Category Archives: Stem Cell Medicine


UC Davis licenses novel compound that helps stem cells regenerate bone – HealthCanal.com (press release) (blog)

The University of California, Davis, has reached a licensing agreement with Regenerative Arthritis and Bone Medicine (RABOME) for a class of drugs developed at the university that hold potential for treating diseases associated with bone loss and inflammatory arthritis.

From Left: Fred Tileston (RABOME), Ruiwu Liu, Nancy Lane, Christy Pifer, Wei Yao, Kit Lam (UC Davis Health), and Jiwei Chen (RABOME).

The license, negotiated by the InnovationAccess team within the UC Davis Office of Research, provides the university-affiliated startup with rights to four families of patents and patent applications related to the novel composition of a hybrid molecule, known as LLP2A-alendronate, which has been found to effectively direct mesenchymal stem cells (MSCs) to induce bone regeneration in animal models. The compound works by guiding transplanted and endogenous MSCs to the surface of the bone where they differentiate into bone-forming cells, thereby increasing bone mass and strength. These cells are also immune-modulating, which helps to reduce inflammation at target sites.

The use of stem cells as therapeutic agents is a growing field, but directing stem cells to travel and adhere to the surface of bone for bone formation has been an elusive goal in regenerative medicine.

There are many stem cells, even in elderly people, but they do not readily migrate to bone, said Wei Yao, co-inventor and associate professor of internal medicine at UC Davis. Finding a molecule that attaches to stem cells and guides them to the targets we need provides a real breakthrough.

Translating discovery into societal and commercial impact

Late last year, RABOME received approval from the U.S. Food and Drug Administration to begin phase I clinical trials to evaluate the safety of the drug in humans. The study sites are currently screening patients for enrollment.

We are pursuing several indications for use, but our initial focus is in developing a treatment for osteonecrosis, a disease caused by reduced blood flow to bones, says Fred Tileston, president and chief executive officer RABOME, which is a California-based company. As many as 20,000 people per year in the United States develop osteonecrosis.

RABOME also plans to pursue other indications for use including fracture healing, osteoporosis and inflammatory arthritis.

We are pleased that this very promising technology is being shepherded by Mr. Tileston, who is an experienced business leader and entrepreneur, said Dushyant Pathak, associate vice chancellor for Technology Management and Corporate Relations at UC Davis. It is exciting to see the teams progress in translating the discovery into commercial and societal impact.

Breaking barriers through cross-discipline collaboration

The development of the novel therapy is the result of a successful research collaboration between two teams at UC Davis: a group of experts on bone health, led by Nancy Lane and Wei Yao from the UC Davis Center for Musculoskeletal Health, and a synergistic group of medicinal chemists led by Kit Lam and Ruiwu Liu from the Department of Biochemistry and Molecular Medicine.

This research was a collaboration of stem cell biologists, biochemists, translational scientists, a bone biologist and clinicians, said Lane, endowed professor of medicine, rheumatology and aging research, anda principal investigator. It was a truly fruitful team effort with remarkable results.

Lane received a Disease Team Therapy Development research grant in 2012 from the California Institute for Regenerative Medicine (CIRM) which, along with federal grants from the National Institutes of Health, supported the preclinical research. CIRM was established in 2004 via California Proposition 71 to fund stem cell research in attempt to accelerate and improve treatments for patients where current needs are unmet.

Conflict of interest disclosure

Because Tileston and Lane are married, UC Davis conducted a conflict of interest review of its licensing agreement with RABOME. The university determined that it did not rise to the level of a financial conflict of interest under NIH rules, which require a finding of a direct and significant impact.

Send email Phone: 916-734-9048

AJ Chelin, Office of Research Send email Phone:530-752-1101

Here is the original post:
UC Davis licenses novel compound that helps stem cells regenerate bone - HealthCanal.com (press release) (blog)

Study of SanBio’s Stem Cell Treatment for Stroke Receives Innovation Award from American Heart Association – Yahoo Finance

MOUNTAIN VIEW, Calif.--(BUSINESS WIRE)--

SanBio, Inc., a scientific leader in regenerative medicine for neurological disorders, today announced that a recent publication of its novel stem cell treatment, SB623, for patients following a stroke, has received a prestigious award from the American Heart Association. The scientific article, Clinical Outcomes of Transplanted Modified Bone Marrow-Derived Mesenchymal Stem Cells in Stroke: A Phase 1/2a Study, was the third prize winner of the 2016 Stroke Progress and Innovation Award.

The Progress and Innovation Awards are offered by Stroke, a leading scientific journal addressing the diagnosis and treatment of cerebrovascular diseases, jointly with the American Heart Association and American Stroke Association. Previous award winners have established important standards of care in neurology, including Activase (alteplase) and induced hypothermia treatment.

Dr. Damien Bates, Chief Medical Officer and Head of Research at SanBio, said, This prize from the American Heart Association recognizes the innovation of our stem cell treatment, SB623, and its potential to treat patients suffering from chronic physical impairments following ischemic stroke. The results of this study are encouraging to all those suffering from the long-term effects of stroke as well as the medical community working to advance treatment options.

