header logo image


Page 19«..10..18192021..»

Archive for the ‘Stem Cell Negative’ Category

Stem cell factor – Wikipedia

Sunday, November 27th, 2016

KITLG Identifiers Aliases KITLG, FPH2, FPHH, KL-1, Kitl, MGF, SCF, SF, SHEP7, DCUA, KIT ligand, DFNA69 External IDs OMIM: 184745 MGI: 96974 HomoloGene: 692 GeneCards: KITLG Genetically Related Diseases testicular germ cell cancer, Testicular cancer[1] RNA expression pattern

Stem cell factor (also known as SCF, KIT-ligand, KL, or steel factor) is a cytokine that binds to the c-KIT receptor (CD117). SCF can exist both as a transmembrane protein and a soluble protein. This cytokine plays an important role in hematopoiesis (formation of blood cells), spermatogenesis, and melanogenesis.

The gene encoding stem cell factor (SCF) is found on the Sl locus in mice and on chromosome 12q22-12q24 in humans.[4] The soluble and transmembrane forms of the protein are formed by alternative splicing of the same RNA transcript,[5][6]

The soluble form of SCF contains a proteolytic cleavage site in exon 6. Cleavage at this site allows the extracellular portion of the protein to be released. The transmembrane form of SCF is formed by alternative splicing that excludes exon 6 (Figure 1). Both forms of SCF bind to c-KIT and are biologically active.

Soluble and transmembrane SCF is produced by fibroblasts and endothelial cells. Soluble SCF has a molecular weight of 18,5 KDa and forms a dimer. It is detected in normal human blood serum at 3.3ng/mL.[7]

SCF plays an important role in the hematopoiesis during embryonic development. Sites where hematopoiesis takes place, such as the fetal liver and bone marrow, all express SCF. Mice that do not express SCF die in utero from severe anemia. Mice that do not express the receptor for SCF (c-KIT) also die from anemia.[8] SCF may serve as guidance cues that direct hematopoietic stem cells (HSCs) to their stem cell niche (the microenvironment in which a stem cell resides), and it plays an important role in HSC maintenance. Non-lethal point mutants on the c-KIT receptor can cause anemia, decreased fertility, and decreased pigmentation.[9]

During development, the presence of the SCF also plays an important role in the localization of melanocytes, cells that produce melanin and control pigmentation. In melanogenesis, melanoblasts migrate from the neural crest to their appropriate locations in the epidermis. Melanoblasts express the KIT receptor, and it is believed that SCF guides these cells to their terminal locations. SCF also regulates survival and proliferation of fully differentiated melanocytes in adults.[10]

In spermatogenesis, c-KIT is expressed in primordial germ cells, spermatogonia, and in primordial oocytes.[11] It is also expressed in the primordial germ cells of females. SCF is expressed along the pathways that the germ cells use to reach their terminal destination in the body. It is also expressed in the final destinations for these cells. Like for melanoblasts, this helps guide the cells to their appropriate locations in the body.[8]

SCF plays a role in the regulation of HSCs in the stem cell niche in the bone marrow. SCF has been shown to increase the survival of HSCs in vitro and contributes to the self-renewal and maintenance of HSCs in-vivo. HSCs at all stages of development express the same levels of the receptor for SCF (c-KIT).[12] The stromal cells that surround HSCs are a component of the stem cell niche, and they release a number of ligands, including SCF.

In the bone marrow, HSCs and hematopoietic progenitor cells are adjacent to stromal cells, such as fibroblasts and osteoblasts (Figure 2). These HSCs remain in the niche by adhering to ECM proteins and to the stromal cells themselves. SCF has been shown to increase adhesion and thus may play a large role in ensuring that HSCs remain in the niche.[8]

A small percentage of HSCs regularly leave the bone marrow to enter circulation and then return to their niche in the bone marrow.[13] It is believed that concentration gradients of SCF, along with the chemokine SDF-1, allow HSCs to find their way back to the niche.[14]

In adult mice, the injection of the ACK2 anti-KIT antibody, which binds to the c-Kit receptor and inactivates it, leads to severe problems in hematopoiesis. It causes a significant decrease in the number HSC and other hematopoietic progenitor cells in the bone marrow.[15] This suggests that SCF and c-Kit plays an important role in hematopoietic function in adulthood. SCF also increases the survival of various hematopoietic progenitor cells, such as megakaryocyte progenitors, in vitro.[16] In addition, it works with other cytokines to support the colony growth of BFU-E, CFU-GM, and CFU-GEMM4. Hematopoietic progenitor cells have also been shown to migrate towards a higher concentration gradient of SCF in vitro, which suggests that SCF is involved in chemotaxis for these cells.

Fetal HSCs are more sensitive to SCF than HSCs from adults. In fact, fetal HSCs in cell culture are 6 times more sensitive to SCF than adult HSCs based on the concentration that allows maximum survival.[17]

Mast cells are the only terminally differentiated hematopoietic cells that express the c-Kit receptor. Mice with SCF or c-Kit mutations have severe defects in the production of mast cells, having less than 1% of the normal levels of mast cells. Conversely, the injection of SCF increases mast cell numbers near the site of injection by over 100 times. In addition, SCF promotes mast cell adhesion, migration, proliferation, and survival.[18] It also promotes the release of histamine and tryptase, which are involved in the allergic response.

The presence of both soluble and transmembrane SCF is required for normal hematopoietic function.[5][19] Mice that produce the soluble SCF but not transmembrane SCF suffer from anemia, are sterile, and lack pigmentation. This suggests that transmembrane SCF plays a special role in vivo that is separate from that of soluble SCF.

SCF binds to the c-KIT receptor (CD 117), a receptor tyrosine kinase.[20] c-Kit is expressed in HSCs, mast cells, melanocytes, and germ cells. It is also expressed in hematopoietic progenitor cells including erythroblasts, myeloblasts, and megakaryocytes. However, with the exception of mast cells, expression decreases as these hematopoietic cells mature and c-KIT is not present when these cells are fully differentiated (Figure 3). SCF binding to c-KIT causes the receptor to homodimerize and auto-phosphorylate at tyrosine residues. The activation of c-Kit leads to the activation of multiple signaling cascades, including the RAS/ERK, PI3-Kinase, Src kinase, and JAK/STAT pathways.[20]

SCF may be used along with other cytokines to culture HSCs and hematopoietic progenitors. The expansion of these cells ex-vivo (outside the body) would allow advances in bone marrow transplantation, in which HSCs are transferred to a patient to re-establish blood formation.[12] One of the problems of injecting SCF for therapeutic purposes is that SCF activates mast cells. The injection of SCF has been shown to cause allergic-like symptoms and the proliferation of mast cells and melanocytes.[8]

Cardiomyocyte-specific overexpression of transmembrane SCF promotes stem cell migration and improves cardiac function and animal survival after myocardial infarction.[21]

Stem cell factor has been shown to interact with CD117.[22][23]

PDB gallery

1exz: STRUCTURE OF STEM CELL FACTOR

1scf: HUMAN RECOMBINANT STEM CELL FACTOR

Unsorted

Go here to see the original:
Stem cell factor - Wikipedia

Read More...

Mesenchymal stem cell – Wikipedia

Wednesday, November 23rd, 2016

Mesenchymal stem cells, or MSCs, are multipotent stromal cells that can differentiate into a variety of cell types,[1] including: osteoblasts (bone cells),[2]chondrocytes (cartilage cells),[3]myocytes (muscle cells)[4] and adipocytes (fat cells). This phenomenon has been documented in specific cells and tissues in living animals and their counterparts growing in tissue culture.

While the terms mesenchymal stem cell and marrow stromal cell have been used interchangeably, neither term is sufficiently descriptive:

In 1924, Russian-born morphologist Alexander A. Maximow used extensive histological findings to identify a singular type of precursor cell within mesenchyme that develops into different types of blood cells.[9]

Scientists Ernest A. McCulloch and James E. Till first revealed the clonal nature of marrow cells in the 1960s.[10][11] An ex vivo assay for examining the clonogenic potential of multipotent marrow cells was later reported in the 1970s by Friedenstein and colleagues.[12][13] In this assay system, stromal cells were referred to as colony-forming unit-fibroblasts (CFU-f).

The first clinical trials of MSCs were completed in 1995 when a group of 15 patients were injected with cultured MSCs to test the safety of the treatment. Since then, over 200 clinical trials have been started. However, most are still in the safety stage of testing.[7]

Subsequent experimentation revealed the plasticity of marrow cells and how their fate could be determined by environmental cues. Culturing marrow stromal cells in the presence of osteogenic stimuli such as ascorbic acid, inorganic phosphate and dexamethasone could promote their differentiation into osteoblasts. In contrast, the addition of transforming growth factor-beta (TGF-b) could induce chondrogenic markers.[citation needed]

The youngest, most primitive MSCs can be obtained from umbilical cord tissue, namely Wharton's jelly and the umbilical cord blood. However MSCs are found in much higher concentration in the Whartons jelly compared to cord blood, which is a rich source of hematopoietic stem cells. The umbilical cord is easily obtained after a birth. It is normally thrown away and poses no risk for collection. The cord MSCs have more primitive properties than other adult MSCs obtained later in life, which might make them a useful source of MSCs for clinical applications.

A rich source for mesenchymal stem cells is the developing tooth bud of the mandibular third molar. While considered multipotent, they may prove to be pluripotent. They eventually form enamel, dentin, blood vessels, dental pulp and nervous tissues, a minimum of 24 other different unique end organs. Because of ease in collection at 810 years of age before calcification and minimal-to-no-morbidity, they probably constitute a major source for research and multiple therapies. These stem cells have been shown capable of producing hepatocytes.

Additionally, amniotic fluid has been shown to be a rich source of stem cells. As many as 1 in 100 cells collected during amniocentesis has been shown to be a pluripotent mesenchymal stem cell.[14]

Adipose tissue is one of the richest sources of MSCs. There are more than 500 times more stem cells in 1 gram of fat than in 1 gram of aspirated bone marrow.[citation needed] Adipose stem cells are actively being researched in clinical trials for treatment of a variety of diseases.

The presence of MSCs in peripheral blood has been controversial. A few groups have successfully isolated MSCs from human peripheral blood and been able to expand them in culture.[15] Australian company Cynata claims the ability to mass-produce MSCs from induced pluripotent stem cells obtained from blood cells.[16][17]

Mesenchymal stem cells are characterized morphologically by a small cell body with a few cell processes that are long and thin. The cell body contains a large, round nucleus with a prominent nucleolus, which is surrounded by finely dispersed chromatin particles, giving the nucleus a clear appearance. The remainder of the cell body contains a small amount of Golgi apparatus, rough endoplasmic reticulum, mitochondria and polyribosomes. The cells, which are long and thin, are widely dispersed and the adjacent extracellular matrix is populated by a few reticular fibrils but is devoid of the other types of collagen fibrils.[18][19]

The International Society for Cellular Therapy (ISCT) has proposed a set of standards to define MSCs. A cell can be classified as an MSC if it shows plastic adherent properties under normal culture conditions and has a fibroblast-like morphology. In fact, some argue that MSCs and fibroblasts are functionally identical.[20] Furthermore, MSCs can undergo osteogenic, adipogenic and chondrogenic differentiation ex-vivo. The cultured MSCs also express on their surface CD73, CD90 and CD105, while lacking the expression of CD11b, CD14, CD19, CD34, CD45, CD79a and HLA-DR surface markers.[21]

MSCs have a great capacity for self-renewal while maintaining their multipotency. Beyond that, there is little that can be definitively said. The standard test to confirm multipotency is differentiation of the cells into osteoblasts, adipocytes and chondrocytes as well as myocytes and neurons. MSCs have been seen to even differentiate into neuron-like cells,[22] but there is lingering doubt whether the MSC-derived neurons are functional.[23] The degree to which the culture will differentiate varies among individuals and how differentiation is induced, e.g., chemical vs. mechanical;[24] and it is not clear whether this variation is due to a different amount of "true" progenitor cells in the culture or variable differentiation capacities of individuals' progenitors. The capacity of cells to proliferate and differentiate is known to decrease with the age of the donor, as well as the time in culture. Likewise, whether this is due to a decrease in the number of MSCs or a change to the existing MSCs is not known.[citation needed]

Numerous studies have demonstrated that human MSCs avoid allorecognition, interfere with dendritic cell and T-cell function and generate a local immunosuppressive microenvironment by secreting cytokines.[25] It has also been shown that the immunomodulatory function of human MSC is enhanced when the cells are exposed to an inflammatory environment characterised by the presence of elevated local interferon-gamma levels.[26] Other studies contradict some of these findings, reflecting both the highly heterogeneous nature of MSC isolates and the considerable differences between isolates generated by the many different methods under development.[27]

The majority of modern culture techniques still take a colony-forming unit-fibroblasts (CFU-F) approach, where raw unpurified bone marrow or ficoll-purified bone marrow Mononuclear cell are plated directly into cell culture plates or flasks. Mesenchymal stem cells, but not red blood cells or haematopoetic progenitors, are adherent to tissue culture plastic within 24 to 48 hours. However, at least one publication has identified a population of non-adherent MSCs that are not obtained by the direct-plating technique.[28]

Other flow cytometry-based methods allow the sorting of bone marrow cells for specific surface markers, such as STRO-1.[29] STRO-1+ cells are generally more homogenous and have higher rates of adherence and higher rates of proliferation, but the exact differences between STRO-1+ cells and MSCs are not clear.[30]

Methods of immunodepletion using such techniques as MACS have also been used in the negative selection of MSCs.[31]

The supplementation of basal media with fetal bovine serum or human platelet lysate is common in MSC culture. Prior to the use of platelet lysates for MSC culture, the pathogen inactivation process is recommended to prevent pathogen transmission.[32]

Mesenchymal stem cells in the body can be activated and mobilized if needed. However, the efficiency is low. For instance, damage to muscles heals very slowly but further study into mechanisms of MSC action may provide avenues for increasing their capacity for tissue repair.[33][34]

A statistical-based analysis of MSC therapy for osteo-diseases (e.g., osteoarthritis) noted that most studies are still underway.[35] Wakitani published a small case series of nine defects in five knees involving surgical transplantation of MSCs with coverage of the treated chondral defects.[36]

At least 218 clinical trials investigating the efficacy of mesenchymal stem cells in treating diseases have been initiated - many of which study autoimmune diseases.[37] Promising results have been shown in conditions such as graft versus host disease, Crohn's disease, multiple sclerosis, systemic lupus erythematosus and systemic sclerosis.[38] While their anti-inflammatory/immunomodulatory effects appear to greatly ameliorate autoimmune disease severity, the durability of these effects are unclear.

However, it is becoming more accepted that diseases involving peripheral tissues, such as inflammatory bowel disease, may be better treated with methods that increase the local concentration of cells.[39]

Many of the early clinical successes using intravenous transplantation came in systemic diseases such as graft versus host disease and sepsis. Direct injection or placement of cells into a site in need of repair may be the preferred method of treatment, as vascular delivery suffers from a "pulmonary first pass effect" where intravenous injected cells are sequestered in the lungs.[40] Clinical case reports in orthopedic applications have been published, though the number of patients treated is small and these methods still lack demonstrated effectiveness.

Scientists have reported that MSCs when transfused immediately a few hours post thawing may show reduced function or show decreased efficacy in treating diseases as compared to those MSCs which are in log phase of cell growth, so cryopreserved MSCs should be brought back into log phase of cell growth in in vitro culture before these are administered for clinical trials or experimental therapies, re-culturing of MSCs will help in recovering from the shock the cells get during freezing and thawing. Various clinical trials on MSCs have failed which used cryopreserved product immediately post thaw as compared to those clinical trials which used fresh MSCs.[41]

Mesenchymal stem cells have been shown to contribute to cancer progression in a number of different cancers, particularly the hematological malignancies because they contact the transformed blood cells in the bone marrow.[42]

See the rest here:
Mesenchymal stem cell - Wikipedia

Read More...

