurodegenerative disease in the inner part of retina. Prevalence models predict an increase of glaucoma incidence to 79.6 million by 2020 worldwide, a jump from 60.5 million in 2010 [11]. Similar to other neurodegenerative disorders, the loss of the nerve cell population from the central nervous system can be used to predict the risk of glaucoma. Additionally, signs of glutamate toxicity, oxidative stress, impaired axonal transport, and reactive glial changes are also well-characterized in glaucoma [146, 147]. However, in glaucoma, retinal ganglion cells (RGC) predominantly die, which leads to the degeneration of the optic nerve and disconnecting the communication of signals from the retina to the brain.

Increases in age and raised intraocular pressure can lead to the occurrence of glaucoma. Diagnosis and prescription of a suitable treatment for glaucoma can be too late as patients may present asymptomatically until the end stage of the disease, which results in significant loss of visual function. Clinically verified treatments such as medication and eye surgery could delay the development of the glaucoma by reducing intraocular pressure but fail to halt the disease entirely to prevent loss of vision [148]. As of the date of this review, two registered clinical trials (NCT01920867 and NCT02330978) are recruiting patients for glaucoma treatment with bone marrow-derived mesenchymal stem cells. The safety of autologous stem cells derived from adipose tissue is also currently being tested in a phase I/II clinical trial (NCT02144103) for glaucomatous neurodegeneration. Additionally, Dr. Goldberg at the University of California has tested the treatment of ciliary neurotrophic factor on primary open angle glaucoma patients at the Bascom Palmer Eye Institute, University of Miami (NCT01408472). Several preclinical models have proven that ciliary neurotrophic factor could augment the survival and renewability of retinal ganglion cells [149, 150].

The optic nerve can lead to various pathologies due to intraorbital, intracranial, intrinsic, or systemic disorders. Optic nerve diseases could also lead to life- and vision-threatening conditions [151]. Neural loss from the optic nerve is a frequently occurring, irreversible blinding pathology that involves optic light-sensing tissue. Similar to the brain, the eye, which is a part of the central nervous system, will not be able to restore neuron loss after the occurrence of disease [148]. The patterns of optic nerve diseases provide information to the researcher to help understand the fundamental pathological activity and establish a method to enhance advanced detection and treatment strategies [148]. Recently, Dr. Jamadar worked in a clinical trial (NCT01834079) at Chaitanya Hospital, Pune, to evaluate the safety and efficacy of using bone marrow-derived autologous cells for treating optic nerve disease. It is hoped that the primary outcome of reducing degeneration of the optic nerve will also lead to improvement in visual function and decreased intracranial hypertension. Neurotech Pharmaceuticals also used similar RPE cell implants to administer CNTF to patients with optic nerve stroke in a separate phase I clinical trial (NCT01411657).

Retinal diseases other than the major ocular diseases discussed above also cause problems. These diseases include retinal detachment and retinal vascular complications. Retinal detachment is a medical condition in which the retina separates from the back of the eye. In a case report by Wilkes et al. [152], one in 10,000 people faces this problem per year. As the detachment period increases, the visual recovery reduces at an exponential rate after macula-off retinal detachment [153]. With modern surgical techniques, such as scleral buckling, pneumatic retinopexy, and pars plana vitrectomy, we can anticipate more than a 90% success rate for anatomical repair [154]. Although these treatments show positive results anatomically, the visual result still remains displeasing due to the enduring functional injury to the macula [155].

Clinical trials for treating retinal detachment began in the 1980s. A report by Brinton states that of 106 cases of eye trauma, 55 eyes (52%) attained final visual acuity of 20/100 after surgery [156]. The researchers also found that patients who engaged in later vitrectomy did not achieve a better final visual outcome than those who engaged in early vitrectomy within 14 days of impairment. In a separate study, Burton found that 53% of patients who experienced macula-off retinal detachments and underwent early surgery reached visual acuity of 20/20 to 20/50 [153]. However, patients with long-standing detachments were not able to reap functional benefits after surgery. A case reported by Suzuki and Hirose in 1997 states that after 3 months of total retinal detachment, vision was recovered in a patient with no light perception (NLP) [157]. After undergoing two surgeries, the patient recovered counting fingers (CF) vision. The scientists hypothesized that some retinal receptors were capable of eluding the failure. Although all of these trials showed a positive result in patient visual function recovery, the treatment is applicable to only early stage impairment and is costly and inconvenient. The use of stem cell-based therapy in retinal detachment cases might be one of the alternative treatments for early or late stage retinal detachment. For instance, fibrovascular scarring in ARMD, DR, ROP, and neovascular glaucoma [158] can be attenuated by introduction of MSCs. The scar tissue could prevent reattachment of retina [159]. MSCs could also neutralize reactive oxygen species in injured eye tissue and secrete various cytokines and growth factors including hepatocyte growth factor (HGF), interleukin 10, and adrenomedullin, which has antifibrotic properties [160].