The clinical trial was a Phase 1/2a, open-label, single-arm, dose escalation study of 18 patients with chronic motor deficits present for at least six months following an ischemic stroke. Patients received precisely targeted injections of SB623 cells directly into the neural tissue surrounding the damaged area of the brain.

Dr. Gary Steinberg, Chairman of the Department of Neurosurgery at the Stanford University School of Medicine and Co-Director of the Stanford Stroke Center, served as Principal Investigator for the clinical trial.

Results for subjects who completed the single arm Phase 1/2a study demonstrated statistically significant improvement in motor function, evaluated using the European Stroke Scale, National Institutes of Health Stroke Scale, the Fugl-Meyer total score and the Fugl-Meyer motor function total score. The data also showed that the treatment was generally safe and well-tolerated by the trial participants.

As lead author of the scientific article, Dr. Steinberg accepted the award at the recent International Stroke Conference in Houston.

About SanBio, Inc. (SanBio)

SanBio is a regenerative medicine company headquartered in Tokyo and Mountain View, California, with cell-based products in various stages of research, development and clinical trials. Its proprietary cell-based product, SB623, is currently in a Phase 2b clinical trial for treatment of chronic motor impairments resulting from stroke, with its joint development partner, Sumitomo Dainippon Pharma Co., Ltd., in the United States and Canada. SanBio is also implementing a global Phase 2 clinical trial using SB623 in the United States and Japan for the treatment of motor impairment resulting from traumatic brain injury. More information about SanBio is available at http://www.sanbio.com.

View source version on businesswire.com: http://www.businesswire.com/news/home/20170226005250/en/

See the rest here:
Study of SanBio's Stem Cell Treatment for Stroke Receives Innovation Award from American Heart Association - Yahoo Finance

World-Renowned Stem Cell Transplantation Expert joins Cellect’s Advisory Board – P&T Community

World-Renowned Stem Cell Transplantation Expert joins Cellect's Advisory Board
P&T Community
"Dr. Cutler is world renowned for his contributions to innovations within the stem cell transplantation industry to drive potential treatments in cancer and many other medical conditions, said Dr. Shai Yarkoni, Cellect's CEO. We look forward to ...

and more »

Here is the original post:
World-Renowned Stem Cell Transplantation Expert joins Cellect's Advisory Board - P&T Community

Early-stage study validates Cellect Bio’s method of stem cell selection; shares ahead 19% – Seeking Alpha

Thinly traded nano cap Cellect Biotechnology Ltd. (APOP +19.4%) jumps on more than a 4x surge in volume in response to its announcement of positive results from a Phase 1 study aimed at validating its proprietary method of stem cell selection called ApoGraft. The process allows for the natural enrichment of stem cells that can be used in cell therapies or transplantation with significantly less risk of rejection.

The study was conducted on blood stem cells donated by 104 healthy subjects. Each sample represented a 5% graft. ApoGraft, used for only a few hours, produced a significant increase in the death of mature immune cells without compromising the quality and quantity of stem cells.

The Companys technology is expected to provide pharma companies, medical research centers and hospitals with the tools to rapidly isolate stem cells for in quantity and quality that will allow stems cell-related treatments and procedures. Cellects technology is applicable to a wide variety of stem cells related treatments in regenerative medicine and that current clinical trials are aimed at the cancer treatment of bone marrow transplantations.

Read the original post:
Early-stage study validates Cellect Bio's method of stem cell selection; shares ahead 19% - Seeking Alpha

Cancer stem cell – Wikipedia

Cancer stem cells (CSCs) are cancer cells (found within tumors or hematological cancers) that possess characteristics associated with normal stem cells, specifically the ability to give rise to all cell types found in a particular cancer sample. CSCs are therefore tumorigenic (tumor-forming), perhaps in contrast to other non-tumorigenic cancer cells. CSCs may generate tumors through the stem cell processes of self-renewal and differentiation into multiple cell types. Such cells are hypothesized to persist in tumors as a distinct population and cause relapse and metastasis by giving rise to new tumors. Therefore, development of specific therapies targeted at CSCs holds hope for improvement of survival and quality of life of cancer patients, especially for patients with metastatic disease.

Existing cancer treatments have mostly been developed based on animal models, where therapies able to promote tumor shrinkage were deemed effective. However, animals do not provide a complete model of human disease. In particular, in mice, whose life spans do not exceed two years, tumor relapse is difficult to study.

The efficacy of cancer treatments is, in the initial stages of testing, often measured by the ablation fraction of tumor mass (fractional kill). As CSCs form a small proportion of the tumor, this may not necessarily select for drugs that act specifically on the stem cells. The theory suggests that conventional chemotherapies kill differentiated or differentiating cells, which form the bulk of the tumor but do not generate new cells. A population of CSCs, which gave rise to it, could remain untouched and cause relapse.