Cell Stem Cell – ScienceDirect.com

Wednesday, November 23rd, 2016

Volume 19, Issue 5 - selected pp. 559-672 (3 November 2016)

Not entitled to full text

Issue contains Open Access articles

Not entitled to full text

Issue contains Open Access articles

Issue contains Open Access articles

Issue contains Open Access articles

Issue contains Open Access articles

Issue contains Open Access articles

Not entitled to full text

Not entitled to full text

Issue contains Open Access articles

Issue contains Open Access articles

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Open Archive Issue

Read more here:
Cell Stem Cell - ScienceDirect.com

Read More...

Home | EMBO Reports

Saturday, October 8th, 2016

You have accessRestricted access

Article

These two authors contributed equally to this work

Using a histone FRAP method, this study identifies Gadd45a as a chromatin relaxer and somatic cell reprogramming enhancer. Gadd45a destabilizes histoneDNA interactions and facilitates the binding of Yamanaka factors to their targets.

Using a histone FRAP method, this study identifies Gadd45a as a chromatin relaxer and somatic cell reprogramming enhancer. Gadd45a destabilizes histoneDNA interactions and facilitates the binding of Yamanaka factors to their targets.

FRAP is used to assess heterochromatin/euchromatin dynamics in somatic cell reprogramming.

Gadd45a is a chromatin relaxer and improves somatic cell reprogramming.

Gadd45a destabilizes histoneDNA interactions and facilitates the binding of Yamanaka factors to their targets.

Keshi Chen, Qi Long, Tao Wang, Danyun Zhao, Yanshuang Zhou, Juntao Qi, Yi Wu, Shengbiao Li, Chunlan Chen, Xiaoming Zeng, Jianguo Yang, Zisong Zhou, Weiwen Qin, Xiyin Liu, Yuxing Li, Yingying Li, Xiaofen Huang, Dajiang Qin, Jiekai Chen, Guangjin Pan, Hans R Schler, Guoliang Xu, Xingguo Liu, Duanqing Pei

The rest is here:
Home | EMBO Reports

Read More...

stem cell negative outcomes Archives – The Niche

Thursday, September 15th, 2016

What can go wrong with unapproved stem cell clinics? The answer including from presentations at the FDA today turns out to be very serious, negative results right here in the U.S.

Thomas Albini, MD gave a talk entitled, Severe Visual Loss After Intravitreal Injection of Autologous Adipose Tissue-derived Stem Cells for Age-related Macular Degeneration.

Stem cell clinic transplants of fat stem cells led to blindness in three women, reported Dr. Albini.

Weve heard encouraging news about how stem cells might help patients regain lost vision or preserve existing vision in the face of a disease like macular degeneration in the future. Theres real potential there with rigorous clinical trials that are ongoing.

Here in this very different case we heard from Dr. Albini about how stem cells inappropriately used by a stem cell clinic in South Florida reportedly caused 3 women to go blind. All had retinal detachment potentially, Dr. Albini said, due to the fat stem cells taking up residence and resulting in pulling of the eye tissue internally. A nurse practitioner reportedly did the transplants rather than a physician. The patients assumed, we were told in the talk, that the listing in clinicaltrials.gov of the trial meant the interventions were legit.

This is such a deeply tragic case we can only hope that more people arent blinded from this kind of stem cell clinic offering.More on this situation here at Nature by Heidi Ledford.

Michael Miller, MD, PhD, spoke next with his talk entitled, Glioproliferative Lesion of the Spinal cord Arising from Exogenous Stem Cells. This case already has had quite a lot ofmedia attention and involves stroke patient Jim Gass, who ended up with a large spinal tumor that dramatically negatively affected his health. We have to give Mr. Gass huge credit for having the courage to go public with this case. He got ES cells and allo MSCs both in China. Then he traveled to Argentina for autologous MSCs and then to Mexico where he got MSCs and neural stem cells. See image above from the talk. The spinal tumor had many weird features of various primitive tumors. It was clearly a malignancy. There were no major cancer-related mutations detected in the OncoPanel assay.

The bottom line. So when those promoting stem cell clinics or wanting much less oversight ask what can go wrong? and they dont really believe much can go wrong, we now know for sure that that view is just not accurate. Intensely bad stem cell clinic outcomes are occurring right here in the U.S.

Follow this link:
stem cell negative outcomes Archives - The Niche

Read More...

Stem Cell Research – Pros and Cons – Explorable.com

Saturday, August 27th, 2016

Pros And Cons in Research

The debate of the pros and cons of stem cell research clearly illustrate the difficult ethics evaluations researchers sometimes must do.

Don't miss these related articles:

All scientists must consider whether the positive effects from their research are likely to be significantly higher than the negative effects.

Stem Cells are crucial to develop organisms. They are nonspecialized cells which have the potential to create other types of specific cells, such as blood-, brain-, tissue- or muscle-cells.

Stem cells are in all of our body and lives, but are far more potent in a fetus (also spelled foetus, ftus, faetus, or ftus) than in an adult body.

Some types of stem cells may be able to create all other cells in the body. Others have the potential to repair or replace damaged tissue or cells.

Embryonic Stem Cells are developed from a female egg after it is fertilized by sperm. The process takes 4-5 days.

Stem cell research is used for investigation of basic cells which develop organisms. The cells are grown in laboratories where tests are carried out to investigate fundamental properties of the cells.

There are stem cells in the both placenta and blood contained in the placenta. Also the primary source of stem cells is from blastocysts. These are fertilized human eggs that were not implanted into a woman.

The controversy surrounding stem cell research led to an intense debate about ethics. Up until the recent years, the research method mainly focused on Embryonic Stem Cells, which involves taking tissue from an aborted embryo to get proper material to study. This is typically done just days after conception or between the 5th and 9th week.

Since then, researchers have moved on to more ethical study methods, such as Induced Pluripotent Stem Cells (iPS). iPS are artificially derived from a non-pluripotent cell, such as adult somatic cells.

This is probably an important advancement in stem cell research, since it allows researchers to obtain pluripotent stem cells, which are important in research, without the controversial use of embryos.

There were two main issues concerning stem cell research with both pros and cons:

The first issue is really not just about stem cell research, as it may be applied to most research about human health.

Since 2007, the second point, concerns about the methods involved, has been less debated, because of scientific developments such as iPS.

As you will most probably notice, the following arguments are not exclusively in use when talking about stem cell research.

Stem cell research can potentially help treat a range of medical problems. It could lead humanity closer to better treatment and possibly cure a number of diseases:

Better treatment of these diseases could also give significant social benefits for individuals and economic gains for society

The controversy regarding the method involved was much tenser when researchers used Embryonic Stem Cells as their main method for stem cell research.

DISCLAIMER: These points are based on the old debate about the methods of stem cells research, from before 2007. Since then, scientists have moved on to use more ethical methods for stem cell research, such as iPS. This section serves as an illustration of the difficult evaluations researchers may have to analyze.

The stem cell-research is an example of the, sometimes difficult, cost-benefit analysis in ethics which scientists need to do. Even though many issues regarding the ethics of stem cell research have now been solved, it serves as a valuable example of ethical cost-benefit analysis.

The previously heated debate seems to have lead to new solutions which makes both sides happier.

Stem Cell pros and cons had to be valued carefully, for a number of reasons.

When you are planning a research project, ethics must always be considered. If you cannot defend a study ethically, you should not and will not be allowed to conduct it. You cannot defend a study ethically unless the presumed cost is lower than expected benefits. The analysis needs to include human/animal discomfort/risks, environmental issues, material costs/benefits, economy etc.

Why was the debate regarding the stem cell research so intense?

First, it was a matter of life - something impossible to measure. And in this case, researchers had to do exactly that: measure life against life.

Both an abortion and someone dying, suffering from a possible curable disease, is a tragedy. Which have the highest value? Does a big breakthrough in the research justify the use of the method in the present?

Would the benefits of studying abortions outweigh the costs? The choice was subjective: Nobody knows all the risks or all the possible outcomes, so we had to value it with our perception of the outcome. Perception is influenced by our individual feelings, morals and knowledge about the issue.

Second, at the time we did not know whether the research was necessary and sufficient to give us the mentioned health benefits.

Third, other consequences of the research are uncertain. Could the research be misused in the future or not? We simply do not know. All knowledge acquired, within research or other arenas, may be used for evil causes in the future - it is impossible to know.

The Stem cell research-debate is an example on how people value various aspects differently. It is also an example of how critics and debate can lead to significant improvements for both sides.

More here:
Stem Cell Research - Pros and Cons - Explorable.com

Read More...

Side effects of bone marrow and stem cell transplants …

Saturday, August 27th, 2016

You will have a low white blood cell count after your treatment. This means you are more at risk of getting an infection. You are likely to get an infection from the normally harmless bacteria we all have in our digestive systems and on our skin.

To try to stop this from happening your nurse may give you tablets called gut sterilisers (antibiotics) and mouthwashes. And they will encourage you to have a shower each day.

You are also at risk of infection from food. The nurses on the ward will tell you and your relatives about the food you can and can't eat. The rules vary from hospital to hospital but you may be told that

Your room will be thoroughly cleaned every day. Your visitors will be asked to wash their hands before they come into your room. They may also have to wear disposable gloves and aprons. Visitors with coughs and colds are not allowed. Some hospitals don't allow you to have plants or flowers in your room because bacteria and fungi can grow in the soil or water, and may cause infection.

Even with all these precautions, most people do get an infection at some point and need to have antibiotics. You can help yourself by trying to do your mouth care properly and getting up to shower and have your bed changed even on the days you don't feel too good.

After a transplant you will have lost immunity to diseases you were vaccinated against as a child. The team caring for you will advise you about the immunisations you need and when. You should only have inactivated immunisations and not live ones. To lower the risk of you getting any of these infections it is important that all your family have the flu vaccine and any children have all their immunisations.

Read more:
Side effects of bone marrow and stem cell transplants ...

Read More...

Risks of Stem Cell Treatments – StemCultures

Saturday, August 27th, 2016

Every day sick patients are asking how canstem cell therapy help them now. These patients are most likely desperate for any help, as the current medicine or prognosis just isnt cutting it. And while one daythere may be aviableanswer ofa yes, right now unfortunately, the field isjust not there yet. But, others do not share this view and are in fact offering to cure peoples diseases with stem cell treatments, a phenomenon known as stem cell tourism as most cases occur outside this country. Below we discuss a little about this.

What are stem cell treatments?

As was mentioned, stem cell treatments have been developed as a way to intervene in the development of and potentially treat a whole host of illnesses and physical maladies. These include baldness, missing teeth, and blindness, as well as degenerative illnesses like Parkinsons disease, type 1 diabetes mellitus, heart failure, and even cancer.

The majority of the advertised stem cell treatments utilize adult stem cells, normally harvested from the patient, and these stem cells are introduced into the damaged part of the body. The stem cells then self-renew within the damaged part, promoting growth of new tissues and subsequently replacing the diseased tissues.

Since the stem cells have been harvested from the body of the patient, theoretically, the odds of rejection or fatal side effects are very minimal. Because this is the case, stem cell treatments essentially provide a less invasive, more viable, and more sustainable therapeutic or treatment approach than similar intervention methods like organ transplantation.

Most stem cell treatments are still in the research phase.

Stem cell treatment clinics have been mushrooming everywhere. They are manifold in medical tourism centers in India, China, Ukraine, and Mexico. Even in the United States, where the oversight of the Food and Drug Administration or FDA is strict, stem cell treatment centers operate.

But while this is the case, it is crucial to keep in mind that most stem cell treatments, with the exception of bone marrow transplantation, are still in the preliminary research stages. In fact, studies of these treatments remain so new that finding published results is next to impossible.

Countries like China that study stem cell treatments on a clinical level do not have adequate and up-to-medical-standard documentation processes either, further putting the public in the dark when it comes to stem cell treatments efficacy and dangers.

There are several potential risks of stem cell treatments.

Even aside from the preliminary research phases and lack of published results, stem cell treatments have many risks. And the worst part is studies on these risks, as on the treatments efficacy, are yet to be explored by the medical community.

For instance, in the case of cancer, there is the danger of further aggravating the progress of the disease. Bear in mind that these treatments involve the introduction of stem cells into the diseased part of the body. Sure, the stem cells will most likely be harvested from the same patient and thus not foreign to the recipients body. However, factors such as uncontrolled growth may still occur and therefore further worsen the disease instead of treat it.

Another danger is the unchecked use of the types of stem cells to be administered. In countries without supervision and regulation of these types of intervention strategies, the use of stem cells harvested from sheep and sharks has been reported for treating human patients; an obviously bad situation.

Think twice before choosing stem cell treatments.

While stem cell treatment clinics are popping up all over most of these are scammers who prey on the desperately ill. Another sector has been cropping up offering stem cell treatments for cosmetic purposes as well. With promises of efficient and unfailing treatments, may they be for cosmetics, mild physical maladies, or serious terminal cases, there is no doubt that these treatments can sometimes be tempting to take.

But bear in mind that stem cell treatmentsthe legitimate ones, that isare mostly in the preliminary research stages. Because of this, you wont really be sure whether the treatment you obtain will work or not. And remember, if sounds to good to be true, it probably is. If there was a miracle treatment out there that really does cure horrible diseases, dont you think every sick patient would be getting it done and being healed? For more information, please visit this website put together by the international society for stem cell research: http://www.closerlookatstemcells.org/

If you enjoyed this post, please consider subscribing below to our monthly newsletter through which we provide stem cell scientists information on the most current research topics and tools to help them constantly improve their stem cell culture experience.

To Successful Research, StemCultures

Information for scientists by scientists on all things related to stem cell growth including stem cell culture, culture medium, tissue culture, fibroblast growth factor (fgf2) and more.

Note: StemCultures facilitates posting on this blog, but the views and accounts expressed herein are those of the author(s) or interviewee(s) and not the views or accounts ofStemCultures its officers or directors whose views and accounts may or may not be similar or identical. StemCultures, its officers and directors do not express any opinion regarding any product or service by virtue of reference to such product or service in this blog.

Read the original post:
Risks of Stem Cell Treatments - StemCultures

Read More...

A Rare Side Effects of Stem Cell Therapy: A Case Study

Thursday, August 4th, 2016

By: Ian Murnaghan BSc (hons), MSc - Updated: 9 Nov 2015 | *Discuss

There is no doubt that stem cell therapy holds enormous potential. Unfortunately, this potential also brings with it side-effects, some particularly severe. Such was the case during a therapy that used human foetal stem cells.

The boy in the case suffered from a rare genetic disease known as Ataxia Telangiectasia. This disorder affects many areas of the body and can cause significant disability. The body does not coordinate properly and those who suffer from the disease have a weak immune system as well as problems with their respiratory system.

While there have been some cases reported where experimentation on rodents resulted in the growth of tumours after stem cell injection, this hadn't been documented in humans after foetal stem cell therapy. Researchers also knew that this risk in rodents could be reduced if the stem cells were differentiated before they were injected. This means that the stem cells were coaxed into the desired body cell for the therapy prior to injection.

In a person who has a healthy immune system, the normal 'checks' on the body would be more likely to prevent a tumour from establishing itself. We have known for some time now that there is the potential for stem cells to trigger the growths of tumours but the reality has been that this is a rarity.

Rather than put a stop to stem cell research, it has been suggested that we need to spend more time looking at the Safety of Stem Cells. We should try to find out more about what can potentially go wrong and then develop safeguards to reduce any risks associated with stem cell therapies. This way, we can get the most benefits from stem cells while minimising any chances of side-effects along the way.

You might also like...