Some of the commonly arising retinal diseases that lead to vision loss are associated with retinal vascular complications. Of these diseases, diabetic retinopathy, retinal vein occlusion (RVO), diabetic macular oedema (DMO), and proliferative diabetic retinopathy (PDR) are of definite epidemiological significance and lead to blindness. Diabetic retinopathy is the third most dominant source of profound visual function impairment and blindness, followed by RVO [161]. In addition to RVO and PDR, ischemic retinopathies are also familiar diseases involved in vasodegeneration. This situation leads to hypoxia, which provokes the release of cytokines and growth factor in neighboring tissues [162] and then leakage of blood vessels and neovascularization, which has a functionally negative effect on optics.

Intravitreal injections of anti-VEGF antibodies and corticosteroids or laser photocoagulation are the contemporary clinical treatments that help attenuate vascular leakage and macular oedema. However, these treatments cause undesirable side effects and do not resolve the fundamental pathology. The vasodegeneration that occurs in the retina is primarily due to the loss of endothelial cells, smooth muscle cells, and pericytes, finally resulting in vascular blockage and hypoxia [162]. An ongoing clinical trial (NCT02119689), which started since 2011, has aimed to study on the impaired function of endothelial progenitor cells in patients of diabetic retinopathy. Stitt et al. hypothesized that the introduction of vascular stem cells such as endothelial progenitor cells can recondition the retinal nerve diseases by repairing and restoring the damaged vessel [163]. EyeCyte Inc. develops endothelial progenitor cells for use as angiogenic therapy in response to clinical indications specific to retinal nerve diseases, particularly those of ischemic diseases [148, 164, 165]. Additionally, the University of Sao Paulo has sponsored Dr. Rubens trial using intravitreal injections of bone marrow-derived hematopoietic stem cells (CD34+ cells) for treating ischemic and diabetic retinopathies (NCT01518842). A subset of CD34+ hematopoietic stem cells, which are proangiogenic, could work in synergy with endothelial cells to repair damaged blood vessels.

Stem cell-based therapy holds an extraordinary prospective in improving the lives of people who suffer from visual disorders. Research in this area will continue to grow to develop new remedies in treating and preventing the problem of vision loss. Interestingly, stem cell-based therapy is not a one-stop general remedy; however, it carries a promising future in producing new biological elements used to treat vision loss.

The authors declare that there are no competing interests regarding the publication of this paper.

Padma Priya Sivan, Sakinah Syed, and Pooi-Ling Mok contributed equally to this paper.

This research was partially supported by the Fundamental Research Grants Scheme (FRGS), Ministry of Educations, Malaysia, under Grant no. 5524401. This work was also supported by the Putra Grant, Universiti Putra Malaysia, Malaysia (9436300), and Ministry of Science and Technology, Taiwan, under Grant no. 104-2221-E-008-107-MY3. Deanship of Scientific Research, College of Science Research Centre, King Saud University, and Kingdom of Saudi Arabia are also acknowledged.

Excerpt from:
Stem Cell Therapy for Treatment of Ocular Disorders - Hindawi

Posted in California Stem Cells | Comments Off on Stem Cell Therapy for Treatment of Ocular Disorders – Hindawi

How Cinnamon Lowers Blood Sugar and Fights Diabetes – Healthline

Posted: December 27, 2022 at 12:23 am

Cinnamon may help support blood sugar management by increasing insulin sensitivity, decreasing blood sugar levels after eating, and reducing the risk of diabetes-related complications.