Cancer stem cells were first identified by John Dick in acute myeloid leukemia in the late 1990s. Since the early 2000s they have been an intense cancer research focus.[1]

In different tumor subtypes, cells within the tumor population exhibit functional heterogeneity and tumors are formed from cells with various proliferative and differentiation capacities.[2] This functional heterogeneity among cancer cells has led to the creation of multiple propagation models to account for heterogeneity and differences in tumor-regenerative capacity: the cancer stem cell (CSC) and stochastic model.

The Cancer Stem Cell Model, also known as the Hierarchical Model proposes that tumors are hierarchically organized (CSCs lying at the apex[3] (Fig. 3).) Within the cancer population of the tumors there are cancer stem cells (CSC) that are tumorigenic cells and are biologically distinct from other subpopulations[4] They have two defining features: their long-term ability to self-renew and their capacity to differentiate into progeny that is non-tumorigenic but still contributes to the growth of the tumor. This model suggests that only certain subpopulations of cancer stem cells have the ability to drive the progression of cancer, meaning that there are specific (intrinsic) characteristics that can be identified and then targeted to destroy a tumor long-term without the need to battle the whole tumor [5]

In order for a cell to become cancerous it must undergo a significant number of alterations to its DNA sequence. This cell model suggests these mutations could occur to any cell in the body resulting in a cancer. Essentially this theory proposes that all cells have the ability to be tumorigenic making all tumor cells equipotent with the ability to self-renew or differentiate, leading to tumor heterogeneity while others can differentiate into non-CSCs [4][6] The cell's potential can be influenced by unpredicted genetic or epigenetic factors, resulting in phenotypically diverse cells in both the tumorigenic and non-tumorigenic cells that compose the tumor.[7]

These mutations could progressively accumulate and enhance the resistance and fitness of cells that allow them to outcompete other tumor cells, better known as the somatic evolution model.[4] The clonal evolution model, which occurs in both the CSC model and stochastic model, postulates that mutant tumor cells with a growth advantage outproliferate others. Cells in the dominant population have a similar potential for initiating tumor growth[8] (Fig. 4).

[9] These two models are not mutually exclusive, as CSCs themselves undergo clonal evolution. Thus, the secondary more dominant CSCs may emerge, if a mutation confers more aggressive properties[10] (Fig. 5).

A study in 2014 argues the gap between these two controversial models can be bridged by providing an alternative explanation of tumor heterogeneity. They demonstrate a model that includes aspects of both the Stochastic and CSC models.[6] They examined cancer stem cell plasticity in which cancer stem cells can transition between non-cancer stem cells (Non-CSC) and CSC via in situ supporting a more Stochastic model.[6][11] But the existence of both biologically distinct non-CSC and CSC populations supports a more CSC model, proposing that both models may play a vital role in tumor heterogeneity.[6]

The existence of CSCs is under debate, because many studies found no cells with their specific characteristics.[12] Cancer cells must be capable of continuous proliferation and self-renewal to retain the many mutations required for carcinogenesis and to sustain the growth of a tumor, since differentiated cells (constrained by the Hayflick Limit[13]) cannot divide indefinitely. If most tumor cells are endowed with stem cell properties, targeting tumor size directly is a valid strategy. If they are a small minority, targeting them may be more effective. Another debate is over the origin of CSCs - whether from disregulation of normal stem cells or from a more specialized population that acquired the ability to self-renew (which is related to the issue of stem cell plasticity).

The first conclusive evidence for CSCs came in 1997. Bonnet and Dick isolated a subpopulation of leukemia cells that expressed surface marker CD34, but not CD38.[14] The authors established that the CD34+/CD38 subpopulation is capable of initiating tumors in NOD/SCID mice that were histologically similar to the donor. The first evidence of a solid tumor cancer stem-like cell followed in 2002 with the discovery of a clonogenic, sphere-forming cell isolated and characterized from human brain gliomas. Human cortical glial tumors contain neural stem-like cells expressing astroglial and neuronal markers in vitro.[15]

In cancer research experiments, tumor cells are sometimes injected into an experimental animal to establish a tumor. Disease progression is then followed in time and novel drugs can be tested for their efficacy. Tumor formation requires thousands or tens of thousands of cells to be introduced. Classically, this was explained by poor methodology (i.e., the tumor cells lose their viability during transfer) or the critical importance of the microenvironment, the particular biochemical surroundings of the injected cells. Supporters of the CSC paradigm argue that only a small fraction of the injected cells, the CSCs, have the potential to generate a tumor. In human acute myeloid leukemia the frequency of these cells is less than 1 in 10,000.[14]

Further evidence comes from histology. Many tumors are heterogeneous and contain multiple cell types native to the host organ. Heterogeneity is commonly retained by tumor metastases. This suggests that the cell that produced them had the capacity to generate multiple cell types, a classical hallmark of stem cells.[14]

The existence of leukemia stem cells prompted research into other cancers. CSCs have recently been identified in several solid tumors, including:

Once the pathways to cancer are hypothesized, it is possible to develop predictive mathematical models,[33] e.g., based on the cell compartment method. For instance, the growths of abnormal cells can be denoted with specific mutation probabilities. Such a model predicted that repeated insult to mature cells increases the formation of abnormal progeny and the risk of cancer.[34] The clinical efficacy of such models[35] remains unestablished.