Share Your Story, Join the Discussion or Seek Advice..

hoka - 9-Nov-15 @ 5:02 PM

non - 25-Sep-15 @ 4:10 AM

non - 25-Sep-15 @ 4:06 AM

vinod - 21-Aug-15 @ 1:06 PM

AdaBrown - 13-Aug-15 @ 9:11 AM

saas - 13-Jul-15 @ 2:26 PM

Bella - 15-Jun-15 @ 12:28 AM

ExploreStemCells - 18-Mar-15 @ 10:13 AM

praju - 15-Mar-15 @ 7:30 AM

mo - 9-Mar-15 @ 2:28 AM

sanju - 13-Jan-15 @ 1:47 PM

ExploreStemCells - 15-Dec-14 @ 11:48 AM

nillu - 13-Dec-14 @ 1:50 PM

Iqbal - 24-Nov-14 @ 7:11 PM

Annu - 9-Nov-14 @ 10:33 PM

ebiiam - 20-Aug-14 @ 3:30 PM

ebiiam - 20-Aug-14 @ 3:21 PM

Pinty - 30-Jul-14 @ 6:49 AM

sam - 24-Jul-14 @ 9:41 PM

dal - 28-May-14 @ 10:58 PM

firoj - 25-Mar-14 @ 4:59 PM

flossy - 12-Jan-14 @ 2:12 PM

Gino - 10-Dec-13 @ 6:49 PM

sam - 5-Dec-13 @ 7:33 AM

Mona - 27-Nov-13 @ 3:57 AM

Sarada - 1-Aug-13 @ 7:19 AM

nicxz - 3-Jul-13 @ 9:54 AM

Title:

(never shown)

Go here to read the rest:
A Rare Side Effects of Stem Cell Therapy: A Case Study

Read More...

Frequently Asked Questions | Stem Cell Of America

Thursday, August 4th, 2016

Does the Stem Cell treatment have any age requirements?

No, Stem Cell Of America accepts patients of all ages.

There are multiple factors in determining the cost of treatment. Please contact us to have your case evaluated.

No, the treatment is not covered by private or public health insurance.

Yes, we routinely accept patients from Canada, England, Australia and other countries all over the world.

Yes, the cells are tested for 14 different criteria, including viral, bacterial, and fungal infections, as well as viability. Moreover, we use PCR DNA testing, which is far more sophisticated and expensive than the screening tests routinely used in the United States for other Stem Cell treatments.

No, Fetal Stem Cells have no antigenicity, which is a cellular fingerprint that can cause a dangerous and sometimes lethal rejection by the body. This phenomenon is called graft-versus-host disease. Our Stem Cells are free of this fingerprint, there is no threat of rejection and therefore no need for immunosuppressive drugs, which can leave the body vulnerable to serious diseases and infection.

Our Fetal Stem Cell treatment has no known negative side effects.

A partial list of disease and conditions that Stem Cell of America has successfully treated includes:

Due to the rapid advances in Stem Cell science, some diseases or conditions may not be listed. Please contact us to get additional information.

Every person is of course different. Each of our bodys healing mechanisms work at a unique pace as they are influenced by many factors. Commonly, significant positive changes are seen between three to six months post treatment. At times, these changes can occur in as little as weeks or even days after receiving treatment.

After the first treatment, the Fetal Stem Cells will continue to proliferate and repair. Some patients choose to receive treatment more than one time to expedite the healing process. The decision is yours. If you decide to repeat the treatment, usually a waiting period of 6 months is recommended.

Fetal Stem Cells are the cellular building blocks of the 220 cell types within the body. The Fetal Stem Cells used by Stem Cell Of America remain in an undifferentiated state and therefore are capable of becoming any tissue, organ or cell type within the body.

Fetal Stem Cells also release Cytokines. Cytokines are cell-derived, hormone-like polypeptides that regulate cellular replication, differentiation, and activation. Cytokines can bring normal cells and tissues to a higher level of function, allowing the bodys own healing mechanisms to partner with the transplanted Fetal Stem Cells for repair and new growth.

In the past 2 decades, Stem Cell Of America has arranged for the treatment of over two thousand patients with Fetal Stem Cells. The number of patients continues to grow. Please contact us to get specific information on a disease or condition.

Stem Cell Of America has offices in the United States and a treatment center in Mexico.

Read the original:
Frequently Asked Questions | Stem Cell Of America

Read More...

Curcumin Inhibits Breast Cancer Stem Cell Migration by …

Thursday, August 4th, 2016

Stem Cell Research & Therapy20145:116

DOI: 10.1186/scrt506

Mukherjee et al.; licensee BioMed Central Ltd.2014

Received: 12April2014

Accepted: 6October2014

Published: 14October2014

The existence of cancer stem cells (CSCs) has been associated with tumor initiation, therapy resistance, tumor relapse, angiogenesis, and metastasis. Curcumin, a plant ployphenol, has several anti-tumor effects and has been shown to target CSCs. Here, we aimed at evaluating (i) the mechanisms underlying the aggravated migration potential of breast CSCs (bCSCs) and (ii) the effects of curcumin in modulating the same.

The migratory behavior of MCF-7 bCSCs was assessed by using cell adhesion, spreading, transwell migration, and three-dimensional invasion assays. Stem cell characteristics were studied by using flow cytometry. The effects of curcumin on bCSCs were deciphered by cell viability assay, Western blotting, confocal microscopy, and small interfering RNA (siRNA)-mediated gene silencing. Evaluations of samples of patients with breast cancer were performed by using immunohistochemistry and flow cytometry.

Here, we report that bCSCs are endowed with aggravated migration property due to the inherent suppression of the tumor suppressor, E-cadherin, which is restored by curcumin. A search for the underlying mechanism revealed that, in bCSCs, higher nuclear translocation of beta-catenin (i) decreases E-cadherin/beta-catenin complex formation and membrane retention of beta-catenin, (ii) upregulates the expression of its epithelial-mesenchymal transition (EMT)-promoting target genes (including Slug), and thereby (iii) downregulates E-cadherin transcription to subsequently promote EMT and migration of these bCSCs. In contrast, curcumin inhibits beta-catenin nuclear translocation, thus impeding trans-activation of Slug. As a consequence, E-cadherin expression is restored, thereby increasing E-cadherin/beta-catenin complex formation and cytosolic retention of more beta-catenin to finally suppress EMT and migration of bCSCs.

Cumulatively, our findings disclose that curcumin inhibits bCSC migration by amplifying E-cadherin/beta-catenin negative feedback loop.

The online version of this article (doi:10.1186/scrt506) contains supplementary material, which is available to authorized users.

Breast cancer is the most common form of cancer diagnosed in women. In 2013, breast cancer accounted for 29% of all new cancer cases and 14% of all cancer deaths among women worldwide[1]. Breast cancer-related mortality is associated with the development of metastatic potential of the primary tumor[2]. Given this high rate of incidence and mortality, it is critical to understand the mechanisms behind metastasis and identify new targets for therapy. For the last few decades, various modalities of cancer therapy were being investigated. But the disease has remained unconquered, largely because of its invasive nature.

Amidst the research efforts to better understand cancer progression, there has been increasing evidence that hints at a role for a subpopulation of tumorigenic cancer cells, termed cancer stem cells (CSCs), in metastasis formation[3]. CSCs are characterized by their preferential ability to initiate and propagate tumor growth and their selective capacity for self-renewal and differentiation into less tumorigenic cancer cells[4]. There are reports which demonstrate that CSCs are enriched among circulating tumor cells in the peripheral blood of patients with breast cancer[5]. Moreover, recent studies show that epithelial-mesenchymal transition (EMT), an early step of tumor cell migration, can induce differentiated cancer cells into a CSC-like state[6]. These observations have established a functional link between CSCs and EMT and suggest that CSCs may underlie local and distant metastases by acquiring mesenchymal features which would greatly facilitate systemic dissemination from the primary tumor mass[7]. Taken together, these studies suggest that CSCs may be a critical factor in the metastatic cascade. Now, the incurability of the malignancy of the disease raises the question of whether conventional anti-cancer therapies target the correct cells since the actual culprits appear to be evasive of current treatment modalities.

Studies focusing on the early steps in the metastatic cascade, such as EMT and altered cell adhesion and motility, have demonstrated that aggressive cancer progression is correlated with the loss of epithelial characteristics and the gain of migratory and mesenchymal phenotype[8], for which downregulation of E-cadherin is a fundamental event[9]. A transcriptional consequence of the presence of E-cadherin in epithelial cells can be inferred from the normal association of E-cadherin with -catenin in adherens junctions. This association prevents -catenin transfer to the nucleus and impedes its role as a transcriptional activator, which occurs through its interaction mainly with the TCF (T-cell factor)-LEF (lymphoid enhancer factor) family of transcription factors but also with other DNA-binding proteins[10]. Accordingly, the involvement of -catenin signaling in EMTs during tumor invasion has been established[11]. Aberrant expression of -catenin has been reported to induce malignant pathways in normal cells[12]. In fact, -catenin acts as an oncogene and modulates transcription of genes to drive cancer initiation, progression, survival, and relapse[12]. All of the existing information regarding abnormal expression and function of -catenin in cancer makes it a putative drug target[12] since its targeting will negatively affect both tumor metastasis and stem cell maintenance. Transcriptional target genes of -catenin involve several EMT-promoting genes, including Slug. Expression of Slug has been shown to be associated with breast tumor recurrence and metastasis[1315]. Pro-migratory transcription factor Slug (EMT-TF), which can repress E-cadherin, triggers the steps of desmosomal disruption, cell spreading, and partial separation at cell-cell borders, which comprise the first and necessary phase of the EMT process[16].

Recently, the use of natural phytochemicals to impede tumor metastasis via multiple targets that regulate the migration potential of tumor cells has gained immense importance[17]. In this regard, curcumin, a dietary polyphenol, has been studied extensively as a chemopreventive agent in a variety of cancers, including those of the breast, liver, prostate, hematological, gastrointestinal, and colorectal cancers, and as an inhibitor of metastasis[18]. In a recent report, curcumin was shown to selectively inhibit the growth and self-renewal of breast CSCs (bCSCs)[19]. However, there are no reports regarding the contribution of curcumin in bCSC migration.

The present study describes (i) the mechanisms governing the augmented migration potential of bCSCs, which (ii) possibly associates with tumor aggressiveness and is largely attributable to the inherent downregulation of the anti-migratory tumor suppressor protein, E-cadherin, in bCSCs, and (iii) the role of curcumin in modulating the same. A search for the upstream mechanism revealed higher nuclear translocation and transcriptional activity of -catenin resulting from disruption of E-cadherin/-catenin complex formation in bCSCs in comparison with non-stem tumor cells. Upregulation of nuclear -catenin resulted in the augmentation of Slug gene expression that, in turn, repressed E-cadherin expression. In contrast, exposure to curcumin inhibited the nuclear translocation of -catenin, thereby hampering the activation of its EMT-promoting target genes, including Slug. Resultant upregulation of E-cadherin led to increase in E-cadherin/-catenin complex formation, which further inhibited nuclear translocation of -catenin. As a consequence, the E-cadherin/-catenin negative feedback loop was amplified upon curcumin exposure, which reportedly inhibits EMT on one hand and promotes cell-cell adherens junction formation on the other. These results suggest that curcumin-mediated inhibition of bCSC migration may be a possible way for achieving CSC-targeted therapy to better fight invasive breast cancers.

Primary human breast cancer tissue samples used in this study were obtained with informed consent from all patients from Department of Surgery, Bankura Sammilani Medical College, Bankura, India, in accordance with the Institutional Human Ethics Committee (approval letter CNMC/ETHI/162/P/2010), and the associated research and analyses were done at Bose Institute, Kolkata, India, in compliance with the Bose Institute Human Ethics Committee (approval letter BIHEC/2010-11/11). These tumors were exclusively primary-site cancers that had not been treated with either chemotherapy or radiation. The selected cases consisted of three primary breast cancer patients of each group. The specimens were washed with phosphate-buffered saline (PBS), cut into small pieces (5 5 mm in size), and immersed in a mixture of colloagenase (10%; Calbiochem, now part of EMD Biosciences, Inc., San Diego, CA, USA) and hyaluronidase (0.5 mg/mL; Calbiochem) for 12 to 16 hours at 37C on orbital shaker. The contents were centrifuged at 80 g for 30 seconds at room temperature. The supernatant, comprising mammary fibroblasts, was discarded, and to the pellet pre-warmed 0.125% trypsin-EDTA was added. The mixture was gently pipetted and kept for 30 minutes at 37C. Finally, the pellet obtained was washed with cold Hanks buffer saline with 2% fetal bovine serum and centrifuged at 450 g for 5 minutes at room temperature. The single cells were seeded on poly-L lysine-coated dishes and cultured in medium containing growth factors, 0.1 ng/mL human recombinant epidermal growth factor, 5 g/mL insulin, 0.5 g/mL hydrocortisone, 50 g/mL gentamycin, 50 ng/mL amphotericin-B, and 15 g/mL bovine pituitary extract at 37C. Medium was replaced every 4 days, and passages were done when the cells reached 80% confluence[20].

Human breast cancer cell lines MCF-7 and T47D were obtained from the National Centre for Cell Science (Pune, India). The cells were routinely maintained in complete Dulbeccos modified Eagles medium (DMEM) supplemented with 10% heat-inactivated fetal bovine serum (FBS), penicillin (100 units/mL), and streptomycin (100 l g/mL) at 37C in a humidified incubator containing 5% CO2. Cells were allowed to reach confluency before use. Cells were maintained in an exponential growth phase for all experiments. All cells were re-plated in fresh complete serum-free medium for 24 hours prior to the experiments. Viable cell numbers were determined by Trypan blue dye exclusion test[21]. Cells were treated with different doses (5, 10, 15, and 20 M) of curcumin (Sigma-Aldrich, St. Louis, MO, USA) for 24 hours to select the optimum non-apoptotic dose of curcumin (15 m) which significantly abrogates migration potential of bCSCs. An equivalent amount of carrier (dimethyl sulfoxide) was added to untreated/control cells. To rule out cell proliferation, all migration assays were performed in the presence of 10 g/mL mitomycin C.

For mammosphere culture, MCF-7/T47D cells were seeded at 2.5 104 cells per well in sixwell Ultralow Adherence plates (Corning Inc., Corning, NY, USA) in DMEM/F12 with 5 g/mL bovine insulin (Sigma-Aldrich), 20 ng/mL recombinant epidermal growth factor, 20 ng/mL basic fibroblast growth factor, B27 supplement (BD Biosciences, San Jose, CA, USA), and 0.4% bovine serum albumin (BSA) as previously described[22]. Primary/1 and secondary/2 mammosphere formation was achieved by using weekly trypsinization and dissociation followed by reseeding in mammosphere media at 2.5 104 cells per well into Ultralow Adherence sixwell plates.

Cell viability assay was performed by using Trypan blue dye exclusion assay. Mammospheres were treated with different doses of curcumin for 24 hours. Thereafter, the numbers of viable cells were counted by Trypan blue dye exclusion by using a hemocytometer. The results were expressed as percentage relative to the control cells.

Expression of human bCSC markers CD44 and CD24 were analyzed by flow cytometric study in different stages of breast cancer tissue as well as in MCF-7/T47D cells and primary and secondary mammospheres by using CD44-FITC and CD24-PE antibodies (BD Biosciences). bCSCs were flow-cytometrically sorted from primary breast tumors on the basis of the cell surface phenotype CD44+/CD24-/low. De-differentiation, drug resistance, and stemness phenomena were quantified flow-cytometrically by measuring mean fluorescence intensities of de-differentiation markers Oct-4-PerCP-Cy5.5, Nanog-PE, and Sox-2-Alexa Fluor-647; drug-resistance markers MRP1-FITC, ABCG2-PE, and ALDH1-FITC (BD Biosciences); and epithelial markers cytokeratin-18-PE and cytokeratin-19-PE (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA). Expression levels of E-cadherin, -catenin, and Slug (Santa Cruz Biotechnology, Inc.) were determined with respective primary antibodies conjugated with PE as previously described[23].