Diabetes is a condition that affects blood sugar management, which can lead to long-term complications like heart disease, kidney disease, and nerve damage (1).

Treatment often includes medications and insulin injections, but many people are also interested in foods that can help lower blood sugar.

One example is cinnamon, a commonly used spice thats added to sweet and savory dishes around the world.

It provides many health benefits, including the ability to help lower blood sugar and manage diabetes (2).

This article tells you everything you need to know about cinnamon and its effects on blood sugar management and diabetes.

Cinnamon is an aromatic spice that comes from the bark of several species of Cinnamomum trees (3).

While you may associate cinnamon with rolls or breakfast cereals, it has actually been used for thousands of years in traditional medicine and food preservation (3).

To obtain cinnamon, the inner bark of Cinnamomum trees must be removed. The bark then undergoes a drying process that causes it to curl up and yield cinnamon sticks, or quills, which can be further processed into powdered cinnamon.

Several different varieties of cinnamon are sold in the United States, and they are typically categorized into two different types (3):

While both types are sold as cinnamon, there are important differences between the two, which will be discussed later in this article.

Cinnamon is made from the dried bark of Cinnamomum trees and is generally categorized into two varieties.

A quick glance at cinnamons nutrition facts may not lead you to believe that its often considered a superfood (4).

A single teaspoon (tsp), the average serving size of cinnamon, doesnt contain a lot of vitamins or minerals. But many recipes call for more than just 1 tsp, and larger amounts of cinnamon do contain a high amount of vitamins and minerals. It also contains larger amounts of antioxidants, which provide many of cinnamons health benefits (5).

In fact, one study in 84 people with polycystic ovary syndrome (PCOS) found that taking 1,500 milligrams (mg) of cinnamon daily led to a significant increase in antioxidant blood levels after 8 weeks (6).

Antioxidants are important because they help the body reduce oxidative stress, a type of damage to cells that is caused by harmful free radicals (7).

One study showed that consuming 1 gram (g) of cinnamon extract daily for 12 weeks reduced fasting blood sugar levels and improved markers of oxidative stress in people with type 2 diabetes (8).

This is significant because oxidative stress has been linked to the development of nearly every chronic disease, including type 2 diabetes (9).

Cinnamon is loaded with antioxidants that decrease oxidative stress. This may be beneficial for several chronic conditions, including type 2 diabetes.

In people with diabetes, either their pancreas cannot produce enough insulin or cells do not respond to insulin properly, leading to high blood sugar levels.

Cinnamon may help lower blood sugar and fight diabetes by imitating the effects of insulin and increasing the movement of sugar from the bloodstream into cells (10).

It can also help lower blood sugar levels by increasing insulin sensitivity, making insulin more efficient at moving sugar into cells (11).

One study in 80 people with PCOS found that taking 1.5 g of cinnamon powder daily for 12 weeks caused a significant reduction in fasting insulin levels and improved insulin sensitivity compared with a placebo (12).

Similarly, another study found that taking 250 mg of cinnamon twice daily for 2 months improved insulin sensitivity in 137 people with high blood sugar levels (13).

Cinnamon may help lower blood sugar by mimicking the effects of insulin and increasing insulins ability to move sugar into cells.

Several studies suggest that cinnamon may help improve blood sugar management.

In fact, a review of 16 studies concluded that cinnamon could significantly reduce fasting blood sugar levels and insulin resistance compared with a placebo in people with type 2 diabetes and prediabetes (14).

Some studies have also found that it could also lower hemoglobin A1c, a measure of long-term blood sugar control.

For instance, a research review reported that cinnamon could reduce hemoglobin A1c in people with type 2 diabetes by 0.27% to 0.83% while also decreasing fasting blood sugar levels by up to 52.2 mg per deciliter (15).

According to another review of 11 studies, cinnamon supplements could lead to modest reductions in fasting blood sugar levels and hemoglobin A1c (16).

However, researchers also noted that more studies are needed and cinnamon should not be used in place of medications or diet and lifestyle changes to manage blood sugar levels (16).

Cinnamon shows promise in lowering fasting blood sugar levels and reducing hemoglobin A1c. However, more research is needed.

Postprandial blood sugar refers to your blood sugar level after eating. Blood sugar levels can increase quite a bit after you eat, depending on the size of the meal and how many carbs it contains (17).

These blood sugar shifts can increase levels of oxidative stress and inflammation, which can damage your bodys cells and contribute to chronic disease (18).

Cinnamon can help keep these blood sugar spikes after meals in check. Some research suggests that it does this by slowing down the rate at which food empties out of your stomach (19).

A study from 2007 found that consuming 1.2 tsp, or 6 g, of cinnamon with a serving of rice pudding slowed the emptying of the stomach and decreased subsequent spikes in blood sugar levels compared with eating rice pudding alone (20).

Other studies suggest that cinnamon may lower blood sugar following meals by blocking digestive enzymes that break down carbs in the small intestine (21).

Cinnamon can help lower blood sugar following meals, possibly by slowing stomach emptying and blocking digestive enzymes.

In addition to supporting blood sugar management, cinnamon may also lower the risk of certain complications associated with diabetes, including heart disease and stroke (22).

For example, one review of 13 studies showed that cinnamon could decrease levels of triglycerides and total cholesterol, both of which are risk factors for heart disease (23).

An analysis from 2020 found that supplementing with at least 2 g of cinnamon per day could significantly lower both systolic and diastolic blood pressure over 8 weeks (24).

Diabetes has also been increasingly linked to the development of Alzheimers disease and other types of dementia, with some people now referring to Alzheimers disease as type 3 diabetes. The classification of type 3 diabetes is highly controversial, and its not widely accepted by the medical community as a clinical diagnosis (25).

Studies suggest that cinnamon extract may decrease the ability of two proteins beta-amyloid and tau to form plaques and tangles, which are routinely linked to the development of Alzheimers disease (26).

However, this research has been completed only in test tubes and animal studies. Further studies in humans are needed to confirm these findings.

Cinnamon may help lower the risk of diseases related to diabetes, such as heart disease and Alzheimers disease. However, more research is needed.

Cinnamon is typically grouped into two different types Ceylon and Cassia.

Cassia cinnamon can be derived from a few different species of Cinnamomum trees. Its generally inexpensive and is found in most food products and the spice aisle of the grocery store.

Ceylon cinnamon, on the other hand, is specifically derived from the Cinnamomum verum tree. Its typically more expensive and less common than Cassia, but studies have shown that Ceylon cinnamon contains more antioxidants (27).

Because it contains more antioxidants, its possible that Ceylon cinnamon may offer more health benefits.

Nevertheless, although several animal and test-tube studies have highlighted the benefits of Ceylon cinnamon, most studies demonstrating health benefits of cinnamon in humans have used the Cassia variety (28).

While both varieties of cinnamon likely lower blood sugar and fight diabetes, most research has focused on the potential benefits of Cassia cinnamon. More research is needed.

Cassia cinnamon is not only lower in antioxidants but also high in a potentially harmful substance called coumarin, an organic substance found in many plants.

Several studies in rats have shown coumarin can be toxic to the liver, leading to concern that it may cause liver damage in humans as well (29).

Accordingly, the European Food Safety Authority has set the tolerable daily intake for coumarin at 0.045 mg per pound (lb.), or 0.1 mg per kilogram (kg) (30).

Using average coumarin levels for Cassia cinnamon, this would be equivalent to about half a tsp (2.5 g) of Cassia cinnamon per day for a 165-pound (75-kg) individual.

Cassia cinnamon is particularly high in coumarin, and you can easily consume more than the upper limit by taking Cassia cinnamon supplements or even eating large amounts of it in foods.

However, Ceylon cinnamon contains much lower amounts of coumarin, and it would be difficult to consume more than the recommended amount of coumarin with this type (3).

Keep in mind that there is limited information on the long-term safety of cinnamon supplements for children and people who are pregnant or nursing (31).

Additionally, people with diabetes who take medications or insulin should talk with a doctor before adding cinnamon to their daily routine, as it could increase the risk of low blood sugar levels, or hypoglycemia (32).

Cassia cinnamon is high in coumarin, which may cause liver damage if consumed in high amounts. People taking medications for diabetes should also talk to a doctor before taking cinnamon supplements to avoid adverse side effects.

Cinnamons benefits for lowering blood sugar have been well-studied.

Yet despite this, no consensus has been reached regarding how much you should consume to reap the benefits while avoiding potential risks.

Generally, most research has studied the effects of 16 g per day, either as a supplement or powder added to foods (15).

However, keep in mind that the coumarin content of Cassia cinnamon can vary. Thats why it may be best to stick to lower doses of around 0.51 g of Cassia cinnamon per day to avoid surpassing the tolerable daily intake of coumarin.

On the other hand, Ceylon cinnamon contains significantly less coumarin and can be consumed safely in doses of up to 1.2 tsp. (6 g) per day (3).

Be sure to talk with a doctor before adding cinnamon supplements to your routine. You may want to start with a lower dose and work your way up to avoid adverse effects on health.

It may be best to limit your intake of Cassia cinnamon to 0.51 g per day. Ceylon cinnamon can be consumed in higher amounts, even though it may not be necessary.

Many studies have suggested that cinnamon has the ability to lower blood sugar and help manage common diabetes complications, among other health benefits.

If you want to take cinnamon supplements or add it to your meals to help lower your blood sugar, it may be best to use Ceylon instead of Cassia.

While it may be more expensive, Ceylon cinnamon contains more antioxidants and lower amounts of coumarin, which can potentially cause liver damage.

Its probably best not to exceed 0.51 g of Cassia daily, but taking up to 1.2 tsp (6 g) daily of Ceylon cinnamon is likely safe.

Read the rest here:
How Cinnamon Lowers Blood Sugar and Fights Diabetes - Healthline

Posted in Diabetes | Comments Off on How Cinnamon Lowers Blood Sugar and Fights Diabetes – Healthline

Diabetic Foot Ulcers: Causes and Treatments – Healthline

Posted: December 27, 2022 at 12:23 am

Foot ulcers are a common complication of diabetes that is not being managed through methods such as diet, exercise, and insulin treatment. Ulcers are formed as a result of skin tissue breaking down and exposing the layers underneath.

Theyre most common under your big toes and the balls of your feet, and they can affect your feet down to the bones.

All people with diabetes can develop foot ulcers, but good foot care can help prevent them. Treatment for diabetic foot ulcers varies depending on their causes.

Discuss any foot concerns with your doctor to ensure its not a serious problem, as infected ulcers can result in amputation if neglected.

One of the first signs of a foot ulcer is drainage from your foot that might stain your socks or leak out in your shoe. Unusual swelling, irritation, redness, and odors from one or both feet are also common early symptoms.

The most visible sign of a serious foot ulcer is black tissue (called eschar) surrounding the ulcer. This forms because of an absence of healthy blood flow to the area around the ulcer.

Partial or complete gangrene, which refers to tissue death due to infections, can appear around the ulcer. In this case, odorous discharge, pain, and numbness can occur.

Signs of foot ulcers are not always obvious. Sometimes, you wont even show symptoms of ulcers until the ulcer has become infected.

Talk with your doctor if you begin to see any skin discoloration, especially tissue that has turned black, or feel any pain around an area that appears callused or irritated.

Your doctor will likely identify the seriousness of your ulcer on a scale of 0 to 5 using the Wagner Ulcer Classification System:

Ulcers in people with diabetes are most commonly caused by:

Poor blood circulation is a form of vascular disease in which blood doesnt flow to your feet efficiently. Poor circulation can also make it more difficult for ulcers to heal.

High glucose levels can slow the healing process of an infected foot ulcer, so blood sugar management is critical. People with type 2 diabetes and other ailments often have a harder time fighting off infections from ulcers.

Nerve damage is a long-term effect and can lead to a loss of feeling in your feet. Damaged nerves can feel tingly and painful. Nerve damage reduces sensitivity to foot pain and results in painless wounds that can cause ulcers.

Ulcers can be identified by drainage from the affected area and sometimes a noticeable lump that isnt always painful.

All people with diabetes are at risk for foot ulcers, which can have multiple causes. Some factors can increase the risk of foot ulcers, including:

Diabetic foot ulcers are also most common in older men.

Stay off your feet to prevent pain from ulcers. This is called off-loading, and its helpful for all forms of diabetic foot ulcers. Pressure from walking can make an infection worse and an ulcer expand.