The origin of CSCs is an active research area. The answer may depend on the tumor type and phenotype. So far the hypothesis that tumors originate from a single "cell of origin" has not been demonstrated using the cancer stem cell model. This is because cancer stem cells are not present in end-stage tumors.

Origin hypotheses include mutants in developing stem or progenitor cells, mutants in adult stem cells or adult progenitor cells and mutant, differentiated cells that acquire stem-like attributes. These theories often focus on a tumor's "cell of origin".

The "mutation in stem cell niche populations during development" hypothesis claims that these developing stem populations are mutated and then reproduce so that the mutation is shared by many descendants. These daughter cells are much closer to becoming tumors and their numbers increase the chance of a cancerous mutation.[36]

Another theory associates adult stem cells (ASC) with tumor formation. This is most often associated with tissues with a high rate of cell turnover (such as the skin or gut). In these tissues, ASCs are candidates because of their frequent cell divisions (compared to most ASCs) in conjunction with the long lifespan of ASCs. This combination creates the ideal set of circumstances for mutations to accumulate: mutation accumulation is the primary factor that drives cancer initiation. Evidence shows that the association represents an actual phenomenon, although specific cancers have been linked to a specific cause.[37][38]

De-differentiation of mutated cells may create stem cell-like characteristics, suggesting that any cell might become a cancer stem cell. In other words, a fully differentiated cell undergoes several mutations that drive it back to a stem-like state.

The concept of tumor hierarchy claims that a tumor is a heterogeneous population of mutant cells, all of which share some mutations, but vary in specific phenotype. A tumor hosts several types of stem cells, one optimal to the specific environment and other less successful lines. These secondary lines may be more successful in other environments, allowing the tumor to adapt, including adaptation to therapeutic intervention. If correct, this concept impacts cancer stem cell-specific treatment regimes.[39] Such a hierarchy would complicate attempts to pinpoint the origin.

CSCs, now reported in most human tumors, are commonly identified and enriched using strategies for identifying normal stem cells that are similar across studies.[40] These procedures include fluorescence-activated cell sorting (FACS), with antibodies directed at cell-surface markers and functional approaches including side population assay or Aldefluor assay.[41] The CSC-enriched result is then implanted, at various doses, in immune-deficient mice to assess its tumor development capacity. This in vivo assay is called a limiting dilution assay. The tumor cell subsets that can initiate tumor development at low cell numbers are further tested for self-renewal capacity in serial tumor studies.[42]

CSC can also be identified by efflux of incorporated Hoechst dyes via multidrug resistance (MDR) and ATP-binding cassette (ABC) Transporters.[41]

Another approach is sphere-forming assays. Many normal stem cells such as hematopoietics or stem cells from tissues, under special culture conditions, form three-dimensional spheres that can differentiate. As with normal stem cells, the CSCs isolated from brain or prostate tumors also have the ability to form anchor-independent spheres.[43]

CSCs have been identified in various solid tumors. Markers specific for normal stem cells are commonly used for isolating CSCs from solid and hematological tumors. Cell surface markers have proved useful for isolation of CSC-enriched populations including CD133 (also known as PROM1), CD44, CD24, EpCAM (epithelial cell adhesion molecule, also known as epithelial specific antigen, ESA), THY1, ATP-binding cassette B5 (ABCB5),[44] and CD200.

CD133 (prominin 1) is a five-transmembrane domain glycoprotein expressed on CD34+ stem and progenitor cells, in endothelial precursors and fetal neural stem cells. It has been detected using its glycosylated epitope known as AC133.

EpCAM (epithelial cell adhesion molecule, ESA, TROP1) is hemophilic Ca2+-independent cell adhesion molecule expressed on the basolateral surface of most epithelial cells.

CD90 (THY1) is a glycosylphosphatidylinositol glycoprotein anchored in the plasma membrane and involved in signal transduction. It may also mediate adhesion between thymocytes and thymic stroma.

CD44 (PGP1) is an adhesion molecule that has pleiotropic roles in cell signaling, migration and homing. It has multiple isoforms, including CD44H, which exhibits high affinity for hyaluronate and CD44V which has metastatic properties.

CD24 (HSA) is a glycosylated glycosylphosphatidylinositol-anchored adhesion molecule, which has co-stimulatory role in B and T cells.

CD200 (OX-2) is a type 1 membrane glycoprotein, which delivers an inhibitory signal to immune cells including T cells, natural killer cells and macrophages.