For immunofluorescence, cells were grown on sterile glass coverslips at 37C for 24 hours. Cells after treatment were washed briefly with PBS and fixed with 4% formaldehyde for 20 minutes at 37C and permeabilized with Triton X100 (for intracellular protein expression analysis). Thereafter, cells were blocked for 2 hours in a blocking buffer (10% BSA in PBS) and incubated for another hour in PBS with 1.5% BSA containing anti-CD44/CD24/E-cadherin/-catenin/phospho-FAK antibody (Santa Cruz Biotechnology, Inc.). After washing in PBS, cells were incubated with FITC/PE-conjugated secondary antibodies in PBS with 1.5% BSA for 45 minutes at 37C in the dark. 4-6-diamidino-2-phenylindole (DAPI) was used for nuclear staining. Coverslips were washed with PBS and mounted on microscopy glass slides with 90% glycerol in PBS. Images were acquired by using a confocal microscope (Carl Zeiss, Jena, Germany)[21].

To determine the expression of bCSC markers in the migrating versus non-migrating fraction of MCF-7 cells, bi-directional wound-healing assay was performed. Briefly, cells were grown to confluency on sterile glass coverslips, after which a sterile 10-L tip was used to scratch the monolayer of cells to form a bi-directional wound. Cells were allowed to migrate for 24 hours and then the coverslips were used for immunofluorescence staining.

Transwell migration assay was performed by using 8.0-m cell culture inserts (BD Biosciences) to test the migratory ability of primary breast cancer cells, MCF-7/T47D cells, and mammosphere-forming cells. Cells were seeded at 2.5 105 cells per well in serum-free DMEM in the upper chamber of 12-well plates and allowed to migrate for 8 hours toward DMEM containing 10% FBS in the lower chamber. After 8 hours, the cells in the upper chamber were removed with a cotton swab and the migrated cells in the lower surface of the membrane were fixed and stained with giemsa or the migrated fraction of 2 mammospheres were collected from the under-surface of the membranes after 24-hour migration assay for flow cytometry. Images were acquired with a brightfield microscope (Leica, Wetzlar, Germany) at 20 magnification. To quantify migratory cells, three independent fields were analyzed by using ImageJ software (National Institutes of Health, Bethesda, MD, USA). Migration was expressed as percentage of cells migrated. For the same, the percentage of cells that migrated in the control set of each relevant experiment was taken as 100%.

For evaluating cell adhesion property, cells were trypsinized by using trypsin-EDTA and resuspended in DMEM at a density of 0.8 106 cells per milliliter. These cell suspensions were allowed to recover from the trypsinization for 1 hour at 37C in a humidified incubator containing 5% CO2. They were mixed gently every 15 minutes during this hour of conditioning. After every 15 minutes of incubation, the dishes were removed from the incubator, and the medium containing unattached cells was removed. Images were acquired with an Olympus BX700 inverted microscope (Olympus, Tokyo, Japan) at 20 magnification. To quantify cell adhesion, the number of unattached cells at 1 hour was determined by counting three independent fields. Attachment (at 1 hour) was expressed as percentage of cells adhered, and the percentage of the control set of each relevant experiment was taken as 100%.

Spreading of the attached cells was monitored. At various time intervals (for every 30 minutes up to 3 hours), cells were imaged by using an Olympus BX700 inverted microscope (Olympus). Images of multiple fields were captured from each experimental set at 40 magnification. From the phase-contrast images, individual cell boundaries were marked with the free-hand tool of ImageJ, and the area within the closed boundary of each cell was quantified by using the analysis tool of ImageJ. Cell spreading (at 3 hours) was expressed as mean circularity of the cells. As confirmation assay for cell adhesion and spreading, MCF-7 cells and 2 mammosphere cells were plated on fibronectin (50 g/mL)-coated surface, and focal adhesions were stained and quantified by immunofluorescence staining for phospho-FAK. In fact, phospho-FAK-enriched clusters at lamellipodia were considered as focal adhesion complex. Focal adhesion segmentation and size measurement were done by using ImageJ software.

Three-dimensional (3D) invasion assay of mammospheres was performed in 96-well plates. Each well was first coated with 80 L matrigel (BD Biosciences) in 3:1 ratio with complete DMEM. Mammospheres with or without curcumin/small interfering RNA (siRNA)/short hairpin RNA (shRNA)/cDNA treatment were mixed with matrigel (6:1) and added to the previously coated wells. Thereafter, the mammospheres were allowed to invade for 48 hours. Images were photographed by using an Olympus BX700 inverted microscope (Olympus) at 20 magnification. Data were analyzed by using ImageJ software as area invaded and were expressed as percentage relative to the control set, the value of which was taken as 100%.

To obtain whole cell lysates, cells were homogenized in buffer (20 mM Hepes, pH 7.5, 10 mM KCl, 1.5 mM MgCl2, 1 mM Na-EDTA, 1 mM Na-EGTA, and 1 mM DTT). All buffers were supplemented with protease and phosphatase inhibitor cocktail[24, 25]. Protein concentrations were estimated by using Lowrys method. An equal amount of protein (50 g) was loaded for Western blotting. For direct Western blot analysis, the cell lysates or the particular fractions were separated by SDS-PAGE, transferred to polyvinylidene difluoride membrane (Millipore, Darmstadt, Germany), and probed with specific antibodies like anti-E-cadherin, anti--catenin, anti-histone H1, anti-cyclin-D1, anti-c-myc, anti-slug, anti-vimentin, anti-MMP-2, anti-MMP-9, anti-twist, anti-Snail, and anti--Actin (Santa Cruz Biotechnology, Inc.). The protein of interest was visualized by chemiluminescence (GE Biosciences, Piscataway, NJ, USA). To study the interaction between E-cadherin and -catenin, -catenin immunocomplex from whole cell lysate was purified by using -catenin antibody and protein A-Sepharose beads (Invitrogen, Frederick, MD, USA). The immunopurified protein was immunoblotted with E-cadherin antibody. The protein of interest was visualized by chemi-luminescence. Equivalent protein loading was verified by using anti--actin/Histone H1 antibody (Santa Cruz Biotechnology, Inc.)[26].

Two micrograms of the total RNA, extracted from cells with TRIzol reagent (Invitrogen, Carlsbad, CA, USA), was reverse-transcribed and subjected to polymerase chain reaction (PCR) with enzymes and reagents of the RTplusPCR system (Eppendorf, Hamburg, Germany) by using GeneAmpPCR 2720 (Applied Biosystems, Foster City, CA, USA). The cDNAs were amplified with specific primers for E-cadherin (forward-CACCTGGAGAGAGGCCATGT, reverse-TGGGAAACAT-GAGCAGCTCT) and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) (forward-CGT-ATTGGGCGCCTGGTCAC, reverse-ATGATGACCCTTT-TGGCTCC).

Cells were transfected separately with 300 pmol of E-cadherin shRNA (Addgene, Cambridge, MA, USA) or Slug siRNA (Santa Cruz Biotechnology, Inc.) by using Lipofectamine 2000 (Invitrogen). The levels of respective proteins were estimated by Western blotting. The Slug cDNA (Addgene) plasmid was used for overexpression studies. The Slug cDNA clone was introduced in cells by using Lipofectamine 2000. Stably expressing clones were isolated by limiting dilution and selection with G418 sulphate (Cellgro, a brand of Mediatech, Inc., Manassas, VA, USA) at a concentration of 400 g/mL, and cells surviving this treatment were cloned and screened by Western blot analysis with specific antibodies.

Tissues were dissected out; fixed in Bouins fixative overnight; cryoprotected in 10% (2 hours), 20% (2 hours), and 30% (overnight) sucrose solution in PBS at 4C; and frozen with expanding CO2, and serial sections were cut on a cryostat (CM1850; Leica) at 15-m thickness. The tissue sections were washed in PBS (pH 7.45) for 15 minutes and treated with 1% BSA in PBS containing 0.1% Triton X-100. Sections were incubated overnight at 25C in a humid atmosphere with primary antibodies against E-cadherin (1:100; Santa Cruz Biotechnology, Inc.) diluted in PBS containing and 1% BSA. Sections were rinsed in PBS for 10 minutes and incubated with biotinylated anti-mouse IgG (Sigma-Aldrich; 1:100) for 1 hour, followed by ExtrAvidin-peroxidase conjugate (Sigma-Aldrich; 1:100) for 40 minutes. 3-Amino-9-ethyl carbazole was used as chromogen (Sigma-Aldrich; 1:100) to visualize the reaction product. Thereafter, sections were counterstained with hematoxylin (1:1; Himedia, Mumbai, India). Finally, sections were washed in distilled water and mounted in glycerol gelatin. Images were acquired with a brightfield microscope (Leica) at 10 magnification.

Values are shown as standard error of mean unless otherwise indicated. Comparison of multiple experimental groups was performed by two-way analysis-of-variance test. Data were analyzed; when appropriate, significance of the differences between mean values was determined by a Students t test. Results were considered significant at a P value of not more than 0.05.

To determine whether CSCs are linked with tumor aggressiveness or malignancy, we performed flow cytometric analyses of bCSC markers CD44

/CD24

in patient-derived tumor samples of different stages. We also tested the migratory potentials of these primary cells of different stages of cancer by performing transwell migration assay. Interestingly, along with the gradual increase in percentage cell migration, that is, 188.67% 9.33% (

A and B), indicating that the CSC population is proportionally related with breast cancer migration. In a parallel experimental set using the razor-wound migration assay method, human breast cancer cell line MCF-7 furnished higher expression of CSC-markers (that is, CD44

/CD24

) in the migrating population as compared with the non-migrating fraction of cells as evident from our confocal data (Figure

C). In line with an earlier report[

], these results revealed that the increase in expression of CSC markers selects for breast cancer cells with enhanced malignant and metastatic ability.

Breast cancer stem cells (CSCs) are highly migratory and are correlated with aggressiveness of the disease. (A) The percentage content of breast CSCs (CD44+/CD24-/low) in different stages of breast cancer was determined by flow cytometry and represented graphically (right panel). The left panel depicts representative flow cytometry data. (B) Migration of primary breast cancer cells of different stages was evaluated by using transwell migration assay. Cells that had migrated to the lower surface of the 8.0-m membrane were stained with Giemsa stain, counted, and represented graphically (right panel). The left panel shows brightfield images of migration assay of different breast cancer stages. (C) Expression of CSC markers (CD44+/CD24-/low) was visualized by immunofluorescence in the migrating front and non-migrating pool of MCF-7 cells after 24-hour wound-healing assay. Data are presented as mean standard error of mean or representative of three independent experiments.

Our next attempt was to evaluate the migratory properties of bCSCs as compared with the non-stem tumor population. For the same, the percentage CSC content of MCF-7 and T47D, as well as of primary/1 and secondary/2 mammospheres generated from these two cell lines, was elucidated by using flow cytometry for the bCSC phenotype, CD44

CD24

. Results of Figure

A depict the presence of 4.3% 0.70% CSCs in MCF-7, 26.72% 2.40% in its 1 mammosphere, and 52.17% 2.86% in 2 mammosphere (

B); de-differentiation and drug-resistance markers, ABCG2 and MRP1 (Figure

C); and ALDH1 (Figure

D). After the presence of higher stemness and CSC enrichment in the mammospheres of both the breast cancer cell lines MCF-7 and T47D was validated, all of our later experiments were performed with mammospheres of MCF-7 cells while re-confirming the key experiments in mammospheres of T47D cells. Next, we compared the migration efficiency of mammospheres with MCF-7 cells. Interestingly, these bCSC-enriched mammospheres were found to be highly migratory as compared with MCF-7 cells within the same time frame. Briefly, mammosphere-forming cells exhibited higher adhesion property than MCF-7 cells; that is, 316% 18.19% mammosphere-forming cells were adhered as compared with MCF-7 cells (100%) (

A). Similarly, mammosphere cells demonstrated lesser circularity (0.503 0.04 mean circularity) than MCF-7 cells (0.873 0.04 mean circularity), thereby depicting higher mesenchymal and migration properties of mammospheres (

B). At this juncture, for more robust assessment of adhesion, we quantified the size of phospho-FAK-enriched focal adhesion area from the lammellipodia of MCF-7 and its 2 mammosphere-forming cells. Our results showed that the mean focal adhesion area of mammosphere-forming cells was significantly higher (

C). Even in transwell migration assay, the percentage migration of mammosphere cells (293.67% 9.56%) was higher than that of MCF-7 cells (taken as 100%) (

D). Results of Figure

D validated the findings of transwell migration assay in the T47D cell line and its mammospheres.

Relative quantification of breast cancer stem cells in MCF-7 and T47D cell lines and their mammospheres along with their characterization for stemness properties. (A) The percentage content of breast cancer stem cells (CD44+/CD24-/low) in MCF-7 and T47D cells, MCF-7/T47D-derived primary/1 and secondary/2 mammospheres, were determined by flow cytometry and represented graphically (right panel). The left panel depicts representative flow cytometry data. (B-D) Graphical representation of relative mean fluorescence intensities (MFIs) in arbitrary units (AU) of de-differentiation markers Oct-4, Sox-2, and Nanog; drug-resistance markers ABCG2 and MRP1; and stemness-related enzyme ALDH1 in MCF-7 and T47D cell lines, along with their respective 2 mammospheres as determined by flow cytometry (right panels). The left panels depict representative flow cytometric histogram overlay data. Data are presented as mean standard error of mean or representative of three independent experiments.

Breast cancer stem cell (CSC)-enriched mammospheres exhibit highly aggravated migratory properties. (A, B) Representative phase-contrast images of cell adhesion and spreading assays of MCF-7 and 2 mammosphere-forming cells (left panels). The right panels demonstrate relative quantification of the data. (C) Confocal images showing focal adhesions in MCF-7 and 2 mammosphere-forming cells, stained with phospho-FAK (PE) (red) and nuclear stain 4-6-diamidino-2-phenylindole (DAPI) (left panel). The right panel illustrates relative quantification data of mean focal adhesion area. (D) Representative brightfield images of transwell migration assays of MCF-7 and T47D cells and their respective 2 mammosphere-forming cells (left and middle panels). The right panel demonstrates relative quantification of the data graphically. (E) The percentage content of breast CSCs (CD44+/CD24-/low) in the migrated fractions of 2 mammospheres of MCF-7 and T47D cell lines as compared with non-stem cancer cells (NSCCs) was determined by flow cytometry and represented graphically (right panel). The left panel depicts representative flow cytometry data. Data are presented as mean standard error of mean or representative of three independent experiments.

At this stage, we considered the possibility that, since the mammosphere is a heterogeneous population of cells consisting of both CSCs and non-stem cancer cells, the migrated population of the mammosphere might be a heterogeneous one. It therefore becomes debatable whether the aggravated migration property of mammospheres is the contribution of bCSCs or of non-stem cancer cells. To get the answer, the migrated cells of the mammospheres were collected from the under-surface of the membranes, and flow cytometric analyses were performed to characterize the migrated cells. Results of Figure3E demonstrated that the majority of the migrating cells of the mammospheres were bCSCs for both the cell lines, that is, 83.67% 2.90% bCSCs for mammospheres of MCF-7 (P <0.001) and 80.33% 3.48% (P <0.001) bCSCs for mammospheres of T47D. These results validate that bCSCs are endowed with aggravated migration potential as compared with the rest of the non-stem tumor population.

Our effort to delineate the mechanism underlying the enhanced migratory behavior of bCSCs revealed suppression of E-cadherin expression, loss of which (a hallmark of EMT) has been reported to promote tumor metastasis[

]. In fact, our immunohistochemical analyses revealed a gradual decrease in the expression levels of E-cadherin protein with increasing stages of breast cancer (Figure

A). Results of our Western blot and reverse transcription-PCR analyses also elucidated lower protein and mRNA levels of E-cadherin in mammospheres than in MCF-7 cells (Figure

B). The same results were obtained in our confocal analyses (Figure

C). In our previous findings, we have shown an increase in CSC percentage with an increase in the stage of breast cancer (Figure

A). Therefore, we postulated that probably bCSCs maintain their aggravated migration property through suppression of the E-cadherin protein expression. As a validation of this hypothesis, shRNA-mediated silencing of E-cadherin protein expression in mammospheres resulted in significant augmentation of the migratory phenotype of these mammospheres, as reflected in our cell-adhesion assay; that is, 316.67% 23.33% E-cadherin-silenced mammosphere cells adhered as compared with the control shRNA-transfected cells (100%) (

D,

). Similarly, E-cadherin-ablated mammospheres demonstrated augmented cell spreading as depicted by loss in mean circularity of cells: that is, 0.45 0.02 and 0.27 0.03 mean circularity of cells of control shRNA-transfected and E-cadherin-silenced mammospheres, respectively (

D,

). In addition, 3D invasion potential of E-cadherin-knocked-down mammospheres was also elevated (161.67% 7.31%) when compared with control shRNA-transfected set (100%) (

E,

). These results were finally confirmed in our transwell migration assay in which E-cadherin-shRNA-transfected mammosphere cells showed 340.67% 26.97% migration as compared with 100% migration of control shRNA-transfected cells (

E,

). Transwell migration assay of mammospheres of T47D cells also rendered similar results: that is, 291.67% 15.41% cell migration in E-cadherin-shRNA transfected mammospheres as compared with 100% cell migration in control shRNA set (

E,

). Taken together, these results validate that suppressed expression of E-cadherin is essential for maintaining accentuated migration potential of bCSCs.