Your doctor may recommend wearing certain items to protect your feet:

Doctors can remove foot ulcers with a debridement, the removal of dead skin or foreign objects that may have caused the ulcer.

An infection is a serious complication of a foot ulcer and requires immediate treatment. Not all infections are treated the same way.

Tissue surrounding the ulcer may be sent to a lab to determine which antibiotic will help. If your doctor suspects a serious infection, they may order an X-ray to look for signs of bone infection.

Infection of a foot ulcer can be prevented with:

Your doctor may prescribe antibiotics, antiplatelets, or anticlotting medications to treat your ulcer if the infection progresses even after preventive or antipressure treatments.

Many of these antibiotics attack Staphylococcus aureus,bacteria known to cause staph infections, or -haemolytic Streptococcus, which is normally found in your intestines.

Talk with your doctor about other health conditions you have that might increase your risk of infections by these harmful bacteria, including HIV and liver problems.

Your doctor may recommend that you seek surgical help for your ulcers. A surgeon can help alleviate pressure around your ulcer by shaving down the bone or removing foot abnormalities such as bunions or hammertoes.

You will likely not need surgery on your ulcer. However, if no other treatment option can help your ulcer heal, surgery can prevent your ulcer from becoming worse or leading to amputation.

According to a 2017 review article in the New England Journal of Medicine, more than half of diabetic foot ulcers become infected. Approximately 20 percent of moderate to severe foot infections in people with diabetes lead to amputation. Preventive care is crucial.

Closely manage your blood glucose, as your chances of diabetes complications remain low when your blood sugar is stable. You can also help prevent foot problems by:

Foot ulcers can return after theyve been treated. Scar tissue can become infected if the area is aggravated again, so your doctor may recommend you wear shoes specially designed for people with diabetes to prevent ulcers from returning.

If you begin to see blackened flesh around an area of numbness, see your doctor right away to seek treatment for an infected foot ulcer. If untreated, ulcers can cause abscesses and spread to other areas on your feet and legs.

At this point, ulcers can often only be treated by surgery, amputation, or replacement of lost skin by synthetic skin substitutes.

When caught early, foot ulcers are treatable. See a doctor right away if you develop a sore on your foot, as the likelihood of infection increases the longer you wait. Untreated infections may require amputations.

While your ulcers heal, stay off your feet and follow your treatment plan. Diabetic foot ulcers can take several weeks to heal.

Ulcers may take longer to heal if your blood sugar is high and constant pressure is applied to the ulcer.

Remaining on a diet that helps you meet your glycemic targets and off-loading pressure from your feet is the most effective way to allow your foot ulcers to heal.

Once an ulcer has healed, consistent preventive care will help you stop an ulcer from ever returning.

Read more:
Diabetic Foot Ulcers: Causes and Treatments - Healthline

Posted in Diabetes | Comments Off on Diabetic Foot Ulcers: Causes and Treatments – Healthline

Mersana Therapeutics Announces Research Collaboration and Commercial License Agreement with Merck KGaA, Darmstadt, Germany to Develop Novel…

Posted: December 26, 2022 at 12:12 am

CAMBRIDGE, Mass., Dec. 22, 2022 (GLOBE NEWSWIRE) -- Mersana Therapeutics, Inc. (NASDAQ: MRSN), a clinical-stage biopharmaceutical company focused on discovering and developing a pipeline of antibody-drug conjugates (ADCs) targeting cancers in areas of high unmet medical need, today announced a research collaboration and commercial license agreement with a subsidiary of Merck KGaA, Darmstadt, Germany to discover novel Immunosynthen ADCs directed against up to two targets. Immunosynthen, Mersana’s proprietary STING-agonist ADC platform, is designed to generate systemically administered ADCs that locally activate STING signaling in both ?tumor-resident immune cells and in antigen-expressing tumor cells, unlocking the anti-tumor potential of innate immune stimulation.?

The rest is here:
Mersana Therapeutics Announces Research Collaboration and Commercial License Agreement with Merck KGaA, Darmstadt, Germany to Develop Novel...

Posted in Global News Feed | Comments Off on Mersana Therapeutics Announces Research Collaboration and Commercial License Agreement with Merck KGaA, Darmstadt, Germany to Develop Novel…

Page 120«..1020..119120121122..130140..»