ALDH is a ubiquitous aldehyde dehydrogenase family of enzymes, which catalyzes the oxidation of aromatic aldehydes to carboxyl acids. For instance, it has a role in conversion of retinol to retinoic acid, which is essential for survival.[45][46]

The first solid malignancy from which CSCs were isolated and identified was breast cancer and they are the most intensely studied. Breast CSCs have been enriched in CD44+CD24/low,[44] SP[47] and ALDH+ subpopulations.[48][49] Breast CSCs are apparently phenotypically diverse. CSC marker expression in breast cancer cells is apparently heterogeneous and breast CSC populations vary across tumors.[50] Both CD44+CD24 and CD44+CD24+ cell populations are tumor initiating cells; however, CSC are most highly enriched using the marker profile CD44+CD49fhiCD133/2hi.[51]

CSCs have been reported in many brain tumors. Stem-like tumor cells have been identified using cell surface markers including CD133,[52] SSEA-1 (stage-specific embryonic antigen-1),[53]EGFR[54] and CD44.[55] The use of CD133 for identification of brain tumor stem-like cells may be problematic because tumorigenic cells are found in both CD133+ and CD133 cells in some gliomas and some CD133+ brain tumor cells may not possess tumor-initiating capacity.[54]

CSCs were reported in human colon cancer.[56] For their identification, cell surface markers such as CD133,[56] CD44[57] and ABCB5,[58] functional analysis including clonal analysis [59] and Aldefluor assay were used.[60] Using CD133 as a positive marker for colon CSCs generated conflicting results. The AC133 epitope, but not the CD133 protein, is specifically expressed in colon CSCs and its expression is lost upon differentiation.[61] In addition, CD44+ colon cancer cells and additional sub-fractionation of CD44+EpCAM+ cell population with CD166 enhance the success of tumor engraftments.[57]

Multiple CSCs have been reported in prostate,[62]lung and many other organs, including liver, pancreas, kidney or ovary.[45][63] In prostate cancer, the tumor-initiating cells have been identified in CD44+[64] cell subset as CD44+21+,[65] TRA-1-60+CD151+CD166+[66] or ALDH+[67] cell populations. Putative markers for lung CSCs have been reported, including CD133+,[68] ALDH+,[69] CD44+[70] and oncofetal protein 5T4+.[71]

Metastasis is the major cause of tumor lethality. However, not every tumor cell can metastasize. This potential depends on factors that determine growth, angiogenesis, invasion and other basic processes.

In epithelial tumors, the epithelial-mesenchymal transition (EMT) is considered to be a crucial event.[72] EMT and the reverse transition from mesenchymal to an epithelial phenotype (MET) are involved in embryonic development, which involves disruption of epithelial cell homeostasis and the acquisition of a migratory mesenchymal phenotype.[73] EMT appears to be controlled by canonical pathways such as WNT and transforming growth factor .[74]

EMT's important feature is the loss of membrane E-cadherin in adherens junctions, where -catenin may play a significant role. Translocation of -catenin from adherens junctions to the nucleus may lead to a loss of E-cadherin and subsequently to EMT. Nuclear -catenin apparently can directly, transcriptionally activate EMT-associated target genes, such as the E-cadherin gene repressor SLUG (also known as SNAI2).[75] Mechanical properties of the tumor microenvironment, such as hypoxia, can contribute to CSC survival and metastatic potential through stabilization of hypoxia inducible factors through interactions with ROS (reactive oxygen species).[76][77]

Tumor cells undergoing an EMT may be precursors for metastatic cancer cells, or even metastatic CSCs.[78] In the invasive edge of pancreatic carcinoma, a subset of CD133+CXCR4+ (receptor for CXCL12 chemokine also known as a SDF1 ligand) cells was defined. These cells exhibited significantly stronger migratory activity than their counterpart CD133+CXCR4 cells, but both showed similar tumor development capacity.[79] Moreover, inhibition of the CXCR4 receptor reduced metastatic potential without altering tumorigenic capacity.[80]

In breast cancer CD44+CD24/low cells are detectable in metastatic pleural effusions.[44] By contrast, an increased number of CD24+ cells have been identified in distant metastases in breast cancer patients.[81] It is possible that CD44+CD24/low cells initially metastasize and in the new site change their phenotype and undergo limited differentiation.[82] The two-phase expression pattern hypothesis proposes two forms of cancer stem cells - stationary (SCS) and mobile (MCS). SCS are embedded in tissue and persist in differentiated areas throughout tumor progression. MCS are located at the tumor-host interface. These cells are apparently derived from SCS through the acquisition of transient EMT (Figure 7).[83]

CSCs have implications for cancer therapy, including for disease identification, selective drug targets, prevention of metastasis and intervention strategies.