The augmented migration potential of breast cancer stem cells (bCSCs) results from the suppression of the epithelial-mesenchymal transition (EMT) marker, E-cadherin. (A) Immunohistological staining for E-cadherin (brown color for antibody staining and counterstained with hematoxylin) of breast tumor samples. (B) Protein and mRNA expression profiles of E-cadherin in MCF-7 cells, 1 and 2 mammospheres, was determined by Western blotting (WB) (upper panel) and reverse transcription-polymerase chain reaction (RT-PCR) (lower panel). (C) Expression of E-cadherin in MCF-7 cells and 2 mammospheres was visualized by immunofluorescence. (D) Graphical representation of relative cell adhesion (left panel) and spreading (right panel) of MCF-7-derived 2 mammospheres with or without transfection with E-cadherin-short hairpin RNA (shRNA). The efficiency of transfection was assessed by evaluating the expression of E-cadherin through WB (inset). (E) A similar experimental setup was scored for three-dimensional (3D) invasion (left panel) and transwell migration (right panel) assays. Transwell migration assay was performed under similar experimental conditions in T47D-derived 2 mammospheres (right panel). -Actin/glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used as an internal loading control. Data are presented as mean standard error of mean or representative of three independent experiments.

There are several reports delineating the pro-migratory role of -catenin protein[

,

]. Moreover, activation of -catenin pathway has been reported in CSCs[

]. Under normal conditions, -catenin exists in physical association with membrane-bound E-cadherin. However, if unbound with surface E-cadherin, -catenin becomes free to translocate to the nucleus and transcriptionally activates several pro-migratory genes necessary for EMT in association with the TCF/LEF transcription factors[

]. Results of our co-immunoprecipitation studies revealed a much lower association between E-cadherin and -catenin proteins in mammospheres as compared with MCF-7 cells (Figure

A). Moreover, although the total -catenin protein level remained unaltered, a significantly higher nuclear level of the protein was observed in mammospheres than MCF-7 cells (Figure

B). Higher nuclear localization of -catenin in mammospheres was confirmed by confocal microscopy (Figure

C). That the transcriptional activity of -catenin was augmented in mammospheres was confirmed in our Western blotting data, in which greater expression of cyclin-D1, c-myc, and Slug proteins (Figure

D), which are direct transcriptional targets of -catenin[

], was observed. However, the expression levels of another important -catenin transcriptional target, Snail, not only was very low in both MCF-7 cells and its mammospheres but also failed to show any significant difference between these two cell types (Figure

D). Cumulatively, these results validate that the higher pro-migratory milieu in bCSCs results from greater transcriptional activity of -catenin.

E-cadherin suppression in breast cancer stem cells (bCSCs) is associated with greater nuclear translocation of -catenin and subsequent trans-activation of Slug. (A) -catenin-associated E-cadherin was assayed by co-immunoprecipitation from cell lysates of MCF-7 and 2 mammospheres by using specific antibodies (left panel) or with normal human immunoglobulin G (IgG) as a negative control (right panel). To ensure comparable protein loading, 20% of supernatant from immunoprecipitation (IP) sample was subjected to determination of -actin by Western blotting (WB). (B) WB was conducted to study the levels of total -catenin and nuclear -catenin in MCF-7 and 2 mammospheres for determining the nuclear translocation of -catenin. (C) The relative nuclear expression of -catenin in MCF-7 and 2 mammospheres was visualized by immunofluorescence. (D) WB was performed to study the expression levels of -catenin target genes Cyclin-D1, c-Myc, Slug and Snail in MCF-7 cells and 2 mammospheres. (E) Protein and mRNA expression profiles of E-cadherin in 2 mammospheres of MCF-7 cells with or without transfection with Slug-short interfering RNA (siRNA) were determined by WB (right panel) and reverse transcription-polymerase chain reaction (RT-PCR) (left panel). The efficiency of transfection was assessed by evaluating the expression of Slug through WB (inset). (F, G) Graphical representation of relative cell adhesion, spreading, three-dimensional invasion, and transwell migration of MCF-7-derived 2 mammospheres with or without transfection with Slug siRNA. Transwell migration assay was also performed under similar experimental conditions in T47D-derived 2 mammospheres (G, right panel). -Actin/histone H1/glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used as an internal loading control. Data are presented as mean standard error of mean or representative of three independent experiments.

It is reported that both the EMT-promoting transcription factors, Slug and Snail, the transcriptional target genes of -catenin, are potent transcriptional repressors of the E-cadherin gene[32]. Our results above, showing significantly greater Slug gene expression in mammospheres than in MCF-7 cells with very low expression levels of Snail in both of the cell types, tempted us to evaluate whether the repression of E-cadherin in bCSCs was mediated through the -catenin/Slug pathway. To that end, siRNA-mediated silencing of Slug in mammospheres resulted in restoration of E-cadherin expression at both protein and mRNA levels (Figure5E). Under such conditions, the migration potential of the mammospheres was simultaneously retarded as was assessed by monitoring (i) adhesion, that is, 52.67% 5.61% cells adhered in Slug-silenced mammospheres as compared with the control set (100%, P <0.01) (Figure5F); (ii) spreading, that is, 0.49 0.03 and 0.7 0.04 mean circularity in control and Slug-ablated mammospheres, respectively (P <0.05; Figure5G, left panel); (iii) invasion, that is, 46.67% 4.05% invasion in Slug-siRNA-transfected mammospheres as compared with control, that is, (100%, P <0.001) (Figure5G, middle panel); and (iv) transwell migration, that is, 37.33% 5.04% in Slug knocked-down mammospheres as compared with 100% migration of the control (P <0.001; Figure5G, right panel) of MCF-7 cells. The effect of Slug silencing in migration potential was further validated in mammospheres of T47D cells (28% 5.69% migration as compared with control, P <0.001, Figure5G, right panel). All of these results confirmed that E-cadherin repression in bCSCs results from the activation of the -catenin/Slug pathway.

The phytochemical curcumin is a known repressor of several tumor properties, including tumor cell migration[

]. Additionally, several recent studies suggest that CSCs could be targeted by using curcumin[

]. However, there are no detailed studies on the anti-migratory role of curcumin in CSCs. Results of our transwell migration assay revealed that 24-hour curcumin treatment inhibits migration of bCSC-enriched mammospheres of both MCF-7 and T47D cells in a dose-dependent manner (Figure

A). Our cell viability assay data showed that curcumin exerted apoptotic effects on mammospheres of both MCF-7 and T47D cells beyond a 15 M dose (Additional file

: Figure S1). Therefore, to avoid the possibility of curcumin-induced cell death in our experimental set-up, further experiments were restricted to the 15 M dose of this phytochemical. Additional validation of the effects of curcumin on adhesion, spreading, and 3D invasion properties of mammospheresthat is, 26% 3.46% cell adhesion,

B) and 44% 4.36% invasion,

D) as compared with 100% value of the respective control sets, and 0.46 0.02 and 0.80 0.05 mean circularity (Figure

C) in control and curcumin-treated mammospheres, respectively (

E). To find out whether curcumin exposure altered only E-cadherin expression or overall epithelial characteristics of these bCSCs, flow cytometric analyses of other epithelial markers cytokeratin-18 and -19 were performed. The results revealed that curcumin augmented the overall epithelial characteristics of these cells (Figure

F). On the other hand, silencing E-cadherin expression by using shRNA significantly nullified the effects of curcumin on the various migratory phenotypes of these CSCs, namely, cell adhesion (351.67% 10.14%), 3D invasion (174% 7.37%), and migration (304.67% 23.79%), as compared with the value of 100% of the respective control sets (

G). The results of mean circularity of control (0.463 0.03) and E-cadherin shRNA-transfected mammospheres (0.276 0.03) of MCF-7 cells (

G) were in line with these findings that silencing E-cadherin expression significantly nullified the effects of curcumin on various migratory phenotypes of these CSCs. These results were validated in T47D cells in which higher migration of E-cadherin shRNA-transfected cells of mammospheres (281.67% 14.81%) was observed in comparison with untransfected ones (100%,

H). These results together indicated that curcumin inhibited bCSC migration property by restoration of the EMT-suppressor, E-cadherin.

See the original post here:
Curcumin Inhibits Breast Cancer Stem Cell Migration by ...

Read More...

Stem cell – Science Daily

Thursday, August 4th, 2016

Stem cells are primal cells found in all multi-cellular organisms.

They retain the ability to renew themselves through mitotic cell division and can differentiate into a diverse range of specialized cell types.

The three broad categories of mammalian stem cells are: embryonic stem cells, derived from blastocysts, adult stem cells, which are found in adult tissues, and cord blood stem cells, which are found in the umbilical cord.

In a developing embryo, stem cells can differentiate into all of the specialized embryonic tissues.

In adult organisms, stem cells and progenitor cells act as a repair system for the body, replenishing specialized cells.

As stem cells can be grown and transformed into specialized cells with characteristics consistent with cells of various tissues such as muscles or nerves through cell culture, their use in medical therapies has been proposed.

In particular, embryonic cell lines, autologous embryonic stem cells generated through therapeutic cloning, and highly plastic adult stem cells from the umbilical cord blood or bone marrow are touted as promising candidates.

Medical researchers believe that stem cell therapy has the potential to change radically the treatment of human disease.

A number of adult stem cell therapies already exist, particularly bone marrow transplants that are used to treat leukemia.

Excerpt from:
Stem cell - Science Daily

Read More...

Hematopoietic Stem Cells, Cell Culture Media, CFC Assays

Thursday, August 4th, 2016

A current view of hematopoiesis is that of a hierarchically organized system, with a rare population of hematopoietic stem cells (HSCs) residing at the top of the hierarchy, giving rise to all blood cell lineages. See MoreHSCs possess the ability of multipotency (i.e. one HSC can differentiate into all functional blood cells) and selfrenewal (i.e. HSCs can divide and give rise to an identical daughter cell, without differentiation).1 Through a series of lineage commitment steps, HSCs give rise to progeny that progressively lose self-renewal potential and successively become more and more restricted in their differentiation capacity, generating multi-potential and lineage-committed progenitor cells, and ultimately mature functional circulating blood cells.

The ability of hematopoietic stem and progenitor cells (HSPCs) to self-renew and differentiate is fundamental for the formation and maintenance of life-long hematopoiesis and deregulation of these processes may lead to severe clinical consequences. HSPCs are also highly valuable for their ability to reconstitute the hematopoietic system when transplanted and this has enabled their use in the clinic to treat a variety of disorders including bone marrow failure, myeloproliferative disorders and other acquired or genetic disorders that affect blood cells.2,3 Given these pivotal roles of HSPCs, much research effort has been directed at developing tools for their detection, enumeration, identification and isolation, and understanding the mechanisms underlying their behavior and fate decisions.4 Exploiting key findings of such research is highly relevant for developing novel methods to obtain clinically relevant numbers of normal HSPCs and to eliminate or inhibit cancer stem cell growth in hematopoietic malignancies.

Read the original here:
Hematopoietic Stem Cells, Cell Culture Media, CFC Assays

Read More...

BMP signaling and stem cell regulation – ScienceDirect

Thursday, August 4th, 2016

Abstract

Stem cells play an essential role in cellular specialization and pattern formation during embryogenesis and in tissue regeneration in adults. This is mainly due to a stem cell's ability to replenish itself (self-renewal) and, at the same time, produce differentiated progeny. Realization of these special stem cell features has changed the prospective of the field. However, regulation of stem cell self-renewal and maintenance of its potentiality require a complicated regulatory network of both extracellular cues and intrinsic programs. Understanding how signaling regulates stem cell behavior will shed light on the molecular mechanisms underlying stem cell self-renewal. In this review, we focus on comparing the progress of recent research regarding the roles of the BMP signaling pathway in different stem cell systems, including embryonic stem cells, germline stem cells, hematopoietic stem cells, and intestinal stem cells. We hope this comparison, together with a brief look at other signaling pathways, will bring a more balanced view of BMP signaling in regulation of stem cell properties, and further point to a general principle that self-renewal of stem cells may require a combination of maintenance of proliferation potential, inhibition of apoptosis, and blocking of differentiation.

Stem cells are the key subset of cells functioning as ancestor cells to produce a variety of types of functionally specialized mature cells in a given tissue, while at the same time undergoing self-renewal, a process of reproducing themselves without losing their developmental potentiality. This self-renewal process is controlled by intrinsic genetic pathways that are subject to regulation by extrinsic signals from the microenvironment (called niche) in which stem cells reside. Stem cells play essential roles ranging from embryonic development and organogenesis (embryonic/fetal stem cells) to tissue regeneration (adult stem cells) (Lin, 2003, Spradling et al., 2001, Watt and Hogan, 2000andWeissman, 2000). To maintain homeostasis, a precise balance between self-renewal and differentiation of stem cells is essential. Loss of this balance tends to lead to uncontrolled cell growth or pre-maturation and thus results in tumors, cancers, or tissue defects. Therefore, understanding the complex signal regulation of stem cell development is crucial for future therapeutic applications. In this review, we will focus on progress that has been made in research studying the bone morphogenesis protein (BMP) signaling pathway in regulation of stem cell properties.

BMPs belong to the transformation growth factor beta (TGF) superfamily. They are involved in regulation of cell proliferation, differentiation, and apoptosis and therefore play essential roles during embryonic development and pattern formation ( Massague, 1998). To maintain homeostasis in adults, the BMP signal also participates in tissue remodeling and regeneration, in which regulation of stem cell behavior is prominent.

There are more than 20 BMPs. Some BMPs have a distinct function while others have overlapping functions, depending on the specificity of their interaction with different types of receptors and the tissues in which they are differentially expressed (Mishina, 2003). Accumulated evidence indicates that BMPs play an important role in regulation of stem cell properties; however, their functions are different in the different stem cell compartments. For instance, in Drosophila germline stem cells (GSCs), Dpp (homolog of BMP2/4) is essential for the maintenance of stem cells ( Xie and Spradling, 1998); in embryonic stem cells (ESCs), BMP signaling appears to be required for ESC self-renewal but this is owing to its ability to block neural differentiation ( Ying et al., 2003a) in addition to its ability to promote non-neural (mesoderm and trophoblast) differentiation ( Xu et al., 2002andYing et al., 2003a); in mesenchymal stem cells, the BMP signal induces osteoblastic differentiation through Bmpr1b but inhibits osteoblastic differentiation through Bmpr1a (Chen et al., 1998); in intestinal stem cells (ISCs), BMP signaling inhibits stem cell activation and expansion (He et al., 2004); and in hematopoietic stem cells (HSCs), BMP signaling through Bmpr1a restricts stem cell number by controlling the niche size (Zhang et al., 2003). A critical and comparative review of the roles of BMPs in different settings and in different stem cell compartments is necessary for a balanced view towards BMP function in the regulation of stem cell properties, and thus will provide important insight into understanding the complex signaling regulation of stem cell self-renewal and fate determination.

The molecular mechanisms that control stem cell self-renewal remain largely unknown, albeit a large body of literature has been published with regard to stem cell self-renewal and the related signaling pathways. In the literature, self-renewal is generally described as a parallel cellular event of proliferation, differentiation, and apoptosis. However, accumulated evidence suggests that self-renewal of stem cells requires a combination of events: maintenance of their proliferation potential, inhibition of apoptosis, and blocking of differentiation.

Multiple signaling pathways have been reported to contribute to the regulation of stem cell self-renewal. However, different molecules and the underlying pathways may play different and overlapping roles in this regard. Maintaining proliferation potential is an obvious principle required for self-renewal of stem cells. However, it is worthwhile to point out that proliferation potential (defined as the capacity of stem cells to undergo continuous division) is different from proliferation per se in that the more the stem cells undergo active proliferation, the more they tend to lose their potential for proliferation. Therefore, stem cell proliferation potential is a functional property which can only be measured by continuous in vitro cell culture, or in vivo repopulation functional assay, rather than by measurement of the rate of proliferation. Recently, several lines of evidence have suggested that the Wnt signaling pathway through -catenin is important for self-renewal of HSCs (Austin et al., 1997, Brandon et al., 2000, Murdoch et al., 2003, Reya et al., 2003, Van Den Berg et al., 1998andWillert et al., 2003), hair follicle stem cells (DasGupta and Fuchs, 1999, Gat et al., 1998andHuelsken et al., 2001), ISCs (He et al., 2004, Sancho et al., 2003andSancho et al., 2004), and ESCs (Sato et al., 2004). In addition to its function in lineage fate determination, the prominent role of Wnt signaling favors cell proliferation and promotes cell growth. Abnormal activation of -catenin leads to over-proliferation of stem cells and results in tumors in the intestines and in hair follicles (Gat et al., 1998andSancho et al., 2004). In contrast, deletion of a Wnt downstream factor, Tcf4, leads to loss of stem cells in the intestines (Korinek et al., 1998). These observations suggest that Wnt/-catenin signaling is important for the proliferation potential of stem cells as -catenin may stimulate Tert (encoding the catalytic subunit of telomerase) expression via activation of Myc (He et al., 1998, Wang et al., 1998andZou et al., 2005). The idea that limiting the proliferation potential affects stem cell self-renewal has been well demonstrated by studies of telomerase (Morrison et al., 1996), HoxB4 (Antonchuk et al., 2002, Helgason et al., 1996, Kyba et al., 2002andSauvageau et al., 1995), p18 (Yuan et al., 2004), P21(Cheng et al., 2000), and Bmi (Lessard and Sauvageau, 2003, Molofsky et al., 2003andPark et al., 2003).

Recent reports indicate that suppression of apoptosis plays an essential role in stem cell self-renewal (Domen and Weissman, 2000, Domen et al., 2000, Opferman et al., 2005andYamane et al., 2005). This idea is further enforced by the fact that the role of -catenin in promoting HSC self-renewal is prominent in the Bcl2-transgenic mouse (Reya et al., 2003), indicating that a coordination between Bcl2, which inhibits apoptosis, and -catenin, which is important for proliferation potential, is required for stem cell self-renewal. Likewise, transgenic expression of the activated form of -catenin alone tends to lead to crypt cell apoptosis, as shown in the intestinal system (Wong et al., 1998). It is also reported that Akt is activated during intestinal stem cell activation and division (He et al., 2004), as well as during hair follicle stem cell activation (Zhang and Li manuscript, submitted). As Akt is a cell survival factor in general, activation of Akt during stem cell activation and division may be necessary to protect stem cells from apoptotic stress including that from partial anoikis, a phenomenon caused by detachment from the extracellular matrix during cell division (Khwaja et al., 1997). Consistent with this conclusion, activation of PI3K/Akt, as a consequence of the loss of PTEN-function, has been reported to result in expansion of embryonic and neural stem cell populations (Groszer et al., 2001andKimura et al., 2003).

More here:
BMP signaling and stem cell regulation - ScienceDirect

Read More...

What Is Stem Cell Research ? (with pictures) – wiseGEEK

Thursday, August 4th, 2016

anon950526 Post 156

Is there any impact due to this?

Obviously, some of you dont have kids. The life of a child is worth so much more than any adult. You got to live. What if that embryo happened to be you? Would you then feel that it is OK to conduct this research?

I am a mother of two, soon to be three. I don't care about any of that just long as my kids at least get a chance at living and there is a God. I had a 50 percent chance of having babies because of a huge benign tumor that grew on my left ovary and killed my left fallopian tube.

I prayed for my babies and got them every time. Besides that, everybody has their

I watched this gruesome abortion video and the lady was 12 weeks along. You could see the child trying to fight for its life. Murder is murder. Helping to save other people or not -- that's like you seeing a man trying to rape a woman and you shoot him dead. It's the same if you were trying to save her life but you get persecuted and convicted for taking matters into your own hands. I am sorry for those people who are sick and have sick babies. I know what it is like to lose loved ones over untreatable diseases. Im against embryo research and I'm not thinking about me. It is about a baby. Sure, it isnt completely formed, but it's still a child, or at least will grow into one, I wish harm on nobody. There is no harm meant and Im not trying to make someone mad. Im just trying to throw some new views into the situation.

Stem cell research can only benefit society and advance us as a species. If your argument is religious, the you are not thinking. You are letting your emotions and beliefs speak for you, not your logic or common sense. A bunch of cells is not a baby, and helping the living is not against "God's will". This is a good thing and it will continue regardless of religious views, because it makes sense.

I'm still kind of learning about this topic, but abortion is something I feel strongly against, but if a baby was taken from it's mother with the mother's okay and they were trying to save people's lives, I would be completely okay with that.

I believe that God does not exist, and that stem cell research is truly phenomenal. This research should not be controversial, nor should it be banned; it is helping the living.

Most of the people who say that stem cell research is bad are religious, but people living in the real world and believe in this thing called 'science' actually make a difference. Religion has only held back society and science. I wonder how many religious people would get angry if they knew that I was a homosexual, atheist physicist who believes in evolution and the big bang theory.

I am writing a persuasive essay on whether stem cell research should be legal or not (even though it already is in the U.S.). I was never a really religious person and stuck mainly to things that I knew for sure were happening. The thing is, most of the stem cells they are using for research are going to be discarded anyway. No one is claiming them, no one is caring about them, and they are just going to be thrown away. It is better for them to be used for a greater cause than just being thrown away and losing the chance to create treatments and cures for cancer and neurodegenerative diseases.

Without trying to offend anyone, please don't

bring God into this, like if you're going to simply take the stem cells and create babies with them. The cells could be considered early life, but you lose cells every day and no one gives it a second thought. I am thirteen years old and sorry if you believe that I am wrong.

This is a terrible thing. Stem cell research is just an excuse for making us Americans pay for other peoples' abortions. This stem cell research crap may have fooled my friends who claim abortion is when the baby isn't fully developed and that it's murder to kill a baby after it's born. What's the difference? Abortion is murder.

This country is so corrupt it will probably start killing the elderly and calling it abortion, or calling every day murder of people abortion. Well, I have had enough of this crap Obama is trying to trick us with. God says life is precious and an undeveloped baby is made of many cells and cells are alive. Think about that, America. Not only that, but abortion is unnecessary. If a girl gets raped, she can put the baby up for adoption instead of murdering the baby. Studies show many women who had abortions regret it.

So what you are all arguing about is if god exists and whats his plan for us, and why or why should we not use pre-embryo stem cells. It's completely your own opinion, but when does life start for a baby -- when the sperm reaches the egg or when you hear his heartbeat?

I, for one, say we should not use embryo stem cells because they are a living being. Also for all of you who say its gods plan for us, who created god? He could not just have created himself out of nowhere. These are just my thoughts. But these are all still questions we do not know the answer to.

I've been researching this Stem Cell subject for a long time, and I'm so amazed at everyone's stories from the news and sites on how stem cells has helped them recover from so many types of sicknesses and diseases. Even cancer can be cured by this type of treatment.

One big factor is that it's not a drug but it will just treat your body in a nice and natural way. Stem cell therapy is nice but I found this Laminine on the market. People keep on talking about it, saying it's the new science breakthrough and that I should give a try. It's not a literal stem cell but it is a stem cell enhancer, and safer than the usual way of Stem Cell Therapy.

I gave it a try and in just a few weeks I felt its proven power that I also recommend it to everyone out there.

How is that clump of cells considered a newborn? Those cells aren't a newborn because there's still a chance that once you implant those embryos they don't hold, so it's not a child. They don't take these people's cells then say, nope you can't have your child -- we're going to use them for someone else. They're leftovers. No one is going to use them and they are going to get discarded. If you consider it a human how is it humane to let it sit there frozen forever, discarded and unloved? People throwing god around are ignorant. Not everyone believes in your "creator" and don't throw it in my face. Believe in your beliefs, but don't force mine.

@post 52: If there is no God, who do you think made the universe? Your dad didn't make it, I didn't make, you didn't make, nor did any person. Only an eternal being must have made the universe.

Look. What none of you guys are realizing is that embryonic stem cell research doesn't focus solely on embryos.

I'm a student getting my masters degree in pediatric nursing, so you guys can't say I don't know this. But maybe if you did some research instead of arguing that everyone except yourself is wrong, you would realize there are valid points to both sides.

Embryonic stem cell research also focuses on umbilical cords and placentas, which does no harm to the baby whatsoever. Now none of you can say that's "murder" or against your religion because I'm Roman Catholic, which is one of the main religions against stem cell research and I am personally all for it.

Now if embryos

Enough about manipulating death and being religiously wrong. Enough said, simple as that.

A novel called "Living Proof" just came out in stores this week that explores the life and death issue of embryonic stem cell research for the first time as a story. It's getting a lot of buzz online and pertains directly to this discussion.

Most say stem cell research is bad because scientists are trying to pay God, but that's a bunch of crap. Like what others have said, God created us, knowing that one day we would come up with this knowledge to maybe find cures. Somebody else stated that we are ungrateful because we want to use this research, but that's not entirely true, because we are grateful for this new research to cure people like me. Yes I said me. I'm a 16 year old diabetic. I may not suffer as much as others with other diseases, but I have.

Also, this same person said that we should be the ones serving overseas. Well, if you've paid attention, we can't because

Anyway, most don't know what it's like to stick a big needle into their own skin every day, but I do. They also don't know what its like to wake up very weak due to a low blood sugar, or to throw up because you get ketones due to not having any more insulin going through their body.

Lastly, most of you probably aren't scared to go to bed, knowing that you might not wake up because you went low, with no one knowing and died. Yes, I know diabetes isn't the worse disease out there, but it's not easy either. I don't really like abortion, but at least the fetus could help cure many people, and not just get thrown away.

In a way, the government not allowing stem cell research, and the people against it can be considered murderers too, because they are standing in the way of curing people, which could save their lives. This is how our country is going downhill, not the other way around.

Finally, you say how a human life is so important, and yes it is, but who's to say that an animal's life isn't? People abuse animals, use them to test new products, that most people use, but I don't see you caring about that. Yes, some people do, but most don't give a crap. And I mean, didn't God create animals too, so shouldn't they be just as important as humans?

Yeah, so that's all I have to say. Hopefully this will make people use their brains a little more, because the people who are against it only really seem to care about themselves, not the people who are actually suffering!

The controversy surrounding the morality of stem cell research is centered around the creation, usage, and destruction of the human embryos. Currently, the limits of technological advancement require the destruction of the human embryo in creating the human embryonic stem cell. Various groups view an embryo as an early-aged human life. As a result, they are concerned with the rights and status of the embryo, and often go so far as to equate such research with murder because of the embryos destruction. However, despite scientific evidence suggesting that the early-stage embryos being used are not early-aged human life, the importance of these embryonic stem cells and their contribution to scientific advancement is tremendous.

Stem cells are cells in the human

John Stuart Mills principle of Utilitarianism also supports the morality of stem cell research. Utilitarianism states that an actions moral worth is determined solely by its contribution to the happiness of all parties involved. The phrase the greatest good for the greatest number of people is often used to describe this principle. But more precisely, the true morality of such research is exhibited in the concept of Negative Utilitarianism. Negative Utilitarianism requires us to promote the least amount of harm, or prevent the greatest amount of suffering for the greatest number of people.

Since science has established that are embryos not yet human, any harm inflicted on them does not weigh in on the moral worth of the action. However, the development of treatments that could potentially cure conditions such as Parkinsons disease and Alzheimers would weigh in on its moral worth. As a result, the prevention of suffering made possible by stem cell research and its potential medical advancements far outweigh any harm inflicted on the embryos, even if the embryos were given moral standing. Thus, by means of Negative Utilitarianism, the morality of stem cell research cannot be called into question.

This is modern day fascism. You shouldn't choose what life has more importance. Speaking as a veteran, people like this make me regret serving an ungrateful country, full of morally degraded people. These people who believe in this should have been the ones overseas. Then tell me how easy it is to choose one life over the other. Those people make me sick, and will be the downfall of this country.

Fundamentalists never fail to amaze me with their ability to only read half the story. The embryos used in stem cell research would be discarded anyway - stem cell research isn't denying them a chance at life, they had no chance at life in the first place. It isn't the same thing as abortion.

And I hope the fundies who are making comments along the lines of "We suffer because God wills it" never take antibiotics when they are sick - surely that would be messing with God's plan for you to die from a disease that modern science can easily cure?

Any opponent to stem cell research on the grounds of all this embryo is a human life crap is nothing but a ignorant idiotic hypocrite and the same goes for anti abortionists.

Why claim to give a crap at all about so called life when none of you seem to give a crap about the starving millions in underdeveloped countries, the starving on the street, those on death row etc.? What about those lives? Aren't they more convincing examples of 'life' than a pile of embryonic goo? Are they not deserving of all the fuss you make over the value of human life?

You people seem more concerned with spouting your ignorant, selfish beliefs and halting progress that could one

Is there any difference between you people in regards to this and those that shared the same beliefs that used to carry out witch hunts all those many years ago? one has to wonder.

i think that stem cell affords advancements to the medical industry. people should stop trying to use the phrase "who are we to play god". if that is the case then don't take medication to relieve pain because under those conditions would that also be playing god?

Remember that some stem cells are taken from the umbilical cord and adult tissue, not just embryos. You wouldn't call it murder if the cells were taken from an inanimate piece of flesh, would you?

I have been reading comments and "playing god" is stupid. Getting and giving shots are playing god implants and anything like that is playing god. you're not letting what happens happen. I read about a wife with four kids with Cystic Fibrosis. finding a cure for that would be playing god. That would be taking his power to save a child.

I think it's all right. People are going to abort fetuses no matter what you say or how you feel. You can say it's wrong and waste it or you can use it to support something new and know you helped to save a life. Would you honestly say that because of what you think you should throw away something that could help people just because its from something not even alive yet?

O.K. so it might be alive, but at an older age in the pregnancy. And people are right: if someone you love was dying, you would not just sit there and watch and say, oh well, too bad for you. You would try to help no matter what the cost.

I don't know what is so bad about trying to save life. Stem cell research has advanced into the stages of using actual cells from adults, (Somatic cells) and this is pushing research today. Take some time and do the "current" research about stem cells and educate yourselves.

As far as the religious perspective goes I am a Christian and "God" gave me the cells in my body and if those cells that "God" gave have a way of saving my life, then that is his will. Helping your body heal is not playing God, it is using what God gave you!

I'm curious; What defines something as "live"? When does life begin? Well, does it not begin at fertilization when the cells go through meiosis? And the DNA is replicated? Well here's what I have to say.

Again, what defines something as a "Live" human? Is it size? Level of development? Environment?

Degree of dependency?

If it's based on size, then isn't that size-ism? Does that mean our society is saying that the unborn aren't human because they aren't as big as us? Yes, an unborn baby isn't as big as a toddler, but a toddler isn't as big as a full grown adult. So does that mean that they toddler isn't human either? Or in any way less human

Level of development: Some argue that since the unborn aren't fully developed yet, they aren't human. I'm 15 and I'm not fully developed; does that mean I'm not human? No. I'm still growing. Development doesn't stop at birth. It starts at conception.