Somatic stem cells are naturally resistant to chemotherapeutic agents. They produce various pumps (such as MDR[citation needed]) that pump out drugs and DNA repair proteins. They have a slow rate of cell turnover (chemotherapeutic agents naturally target rapidly replicating cells).[citation needed] CSCs that develop from normal stem cells may also produce these proteins, which could increase their resistance towards chemotherapy. The surviving CSCs then repopulate the tumor, causing a relapse.[84]

Selectively targeting CSCs may allow treatment of aggressive, non-resectable tumors, as well as prevent metastasis and relapse.[84] The hypothesis suggests that upon CSC elimination, cancer could regress due to differentiation and/or cell death.[citation needed] The fraction of tumor cells that are CSCs and therefore need to be eliminated is unclear.[85]

Studies looked for specific markers[17] and for proteomic and genomic tumor signatures that distinguish CSCs from others.[86] In 2009, scientists identified the compound salinomycin, which selectively reduces the proportion of breast CSCs in mice by more than 100-fold relative to Paclitaxel, a commonly used chemotherapeutic agent.[87] Some types of cancer cells can survive treatment with salinomycin through autophagy,[88] whereby cells use acidic organelles such as lysosomes to degrade and recycle certain types of proteins. The use of autophagy inhibitors can kill cancer stem cells that survive by autophagy.[89]

The cell surface receptor interleukin-3 receptor-alpha (CD123) is overexpressed on CD34+CD38- leukemic stem cells (LSCs) in acute myelogenous leukemia (AML) but not on normal CD34+CD38- bone marrow cells.[90] Treating AML-engrafted NOD/SCID mice with a CD123-specific monoclonal antibody impaired LSCs homing to the bone marrow and reduced overall AML cell repopulation including the proportion of LSCs in secondary mouse recipients.[91]

A 2015 study packaged nanoparticles with miR-34a and ammonium bicarbonate and delivered them to prostate CSCs in a mouse model. Then they irradiated the area with near-infrared laser light. This caused the nanoparticles to swell three times or more in size bursting the endosomes and dispersing the RNA in the cell. miR-34a can lower the levels of CD44.[92][93]

The design of new drugs for targeting CSCs requires understanding the cellular mechanisms that regulate cell proliferation. The first advances in this area were made with hematopoietic stem cells (HSCs) and their transformed counterparts in leukemia, the disease for which the origin of CSCs is best understood. Stem cells of many organs share the same cellular pathways as leukemia-derived HSCs.

A normal stem cell may be transformed into a CSC through disregulation of the proliferation and differentiation pathways controlling it or by inducing oncoprotein activity.

The Polycomb group transcriptional repressor Bmi-1 was discovered as a common oncogene activated in lymphoma[94] and later shown to regulate HSCs.[95] The role of Bmi-1 has been illustrated in neural stem cells.[96] The pathway appears to be active in CSCs of pediatric brain tumors.[97]

The Notch pathway plays a role in controlling stem cell proliferation for several cell types including hematopoietic, neural and mammary[98] SCs. Components of this pathway have been proposed to act as oncogenes in mammary[99] and other tumors.

A branch of the Notch signaling pathway that involves the transcription factor Hes3 regulates a number of cultured cells with CSC characteristics obtained from glioblastoma patients.[100]

These developmental pathways are SC regulators.[101] Both Sonic hedgehog (SHH) and Wnt pathways are commonly hyperactivated in tumors and are necessary to sustain tumor growth. However, the Gli transcription factors that are regulated by SHH take their name from gliomas, where they are highly expressed. A degree of crosstalk exists between the two pathways and they are commonly activated together.[102] By contrast, in colon cancer hedgehog signalling appears to antagonise Wnt.[103]

Sonic hedgehog blockers are available, such as cyclopamine. A water-soluble cyclopamine may be more effective in cancer treatment. DMAPT, a water-soluble derivative of parthenolide, induces oxidative stress and inhibits NF-B signaling[104] for AML (leukemia) and possibly myeloma and prostate cancer. Telomerase is a study subject in CSC physiology.[105] GRN163L (Imetelstat) was recently started in trials to target myeloma stem cells.

Wnt signaling can become independent of regular stimuli, through mutations in downstream oncogenes and tumor suppressor genes that become permanently activated even though the normal receptor has not received a signal. -catenin binds to transcription factors such as the protein TCF4 and in combination the molecules activate the necessary genes. LF3 strongly inhibits this binding in vitro, in cell lines and reduced tumor growth in mouse models. It prevented replication and reduced their ability to migrate, all without affecting healthy cells. No cancer stem cells remained after treatment. The discovery was the product of "rational drug design", involving AlphaScreens and ELISA technologies.[106]

Here is the original post:
Cancer stem cell - Wikipedia

Adult Stem Cell Medicine Technology | Asymmetrex

Asymmetrex is a life sciences biotechnology company with a focus on innovating adult stem cell medicine technology that will advance the potential of adult tissue stem cells into routine medical practice. Adult tissue stem cells are found in the bodies of children and adults. They are a small fraction of the cells (less than 1 per 1000) that make up organs and tissues like the liver, cornea, skin, muscles, hair, brain, and bone marrow. Despite their small fraction, they are responsible for continuously renewing and repairing the body.

Because of their normal role in maintaining and restoring organs and tissues, tissue stem cells obtained from a donating person have an inherent ability to reconstitute severely damaged tissues due to injury or disease in another recipient person. Currently, tissue stem cell transplantation treatments of this type are only available for a few tissues, e.g., bone marrow and the cornea of the eye. There are many, many more tissues in the body for which stem cell transplantation therapies are needed, but not possible. The major cause of this shortcoming insufficient quantities of donor stem cells often also undermines the effectiveness of the tissue stem cell treatments that are available.

Asymmetrex holds adult stem cell patents for technologies that promotethe multiplication of adult tissue stem cells. Tested so far for tissue stem cells found in the liver, lung, pancreas, muscle, skin and hair follicle, the technologies have the potential to produce therapeutic human tissue stem cells by the pound, trillions of cells at a time. Unlike other presently popularized strategies based on pluripotent stem cells, Asymmetrexsadult stem cell medicine technologyproducesnormal cells without high rates of mutation or tumor-forming properties. A major pursuit of Asymmetrex is collaboration with strategic partners to develop robust manufacturing processes for producing medically important tissue stem cells and their differentiated derivative cells for use in transplantation therapies and drug development.

Another long-standing challenge in stem cell biomedicine is lack of means to identify and count tissue stem cells. Because of this need, even the available tissue stem cell therapies like bone marrow transplantation cannot be reliably optimized to achieve better treatment outcomes. This problem has existed for half a century because of the failure to discover biological markers found exclusively in or on adult tissue stem cells.

Employing its internationally recognized, special research expertise in unique adult tissue stem cell properties, Asymmetrex has developed several technologies that make it now possible to either count tissue stem cells directly or estimate their number precisely. This adult stem cell medicine technologyand innovation provide, for the first time, the means to monitor tissue stem cell number and quality for applications in regenerative medicine and drug development.

By continuing to discover and develop adult stem cell medicine technology for the production, identification, and quantification of restorative adult tissue stem cells, Asymmetrex will set the direction and pace of modern stem cell biomedicine. In addition to our current focus in developing stem cell toxicology assays for the pharmaceutical industry, we also license technologies for stem cell detection (including cancer stem cells) and stem cell expansion for user-exclusive applications.

View original post here:
Adult Stem Cell Medicine Technology | Asymmetrex

Duke Stem Cell and Regenerative Medicine Program

Overview Our program brings together basic scientists and clinicians studying stem cells in a variety of adult and developing organ systems. The goal is to understand and exploit their remarkable capacity to maintain healthy tissues and to replace cells lost by disease or injury. Program highlights include:

Faculty Search Cell Biology is hiring a tenure-track Assistant or Associate Professor with a strong record of creativity and productivity in developmental and/or regenerative biology. Applicants should submit a curriculum vitae, a 3-page summary of accomplishments and research plans, a teaching statement, and at least 3 letters of recommendation by November 15, 2015. Applications should be submitted via Academic Jobs Online. Questions may be directed to Ken Poss or Brigid Hogan.

Executive Director Search The new tissueregenerationinitiative at Duke is hiring an Executive Director, an Associate in Research position at Duke University, to work closely with the Director, Co-Directors, and faculty members to promote and integrate discovery research, training, and applications in the broad field of tissue regeneration.We invite applications from candidates who have a Ph.D. and postdoctoral research experience in the relevant areas of developmental biology, stem cell biology, or tissue regeneration tosubmit a cover letter, curriculum vitae, summary of research accomplishments and any administrative leadership experience, and a list of at least three references to Academic Jobs Online. Questions may be directed toKen Poss.

Niche regulation of new neurons production in the adult brain Robust production of new neurons continues in the adult rodent brain, but how this is sustained remains unknown. Researchers in Dr. Chay T. Kuos laboratory found that self-assembly of radial glia into support structures for adult stem cells is critical for continued neurogenesis. More...

Zebrafish heart regeneration During heart regeneration in zebrafish, retinoic production in endocardial and epicardial cells localizes to areas of tissue damage, where it promotes cardiomyocyte proliferation. More...

Intestinal Crypt Proliferation Stem cell/transit amplifying compartments (green) reside in the base of each mouse intestinal crypt. These cells give rise to the multiple lineages of the intestinal epithelium (Lechler lab). More...

Lung epithelial stem cell regulationThe airways of the lung are lined by an epithelium that contains large numbers of cells specialized for making and secreting glycoproteins and mucus, as well as multiciliated cells that remove the mucus and the particles trapped in it. More...

Role of immune cells in the spermatogonial stem cell niche In addition to their roles in immune and inflammatory responses, macrophages have diverse functions in development. In reproductive biology, macrophages have been implicated in ovarian follicular growth and in Leydig cell function, but their role in spermatogonial differentiation has not been examined. More...

Drosophila hindgut repairThe fruit fly Drosophila has long been a leading genetic model for stem cell research. However, until recently no Drosophila models existed for study of mechanisms by which adult organs lacking active stem cells repair damaged tissue. More...

Indispensible pre-mitotic endocycles promote aneuploidy in the Drosophila rectum

Time lapse imaging of a tripolar division during developmental organ regeneration in the Drosophila hindgut. These divisions occur in cells with extra copies of the genome (polyploid cells) and produce an adult organ in which many of the cells have variable, imbalanced chromosome numbers (aneuploid cells). DNA is in purple, and centrosomes and cell membranes are in green.

Fox Lab. Schoenfelder et al. (2014) Development 141:3551-3560

Read the original:
Duke Stem Cell and Regenerative Medicine Program

Eli and Edythe Broad Center for Regenerative Medicine and …

Welcome

Welcome to the Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, located on the University of Southern Californias Health Sciences Campus.

Our investigators are exploring the normal mechanisms that build, maintain and repair our body systems, to develop knowledge-based approaches for regenerative medicine. Scientists are researching kidney, liver, neural, blood, cardiovascular, skeletal and skin disease models.

The center serves as a hub for USC Stem Cell, which connects researchers in stem cell biology and regenerative medicine across USC.

Oct 9, 2015

As a winner of the NIH Directors New Innovator Award, USC Stem Cell principal investigator Min Yu will strive to develop individualized medicine targeting rare and deadly breast cancer stem cells. The five-year, $2.475 million award is part of the High-Risk, High-Reward Research program supported by the NIH Common Fund.

Sep 22, 2015

How do you turn stem cells into nephrons, the functional unit of the kidney? Albert D. Kim, PhD, a postdoctoral fellow in the laboratory of Andy McMahon, PhD, is exploring this question with support from a Hearst Fellowship, an award recognizing an exceptional junior postdoctoral fellow pursuing stem cell research at USC.

Sep 21, 2015

Once the stuff of science fiction, genetic engineering is now offered on a fee-for-service basis at USC. On September 19, USC Stem Cell faculty and staff welcomed their supporters, the Chang and Choi families, and nearly 100 of their friends to celebrate the grand opening of the Chang Stem Cell Engineering Facility, located on the second floor of the Eli and Edythe Broad Center (BCC) for Regenerative Medicine and Stem Cell Research at USC on the Health Sciences Campus. Established with a generous gift from the Chang family, the stem cell engineering facility will serve researchers at USC as well as at other institutions.

See the rest here:
Eli and Edythe Broad Center for Regenerative Medicine and ...

California Stem Cell Report

Highlights Outrage about prices Industry euphemisms Handy demons California's stem cell direction

Halloween is just around the corner and some stem cell folks here in California are doing their best to wish away a particularly frightening specter.

Some of the rhetoric amounted to no more than whistling in the dark. Investors, researchers and business executives danced around what almost certainly appear to be extremely high treatment costs for stem cell treatments.

Those costs are the type that have stirred recent outrage among consumers and among some physicians. The controversy has emerged anew in the presidential race and last week knocked the stock market around a bit.

yesterday featured a woman with cystic fibrosis who said about a drug maker,

Inevitably meetings like the Mesa conference rarely deal directly with the tough and emotional issues that are typified by the ire expressed by that woman,Klyn Elsbury, who lives a few miles north of the Mesa meeting.Instead biotech executives retreat behind such euphemisms as reimbursement, which is a catch-all term for how do we make a profit.

Yesterday the matter of pricing did come before one panel. While this writer came in late and did not hear all the names, the general response could be called if we build it, they will come.

Many of the potential products being tested now involve unmet medical needs, and thus the demand could be extraordinarily high. In other words, if you want to live, you will have to pay our price.

It would be super transformative in the market place, one speaker said, if a company has produced the only drug that will save a persons life. Another said the system will eventually find a (pricing) model. Which is where whistling in the dark comes in. But if the industry doesnt directly face the emotional and medical concerns about predatory business actions, the industry, in all likelihood, will be hoist on its own pricing petard.

Lawmakers and regulators fueled by public outrage may well react to overly aggressive prices and begin to impose what could amount to some sort of profit rationing. After all good public health is a virtuous thing. And if prices stand in the way, something needs to be done about it. Or so the reasoning will go. Every politician needs a demon to rail against. Big Pharma and related stem cell firms could be that handy demon.

The argument in some circles maintains that prices will start out sky high and then decrease over time. But that does not mean the public and other payers will wait for decades and patiently pay $1 million per treatment.

That figure popped up this week in an item by UC Davis stem cell scientist Paul Knoepfler. He wrote on his blog, ipscell.com, about a pricing model that did, in fact, run as high as $1 million.

Knoepfler said the stem cell community needs to answer following question and soon.

Californias $3 billion stem cell agency, in particular, has an economic dog in the pricing hooha. The agency is in the midst of determining how to spend its last $800 million or so. It can decide to put that money into research that offers the likelihood of relatively affordable treatments or instead into $1 million cash cow therapies for Big Pharma.

What the agency does now will affect whether it vanishes in a few years for lack of funding or can find additional support from the state and/or private sources. If its only product after running through $6 billion (including interest) is a $1 million therapy, some might look askance at providing additional cash.

Read this article:
California Stem Cell Report