The most common argument in this category is the baby can't think, or feel pain, or even know that they exist. I beg to differ. There was an article published in a newspaper that said a doctor was performing an abortion, and on the screen, you could see the baby trying to get away from the tool trying to pull it out. In another, there was a case where the baby stuck its hand out and held onto the doctor's finger. Look it up.

We say that they can't feel pain, so they aren't human. But what about those with Sepa disease? They are born unable to feel pain; can we go and kill them too? They can't feel pain so they aren't human, so it's okay, right? Wrong.

Environment: Most common argument: The unborn baby isn't in the world yet, it's in the mother's body, and it doesn't even breathe air. This argument seems to be saying that the unborn child isn't human because it's in a different environment then we are. But, since when does where we are, determine who we are? In our day to day lives, we change our environment multiple times. But it doesn't change who we are as a person, unless you have a multiple personality disorder.

So here's a question; How does the eight-inch trip down the birth canal change who you are as a blob of tissue, into a valued human being with rights? Truth be told, it doesn't. Another argument is the unborn baby is in the mother's body, which is her body, so the mother should be able to do what ever she wants with that baby. So what's the difference between a baby the day before it's born, and one day after?

A day before: Not fully developed; dependent on the mother; in the mother's body -- her property.

A day after: Not fully developed; dependent on the mother; in the mother's house -- her property

What makes it okay to kill the one but not the other?

There was a case where a man went and murdered a pregnant woman and was charged with double murder. In that case, the government and court are considering that fetus is a life with value. However, in the same time, something like 32 abortions were performed under the protection of the law. How come those babies don't get the same justification? Is it because they aren't wanted? If an orphan was murdered, would no one care because they weren't wanted? Of course not. It's absurd to me the double standard in our society.

Degree of Dependency: Arguments are that if the unborn baby is still dependent on the mother, and can't survive on their own yet, they aren't human. Even a one week old baby is still dependent on the mother. It will be for a while.

Once again, I'm 15 and I can't survive on my own. I depend on my parents. I depend on my government and school system, I depend on my friends. I'm dependent. But do I not have value and rights?

What about those who depend on medical instruments? And life support?

Parents depend on others to provide them with jobs, food and money, with places for their children to go to get an education. Our classrooms are getting smaller and smaller due to the number of abortions each year. But again, does this affect a person's humanity in some way because they depend on someone? You're going through a divorce and you need someone to lean on; you're depending on them. Oh, yeah, sorry for the bad timing, but oh geez this is tough, you're not human, so we're going to have to kill you. No, I don't think so.

What about those on welfare? Can we kill them too because they depend on the government to provide them with money? I don't think so. We are all dependent on someone to a degree. But who goes around saying that those who depend on someone are less human or not human at all? No one. Why? Because it's hypocritical and illogical. But somehow, our society is able to accept this argument when it comes to unborn children. The faces of tomorrow. We are the people are today, and we're killing tomorrow's people.

So if we use these arguments to allow the killing of the unborn, then we should be allowed to kill: Any child; those on welfare; those with medical tools and medications; those with mental disabilities. They aren't what people consider "the norm"- are very dependent, usually have a difference in size, aren't developed as much as those who are not disabled, and depending on the case, their environment may be different then ours. And so on and so forth.

Of course not. We would never dream of doing that. It upsets many many people even using those cases as examples. So it should be clear that the unborn are human, as well as those with developmental disabilities, differences, different circumstances- welfare, etc., and the sick.

For people who don't understand, here is what I'm saying. I am pro choice- that women should have choices to do what they want in life from the unimportant (what flavor of ice cream I want) to the extremely important (what career I want to pursue) but should not have the "right" to make a choice about another person's life. You don't get to decide who lives or dies.

I do not believe we should use stem cells. If we can cure all diseases and grow back body parts then we will evidently live a very, very, very long time. This could potentially result in an overpopulation problem which we are currently starting to experience.

Humans were not meant to live forever, and maybe you should ask yourself: do you really want to live forever?

To those people who say that it's okay for scientists to do stem cell research, yes it's okay for them to do research to improve other people's lives without using stupid, dangerous chemicals on the cells. Some aspects of stem cell research, is not just against religious values, but also against our morals in general.

Thank you anon332, for trying to knock some sense into these people, who think that science always does good things. Wake up!

Religion is a vessel of our hopes, fears and a face of the unexplained. Without religion, science would grow like a cancer, and without science the other way around. Balance is what is needed.

For those who believe in God, it would not be that with stem cells we are playing God. The man or woman will live because God had willed it.

For those who don't, embryos are lives. A full life. Of course, this is only my opinion.

The world makes us what we are. It influences our choices and our minds. Everything around us at every moment is changing us.

In short, the world makes us what we are, but we make the world as how it is.

Science without religion is blasphemous. Religion without science is idiocy.

I believe stem cell research is definitely a good thing for people that are sick. I don't think people should be allowed to use a fetus for the research, but embryonic research is ethical. The embryo isn't yet developing human characteristics the way a fetus is.

I will begin by saying that I am a 14 year old girl of undecided religious beliefs. I have read many of these comments, and I have a few ideas which may help/clear up some misconceptions.

As of now, I do not officially believe in God. I do, however, understand many religions and I have accepted religious beliefs and ideas, especially those pertaining to abortion and stem cell research. Here are some of my views on the subject. I have tried to incorporate all the different positions on the subject:

1. Embryos are not fetuses and stem cell research is not the same as abortion. Fetuses are developed forms in which the cells have begun to specialize. Embryos are clusters

2. If an embryo is not used, it can be donated, either to research or to another parent, or it can be thrown away. Based on the dilemmas that seem to be arising, i am assuming that no one is of the belief that throwing away the embryos is in the best interest of anyone. That leaves donation to a parent or donation to research. If the decision is made to donate to another parent, then I think that is fine. If that idea is declined, then I don't see why a group of cells shouldn't be used to potentially help others. If a leftover embryo is just going to be thrown away, then why would the people throwing it away care if it were used. Remember, an embryo does not have even the most remote form of a brain or a heart.

3. If the argument is about whether or not the embryo has a soul, I cannot help. I do not believe that a random grouping of cells has a soul. I do not even necessarily believe in souls.

4. In this article, it says that the most common argument against stem cells is the belief in man not manipulating human life. I cannot say whether or not this is actually the big argument, but for those people who do believe that i will ask, "Have you been vaccinated? Have you ever taken any medicine for an illness? Is this not manipulating life?". If one believes that Man should not be allowed to manipulate life, then they should also believe that medicines and known cures should not be used. if a person gets pneumonia, should the doctors just let them die because they don't believe in manipulating life? Isn't that murder?

As for manipulating the embryo, I can only repeat that I do not personally believe that a group of unspecialized cells should be treated as humans.

5. I do not believe that stem cell research should be used for cloning. As for "creating another you" in case you get diseases later in life, watch or read "My Sister's Keeper".

6. As for the ethics, there are generally five ethical approaches: utilitarian-whatever does the most good and the least harm, Rights-whatever considers the rights of everyone involved, Fairness of Justice-treats all equally and proportionally, Common good-the values of Confucianism or putting the group before the individual, and Virtue-what will make me more of the person i want to be? If using an organism that is displaying the characteristics of life, but that is not developed into a fetus will help others, I don't see how it contradicts any of the ethical approaches.

6. If I decidedly believed in God, I would say, "God made us. I believe he made us for a reason. He gave us prayer and life. He also gave us doctors and medicine. we should use them".

7. For those who say "if your loved one was dying, then your view would change" you are correct. Their view would change, but do we really want a society where people make official decisions based on the health status of their loved ones, when they are stressed and not thinking clearly?

These are the things I have come up with. I didn't mean to offend anyone with my statements, and if I did, I am sorry.

Yeah, they are not using a fetus. they are using an embryo, which there is a huge difference.

Number one, they are not using a fetus; which i agree is human. The embryos used for stem cell research are four to five days old and have no specialized tissues, no nervous system, or heart. Each embryo contains about one-hundred cells, the cells of which are still undifferentiated (meaning that the cell has not decided what it is going to be).

For those of you saying that we are playing god. Is not God's greatest gift the gift of life? After IVF a woman has limited choices what to do with her leftover embryos. She may donate to another couple, donate to research, keep the embryos for maybe future implantation, or she may discard them.

Read more:
What Is Stem Cell Research ? (with pictures) - wiseGEEK

Read More...

Stem Cell Transplantation | MD Anderson Cancer Center

Thursday, August 4th, 2016

A stem cell transplant is a procedure that replaces defective or damaged cells in patients whose normal blood cells have been affected by cancer. Transplants also are used to help patients recover from aggressive radiation and chemotherapy treatments.

Stem cells are immature cells that begin life in the bone marrow and eventually develop into the various types of mature blood cells:

There are three types of stem cell transplantation:

Autologous transplant: cells are harvested from the patient's own bone marrow before chemotherapy and are replaced after cancer treatment.

Allogeneic transplant: stem cells come from a donor whose tissue most closely matches the patient.

Umbilical cord blood from newborn infants is extracted from the placenta after birth and saved in special cord blood banks for future use. MDAnderson's Cord Blood Bank actively seeks donations of umbilical cords.

Stem cell transplants commonly are used to treat leukemia and lymphoma, cancers which affect the blood and lymphatic system. Transplants also can be used to help patients recover from or better tolerate cancer treatment, and to treat hereditary blood disorders such as sickle cell anemia.

Stem cell transplant patients are matched with eligible donors by human leukocyte antigen (HLA) typing. HLA are proteins that exist on the surface of most cells in the body. HLA markers help the body distinguish normal cells from foreign cells, such as cancer cells.

HLA typing is done with a patient blood sample, which is then compared with samples from a family member or a donor registry. It can sometimes take several weeks or longer to find a suitable donor.

The closest possible match between the HLA markers of the donor and the patient reduces the risk of the body rejecting the new stem cells (graft versus host disease).

The best match is usually a first degree relative (children, siblings or parents). However, about 75% of patients do not have a suitable donor in their family and require cells from matched unrelated donors (MUD), who are located through registries such as the National Marrow Donor Program.

Because the patients immune system is wiped out before a stem cell transplant, it takes about six months to a year for the immune system to recover and start producing healthy new blood cells. Transplant patients are at increased risk for infections during this time, and must take precautions. Other side effects include:

Read more:
Stem Cell Transplantation | MD Anderson Cancer Center

Read More...

Cancer stem cell – Wikipedia, the free encyclopedia

Friday, October 23rd, 2015

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 focus of cancer research[1]

In different tumor subtypes, cells within the tumor population exhibit functional heterogeneity, and tumors are formed from cells with various proliferative and differentiate capacities.[2] This functional tumour heterogeneity among cancer cells has led to the creation of at least two models, which have been put forward to account for heterogeneity and differences in tumor-regenerative capacity: the cancer stem cell (CSC) and clonal evolution models[3]

The cancer stem cell model refers to a subset of tumor cells that have the ability to self-renew and are able to generate the diverse tumor cells.[3] These cells have been termed cancer stem cells to reflect their stem-like properties. One implication of the CSC model and the existence of CSCs is that the tumor population is hierarchically arranged with CSCs lying at the apex of the hierarchy[4] (Fig. 3).

The clonal evolution model postulates that mutant tumor cells with a growth advantage are selected and expanded. Cells in the dominant population have a similar potential for initiating tumor growth[5] (Fig. 4).

[6] 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[7] (Fig. 5).

The existence of CSCs is a subject of debate within medical research, because many studies have not been successful in discovering the similarities and differences between normal tissue stem cells and cancer (stem) cells.[8] Cancer cells must be capable of continuous proliferation and self-renewal in order to retain the many mutations required for carcinogenesis, and to sustain the growth of a tumor since differentiated cells (constrained by the Hayflick Limit[9]) cannot divide indefinitely. However, it is debated whether such cells represent a minority. If most cells of the tumor are endowed with stem cell properties, there is no incentive to focus on a specific subpopulation. There is also debate on the cell of origin of CSCs - whether they originate from normal stem cells that have lost the ability to regulate proliferation, or from more differentiated population of progenitor cells that have acquired abilities to self-renew (which is related to the issue of stem cell plasticity).

The first conclusive evidence for CSCs was published in 1997 in Nature Medicine. Bonnet and Dick[10] isolated a subpopulation of leukaemic cells that expressed a specific surface marker CD34, but lacked the CD38 marker. The authors established that the CD34+/CD38 subpopulation is capable of initiating tumors in NOD/SCID mice that are 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.[11]

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 ability to inhibit it. However, efficient tumor formation requires thousands or tens of thousands of cells to be introduced. Classically, this has been 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.[10]

Further evidence comes from histology, the study of the tissue structure of tumors. Many tumors are very heterogeneous and contain multiple cell types native to the host organ. Heterogeneity is commonly retained by tumor metastases. This implies that the cell that produced them had the capacity to generate multiple cell types. In other words, it possessed multidifferentiative potential, a classical hallmark of stem cells.[10]

The existence of leukaemic stem cells prompted further research into other types of cancer. CSCs have recently been identified in several solid tumors, including cancers of the:

Once the pathways to cancer are hypothesized, it is possible to develop predictive mathematical biology models,[29] e.g., based on the cell compartment method. For instance, the growths of the abnormal cells from their normal counterparts can be denoted with specific mutation probabilities. Such a model has been employed to predict that repeated insult to mature cells increases the formation of abnormal progeny, and hence the risk of cancer.[30] Considerable work needs to be done, however, before the clinical efficacy of such models[31] is established.

The origin of cancer stem cells is still an area of ongoing research. Several camps have formed within the scientific community regarding the issue, and it is possible that several answers are correct, depending on the tumor type and the phenotype the tumor presents. One important distinction that will often be raised is that the cell of origin for a tumor can not be demonstrated using the cancer stem cell as a model. This is because cancer stem cells are isolated from end-stage tumors. Therefore, describing a cancer stem cell as a cell of origin is often an inaccurate claim, even though a cancer stem cell is capable of initiating new tumor formation.

With that caveat mentioned, various theories define the origin of cancer stem cells. In brief, CSC can be generated as: mutants in developing stem or progenitor cells, mutants in adult stem cells or adult progenitor cells, or mutant differentiated cells that acquire stem-like attributes. These theories often do focus on a tumor's cell of origin and as such must be approached with skepticism.

Some researchers favor the theory that the cancer stem cell is generated by a mutation in stem cell niche populations during development. The logical progression claims that these developing stem populations are mutated and then expand such that the mutation is shared by many of the descendants of the mutated stem cell. These daughter stem cells are then much closer to becoming tumors, and since there are many of them there is more chance of a mutation that can cause cancer.[32]

Another theory associates adult stem cells with the formation of tumors. This is most often associated with tissues with a high rate of cell turnover (such as the skin or gut). In these tissues, it has long been expected that stem cells are responsible for tumor formation. This is a consequence of the frequent cell divisions of these stem cells (compared to most adult stem cells) in conjunction with the extremely long lifespan of adult stem cells. This combination creates the ideal set of circumstances for mutations to accumulate; accumulation of mutations is the primary factor that drives cancer initiation. In spite of the logical backing of the theory, only recently has any evidence appeared showing association represents an actual phenomenon. It is important to bear in mind that due to the heterogeneous nature of evidence it is possible that any individual cancer could come from an alternative origin. Recent evidence supports the idea that cancer stem cells, and cancer, arise from normal stem cells.[33][34]

A third possibility often raised is the potential de-differentiation of mutated cells such that these cells acquire stem cell like characteristics. This is often used as a potential alternative to any specific cell of origin, as it suggests that any cell might become a cancer stem cell.

Another related concept is the concept of tumor hierarchy. This concept claims that a tumor is a heterogeneous population of mutant cells, all of which share some mutations but vary in specific phenotype. In this model, the tumor is made up of several types of stem cells, one optimal to the specific environment and several less successful lines. These secondary lines can become more successful in some environments, allowing the tumor to adapt to its environment, including adaptation to tumor treatment. If this situation is accurate, it has severe repercussions on cancer stem cell specific treatment regime.[35] Within a tumor hierarchy model, it would be extremely difficult to pinpoint the cancer stem cell's origin.

CSC, now reported in most human tumors, are commonly identified and enriched using strategies for identifying normal stem cells that are similar across various studies.[36] The procedures include fluorescence-activated cell sorting (FACS), with antibodies directed at cell-surface markers, and functional approaches including SP analysis (side population assay) or Aldefluor assay.[37] The CSC-enriched population purified by these approaches is then implanted, at various cell doses, in immune-deficient mice to assess its tumor development capacity. This in vivo assay is called 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.[38]

CSC can also be identified by efflux of incorporated Hoechst dyes via multidrug resistance (MDR) and ATP-binding cassette (ABC) Transporters.[37]

Another approach which has also been used for identification of cell subsets enriched with CSCs in vitro is sphere-forming assays. Many normal stem cells such as hematopoietics or stem cells from tissues are capable, under special culture conditions, to form three-dimensional spheres, which can differentiate into multiple cell types. As with normal stem cells, the CSCs isolated from brain or prostate tumors also have the ability to form anchorage-independent spheres.[39]

Data over recent years have indicated the existence of CSCs in various solid tumors. For isolating CSCs from solid and hematological tumors, markers specific for normal stem cells of the same organ are commonly used. Nevertheless, a number of cell surface markers have proved useful for isolation of subsets enriched for CSC 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).,[40] 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, NK 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 role in conversion of retinol to retinoic acid, which is essential for survival.[41][42]

The first solid malignancy from which CSCs were isolated and identified was breast cancer. Therefore, these CSCs are the most intensely studied. Breast CSCs have been enriched in CD44+CD24/low,[40] SP,[43]ALDH+ subpopulations.[44][45] However, recent evidence indicates that breast CSCs are very phenotypically diverse, and there is evidence that not only CSC marker expression in breast cancer cells is heterogeneous but also there exist many subsets of breast CSC.[46] Last studies provide further support to this point. 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.[47]

CSCs have been reported in many brain tumors. Stem-like tumor cells have been identified using cell surface markers including CD133,[48]SSEA-1 (stage-specific embryonic antigen-1),[49]EGFR[50] and CD44.[51] However, there is uncertainty about the use of CD133 for identification of brain tumor stem-like cells 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.[50]

Similarly, CSCs have also been reported in human colon cancer.[52] For their identification, cell surface markers such as CD133,[52] CD44[53] and ABCB5,[54] or functional analysis including clonal analysis [55] or Aldefluor assay were used.[56] Using CD133 as a positive marker for colon CSCs has generated conflicting results. Nevertheless, recent studies indicated that the AC133 epitope, but not the CD133 protein, is specifically expressed in colon CSCs and its expression is lost upon differentiation.[57] In addition, using CD44+ colon cancer cells and additional sub-fractionation of CD44+EpCAM+ cell population with CD166 enhance the success of tumor engraftments.[53]

Multiple CSCs have been reported in prostate,[58]lung and many other organs, including liver, pancreas, kidney or ovary.[41][59] In prostate cancer, the tumor-initiating cells have been identified in CD44+[60] cell subset as CD44+21+,[61] TRA-1-60+CD151+CD166+[62] or ALDH+[63] cell populations. Putative markers for lung CSCs have been reported, including CD133+,[64] ALDH+,[65] CD44+[66] and oncofetal protein 5T4+.[67]

Metastasis is the major cause of tumor lethality in patients. However, not every cell in the tumor has the ability to metastasize. This potential depends on factors that determine growth, angiogenesis, invasion and other basic processes of tumor cells. In the many epithelial tumors, the epithelial-mesenchymal transition (EMT) is considered as a crucial events in the metastatic process.[68] 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.[69] The EMT appears to be controlled by canonical pathways such as WNT and transforming growth factor pathway.[70] The important feature of EMT is the loss of membrane E-cadherin in adherens junctions, where the -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. There is evidence that nuclear -catenin can directly transcriptionally activate EMT-associated target genes, such as the E-cadherin gene repressor SLUG (also known as SNAI2).[71]

Recent data have supported the concept, that tumor cells undergoing an EMT could be precursors for metastatic cancer cells, or even metastatic CSCs.[72] In the invasive edge of pancreatic carcinoma a subset of CD133+CXCR4+ (receptor for CXCL12 chemokine also known as a SDF1 ligand) cells has been defined. These cells exhibited significantly stronger migratory activity than their counterpart CD133+CXCR4 cells, but both cell subsets showed similar tumor development capacity.[73] Moreover, inhibition of the CXCR4 receptor led to the reduced metastatic potential without altering tumorigenic capacity.[74]

On the other hand, in the breast cancer CD44+CD24/low cells are detectable in metastatic pleural effusions.[40] By contrast, an increased number of CD24+ cells have been identified in distant metastases in patients with breast cancer.[75] Although, there are only few data on mechanisms mediating metastasis in breast cancer, it is possible that CD44+CD24/low cells initially metastasize and in the new site they change their phenotype and undergo limited differentiation.[76] These findings led to new dynamic two-phase expression pattern concept based on the existence of two forms of cancer stem cells - stationary cancer stem cells (SCS) and mobile cancer stem cells (MCS). SCS are embedded in tissue and persist in differentiated areas throughout all tumor progression. The term MCS describes cells that are located at the tumor-host interface. There is an evidence that these cells are derived from SCS through the acquisition of transient EMT [77] (Fig. 7)

The existence of CSCs has several implications in terms of future cancer treatment and therapies. These include disease identification, selective drug targets, prevention of metastasis, and development of new intervention strategies.

Normal 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 and they also 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 chemotherapeutic agents. The surviving CSCs then repopulate the tumor, causing a relapse.[78] By selectively targeting CSCs, it would be possible to treat patients with aggressive, non-resectable tumors, as well as preventing patients from metastasizing and relapsing.[78] The hypothesis suggests that upon CSC elimination, cancer could regress due to differentiation and/or cell death[citation needed]. What fraction of tumor cells are CSCs and therefore need to be eliminated is not clear yet.[79]

A number of studies have investigated the possibility of identifying specific markers that may distinguish CSCs from the bulk of the tumor (as well as from normal stem cells).[13] Proteomic and genomic signatures of tumors are also being investigated.[80][citation needed]. In 2009, scientists identified one compound, Salinomycin, that selectively reduces the proportion of breast CSCs in mice by more than 100-fold relative to Paclitaxel, a commonly used chemotherapeutic agent.[81] Some types of cancer cells can survive treatment with salinomycin through autophagy,[82] whereby cells use acidic organelles like lysosomes, to degrade and recycle certain types of proteins. The use of autophagy inhibitors can enable killing of cancer stem cells that survive by autophagy.[83]

The cell surface receptor interleukin-3 receptor-alpha (CD123) was shown to be overexpressed on CD34+CD38- leukemic stem cells (LSCs) in acute myelogenous leukemia (AML) but not on normal CD34+CD38- bone marrow cells.[84] Jin et al., then demonstrated that 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.[85]

The design of new drugs for the treatment of CSCs will likely require an understanding of 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. It is now becoming increasingly clear that stem cells of many organs share the same cellular pathways as leukemia-derived HSCs.

Additionally, a normal stem cell may be transformed into a cancer stem cell 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[86] and later shown to specifically regulate HSCs.[87] The role of Bmi-1 has also been illustrated in neural stem cells.[88] The pathway appears to be active in CSCs of pediatric brain tumors.[89]

The Notch pathway has been known to developmental biologists for decades. Its role in control of stem cell proliferation has now been demonstrated for several cell types including hematopoietic, neural and mammary[90] stem cells. Components of the Notch pathway have been proposed to act as oncogenes in mammary[91] and other tumors.

A particular branch of the Notch signaling pathway that involves the transcription factor Hes3 has been shown to regulate a number of cultured cells with cancer stem cell characteristics obtained from glioblastoma patients.[92]

These developmental pathways are also strongly implicated as stem cell regulators.[93] Both Sonic hedgehog (SHH) and Wnt pathways are commonly hyperactivated in tumors and are required to sustain tumor growth. However, the Gli transcription factors that are regulated by SHH take their name from gliomas, where they are commonly expressed at high levels. A degree of crosstalk exists between the two pathways and their activation commonly goes hand-in-hand.[94] This is a trend rather than a rule. For instance, in colon cancer hedgehog signalling appears to antagonise Wnt.[95]

Sonic hedgehog blockers are available, such as cyclopamine. There is also a new water-soluble cyclopamine that may be more effective in cancer treatment. There is also DMAPT, a water-soluble derivative of parthenolide (induces oxidative stress, inhibits NF-B signaling[96]) for AML (leukemia), and possibly myeloma and prostate cancer. A clinical trial of DMAPT is to start in England in late 2007 or 2008[citation needed]. Finally, the enzyme telomerase may qualify as a study subject in CSC physiology.[97] GRN163L (Imetelstat) was recently started in trials to target myeloma stem cells. If it is possible to eliminate the cancer stem cell, then a potential cure may be achieved if there are no more CSCs to repopulate a cancer.

The monolayer of CSCs grown as spheroids showed better growth rate than the MDA-MB 231 cells, which shows the efficacy of 3D spheroid format of growing CSCs. CD44 show increased expression in spheroids compared to 2D culture of MDA-MB 231. ALDH1 a key marker of breast stem cells was highly expressed in BCSCs and MDA-MB 231 grown in 3D, while being absent in CSCs and MDA-MB 231 cells grown in 2D.

The CSCs grown as spheroids showed better growth rate, which showed the efficacy of 3D spheroid format for CSCs culture. Since the association between BCSCs prevalence and clinical outcome and the evidence presented in this study support key roles of CSCs in breast cancer metastasis and drug resistance, it has been proposed that new therapies must target these cells[98]

Read the rest here:
Cancer stem cell - Wikipedia, the free encyclopedia

Read More...

Hepatitis B reactivation in HBsAg-negative/HBcAb-positive …

Sunday, October 11th, 2015

HBsAg-negative/HBcAb-positive haematopoietic stem cell transplant (HSCT) recipients are at high risk of hepatitis B virus (HBV) reactivation. Allogeneic HSCT recipients from years 2000 to 2010 were evaluated in order to study the impact of being HBsAg-negative/HBcAb-positive in this population. Overall, 137 of 764 patients (18%) were HBsAg-negative/HBcAb-positive before HSCT. Overall survival, non-relapse mortality (NRM), acute and chronic graft-vs.-host disease were similar in HBcAb-positive and HBcAb-negative patients. Reactivation occurred in 14 patients (10%) within a median of 19 months after HSCT (range 9-77). Cause-specific hazard for reactivation was decreased in the case of an HBV-immune/exposed donor (HRadjusted = 0.12; 95% CI, 0.02-0.96; p 0.045) and increased in patients who received rituximab treatment (HRadjusted = 2.91; 95%CI, 0.77-10.97; p 0.11). Competing risk analyses documented a protective role of an HBV-immune/exposed donor (p 0.041) and an increased probability associated with the length of treatment with cyclosporine (p <0.001) and treatment with rituximab (but not with low-dose rituximab prophylaxis, p <0.001 at each landmark point). No differences in overall survival and NRM were found between patients with and without HBV reactivation. The donor's immunity was independently and consistently associated with a decreased risk of HBV reactivation, while rituximab and cyclosporine treatments increased the probability.

2014 The Authors Clinical Microbiology and Infection 2014 European Society of Clinical Microbiology and Infectious Diseases.

See the original post here:
Hepatitis B reactivation in HBsAg-negative/HBcAb-positive ...

Read More...

Stem-Kine | The World’s First Clinically Proven Stem Cell …

Sunday, September 13th, 2015

Clinical Trials on Stem-Kine 1. Circulating endothelial progenitor cells: a new approach to anti-aging medicine?

Journal of Translational Medicine 2009, 7:106. Mikirova NA, Jackson JA, Hunninghake R, Kenyon J, Chan KWH, Swindlehurst CA, Minev B, Patel A, Murphy MP, Smith L, Alexandrescu DT, Ichim TE, Riordan NH.

ABSTRACT: Endothelial dysfunction is associated with major causes of morbidity and mortality, as well as numerous age-related conditions. The possibility of preserving or even rejuvenating endothelial function offers a potent means of preventing/treating some of the most fearful aspects of aging such as loss of mental, cardiovascular, and sexual function. Endothelial precursor cells (EPC) provide a continual source of replenishment for damaged or senescent blood vessels. In this review we discuss the biological relevance of circulating EPC in a variety of pathologies in order to build the case that these cells act as an endogenous mechanism of regeneration. Factors controlling EPC mobilization, migration, and function, as well as therapeutic interventions based on mobilization of EPC will be reviewed. We conclude by discussing several clinically-relevant approaches to EPC mobilization and provide preliminary data on a food supplement, Stem-Kine, which enhanced EPC mobilization in human subjects

Link to Full Text

Journal of Translational Medicine 2010, Apr 8;8:34. Mikirova NA, Jackson JA, Hunninghake R, Kenyon J, Chan KW, Swindlehurst CA, Minev B, Patel AN, Murphy MP, Smith L, Ramos F, Ichim TE, Riordan NH.

ABSTRACT: The medical significance of circulating endothelial or hematopoietic progenitors is becoming increasing recognized. While therapeutic augmentation of circulating progenitor cells using G-CSF has resulted in promising preclinical and early clinical data for several degenerative conditions, this approach is limited by cost and inability to perform chronic administration. Stem-Kine is a food supplement that was previously reported to augment circulating EPC in a pilot study. Here we report a trial in 18 healthy volunteers administered Stem-Kine twice daily for a 2 week period. Significant increases in circulating CD133 and CD34 cells were observed at days 1, 2, 7, and 14 subsequent to initiation of administration, which correlated with increased hematopoietic progenitors as detected by the HALO assay

Link to Full Text

Stem-Kine nutritionally increases the release of your bodys own stem cells into your blood stream, where they can be used to help your body heal

Stem cells are your bodys natural healing mechanism. By taking Stem-Kine you are increasing your bodys ability to heal itself quicker and more effectively

Read the original here:
Stem-Kine | The World's First Clinically Proven Stem Cell ...

Read More...

Characterization and Differentiation of Stem Cells …

Monday, August 24th, 2015

Date: 25 Aug 2015

Rent the article at a discount

* Final gross prices may vary according to local VAT.

Circumcision is described as a cultural, medical, and religious process which states surgical removal of the foreskin either partly or fully. Cells isolated from the circumcised tissues are referred as foreskin cells. They have been thought as feeder cell lines for embryonic stem cells. Their fibroblastic properties were also utilized for several experiments. The waste tissues that remain after the circumcision thought to have stem cell properties. Therefore, there have been very few attempts to expose their stem cell properties without turning them into induced pluripotent stem cells. Although stem cell isolation from prepuce and their mesenchymal multilineage differentiation potential have been presented many times in the literature, the current study explored hematopoietical phenotype of newborn foreskin stem cells for the first time. According to the results, human newborn foreskin stem cells (hnFSSCs) were identified by their capability to turn into all three germ layer cell types under in vitro conditions. In addition, these cells have exhibited a stable phenotype and have remained as a monolayer in vitro. hnFSSCs suggested to carry different treatment potentials for bone damages, cartilage problems, nerve damages, lesion formations, and other diseases that are derive from mesodermal, endodermal, and ectodermal origins. Owing to the location of the tissue in the body and differentiation capabilities of hnFSSCs, these cells can be considered as easily obtainable and utilizable even better than the other stem cell sources. In addition, hnFSSCs offers a great potential for tissue engineering approaches due to exhibiting embryonic stem cell-like characteristics, not having any ethical issues, and teratoma induction as in embryonic stem cell applications.

Go here to read the rest:
Characterization and Differentiation of Stem Cells ...

Read More...

Page 19«..10..18192021..»


2024 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick