Author Archives: admin

Top Stem Cell Therapy Clinic in Vail & Denver, Colorado …

Do you have an idea of the natural healing potential that is available in your body?

Read on to find out where your body stores these powerful stem cells.

Adult stem cells are found in the highest concentration in adipose (fat) tissue. In smaller concentrations, they are additionally found in your bone marrow. Beyond what is used for harvesting, stem cells are also found in blood, skin, muscles, and organs.

Adipose tissue provides the largest volume of adult stem cells (1,000 to 2,000 times the number of cells per volume found in bone marrow). Bone marrow provides some stem cells but more importantly provides a large volume of growth factors to aid in the repair process. In addition to adult stem cells, fat tissue also contains numerous other regenerative cells that are important to the healing process.

Stem cells derived from adipose fat tissue have been shown to be a much better source for the repair of cartilage degeneration and recent studies have demonstrated its superior ability to differentiate into cartilage.

There are some myths and misconceptions about stem cells and where the cells come from. Dr. Brandt has dedicated a blog post to the important topic.

Read this article:
Top Stem Cell Therapy Clinic in Vail & Denver, Colorado ...

Stem Cell Therapy – American Regenerative Clinic, Stem …

Stem Cell Therapy employs the bodys own healing potential by isolating stem cells from one location of the body (fat tissue or bone marrow) and relocating them to an area of disease, injury, or inflammation. The main function of stem cells is to maintain and repair tissue. Stem Cell Therapy enhances this natural function by getting stem cells more directly to an area of need within the body. As soon, as stem cells get in there, they will grow to type of cells the body needs to be repaired. Stem cells have the unique ability to form many different types of tissue including bone, cartilage, muscle, etc. They are naturally anti-inflammatory, and can therefore help in the bodys own healing process and potentially reverse the effects of many diseases.

Some of the conditions currently being successfully treated include arthritis, sport related injuries, COPD, CHF, CAD, DM, auto-immune disease such as Lupus Erythematosus and Crohns disease, neurological disorders such as multiple sclerosis and Parkinson disease, spinal cord injuries, autism, critical limb ischemia, and many others.

American Regenerative Clinic will perform outpatient procedures using a process in which we isolate a patients own stem cells from either their own adipose (fat) tissue, or bone marrow. After mini-liposuction or bone marrow harvest from pelvic bone under local anesthesia, the patient tissue will be specially processed. Then immediately Isolated Stem Cells will be delivered to the same patient. Depending on nature of the disease, the cells may be injected into the joint, IV, intrathecal (lumbar puncture), intramuscular, or as a combination of above. The entire process will take just a few hours.

In many old injuries and degenerative diseases, a phenomenon called cellular depletion occurs. Because Prolotherapy and PRP rely on the bodys available repair cells locally, these two methods may produce little results. Prolotherapy starts repair process by mobilizing growth factors in the area via lengthy process of multiple injections. PRP brings them right away, but lacks bricks for repair. More advanced Stem Cell Therapy delivers everything the body needs for promising treatment.

Please read the instructions below carefully.

Discontinue Anti-Inflammatory medication (Advil, Motrin, Ibuprofen, Aleve, and Aspirin) at least 3-4 days before your procedure. Discontinue any blood thinners or any herbs, supplements or vitamins 1-2 weeks before your procedure. Discontinue Systemic Steroids (Prednisone, Hydrocortisone, etc.) 1-2 weeks before your procedure. Steroid Injections (cortisone) should be discontinued at least 1 month before your procedure. Eat a light, healthy breakfast the day of your procedure. Drink plenty of water, especially the day of your procedure.

If you have any questions, you are welcome to call our office at 248-876-4242.

Please read the following steps carefully.

0-3 Days Post Stem Cell Therapy: It is highly recommended that the patient rest the day of the procedure. The next two days the patient should focus on keeping the affected joint(s) immobilized. We strongly encourage you to use connective tissue support vitamins and/or cream. It is imperative that you take this as directed by the doctor; this will enhance the healing process. Ice can be applied to the area of injection for 15-20 minutes, 3-4 times a day, for the first 48 hours. Mild to moderate post-procedure pain can happen. Significant post procedure pain will typically resolve during the first few days after the procedure. If you are experiencing post procedure pain, you can take Tylenol as needed. If necessary, pain medication will be prescribed during your visit. If antibiotics are prescribed, please take them as directed. They will only be prescribed if they are needed. DO NOT TAKE anti-inflammatory medications such as; Advil, Motrin, Ibuprofen, Aleve, and Aspirin, for at least 2 weeks after your procedure. DO NOT TAKE blood thinners or any herbs, supplements or vitamins 3-4 days after your procedure. DO NOT TAKE systemic steroids such as; Prednisone, Hydrocortisone, etc. for at least 2 weeks following your procedure. Do not take hot baths or go to saunas during the first few days following your procedure. Avoid showering for 24 hours following your procedure. Do not consume alcoholic beverages for the first 7 days following your procedure. Avoid smoking. Smoking delays healing and can increase the risk of complications. Drink at least 64 ounces of water daily to help your heal properly. Water does not mean tea, coffee, soda or juice. Wound Care: Keep the area clean, apply a dash antibiotic ointment and a Band-Aid to the post-harvest site. If you see signs of: excessive redness, swelling or experience excessive pain, please call us at 248.876.4242.

3-7 Days Post Stem Cell Therapy: At this point you should gradually start increasing your daily activities and increase your exercise. To maximize the effects of the procedure, proper exercise is necessary. If you are still experiencing pain, continue you can take Tylenol as needed. If prescribed antibiotics please continue to take them as directed. They will only be prescribed if they are needed. Continue to use the connective tissue support vitamins and/or cream to enhance healing. Continue to avoid alcohol for at least 7 days after your procedure. DO NOT TAKE systemic steroids such as; Prednisone, Hydrocortisone, etc. for at least 2 weeks following your procedure. DO NOT TAKE anti-inflammatory medications such as; Advil, Motrin, Ibuprofen, Aleve, and Aspirin, for at least 2 weeks after your procedure.

7 Days Post Stem Cell Therapy: A follow up appointment must be schedule for 1 week (7 days) after your procedure. During the follow up the doctor will assess your healing and determine the next best course of action. If sutures were placed, this will be the time when they will be removed.

1-4 Weeks Post Stem Cell Therapy: Continue to use the connective tissue support vitamins and/or cream to enhance healing. At this point after your procedure, we highly recommend starting physical therapy to aid you in the healing process and help you regain full range of motion to the affected joint(s). DO NOT TAKE systemic steroids such as; Prednisone, Hydrocortisone, etc. for at least 2 weeks following your procedure. DO NOT TAKE anti-inflammatory medications such as; Advil, Motrin, Ibuprofen, Aleve, and Aspirin, for at least 2 weeks after your procedure.

If you have any questions, at any time after your procedure, you are welcome to call our office at 248-876-4242.

Go here to read the rest:
Stem Cell Therapy - American Regenerative Clinic, Stem ...

4. The Adult Stem Cell | stemcells.nih.gov

For many years, researchers have been seeking to understand the body's ability to repair and replace the cells and tissues of some organs, but not others. After years of work pursuing the how and why of seemingly indiscriminant cell repair mechanisms, scientists have now focused their attention on adult stem cells. It has long been known that stem cells are capable of renewing themselves and that they can generate multiple cell types. Today, there is new evidence that stem cells are present in far more tissues and organs than once thought and that these cells are capable of developing into more kinds of cells than previously imagined. Efforts are now underway to harness stem cells and to take advantage of this new found capability, with the goal of devising new and more effective treatments for a host of diseases and disabilities. What lies ahead for the use of adult stem cells is unknown, but it is certain that there are many research questions to be answered and that these answers hold great promise for the future.

Adult stem cells, like all stem cells, share at least two characteristics. First, they can make identical copies of themselves for long periods of time; this ability to proliferate is referred to as long-term self-renewal. Second, they can give rise to mature cell types that have characteristic morphologies (shapes) and specialized functions. Typically, stem cells generate an intermediate cell type or types before they achieve their fully differentiated state. The intermediate cell is called a precursor or progenitor cell. Progenitor or precursor cells in fetal or adult tissues are partly differentiated cells that divide and give rise to differentiated cells. Such cells are usually regarded as "committed" to differentiating along a particular cellular development pathway, although this characteristic may not be as definitive as once thought [82] (see Figure 4.1. Distinguishing Features of Progenitor/Precursor Cells and Stem Cells).

Figure 4.1. Distinguishing Features of Progenitor/Precursor Cells and Stem Cells. A stem cell is an unspecialized cell that is capable of replicating or self renewing itself and developing into specialized cells of a variety of cell types. The product of a stem cell undergoing division is at least one additional stem cell that has the same capabilities of the originating cell. Shown here is an example of a hematopoietic stem cell producing a second generation stem cell and a neuron. A progenitor cell (also known as a precursor cell) is unspecialized or has partial characteristics of a specialized cell that is capable of undergoing cell division and yielding two specialized cells. Shown here is an example of a myeloid progenitor/precursor undergoing cell division to yield two specialized cells (a neutrophil and a red blood cell).

( 2001 Terese Winslow, Lydia Kibiuk)

Adult stem cells are rare. Their primary functions are to maintain the steady state functioning of a cellcalled homeostasisand, with limitations, to replace cells that die because of injury or disease [44, 58]. For example, only an estimated 1 in 10,000 to 15,000 cells in the bone marrow is a hematopoietic (bloodforming) stem cell (HSC) [105]. Furthermore, adult stem cells are dispersed in tissues throughout the mature animal and behave very differently, depending on their local environment. For example, HSCs are constantly being generated in the bone marrow where they differentiate into mature types of blood cells. Indeed, the primary role of HSCs is to replace blood cells [26] (see Chapter 5. Hematopoietic Stem Cells). In contrast, stem cells in the small intestine are stationary, and are physically separated from the mature cell types they generate. Gut epithelial stem cells (or precursors) occur at the bases of cryptsdeep invaginations between the mature, differentiated epithelial cells that line the lumen of the intestine. These epithelial crypt cells divide fairly often, but remain part of the stationary group of cells they generate [93].

Unlike embryonic stem cells, which are defined by their origin (the inner cell mass of the blastocyst), adult stem cells share no such definitive means of characterization. In fact, no one knows the origin of adult stem cells in any mature tissue. Some have proposed that stem cells are somehow set aside during fetal development and restrained from differentiating. Definitions of adult stem cells vary in the scientific literature range from a simple description of the cells to a rigorous set of experimental criteria that must be met before characterizing a particular cell as an adult stem cell. Most of the information about adult stem cells comes from studies of mice. The list of adult tissues reported to contain stem cells is growing and includes bone marrow, peripheral blood, brain, spinal cord, dental pulp, blood vessels, skeletal muscle, epithelia of the skin and digestive system, cornea, retina, liver, and pancreas.

In order to be classified as an adult stem cell, the cell should be capable of self-renewal for the lifetime of the organism. This criterion, although fundamental to the nature of a stem cell, is difficult to prove in vivo. It is nearly impossible, in an organism as complex as a human, to design an experiment that will allow the fate of candidate adult stem cells to be identified in vivo and tracked over an individual's entire lifetime.

Ideally, adult stem cells should also be clonogenic. In other words, a single adult stem cell should be able to generate a line of genetically identical cells, which then gives rise to all the appropriate, differentiated cell types of the tissue in which it resides. Again, this property is difficult to demonstrate in vivo; in practice, scientists show either that a stem cell is clonogenic in vitro, or that a purified population of candidate stem cells can repopulate the tissue.

An adult stem cell should also be able to give rise to fully differentiated cells that have mature phenotypes, are fully integrated into the tissue, and are capable of specialized functions that are appropriate for the tissue. The term phenotype refers to all the observable characteristics of a cell (or organism); its shape (morphology); interactions with other cells and the non-cellular environment (also called the extracellular matrix); proteins that appear on the cell surface (surface markers); and the cell's behavior (e.g., secretion, contraction, synaptic transmission).

The majority of researchers who lay claim to having identified adult stem cells rely on two of these characteristicsappropriate cell morphology, and the demonstration that the resulting, differentiated cell types display surface markers that identify them as belonging to the tissue. Some studies demonstrate that the differentiated cells that are derived from adult stem cells are truly functional, and a few studies show that cells are integrated into the differentiated tissue in vivo and that they interact appropriately with neighboring cells. At present, there is, however, a paucity of research, with a few notable exceptions, in which researchers were able to conduct studies of genetically identical (clonal) stem cells. In order to fully characterize the regenerating and self-renewal capabilities of the adult stem cell, and therefore to truly harness its potential, it will be important to demonstrate that a single adult stem cell can, indeed, generate a line of genetically identical cells, which then gives rise to all the appropriate, differentiated cell types of the tissue in which it resides.

Adult stem cells have been identified in many animal and human tissues. In general, three methods are used to determine whether candidate adult stem cells give rise to specialized cells. Adult stem cells can be labeled in vivo and then they can be tracked. Candidate adult stem cells can also be isolated and labeled and then transplanted back into the organism to determine what becomes of them. Finally, candidate adult stem cells can be isolated, grown in vitro and manipulated, by adding growth factors or introducing genes that help determine what differentiated cells types they will yield. For example, currently, scientists believe that stem cells in the fetal and adult brain divide and give rise to more stem cells or to several types of precursor cells, which give rise to nerve cells (neurons), of which there are many types.

It is often difficultif not impossibleto distinguish adult, tissue-specific stem cells from progenitor cells, which are found in fetal or adult tissues and are partly differentiated cells that divide and give rise to differentiated cells. These are cells found in many organs that are generally thought to be present to replace cells and maintain the integrity of the tissue. Progenitor cells give rise to certain types of cellssuch as the blood cells known as T lymphocytes, B lymphocytes, and natural killer cellsbut are not thought to be capable of developing into all the cell types of a tissue and as such are not truly stem cells. The current wave of excitement over the existence of stem cells in many adult tissues is perhaps fueling claims that progenitor or precursor cells in those tissues are instead stem cells. Thus, there are reports of endothelial progenitor cells, skeletal muscle stem cells, epithelial precursors in the skin and digestive system, as well as some reports of progenitors or stem cells in the pancreas and liver. A detailed summary of some of the evidence for the existence of stem cells in various tissues and organs is presented later in the chapter.

It was not until recently that anyone seriously considered the possibility that stem cells in adult tissues could generate the specialized cell types of another type of tissue from which they normally resideeither a tissue derived from the same embryonic germ layer or from a different germ layer (see Table 1.1. Embryonic Germ Layers From Which Differentiated Tissues Develop). For example, studies have shown that blood stem cells (derived from mesoderm) may be able to generate both skeletal muscle (also derived from mesoderm) and neurons (derived from ectoderm). That realization has been triggered by a flurry of papers reporting that stem cells derived from one adult tissue can change their appearance and assume characteristics that resemble those of differentiated cells from other tissues.

The term plasticity, as used in this report, means that a stem cell from one adult tissue can generate the differentiated cell types of another tissue. At this time, there is no formally accepted name for this phenomenon in the scientific literature. It is variously referred to as "plasticity" [15, 52], "unorthodox differentiation" [10] or "transdifferentiation" [7, 54].

To be able to claim that adult stem cells demonstrate plasticity, it is first important to show that a cell population exists in the starting tissue that has the identifying features of stem cells. Then, it is necessary to show that the adult stem cells give rise to cell types that normally occur in a different tissue. Neither of these criteria is easily met. Simply proving the existence of an adult stem cell population in a differentiated tissue is a laborious process. It requires that the candidate stem cells are shown to be self-renewing, and that they can give rise to the differentiated cell types that are characteristic of that tissue.

To show that the adult stem cells can generate other cell types requires them to be tracked in their new environment, whether it is in vitro or in vivo. In general, this has been accomplished by obtaining the stem cells from a mouse that has been genetically engineered to express a molecular tag in all its cells. It is then necessary to show that the labeled adult stem cells have adopted key structural and biochemical characteristics of the new tissue they are claimed to have generated. Ultimatelyand most importantlyit is necessary to demonstrate that the cells can integrate into their new tissue environment, survive in the tissue, and function like the mature cells of the tissue.

In the experiments reported to date, adult stem cells may assume the characteristics of cells that have developed from the same primary germ layer or a different germ layer (see Figure 4.2. Preliminary Evidence of Plasticity Among Nonhuman Adult Stem Cells). For example, many plasticity experiments involve stem cells derived from bone marrow, which is a mesodermal derivative. The bone marrow stem cells may then differentiate into another mesodermally derived tissue such as skeletal muscle [28, 43], cardiac muscle [51, 71] or liver [4, 54, 97].

Figure 4.2. Preliminary Evidence of Plasticity Among Nonhuman Adult Stem Cells.

( 2001 Terese Winslow, Lydia Kibiuk, Caitlin Duckwall)

Alternatively, adult stem cells may differentiate into a tissue thatduring normal embryonic developmentwould arise from a different germ layer. For example, bone marrow-derived cells may differentiate into neural tissue, which is derived from embryonic ectoderm [15, 65]. Andreciprocallyneural stem cell lines cultured from adult brain tissue may differentiate to form hematopoietic cells [13], or even give rise to many different cell types in a chimeric embryo [17]. In both cases cited above, the cells would be deemed to show plasticity, but in the case of bone marrow stem cells generating brain cells, the finding is less predictable.

In order to study plasticity within and across germ layer lines, the researcher must be sure that he/she is using only one kind of adult stem cell. The vast majority of experiments on plasticity have been conducted with adult stem cells derived either from the bone marrow or the brain. The bone marrow-derived cells are sometimes sortedusing a panel of surface markersinto populations of hematopoietic stem cells or bone marrow stromal cells [46, 54, 71]. The HSCs may be highly purified or partially purified, depending on the conditions used. Another way to separate population of bone marrow cells is by fractionation to yield cells that adhere to a growth substrate (stromal cells) or do not adhere (hematopoietic cells) [28].

To study plasticity of stem cells derived from the brain, the researcher must overcome several problems. Stem cells from the central nervous system (CNS), unlike bone marrow cells, do not occur in a single, accessible location. Instead, they are scattered in three places, at least in rodent brainthe tissue around the lateral ventricles in the forebrain, a migratory pathway for the cells that leads from the ventricles to the olfactory bulbs, and the hippocampus. Many of the experiments with CNS stem cells involve the formation of neurospheres, round aggregates of cells that are sometimes clonally derived. But it is not possible to observe cells in the center of a neurosphere, so to study plasticity in vitro, the cells are usually dissociated and plated in monolayers. To study plasticity in vivo, the cells may be dissociated before injection into the circulatory system of the recipient animal [13], or injected as neurospheres [17].

The differentiated cell types that result from plasticity are usually reported to have the morphological characteristics of the differentiated cells and to display their characteristic surface markers. In reports that transplanted adult stem cells show plasticity in vivo, the stem cells typically are shown to have integrated into a mature host tissue and assumed at least some of its characteristics [15, 28, 51, 65, 71]. Many plasticity experiments involve injury to a particular tissue, which is intended to model a particular human disease or injury [13, 54, 71]. However, there is limited evidence to date that such adult stem cells can generate mature, fully functional cells or that the cells have restored lost function in vivo [54]. Most of the studies that show the plasticity of adult stem cells involve cells that are derived from the bone marrow [15, 28, 54, 65, 77] or brain [13, 17]. To date, adult stem cells are best characterized in these two tissues, which may account for the greater number of plasticity studies based on bone marrow and brain. Collectively, studies on plasticity suggest that stem cell populations in adult mammals are not fixed entities, and that after exposure to a new environment, they may be able to populate other tissues and possibly differentiate into other cell types.

It is not yet possible to say whether plasticity occurs normally in vivo. Some scientists think it may [14, 64], but as yet there is no evidence to prove it. Also, it is not yet clear to what extent plasticity can occur in experimental settings, and howor whetherthe phenomenon can be harnessed to generate tissues that may be useful for therapeutic transplantation. If the phenomenon of plasticity is to be used as a basis for generating tissue for transplantation, the techniques for doing it will need to be reproducible and reliable (see Chapter 10. Assessing Human Stem Cell Safety). In some cases, debate continues about observations that adult stem cells yield cells of tissue types different than those from which they were obtained [7, 68].

More than 30 years ago, Altman and Das showed that two regions of the postnatal rat brain, the hippocampus and the olfactory bulb, contain dividing cells that become neurons [5, 6]. Despite these reports, the prevailing view at the time was that nerve cells in the adult brain do not divide. In fact, the notion that stem cells in the adult brain can generate its three major cell typesastrocytes and oligodendrocytes, as well as neuronswas not accepted until far more recently. Within the past five years, a series of studies has shown that stem cells occur in the adult mammalian brain and that these cells can generate its three major cell lineages [35, 48, 63, 66, 90, 96, 104] (see Chapter 8. Rebuilding the Nervous System with Stem Cells).

Today, scientists believe that stem cells in the fetal and adult brain divide and give rise to more stem cells or to several types of precursor cells. Neuronal precursors (also called neuroblasts) divide and give rise to nerve cells (neurons), of which there are many types. Glial precursors give rise to astrocytes or oligodendrocytes. Astrocytes are a kind of glial cell, which lend both mechanical and metabolic support for neurons; they make up 70 to 80 percent of the cells of the adult brain. Oligodendrocytes make myelin, the fatty material that ensheathes nerve cell axons and speeds nerve transmission. Under normal, in vivo conditions, neuronal precursors do not give rise to glial cells, and glial precursors do not give rise to neurons. In contrast, a fetal or adult CNS (central nervous systemthe brain and spinal cord) stem cell may give rise to neurons, astrocytes, or oligodendrocytes, depending on the signals it receives and its three-dimensional environment within the brain tissue. There is now widespread consensus that the adult mammalian brain does contain stem cells. However, there is no consensus about how many populations of CNS stem cells exist, how they may be related, and how they function in vivo. Because there are no markers currently available to identify the cells in vivo, the only method for testing whether a given population of CNS cells contains stem cells is to isolate the cells and manipulate them in vitro, a process that may change their intrinsic properties [67].

Despite these barriers, three groups of CNS stem cells have been reported to date. All occur in the adult rodent brain and preliminary evidence indicates they also occur in the adult human brain. One group occupies the brain tissue next to the ventricles, regions known as the ventricular zone and the sub-ventricular zone (see discussion below). The ventricles are spaces in the brain filled with cerebrospinal fluid. During fetal development, the tissue adjacent to the ventricles is a prominent region of actively dividing cells. By adulthood, however, this tissue is much smaller, although it still appears to contain stem cells [70].

A second group of adult CNS stem cells, described in mice but not in humans, occurs in a streak of tissue that connects the lateral ventricle and the olfactory bulb, which receives odor signals from the nose. In rodents, olfactory bulb neurons are constantly being replenished via this pathway [59, 61]. A third possible location for stem cells in adult mouse and human brain occurs in the hippocampus, a part of the brain thought to play a role in the formation of certain kinds of memory [27, 34].

Central Nervous System Stem Cells in the Subventricular Zone. CNS stem cells found in the forebrain that surrounds the lateral ventricles are heterogeneous and can be distinguished morphologically. Ependymal cells, which are ciliated, line the ventricles. Adjacent to the ependymal cell layer, in a region sometimes designated as the subependymal or subventricular zone, is a mixed cell population that consists of neuroblasts (immature neurons) that migrate to the olfactory bulb, precursor cells, and astrocytes. Some of the cells divide rapidly, while others divide slowly. The astrocyte-like cells can be identified because they contain glial fibrillary acidic protein (GFAP), whereas the ependymal cells stain positive for nestin, which is regarded as a marker of neural stem cells. Which of these cells best qualifies as a CNS stem cell is a matter of debate [76].

A recent report indicates that the astrocytes that occur in the subventricular zone of the rodent brain act as neural stem cells. The cells with astrocyte markers appear to generate neurons in vivo, as identified by their expression of specific neuronal markers. The in vitro assay to demonstrate that these astrocytes are, in fact, stem cells involves their ability to form neurospheresgroupings of undifferentiated cells that can be dissociated and coaxed to differentiate into neurons or glial cells [25]. Traditionally, these astrocytes have been regarded as differentiated cells, not as stem cells and so their designation as stem cells is not universally accepted.

A series of similar in vitro studies based on the formation of neurospheres was used to identify the subependymal zone as a source of adult rodent CNS stem cells. In these experiments, single, candidate stem cells derived from the subependymal zone are induced to give rise to neurospheres in the presence of mitogenseither epidermal growth factor (EGF) or fibroblast growth factor-2 (FGF-2). The neurospheres are dissociated and passaged. As long as a mitogen is present in the culture medium, the cells continue forming neurospheres without differentiating. Some populations of CNS cells are more responsive to EGF, others to FGF [100]. To induce differentiation into neurons or glia, cells are dissociated from the neurospheres and grown on an adherent surface in serum-free medium that contains specific growth factors. Collectively, the studies demonstrate that a population of cells derived from the adult rodent brain can self-renew and differentiate to yield the three major cell types of the CNS cells [41, 69, 74, 102].

Central Nervous System Stem Cells in the Ventricular Zone. Another group of potential CNS stem cells in the adult rodent brain may consist of the ependymal cells themselves [47]. Ependymal cells, which are ciliated, line the lateral ventricles. They have been described as non-dividing cells [24] that function as part of the blood-brain barrier [22]. The suggestion that ependymal cells from the ventricular zone of the adult rodent CNS may be stem cells is therefore unexpected. However, in a recent study, in which two molecular tagsthe fluorescent marker Dil, and an adenovirus vector carrying lacZ tagswere used to label the ependymal cells that line the entire CNS ventricular system of adult rats, it was shown that these cells could, indeed, act as stem cells. A few days after labeling, fluorescent or lacZ+ cells were observed in the rostral migratory stream (which leads from the lateral ventricle to the olfactory bulb), and then in the olfactory bulb itself. The labeled cells in the olfactory bulb also stained for the neuronal markers III tubulin and Map2, which indicated that ependymal cells from the ventricular zone of the adult rat brain had migrated along the rostral migratory stream to generate olfactory bulb neurons in vivo [47].

To show that Dil+ cells were neural stem cells and could generate astrocytes and oligodendrocytes as well as neurons, a neurosphere assay was performed in vitro. Dil-labeled cells were dissociated from the ventricular system and cultured in the presence of mitogen to generate neurospheres. Most of the neurospheres were Dil+; they could self-renew and generate neurons, astrocytes, and oligodendrocytes when induced to differentiate. Single, Dil+ ependymal cells isolated from the ventricular zone could also generate self-renewing neurospheres and differentiate into neurons and glia.

To show that ependymal cells can also divide in vivo, bromodeoxyuridine (BrdU) was administered in the drinking water to rats for a 2- to 6-week period. Bromodeoxyuridine (BrdU) is a DNA precursor that is only incorporated into dividing cells. Through a series of experiments, it was shown that ependymal cells divide slowly in vivo and give rise to a population of progenitor cells in the subventricular zone [47]. A different pattern of scattered BrdU-labeled cells was observed in the spinal cord, which suggested that ependymal cells along the central canal of the cord occasionally divide and give rise to nearby ependymal cells, but do not migrate away from the canal.

Collectively, the data suggest that CNS ependymal cells in adult rodents can function as stem cells. The cells can self-renew, and most proliferate via asymmetrical division. Many of the CNS ependymal cells are not actively dividing (quiescent), but they can be stimulated to do so in vitro (with mitogens) or in vivo (in response to injury). After injury, the ependymal cells in the spinal cord only give rise to astrocytes, not to neurons. How and whether ependymal cells from the ventricular zone are related to other candidate populations of CNS stem cells, such as those identified in the hippocampus [34], is not known.

Are ventricular and subventricular zone CNS stem cells the same population? These studies and other leave open the question of whether cells that directly line the ventriclesthose in the ventricular zoneor cells that are at least a layer removed from this zonein the subventricular zone are the same population of CNS stem cells. A new study, based on the finding that they express different genes, confirms earlier reports that the ventricular and subventricular zone cell populations are distinct. The new research utilizes a technique called representational difference analysis, together with cDNA microarray analysis, to monitor the patterns of gene expression in the complex tissue of the developing and postnatal mouse brain. The study revealed the expression of a panel of genes known to be important in CNS development, such as L3-PSP (which encodes a phosphoserine phosphatase important in cell signaling), cyclin D2 (a cell cycle gene), and ERCC-1 (which is important in DNA excision repair). All of these genes in the recent study were expressed in cultured neurospheres, as well as the ventricular zone, the subventricular zone, and a brain area outside those germinal zones. This analysis also revealed the expression of novel genes such as A16F10, which is similar to a gene in an embryonic cancer cell line. A16F10 was expressed in neurospheres and at high levels in the subventricular zone, but not significantly in the ventricular zone. Interestingly, several of the genes identified in cultured neurospheres were also expressed in hematopoietic cells, suggesting that neural stem cells and blood-forming cells may share aspects of their genetic programs or signaling systems [38]. This finding may help explain recent reports that CNS stem cells derived from mouse brain can give rise to hematopoietic cells after injection into irradiated mice [13].

Central Nervous System Stem Cells in the Hippocampus. The hippocampus is one of the oldest parts of the cerebral cortex, in evolutionary terms, and is thought to play an important role in certain forms of memory. The region of the hippocampus in which stem cells apparently exist in mouse and human brains is the subgranular zone of the dentate gyrus. In mice, when BrdU is used to label dividing cells in this region, about 50% of the labeled cells differentiate into cells that appear to be dentate gyrus granule neurons, and 15% become glial cells. The rest of the BrdU-labeled cells do not have a recognizable phenotype [90]. Interestingly, many, if not all the BrdU-labeled cells in the adult rodent hippocampus occur next to blood vessels [33].

In the human dentate gyrus, some BrdU-labeled cells express NeuN, neuron-specific enolase, or calbindin, all of which are neuronal markers. The labeled neuron-like cells resemble dentate gyrus granule cells, in terms of their morphology (as they did in mice). Other BrdU-labeled cells express glial fibrillary acidic protein (GFAP) an astrocyte marker. The study involved autopsy material, obtained with family consent, from five cancer patients who had been injected with BrdU dissolved in saline prior to their death for diagnostic purposes. The patients ranged in age from 57 to 72 years. The greatest number of BrdU-labeled cells were identified in the oldest patient, suggesting that new neuron formation in the hippocampus can continue late in life [27].

Fetal Central Nervous System Stem Cells. Not surprisingly, fetal stem cells are numerous in fetal tissues, where they are assumed to play an important role in the expansion and differentiation of all tissues of the developing organism. Depending on the developmental stage of an animal, fetal stem cells and precursor cellswhich arise from stem cellsmay make up the bulk of a tissue. This is certainly true in the brain [48], although it has not been demonstrated experimentally in many tissues.

It may seem obvious that the fetal brain contains stem cells that can generate all the types of neurons in the brain as well as astrocytes and oligodendrocytes, but it was not until fairly recently that the concept was proven experimentally. There has been a long-standing question as to whether or not the same cell type gives rise to both neurons and glia. In studies of the developing rodent brain, it has now been shown that all the major cell types in the fetal brain arise from a common population of progenitor cells [20, 34, 48, 80, 108].

Neural stem cells in the mammalian fetal brain are concentrated in seven major areas: olfactory bulb, ependymal (ventricular) zone of the lateral ventricles (which lie in the forebrain), subventricular zone (next to the ependymal zone), hippocampus, spinal cord, cerebellum (part of the hindbrain), and the cerebral cortex. Their number and pattern of development vary in different species. These cells appear to represent different stem cell populations, rather than a single population of stem cells that is dispersed in multiple sites. The normal development of the brain depends not only on the proliferation and differentiation of these fetal stem cells, but also on a genetically programmed process of selective cell death called apoptosis [76].

Little is known about stem cells in the human fetal brain. In one study, however, investigators derived clonal cell lines from CNS stem cells isolated from the diencephalon and cortex of human fetuses, 10.5 weeks post-conception [103]. The study is unusual, not only because it involves human CNS stem cells obtained from fetal tissue, but also because the cells were used to generate clonal cell lines of CNS stem cells that generated neurons, astrocytes, and oligodendrocytes, as determined on the basis of expressed markers. In a few experiments described as "preliminary," the human CNS stem cells were injected into the brains of immunosuppressed rats where they apparently differentiated into neuron-like cells or glial cells.

In a 1999 study, a serum-free growth medium that included EGF and FGF2 was devised to grow the human fetal CNS stem cells. Although most of the cells died, occasionally, single CNS stem cells survived, divided, and ultimately formed neurospheres after one to two weeks in culture. The neurospheres could be dissociated and individual cells replated. The cells resumed proliferation and formed new neurospheres, thus establishing an in vitro system that (like the system established for mouse CNS neurospheres) could be maintained up to 2 years. Depending on the culture conditions, the cells in the neurospheres could be maintained in an undifferentiated dividing state (in the presence of mitogen), or dissociated and induced to differentiate (after the removal of mitogen and the addition of specific growth factors to the culture medium). The differentiated cells consisted mostly of astrocytes (75%), some neurons (13%) and rare oligodendrocytes (1.2%). The neurons generated under these conditions expressed markers indicating they were GABAergic, [the major type of inhibitory neuron in the mammalian CNS responsive to the amino acid neurotransmitter, gammaaminobutyric acid (GABA)]. However, catecholamine-like cells that express tyrosine hydroxylase (TH, a critical enzyme in the dopamine-synthesis pathway) could be generated, if the culture conditions were altered to include different medium conditioned by a rat glioma line (BB49). Thus, the report indicates that human CNS stem cells obtained from early fetuses can be maintained in vitro for a long time without differentiating, induced to differentiate into the three major lineages of the CNS (and possibly two kinds of neurons, GABAergic and TH-positive), and engraft (in rats) in vivo [103].

Central Nervous System Neural Crest Stem Cells. Neural crest cells differ markedly from fetal or adult neural stem cells. During fetal development, neural crest cells migrate from the sides of the neural tube as it closes. The cells differentiate into a range of tissues, not all of which are part of the nervous system [56, 57, 91]. Neural crest cells form the sympathetic and parasympathetic components of the peripheral nervous system (PNS), including the network of nerves that innervate the heart and the gut, all the sensory ganglia (groups of neurons that occur in pairs along the dorsal surface of the spinal cord), and Schwann cells, which (like oligodendrocytes in the CNS) make myelin in the PNS. The non-neural tissues that arise from the neural crest are diverse. They populate certain hormone-secreting glandsincluding the adrenal medulla and Type I cells in the carotid bodypigment cells of the skin (melanocytes), cartilage and bone in the face and skull, and connective tissue in many parts of the body [76].

Thus, neural crest cells migrate far more extensively than other fetal neural stem cells during development, form mesenchymal tissues, most of which develop from embryonic mesoderm as well as the components of the CNS and PNS which arises from embryonic ectoderm. This close link, in neural crest development, between ectodermally derived tissues and mesodermally derived tissues accounts in part for the interest in neural crest cells as a kind of stem cell. In fact, neural crest cells meet several criteria of stem cells. They can self-renew (at least in the fetus) and can differentiate into multiple cells types, which include cells derived from two of the three embryonic germ layers [76].

Recent studies indicate that neural crest cells persist late into gestation and can be isolated from E14.5 rat sciatic nerve, a peripheral nerve in the hindlimb. The cells incorporate BrdU, indicating that they are dividing in vivo. When transplanted into chick embryos, the rat neural crest cells develop into neurons and glia, an indication of their stem cell-like properties [67]. However, the ability of rat E14.5 neural crest cells taken from sciatic nerve to generate nerve and glial cells in chick is more limited than neural crest cells derived from younger, E10.5 rat embryos. At the earlier stage of development, the neural tube has formed, but neural crest cells have not yet migrated to their final destinations. Neural crest cells from early developmental stages are more sensitive to bone morphogenetic protein 2 (BMP2) signaling, which may help explain their greater differentiation potential [106].

The notion that the bone marrow contains stem cells is not new. One population of bone marrow cells, the hematopoietic stem cells (HSCs), is responsible for forming all of the types of blood cells in the body. HSCs were recognized as a stem cells more than 40 years ago [9, 99]. Bone marrow stromal cellsa mixed cell population that generates bone, cartilage, fat, fibrous connective tissue, and the reticular network that supports blood cell formationwere described shortly after the discovery of HSCs [30, 32, 73]. The mesenchymal stem cells of the bone marrow also give rise to these tissues, and may constitute the same population of cells as the bone marrow stromal cells [78]. Recently, a population of progenitor cells that differentiates into endothelial cells, a type of cell that lines the blood vessels, was isolated from circulating blood [8] and identified as originating in bone marrow [89]. Whether these endothelial progenitor cells, which resemble the angioblasts that give rise to blood vessels during embryonic development, represent a bona fide population of adult bone marrow stem cells remains uncertain. Thus, the bone marrow appears to contain three stem cell populationshematopoietic stem cells, stromal cells, and (possibly) endothelial progenitor cells (see Figure 4.3. Hematopoietic and Stromal Stem Cell Differentiation).

Figure 4.3. Hematopoietic and Stromal Stem Cell Differentiation.

( 2001 Terese Winslow, Lydia Kibiuk)

Two more apparent stem cell types have been reported in circulating blood, but have not been shown to originate from the bone marrow. One population, called pericytes, may be closely related to bone marrow stromal cells, although their origin remains elusive [12]. The second population of blood-born stem cells, which occur in four species of animals testedguinea pigs, mice, rabbits, and humansresemble stromal cells in that they can generate bone and fat [53].

Hematopoietic Stem Cells. Of all the cell types in the body, those that survive for the shortest period of time are blood cells and certain kinds of epithelial cells. For example, red blood cells (erythrocytes), which lack a nucleus, live for approximately 120 days in the bloodstream. The life of an animal literally depends on the ability of these and other blood cells to be replenished continuously. This replenishment process occurs largely in the bone marrow, where HSCs reside, divide, and differentiate into all the blood cell types. Both HSCs and differentiated blood cells cycle from the bone marrow to the blood and back again, under the influence of a barrage of secreted factors that regulate cell proliferation, differentiation, and migration (see Chapter 5. Hematopoietic Stem Cells).

HSCs can reconstitute the hematopoietic system of mice that have been subjected to lethal doses of radiation to destroy their own hematopoietic systems. This test, the rescue of lethally irradiated mice, has become a standard by which other candidate stem cells are measured because it shows, without question, that HSCs can regenerate an entire tissue systemin this case, the blood [9, 99]. HSCs were first proven to be blood-forming stem cells in a series of experiments in mice; similar blood-forming stem cells occur in humans. HSCs are defined by their ability to self-renew and to give rise to all the kinds of blood cells in the body. This means that a single HSC is capable of regenerating the entire hematopoietic system, although this has been demonstrated only a few times in mice [72].

Over the years, many combinations of surface markers have been used to identify, isolate, and purify HSCs derived from bone marrow and blood. Undifferentiated HSCs and hematopoietic progenitor cells express c-kit, CD34, and H-2K. These cells usually lack the lineage marker Lin, or express it at very low levels (Lin-/low). And for transplant purposes, cells that are CD34+ Thy1+ Lin- are most likely to contain stem cells and result in engraftment.

Two kinds of HSCs have been defined. Long-term HSCs proliferate for the lifetime of an animal. In young adult mice, an estimated 8 to 10 % of long-term HSCs enter the cell cycle and divide each day. Short-term HSCs proliferate for a limited time, possibly a few months. Long-term HSCs have high levels of telomerase activity. Telomerase is an enzyme that helps maintain the length of the ends of chromosomes, called telomeres, by adding on nucleotides. Active telomerase is a characteristic of undifferentiated, dividing cells and cancer cells. Differentiated, human somatic cells do not show telomerase activity. In adult humans, HSCs occur in the bone marrow, blood, liver, and spleen, but are extremely rare in any of these tissues. In mice, only 1 in 10,000 to 15,000 bone marrow cells is a long-term HSC [105].

Short-term HSCs differentiate into lymphoid and myeloid precursors, the two classes of precursors for the two major lineages of blood cells. Lymphoid precursors differentiate into T cells, B cells, and natural killer cells. The mechanisms and pathways that lead to their differentiation are still being investigated [1, 2]. Myeloid precursors differentiate into monocytes and macrophages, neutrophils, eosinophils, basophils, megakaryocytes, and erythrocytes [3]. In vivo, bone marrow HSCs differentiate into mature, specialized blood cells that cycle constantly from the bone marrow to the blood, and back to the bone marrow [26]. A recent study showed that short-term HSCs are a heterogeneous population that differ significantly in terms of their ability to self-renew and repopulate the hematopoietic system [42].

Attempts to induce HSC to proliferate in vitroon many substrates, including those intended to mimic conditions in the stromahave frustrated scientists for many years. Although HSCs proliferate readily in vivo, they usually differentiate or die in vitro [26]. Thus, much of the research on HSCs has been focused on understanding the factors, cell-cell interactions, and cell-matrix interactions that control their proliferation and differentiation in vivo, with the hope that similar conditions could be replicated in vitro. Many of the soluble factors that regulate HSC differentiation in vivo are cytokines, which are made by different cell types and are then concentrated in the bone marrow by the extracellular matrix of stromal cellsthe sites of blood formation [45, 107]. Two of the most-studied cytokines are granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-3 (IL-3) [40, 81].

Also important to HSC proliferation and differentiation are interactions of the cells with adhesion molecules in the extracellular matrix of the bone marrow stroma [83, 101, 110].

Bone Marrow Stromal Cells. Bone marrow (BM) stromal cells have long been recognized for playing an important role in the differentiation of mature blood cells from HSCs (see Figure 4.3. Hematopoietic and Stromal Stem Cell Differentiation). But stromal cells also have other important functions [30, 31]. In addition to providing the physical environment in which HSCs differentiate, BM stromal cells generate cartilage, bone, and fat. Whether stromal cells are best classified as stem cells or progenitor cells for these tissues is still in question. There is also a question as to whether BM stromal cells and so-called mesenchymal stem cells are the same population [78].

BM stromal cells have many features that distinguish them from HSCs. The two cell types are easy to separate in vitro. When bone marrow is dissociated, and the mixture of cells it contains is plated at low density, the stromal cells adhere to the surface of the culture dish, and the HSCs do not. Given specific in vitro conditions, BM stromal cells form colonies from a single cell called the colony forming unit-F (CFU-F). These colonies may then differentiate as adipocytes or myelosupportive stroma, a clonal assay that indicates the stem cell-like nature of stromal cells. Unlike HSCs, which do not divide in vitro (or proliferate only to a limited extent), BM stromal cells can proliferate for up to 35 population doublings in vitro [16]. They grow rapidly under the influence of such mitogens as platelet-derived growth factor (PDGF), epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), and insulin-like growth factor-1 (IGF-1) [12].

To date, it has not been possible to isolate a population of pure stromal cells from bone marrow. Panels of markers used to identify the cells include receptors for certain cytokines (interleukin-1, 3, 4, 6, and 7) receptors for proteins in the extracellular matrix, (ICAM-1 and 2, VCAM-1, the alpha-1, 2, and 3 integrins, and the beta-1, 2, 3 and 4 integrins), etc. [64]. Despite the use of these markers and another stromal cell marker called Stro-1, the origin and specific identity of stromal cells have remained elusive. Like HSCs, BM stromal cells arise from embryonic mesoderm during development, although no specific precursor or stem cell for stromal cells has been isolated and identified. One theory about their origin is that a common kind of progenitor cellperhaps a primordial endothelial cell that lines embryonic blood vesselsgives rise to both HSCs and to mesodermal precursors. The latter may then differentiate into myogenic precursors (the satellite cells that are thought to function as stem cells in skeletal muscle), and the BM stromal cells [10].

In vivo, the differentiation of stromal cells into fat and bone is not straightforward. Bone marrow adipocytes and myelosupportive stromal cellsboth of which are derived from BM stromal cellsmay be regarded as interchangeable phenotypes [10, 11]. Adipocytes do not develop until postnatal life, as the bones enlarge and the marrow space increases to accommodate enhanced hematopoiesis. When the skeleton stops growing, and the mass of HSCs decreases in a normal, age-dependent fashion, BM stromal cells differentiate into adipocytes, which fill the extra space. New bone formation is obviously greater during skeletal growth, although bone "turns over" throughout life. Bone forming cells are osteoblasts, but their relationship to BM stromal cells is not clear. New trabecular bone, which is the inner region of bone next to the marrow, could logically develop from the action of BM stromal cells. But the outside surface of bone also turns over, as does bone next to the Haversian system (small canals that form concentric rings within bone). And neither of these surfaces is in contact with BM stromal cells [10, 11].

It is often difficultif not impossibleto distinguish adult, tissue-specific stem cells from progenitor cells. With that caveat in mind, the following summary identifies reports of stem cells in various adult tissues.

Endothelial Progenitor Cells. Endothelial cells line the inner surfaces of blood vessels throughout the body, and it has been difficult to identify specific endothelial stem cells in either the embryonic or the adult mammal. During embryonic development, just after gastrulation, a kind of cell called the hemangioblast, which is derived from mesoderm, is presumed to be the precursor of both the hematopoietic and endothelial cell lineages. The embryonic vasculature formed at this stage is transient and consists of blood islands in the yolk sac. But hemangioblasts, per se, have not been isolated from the embryo and their existence remains in question. The process of forming new blood vessels in the embryo is called vasculogenesis. In the adult, the process of forming blood vessels from pre-existing blood vessels is called angiogenesis [50].

Evidence that hemangioblasts do exist comes from studies of mouse embryonic stem cells that are directed to differentiate in vitro. These studies have shown that a precursor cell derived from mouse ES cells that express Flk-1 [the receptor for vascular endothelial growth factor (VEGF) in mice] can give rise to both blood cells and blood vessel cells [88, 109]. Both VEGF and fibroblast growth factor-2 (FGF-2) play critical roles in endothelial cell differentiation in vivo [79].

Several recent reports indicate that the bone marrow contains cells that can give rise to new blood vessels in tissues that are ischemic (damaged due to the deprivation of blood and oxygen) [8, 29, 49, 94]. But it is unclear from these studies what cell type(s) in the bone marrow induced angiogenesis. In a study which sought to address that question, researchers found that adult human bone marrow contains cells that resemble embryonic hemangioblasts, and may therefore be called endothelial stem cells.

In more recent experiments, human bone marrow-derived cells were injected into the tail veins of rats with induced cardiac ischemia. The human cells migrated to the rat heart where they generated new blood vessels in the infarcted muscle (a process akin to vasculogenesis), and also induced angiogenesis. The candidate endothelial stem cells are CD34+(a marker for HSCs), and they express the transcription factor GATA-2 [51]. A similar study using transgenic mice that express the gene for enhanced green fluorescent protein (which allows the cells to be tracked), showed that bone-marrow-derived cells could repopulate an area of infarcted heart muscle in mice, and generate not only blood vessels, but also cardiomyocytes that integrated into the host tissue [71] (see Chapter 9. Can Stem Cells Repair a Damaged Heart?).

And, in a series of experiments in adult mammals, progenitor endothelial cells were isolated from peripheral blood (of mice and humans) by using antibodies against CD34 and Flk-1, the receptor for VEGF. The cells were mononuclear blood cells (meaning they have a nucleus) and are referred to as MBCD34+ cells and MBFlk1+ cells. When plated in tissue-culture dishes, the cells attached to the substrate, became spindle-shaped, and formed tube-like structures that resemble blood vessels. When transplanted into mice of the same species (autologous transplants) with induced ischemia in one limb, the MBCD34+ cells promoted the formation of new blood vessels [8]. Although the adult MBCD34+ and MBFlk1+ cells function in some ways like stem cells, they are usually regarded as progenitor cells.

Skeletal Muscle Stem Cells. Skeletal muscle, like the cardiac muscle of the heart and the smooth muscle in the walls of blood vessels, the digestive system, and the respiratory system, is derived from embryonic mesoderm. To date, at least three populations of skeletal muscle stem cells have been identified: satellite cells, cells in the wall of the dorsal aorta, and so-called "side population" cells.

Satellite cells in skeletal muscle were identified 40 years ago in frogs by electron microscopy [62], and thereafter in mammals [84]. Satellite cells occur on the surface of the basal lamina of a mature muscle cell, or myofiber. In adult mammals, satellite cells mediate muscle growth [85]. Although satellite cells are normally non-dividing, they can be triggered to proliferate as a result of injury, or weight-bearing exercise. Under either of these circumstances, muscle satellite cells give rise to myogenic precursor cells, which then differentiate into the myofibrils that typify skeletal muscle. A group of transcription factors called myogenic regulatory factors (MRFs) play important roles in these differentiation events. The so-called primary MRFs, MyoD and Myf5, help regulate myoblast formation during embryogenesis. The secondary MRFs, myogenin and MRF4, regulate the terminal differentiation of myofibrils [86].

With regard to satellite cells, scientists have been addressing two questions. Are skeletal muscle satellite cells true adult stem cells or are they instead precursor cells? Are satellite cells the only cell type that can regenerate skeletal muscle. For example, a recent report indicates that muscle stem cells may also occur in the dorsal aorta of mouse embryos, and constitute a cell type that gives rise both to muscle satellite cells and endothelial cells. Whether the dorsal aorta cells meet the criteria of a self-renewing muscle stem cell is a matter of debate [21].

Another report indicates that a different kind of stem cell, called an SP cell, can also regenerate skeletal muscle may be present in muscle and bone marrow. SP stands for a side population of cells that can be separated by fluorescence-activated cell sorting analysis. Intravenously injecting these muscle-derived stem cells restored the expression of dystrophin in mdx mice. Dystrophin is the protein that is defective in people with Duchenne's muscular dystrophy; mdx mice provide a model for the human disease. Dystrophin expression in the SP cell-treated mice was lower than would be needed for clinical benefit. Injection of bone marrow- or muscle-derived SP cells into the dystrophic muscle of the mice yielded equivocal results that the transplanted cells had integrated into the host tissue. The authors conclude that a similar population of SP stem cells can be derived from either adult mouse bone marrow or skeletal muscle, and suggest "there may be some direct relationship between bone marrow-derived stem cells and other tissue- or organ-specific cells" [43]. Thus, stem cell or progenitor cell types from various mesodermally-derived tissues may be able to generate skeletal muscle.

Epithelial Cell Precursors in the Skin and Digestive System. Epithelial cells, which constitute 60 percent of the differentiated cells in the body are responsible for covering the internal and external surfaces of the body, including the lining of vessels and other cavities. The epithelial cells in skin and the digestive tract are replaced constantly. Other epithelial cell populationsin the ducts of the liver or pancreas, for exampleturn over more slowly. The cell population that renews the epithelium of the small intestine occurs in the intestinal crypts, deep invaginations in the lining of the gut. The crypt cells are often regarded as stem cells; one of them can give rise to an organized cluster of cells called a structural-proliferative unit [93].

The skin of mammals contains at least three populations of epithelial cells: epidermal cells, hair follicle cells, and glandular epithelial cells, such as those that make up the sweat glands. The replacement patterns for epithelial cells in these three compartments differ, and in all the compartments, a stem cell population has been postulated. For example, stem cells in the bulge region of the hair follicle appear to give rise to multiple cell types. Their progeny can migrate down to the base of the follicle where they become matrix cells, which may then give rise to different cell types in the hair follicle, of which there are seven [39]. The bulge stem cells of the follicle may also give rise to the epidermis of the skin [95].

Another population of stem cells in skin occurs in the basal layer of the epidermis. These stem cells proliferate in the basal region, and then differentiate as they move toward the outer surface of the skin. The keratinocytes in the outermost layer lack nuclei and act as a protective barrier. A dividing skin stem cell can divide asymmetrically to produce two kinds of daughter cells. One is another self-renewing stem cell. The second kind of daughter cell is an intermediate precursor cell which is then committed to replicate a few times before differentiating into keratinocytes. Self-renewing stem cells can be distinguished from this intermediate precusor cell by their higher level of 1 integrin expression, which signals keratinocytes to proliferate via a mitogen-activated protein (MAP) kinase [112]. Other signaling pathways include that triggered by -catenin, which helps maintain the stem-cell state [111], and the pathway regulated by the oncoprotein c-Myc, which triggers stem cells to give rise to transit amplifying cells [36].

Stem Cells in the Pancreas and Liver. The status of stem cells in the adult pancreas and liver is unclear. During embryonic development, both tissues arise from endoderm. A recent study indicates that a single precursor cell derived from embryonic endoderm may generate both the ventral pancreas and the liver [23]. In adult mammals, however, both the pancreas and the liver contain multiple kinds of differentiated cells that may be repopulated or regenerated by multiple types of stem cells. In the pancreas, endocrine (hormone-producing) cells occur in the islets of Langerhans. They include the beta cells (which produce insulin), the alpha cells (which secrete glucagon), and cells that release the peptide hormones somatostatin and pancreatic polypeptide. Stem cells in the adult pancreas are postulated to occur in the pancreatic ducts or in the islets themselves. Several recent reports indicate that stem cells that express nestinwhich is usually regarded as a marker of neural stem cellscan generate all of the cell types in the islets [60, 113] (see Chapter 7. Stem Cells and Diabetes).

The identity of stem cells that can repopulate the liver of adult mammals is also in question. Recent studies in rodents indicate that HSCs (derived from mesoderm) may be able to home to liver after it is damaged, and demonstrate plasticity in becoming into hepatocytes (usually derived from endoderm) [54, 77, 97]. But the question remains as to whether cells from the bone marrow normally generate hepatocytes in vivo. It is not known whether this kind of plasticity occurs without severe damage to the liver or whether HSCs from the bone marrow generate oval cells of the liver [18]. Although hepatic oval cells exist in the liver, it is not clear whether they actually generate new hepatocytes [87, 98]. Oval cells may arise from the portal tracts in liver and may give rise to either hepatocytes [19, 55] and to the epithelium of the bile ducts [37, 92]. Indeed, hepatocytes themselves, may be responsible for the well-know regenerative capacity of liver.

Chapter 3|Table of Contents|Chapter 5

Historical content: June 17, 2001

View original post here:
4. The Adult Stem Cell | stemcells.nih.gov

Human skin cells transformed directly into motor neurons – Washington University School of Medicine in St. Louis

Visit the News Hub

New technique could aid treatments for diseases that lead to paralysis

Scientists have discovered a new way to convert human skin cells directly into motor neurons (above). The technique, developed at Washington University School of Medicine in St. Louis, could help researchers better understand diseases of motor neurons, such as amyotrophic lateral sclerosis. Human motor neurons are difficult to study since they can't be taken from living patients. The motor neurons pictured were converted from skin cells sampled from a healthy 42-year-old woman.

Scientists working to develop new treatments for neurodegenerative diseases have been stymied by the inability to grow human motor neurons in the lab. Motor neurons drive muscle contractions, and their damage underlies devastating diseases such as amyotrophic lateral sclerosis and spinal muscular atrophy, both of which ultimately lead to paralysis and early death.

In new research, scientists at Washington University School of Medicine in St. Louis have converted skin cells from healthy adults directly into motor neurons without going through a stem cell state.

The technique makes it possible to study motor neurons of the human central nervous system in the lab. Unlike commonly studied mouse motor neurons, human motor neurons growing in the lab would be a new tool since researchers cant take samples of these neurons from living people but can easily take skin samples.

The study is published Sept. 7 in the journal Cell Stem Cell.

Avoiding the stem cell phase eliminates ethical concerns raised when producing what are called pluripotent stem cells, which are similar to embryonic stem cells in their ability to become all adult cell types. And importantly, avoiding a stem cell state allows the resulting motor neurons to retain the age of the original skin cells and, therefore, the age of the patient. Maintaining the chronological age of these cells is vital when studying neurodegenerative diseases that develop in people at different ages and worsen over decades.

In this study, we only used skin cells from healthy adults ranging in age from early 20s to late 60s, said senior author Andrew S. Yoo, PhD, an assistant professor of developmental biology. Our research revealed how small RNA molecules can work with other cell signals called transcription factors to generate specific types of neurons, in this case motor neurons. In the future, we would like to study skin cells from patients with disorders of motor neurons. Our conversion process should model late-onset aspects of the disease using neurons derived from patients with the condition.

Going back through a pluripotent stem cell phase is a bit like demolishing a house and building a new one from the ground up, Yoo said. What were doing is more like renovation. We change the interior but leave the original structure, which retains the characteristics of the aging adult neurons that we want to study.

The ability of scientists to convert human skin cells into other cell types, such as neurons, has the potential to enhance understanding of disease and lead to finding new ways to heal damaged tissues and organs, a field called regenerative medicine.

Human skin cells (above) sampled from a healthy adult and then converted into different types of neurons have the potential to be a valuable research tool. Similar skin samples from patients with neurodegenerative diseases could allow scientists to study the disease in its native cell type.

To convert skin cells into motor neurons, the researchers exposed the skin cells to molecular signals that are usually present at high levels in the brain. Past work by Yoo and his colleagues then at Stanford University showed that exposure to two short snippets of RNA turned human skin cells into neurons. These two microRNAs called miR-9 and miR-124 are involved with repackaging the genetic instructions of the cell.

In the new study, the researchers extensively characterized this repackaging process, detailing how skin cells reprogrammed into generic neurons then can be guided into specific types of neurons. They found that genes involved in this process become poised for expression but remain inactive until the correct combination of molecules is provided. After much experimentation with multiple combinations, the researchers found that adding two more signals to the mix transcription factors called ISL1 and LHX3 turned the skin cells into spinal cord motor neurons in about 30 days.

The combination of signals microRNAs miR-9 and miR-124 plus transcription factors ISL1 and LHX3 tells the cell to fold up the genetic instructions for making skin and unfurl the instructions for making motor neurons, according to Yoo and the studys co-first authors, Daniel G. Abernathy and Matthew J. McCoy, doctoral students in Yoos lab; and Woo Kyung Kim, PhD, a postdoctoral research associate.

Another past study from Yoos team showed that exposure to the same two microRNAs, miR-9 and miR-124, plus a different mix of transcription factors could turn skin cells into a different type of neuron. In that case, the skin cells became striatal medium spiny neurons, which are affected in Huntingtons disease an inherited, eventually fatal genetic disorder that causes involuntary muscle movements and cognitive decline beginning in middle adulthood.

In the new study, the researchers said the converted motor neurons compared favorably to normal mouse motor neurons, in terms of the genes that are turned on and off and how they function. But the scientists cant be certain these cells are perfect matches for native human motor neurons since its difficult to obtain samples of cultured motor neurons from adult individuals. Future work studying neuron samples donated from patients after death is required to determine how precisely these cells mimic native human motor neurons.

This work is supported by the National Institutes of Health (NIH) Directors Innovator Award, grant number DP2NS083372-01; the Presidential Early Career Award for Scientists and Engineers; the Missouri Spinal Cord Injury/Disease Research Program; the Cure Alzheimers Fund; Andrew B. and Virginia C. Craig Faculty Fellowship; a Philip and Sima Needleman Graduate Student Fellowship; a Ruth L. Kirschstein National Research Service Award Institutional Predoctoral Fellowship, grant number T32GM081739; and the Childrens Discovery Institute.

Abernathy DG, Kim WK, McCoy MJ, Lake AM, Ouwenga R, Lee SW, Xing X, Li D, Lee HJ, Heuckeroth RO, Dougherty JD, Wang T, Yoo AS. MicroRNAs induce a permissive chromatin environment that enables neuronal subtype-specific reprogramming of adult human fibroblasts. Cell Stem Cell. Sept. 7, 2017.

Washington University School of Medicines 2,100 employed and volunteer faculty physicians also are the medical staff of Barnes-Jewish and St. Louis Childrens hospitals. The School of Medicine is one of the leading medical research, teaching and patient-care institutions in the nation, currently ranked seventh in the nation by U.S. News & World Report. Through its affiliations with Barnes-Jewish and St. Louis Childrens hospitals, the School of Medicine is linked to BJC HealthCare.

Read more:
Human skin cells transformed directly into motor neurons - Washington University School of Medicine in St. Louis

Hacking Your Genes Has Never Been Easier – Outside Magazine

Josiah Zayner and I are drinking fluorescent green beer at the ODIN, his Oakland lab. The tables are scattered with pipettes and disposable blue gloves, cases of Red Bull and Slim Jims are near at hand, and Drake is pulsing on the sound system. Its not St. Patricks Day, and the beer isnt really all that green. Its the ghostly luminescence of jellyfish pulsing through the depths. Thats because its chock full of glowing jellyfish protein.

But no jellyfish were harmed in the making of this beer. Zayner is the worlds most notorious biohackera new breed of garage tinkerer experimenting with DNA and biological systems outside the confines of traditional research. In this case, he genetically engineered a common brewers yeast by adding a jellyfishs green fluorescent protein (GFP) gene that he ordered online. As long as you know the DNA sequence of the gene you wantthe As, Cs, Gs, and Ts of the genetic codeyou no longer need the actual critter the gene came from. You just run off the code on a special DNA printer containing cartridges filled with liquid As, Cs, Gs, and Ts. Then you insert the new DNA into whichever organism you want to modify. The process is shockingly easy.

Download the Audm app for your iPhone to listen to more longform titles.

I raise my glass and pause. Zayners yeast suffuses the beer with a gauzy haze. I have no idea which species of jellyfish the GFP gene came from, but my hunch is that it has never been a regular part of the human diet. Zayner assures me its safe. Genetic engineers love GFP because its such an easy visual. They include it with whichever other gene theyre trying to insert, and if their organism glows, they know the experiment worked without having to send off a sample for DNA sequencing. Scientists have engineered glowing cats and mice using GFP, he points out, and the creatures lived just fine.

I eye Zayner. He has drunk a fair amount of GFP beer himself, and while I wouldnt say he looks normalhe sports dozens of piercings, plugs in both earlobes, and a spike of bleached hair that is sometimes blue and sometimes whitehe seems healthy enough.

Dude, he assures me, we did all the normal FDA tests. Its nontoxic, nonallergenic. As further proof, he shows me his left forearm. Right next to the tattoo that says CREATE SOMETHING BEAUTIFUL is a row of four tiny wounds. I modified myself with it. Its fine.

Agar plates and vials of microbes at the ODIN lab. (Justin Kaneps)

Zayner claims he was the first to genetically modify himself with another speciess DNA. For what he would call a science experiment and I would call conceptual art, he removed dead skin cells from his forearm (just rub the same spot with a toothbrush 200 times) and used a tattoo needle to punch jellyfish DNA into his skin. The DNA was attached to a common virus that specializes in infiltrating human cells and parking itself there. Those skin cells then began manufacturing the GFP along with all their regular proteinsthough, to Zayners disappointment, not enough to see the glow with the naked eye. He also performed a DIY fecal transplant on himself, which was chronicled in the recent documentary Gut Hack, curing himself of years of irritable bowel syndrome.

Im not sure what I think about any of this, starting with my beer. I tend to favor pilsner over jellybrew, but Im trying to maintain my chill biohacker persona, so I chug. Weve spiked it with enough blood orange juice to cover any weirdness, and frankly it goes down pretty easy. Just like that, this crunchy Vermonter who always shunned GMOs filled his belly with them, and starts looking forward to the week ahead.

Id always thought of genetic engineering as something done in million-dollar labs by corporate powerhouses like Monsanto. Extracting the DNA from life forms and inserting it into other life forms seemed like the kind of thing that required high-tech machines and years of trial and error. And it used to. But that was before Crispr, Science magazines 2015 Breakthrough of the Year, an engineered protein that can snip out sequences of DNA wherever you want. Its like a search and replace function for genes. It works on bacterial cells, it works on mouse cells, and it works on human cells. Its been used to engineer immune cells that kill cancer, viruses that kill antibiotic-resistant bacteria, female mosquitoes that cant reproduce (to crash the population), and a yeast infused with genetic code from poppies and rats that makes opioids out of sugar in a tank. But the crazy thing about Crispr is that its so easy to use and cheap to make that it also allows any budding hacker with some basic biology and a mischievous mind to play God in their garage.

The only thing missing is someone to share this knowledge with the multitudes, and thats where Zayner comes in. He started out traditionally enough: wunderkind Ph. D. candidate at the University of Chicago and then research fellow at NASA, where he adapted organisms for life on Mars. But then, in 2015, he veered off to become the pierced Prometheus of genetic engineering, bringing it down to us mortals from the labs of academia. In this field, there are a bunch of people with a lot of knowledge and a bunch of people with a lot of crazy, he says with a smile, but there are very few with a lot of knowledge and a lot of crazy.

Not for the first time, I smile back at Zayner and try to gauge the crazy. For now Im coming down on the side of like a fox. Hes made a huge success of the ODINshort for Open Discovery Institute and inspired by the Norse godthe combination lab and mail-order business he founded in 2013 to make DIY bio accessible to everyone. The ODIN sells pre-engineered GFP yeast ($80) online, along with DIY Crispr kits ($150), fluorescent-yeast-engineering kits ($160), something called the Amino DNA Playground ($349), and a complete Genetic Engineering Home Lab Kit ($999) stocked with pipettes, tubes, scales, antibiotics, agar, light-activated bacteria, bioluminescent bacteria, Crispr, and a PCR machine, which makes copies of DNA through polymerase chain reaction. The ODINs clients include community colleges, high school kids, and mysterious individuals.

Jars of Crispr. (Justin Kaneps)

All ODIN kits are designed to engineer bacteria or yeast, the cheapest and simplest critters to work with, and they focus on obvious visuals like GFP. They are the Easy-Bake Ovens of genetic engineering. They offer quick success to rank amateurs like me and a tantalizing taste of the endless possibilities. Where we take it from there is up to us.

Zayner and his fellow biohackers are big on genetic freedom. Everything your body makes or does is encoded by a gene. And the more we learn about the genetic basis of human processesfrom disease and life expectancy to athletic and mental performancethe closer we get to being able to reprogram our bodies. I think we could do substantial changes to ourselves right now, Zayner says. You could go a little more crazy than scientists have been willing to let on.

For years there have been rumors that people already are. Gene doping, as its called, could theoretically give anybody the ability to burn oxygen like a Tibetan mountaineer, to build muscle like LeBron James, and to never get heart disease. Its all in the genes. Its in the hard work and good habits, too, but without certain tools you can only go so far. And in either the shady present or the not so distant future, well all have access to those tools, which Zayner finds pretty exciting. This is the first time in human history that were no longer stuck with the genes we had at birth. It fucking blows your mind.

He sees no reason to let corporations and ivory-tower institutions have all the fun. Hence the Easy-Bake Ovens. Give a man a cookie and he eats for a day. Teach a man to cook and youve stolen fire from the gods.

Josiah Zayner. The name screams Marvel Comics. The backstory, too: Country childhood on an Indiana farm. Pentecostal parents. (His brothers are Micah, Zachariah, and Jedediah; the dog was named Jeremiah.) Missionary in Peru. Teenage member of the late-nineties hacker collective Legions of the Underground. Biophysics Ph.D. from the University of Chicago. Synthetic-biology fellowship at NASAs Ames Research Center. Then something goes horribly wrong.

In Zayners case, there was no lab explosion. No rampaging through the streets of Mountain View, paralyzing Google employees with jellyfish tentacles sprouting from his back. No, what went wrong is that Zayner discovered that NASA was deadly dull. Empty offices. Stultifying bureaucracy. A supervisor who actually told him to spend less time in the lab. Not the place for someone who wanted to change the universe. So he did what any budding superhero would do: he went rogue.

Crispr and pipettes. (Justin Kaneps)

As his two-year NASA fellowship neared its end in 2015, Zayner launched an Indiegogo campaign offering contributors their own DIY gene-editing kit. Hed learned just enough while getting his Ph.D. to realize that genetic engineering was way more accessible than most people knew, and he couldnt wait to liberate it from the elite labs he loathed and bring it to the people, because, as he told me, I was always that poor-as-dirt kid dreaming that he could do some great experiment. The pitch video featured shots of Zayner swigging from a flask at the lab bench (his kitchen counter) while the voiceover asked, If you had access to cutting-edge syntheticbiology tools, what would you create? The campaign raised more than $70,000.

It also freaked out critics. Zayners campaign is worrisome because it does not seem to comply with the DIYbio.org code of conduct, Todd Kuiken, a scholar in the Genetic Engineering and Society Center at North Carolina State University, wrote in Nature in 2016. He was referring to the nonprofit founded in 2008 to foster safe practices in DIY biology. For example, he noted, The video that accompanies his campaign zooms in on petri dishes containing samples that are stored next to food in a refrigerator. Kuiken also believes there needs to be a robust public dialogue about the responsible use of Crispr.

The refrigerator comment still annoys Zayner. So are you saying that being able to do science is a class thing? Only people who can afford second fridges should do science? But he got his act together and bought another fridge, in part because he was already under scrutiny from the FDA, which had threatened to seize his equipment because of his Internet sales. Zayner has also been warned of possible prosecution by officials in Germany, where biohacking is banned. But the practice is perfectly legal throughout the United States, mostly because it has never occurred to legislators to outlaw such a thing, and the ODIN is doing well. Zayner sells thousands of gene-editing kits globally every year, and he expects to gross at least $400,000 in 2017. The world wants this.

The workday at the ODIN starts late-morning. One employee is multi-tasking, packing kits for the days orders while he propagates new batches of microbes. Zayners brother Micah is scarfing Chinese takeout on the couch. The air is redolent with the funk of E. coli bacteria and young male. Zayner solders new wiring onto used PCR machines (There are few things Im one of the worlds leading experts on, but finding functional lab equipment on eBay is one of them, he says) while guiding me through an attempt to engineer antibiotic resistance into E. coli using Crispr. Despite the punk trappings, Zayner is gentle, kind, and a very good teacher.

We rehydrate some dried E. coli in a test tube, pour it into a petri plate containing nutrients, and set it aside overnight. In the morning, we have a flourishing colony of fuzzy white bacteria. We scrape it up, divide it into two plastic tubes of liquid, and to one tube add a few drops of Crispr programmed to change a single A to a C, which will flip the electrical charge of a protein in the bacteria from positive to negative at the point where streptomycin normally attacks it, repelling the antibiotic molecules. Then we pour the two batches onto fresh agar plates laced with streptomycin and incubate everything at 99 degrees for 24 hours.

Genetically modified beer. (Justin Kaneps)

The next day, I pull our agar plates out of the incubator and examine them. Eureka! The normal bacteria is stone-cold dead. But the plate with the modified bacteria is studded with survivor colonies. Weve created GMOs in a day. They and their trillions of descendants will be immune to streptomycin.

Or they would have been if we hadnt killed the whole colony with bleach and thrown it in the trash. As crazy as our creation sounds, it turns out that it was pretty innocuous. This particular version of antibiotic resistance is so simplejust a single changed letter of DNAthat bacteria come up with it on their own all the time. We werent introducing anything the world hadnt seen before, and anyway our weak lab strain was about as dangerous as a cocker spaniel. Yet I cant help but wonder about all the biohackers out there who arent bleaching their experiments. What could the wrong person do with this knowledge?

Thats what I asked Ed You, the biological-countermeasures specialist at the FBIs Weapons of Mass Destruction directorate. You is the governments point person on bioweapons; its his job to worry about this stuff, but he had bigger things on his mind than the ODIN. The most dangerous bioterrorist out there is Mother Nature, he told me over the phone. Were getting hit with emerging and reemerging infectious diseases all the time. Bird flu, MERS, SARS, Zika, West Nile. If you think about a clear and present danger, its that. So we absolutely need the innovation that comes from the life sciences, from DIY bio, to make sure we develop the right counters.

Wait a minute, I said. You actually want them out there tinkering? Yes, he replied. Biology is proliferating quickly, but how do we address security in a way that doesnt handicap forward progress? If you shut down DIY bio, then you run a completely different national-security problem. If you stifle innovation, then youre going to be missing out on opportunities to come up with new vaccines, new biodefense, new countermeasures, new businesses. And if that happens, then youve developed a whole different kind of vulnerability.

You pointed out that the field was moving so fast that agents could never keep up with the pace of the advances. Instead, hes cultivated a neighborhood-watch mentality among the countrys scientists and biohackers. Theyre best positioned to see where the advances are coming from, he said. If someone like Josiah gets a suspicious order of some kind, he knows that hes got a local coordinator in the San Francisco field office he can contact.

Agar plates. (Justin Kaneps)

It all sounded strangely progressive for a bunch of G-men, but every expert I consulted told me that they had no concerns about Zayner. Forget the garagistas, they told me; worry about the academics. Many labs now have the technology and know-how to make some fearsome beasties. Last year, a scientist in Canada shocked the world when he managed to bring to life horsepox, a smallpox cousin that went extinct in the 1980s, by synthesizing its DNA from a sequence stored in a computer database. Are we entering a new era of bioterror?

Probably not, Zayner told me. Lets imagine youre the worst person in the world and you want to hurt people with biologicals. First you have to have the knowledge. Then you have to have the facility. Then you have to think about how its going to spread. It would be an astounding feat. Could you kill one or two people? Sure. But you can do that with a fucking kitchen knife.

That night, Zayner and I celebrate our successful biohack over pig-ear fries and sake at a Korean joint before heading over to Counter Culture Labs, a communal biohacker space where he occasionally teaches. Amid the lab benches and anarchist posters are shelves of strange plants under grow lights and a pig heart in a vat. One woman is attempting to create vegan cheese by inserting cow milk-producing genes into yeast, while another man is quietly sequencing the DNA of the mushrooms he collects in Mexico each summer. A small team are hard at work designing an organism that can produce human insulin. In keeping with the hacker ethos, they will gift it to the world open-source.

There are dozens of biohacker enclaves like this around the globe, such as Genspace in Brooklyn, New York, where hipsters can take Crispr classes and attend Biohacker Boot Camp. The U.S. has been the hub, but now Europe is coming on strong. DIYbio.org has nearly 5,000 members in its Google Group and boasts 99 local chapters, from Madison to Mumbai. Most biohackers never get beyond simple experiments with microbes, but a few have taken it further. David Ishee, a dog breeder in Mississippi, is editing heritable diseases out of his dalmatians. Sebastian Cocioba, a plant hacker in New York, engineered a pioneering blue rose gene, using a DNA sequence from a tropical clam that produces an intensely blue protein, as well as a beefsteak tomato that produces cow protein in its flesh. Cocioba, who operates out of his 12th-floor apartment in Long Island City, is so skilled that he has been asked by MIT to spearhead a top-secret flower project, the details of which cant be shared except to say that in a few years it will capture the worlds attention.

And what about people? I ask. How long before cyclists start giving themselves the EPO gene to produce more red blood cells, or lifters start playing around with the gene for human growth factor?

Zayner laughs. Dude, either people are already doing that shit, or its going to start immediately. Id be very surprised if there isnt somebody out there doing it already. Its so hard to test for. What are you going to do, look for DNA? If a professional athlete came to me right now and said, Ill give you $100,000 to make me a piece of DNA, Id be like, Hell yeah.

Zayner believes we should all have access to DIY bio. (Justin Kaneps)

Surprisingly, this is perfectly legal, though its long been banned by sporting organizations. Athletes and life-extension buffs have been sniffing around gene-therapy clinics for years, ever since pioneering physiologist Lee Sweeney, from the University of Pennsylvania, showed that mice injected with the gene IGF-1, or insulin-like growth factor, significantly increased their muscle mass. Sweeney has also shown that mice injected with endurance genes were able to run 70 percent farther on the wheel than their unmodified peers, and that couch-potato mice ran 44 percent farther.

Just this June, a team of U.S. and Israeli scientists announced the discovery of a rare genetic mutation linked to ten years of extra longevity in men. And in 2015, Liz Parrish, the CEO of the startup BioViva, announced that she was the first person to attempt to reverse her own aging with gene therapy. I am patient zero, she wrote on Reddit. I will be 45 in January. I have aging as a disease. Parrish traveled to a clinic in Colombia (the therapy isnt approved in the U.S.) and received injections of one gene to extend the lifespan of her individual cells and another to block myostatin, the hormone that regulates muscle deterioration.

Myostatin is the holy grail of potential dopers who believe they can both arrest the natural deterioration of muscle and build more in their youth. Muscle is metabolically expensive to maintain, so myostatins job is to stop new muscle from being made once youve got enough and to atrophy muscle you arent using. You can find images online of dogs, cows, and people with a rare mutation that shuts down the myostatin gene and turns them into Incredible Hulks. Scientists in China recently used Crispr to turn off the myostatin gene in two beagles. The dogs look healthy, happyand ripped.

But Im less interested in what athletes are doing than in something Zayner said to me on my first day in the lab: This is the first time in history that were no longer stuck with the genes we had at birth. If Zayner has his way, well all be sculpting our own evolution.

Lets be clear: dont try this at home! Although hundreds of gene-therapy trials are under way, and many experts believe they will eventually transform almost every aspect of human health, few have been proven safe. When you start scrambling your DNA, very bad things can happen. You can get cancer. Your immune system can attack the unfamiliar DNA, as happened when an 18-year-old with a rare metabolic disorder died during a University of Pennsylvania gene-therapy trial in 1999.

But sick people wont wait for years of trials, Zayner says. He hears regularly from people willing to roll the dice. Hes been consulting pro bono for a man using Crispr to treat his own Huntingtons disease and another who is treating his 32-year-old wifes advanced lung carcinoma with genetically engineered DNA vaccines. A lot of people contact me with stuff like thatIm suffering. Can you help?

Zayner sticks to the free advice, helping people figure out the sequence of the DNA they need without supplying anything himself, but he knows where this is headed. The only thing holding people back is morality. I have no doubt there are places in Singapore or Thailand or the Philippines doing it. They could totally create individualized cancer treatments right now. Clinics will pop up. Youll go to shops in the back alleys of Bangkok and hand $10,000 to a synthetic biologist and hell take a blood sample and make you up a vaccine in a couple of days.

Im flashing back to Blade Runners replicant shopsI just do eyeswhen Zayner gets a funny smile and cocks his head. Want to try something kind of creepy Ive been thinking about?

For our final piece of conceptual art, Zayner and I swab the crevices of our skin and inside our mouths with Q-tips and swirl the gunk into tubes of distilled water. We spread the contents over agar plates and incubate them overnight.

The next morning, Josiahthing is nearly barren, but Rowanthing is crawling with cells. Look at those big fat yeasties! Zayner mutters with envy. All I can think is, if this works, it will give new meaning to the term homebrew.

We scrape up some Josiahthing and Rowanthing and put each in its own microcentrifuge tube with some chemicals that soften up cell walls so new DNA can get inside. We pipette ten microliters of the jellyfish DNA into each tube, shake them up, let them sit for a few hours, then pour them across new agar plates and cross our fingers. If this actually works, I might make it a kit, Zayner muses.

By then I have to catch a flight home, so I tape up my petri plate and pack it, along with yellow-tint glasses and a blue LED, which makes the fluorescence easier to see. TSA doesnt bat an eye.

The next day I get an e-mail from Zayner: Any growth on that plate?

Yep! Four or five nice, puffy little white colonies.

Put on the glasses and shine blue light on them. Do they glow?

I don the glasses and hit the plate with the blue LED. There are a dozen tiny colonies that stay dull under the light, but there are also five large conical colonies fluorescing like the Green Goblin. Totally! I write back, and send a photo.

Amazing! So cool! So jealous. Mine didnt work.

I feel as proud as Victor Frankenstein. Ive created life from my own spit. In the following weeks, Rowanthing develops an apex so green you dont even need the glasses to see it. Whatever it is, its new to this planet, and its burbling away in my basement, waiting to meet the world.

Contributing editor Rowan Jacobsen (@rowanjacobsen) is a Knight Science Journalism Fellow at MIT. Justin Kaneps(@Justkaneps) is anOutsidecontributing photographer.

See the original post:
Hacking Your Genes Has Never Been Easier - Outside Magazine

Quick Hits: Withdrawn Leukemia Drug Returning, Drugmaker in $58M Settlement Over Sales Reps, and More – MedShadow (registration) (blog)

The leukemia drug Mylotarg (gemtuzumab ozogamicin), which was voluntarily withdrawn from the market in 2010 over safety concerns and questions about its efficacy, is available again. The FDA has approved the biologic for adults with newly diagnosed acute myeloid leukemia and for patients at least 2 years old who have had a relapse or didnt respond to prior treatment. Mylotarg originally received accelerated approval in 2000 for older adults who had experienced a relapse. But it was removed from the market after patient deaths and a lack of clinical benefit were observed in confirmatory trials. With the new approval, Mylotarg now has a lower recommended dose, a different schedule of how often the drug is given, and a different patient population. Pfizer, Mylotargs manufacturer, presented the FDA with additional data, analysis and research from clinical trials that lasted for 10 years in support of re-approval. The FDA said it allowed Mylotargs return after careful examination and based on the benefits outweighing the risks. Posted September 1, 2017. Via FDA.

Novo Nordisk has agreed to pay $58 million over allegations that some of its sales representatives downplayed a cancer risk associated with its diabetes drug Victoza (liraglutide) in marketing to doctors. When Victoza was approved in 2010, the FDA mandated it come with a Risk Evaluation and Mitigation Strategy (REMS) that required the drugmaker to provide information regarding Victozas potential risk of medullary thyroid carcinoma (MTC) a rare form of cancer associated with the drug to physicians. According to a complaint filed by the US Department of Justice (DOJ), some Novo Nordisk sales representatives created a misleading impression that the cancer risk associated with Victoza was incorrect or unimportant. In addition, they failed to accurately report important data regarding the drugs safety and efficacy. The DOJ also noted that a survey in 2011 found that half of primary care doctors polled said they did not know about the MTC risk. Posted September 5, 2017. Via US Department of Justice.

The FDA has approved the first-ever gene therapy, Kymriah (tisagenlecleucel), as a treatment for children and young adults for a type of leukemia. The immunotherapy is considered a breakthrough since it is made using the patients own T-cells, white blood cells that are part of the bodys immune system that fight infections and cancer. Kymriah is custom-made for each patient. A patients T-cells are sent to a manufacturing facility and then modified genetically to include a gene that has a specific protein that tells the T-cells to find and kill leukemia cells. After the modification, the T-cells are infused back into the patient. Researchers found that Kymriah led to remission of acute lymphoblastic leukemia in 83% of 63 children and young adult patients within 3 months. Despite the benefits, the treatment carries risks, including a boxed warning about a potentially fatal immune reaction known as cytokine-release syndrome. Other severe side effects seen with Kymriah include serious infections, low blood pressure (hypotension), acute kidney injury, fever and decreased oxygen (hypoxia). Because of these risks, the FDA is requiring that hospitals and clinics that want to dispense Kymriah need to be certified and their staff involved in the treatment trained to recognize and manage symptoms. Posted August 30, 2017. Via FDA.

Alanna McCatty is a recent graduate of Pace University with a degree in communications. At MedShadow, she reports on new findings and research on the side effects of prescription drugs.

See more here:
Quick Hits: Withdrawn Leukemia Drug Returning, Drugmaker in $58M Settlement Over Sales Reps, and More - MedShadow (registration) (blog)

FDA approves first cell-based gene therapy for use in the United States – Gears Of Biz

The U.S. Food and Drug Administration issued a historic action today making the first gene therapy available in the United States, ushering in a new approach to the treatment of cancer and other serious and life-threatening diseases.

The FDA approved Kymriah (tisagenlecleucel) for certain pediatric and young adult patients with a form of acute lymphoblastic leukemia (ALL).

Were entering a new frontier in medical innovation with the ability to reprogram a patients own cells to attack a deadly cancer, said FDA Commissioner Scott Gottlieb, M.D. New technologies such as gene and cell therapies hold out the potential to transform medicine and create an inflection point in our ability to treat and even cure many intractable illnesses. At the FDA, were committed to helping expedite the development and review of groundbreaking treatments that have the potential to be life-saving.

Kymriah, a cell-based gene therapy, is approved in the United States for the treatment of patients up to 25 years of age with B-cell precursor ALL that is refractory or in second or later relapse.

Kymriah is a genetically-modified autologous T-cell immunotherapy. Each dose of Kymriah is a customized treatment created using an individual patients own T-cells, a type of white blood cell known as a lymphocyte. The patients T-cells are collected and sent to a manufacturing center where they are genetically modified to include a new gene that contains a specific protein (a chimeric antigen receptor or CAR) that directs the T-cells to target and kill leukemia cells that have a specific antigen (CD19) on the surface. Once the cells are modified, they are infused back into the patient to kill the cancer cells.

ALL is a cancer of the bone marrow and blood, in which the body makes abnormal lymphocytes. The disease progresses quickly and is the most common childhood cancer in the U.S. The National Cancer Institute estimates that approximately 3,100 patients aged 20 and younger are diagnosed with ALL each year. ALL can be of either T- or B-cell origin, with B-cell the most common. Kymriah is approved for use in pediatric and young adult patients with B-cell ALL and is intended for patients whose cancer has not responded to or has returned after initial treatment, which occurs in an estimated 15-20 percent of patients.

Kymriah is a first-of-its-kind treatment approach that fills an important unmet need for children and young adults with this serious disease, said Peter Marks, M.D., Ph.D., director of the FDAs Center for Biologics Evaluation and Research (CBER). Not only does Kymriah provide these patients with a new treatment option where very limited options existed, but a treatment option that has shown promising remission and survival rates in clinical trials.

The safety and efficacy of Kymriah were demonstrated in one multicenter clinical trial of 63 pediatric and young adult patients with relapsed or refractory B-cell precursor ALL. The overall remission rate within three months of treatment was 83 percent.

Treatment with Kymriah has the potential to cause severe side effects. It carries a boxed warning for cytokine release syndrome (CRS), which is a systemic response to the activation and proliferation of CAR T-cells causing high fever and flu-like symptoms, and for neurological events. Both CRS and neurological events can be life-threatening. Other severe side effects of Kymriah include serious infections, low blood pressure (hypotension), acute kidney injury, fever, and decreased oxygen (hypoxia). Most symptoms appear within one to 22 days following infusion of Kymriah. Since the CD19 antigen is also present on normal B-cells, and Kymriah will also destroy those normal B cells that produce antibodies, there may be an increased risk of infections for a prolonged period of time.

The FDA today also expanded the approval of Actemra (tocilizumab) to treat CAR T-cell-induced severe or life-threatening CRS in patients 2 years of age or older. In clinical trials in patients treated with CAR-T cells, 69 percent of patients had complete resolution of CRS within two weeks following one or two doses of Actemra.

Because of the risk of CRS and neurological events, Kymriah is being approved with a risk evaluation and mitigation strategy (REMS), which includes elements to assure safe use (ETASU). The FDA is requiring that hospitals and their associated clinics that dispense Kymriah be specially certified. As part of that certification, staff involved in the prescribing, dispensing, or administering of Kymriah are required to be trained to recognize and manage CRS and neurological events. Additionally, the certified health care settings are required to have protocols in place to ensure that Kymriah is only given to patients after verifying that tocilizumab is available for immediate administration. The REMS program specifies that patients be informed of the signs and symptoms of CRS and neurological toxicities following infusion and of the importance of promptly returning to the treatment site if they develop fever or other adverse reactions after receiving treatment with Kymriah.

To further evaluate the long-term safety, Novartis is also required to conduct a post-marketing observational study involving patients treated with Kymriah.

The FDA granted Kymriah Fast Track, Priority Review, and Breakthrough Therapy designations. The Kymriah application was reviewed using a coordinated, cross-agency approach. The clinical review was coordinated by the FDAs Oncology Center of Excellence, while CBER conducted all other aspects of review and made the final product approval determination.

The FDA granted approval of Kymriah to Novartis Pharmaceuticals Corp. The FDA granted the expanded approval of Actemra to Genentech Inc.

Read the original here:
FDA approves first cell-based gene therapy for use in the United States - Gears Of Biz

Researchers point way to improved stem cell transplantation therapies – Medical Xpress

September 7, 2017 When transiently expressed in HSCs, BCL-XL temporarily protects the cells from apoptosis and enhances their ability to be transplanted from one mouse to another. This microscopy image shows the successful introduction of BCL-XL (green) into a cell labeled for its nucleus (blue) and mitochondria (red). Credit: Kollek et al., 2017

Researchers in Germany have demonstrated that hematopoietic stem cell (HSC) transplants can be improved by treatments that temporarily prevent the stem cells from dying. The approach, which is described in a paper to be published September 7 in The Journal of Experimental Medicine, could allow those in need of such transplants, including leukemia and lymphoma patients, to be treated with fewer donor stem cells while limiting potential adverse side effects.

HSCs give rise to the many different cell types found in blood and can be used to treat a variety of diseases, including multiple myeloma, leukemia, and blood disorders such as sickle cell anemia. HSCs can be harvested from a suitable donor and then transplanted into a patient, where, after establishing themselves in the bone marrow, they can generate healthy blood cells.

The transplantation process is stressful for HSCs, however, and many of them die before they can successfully ensconce themselves in the patient's bone marrow. This limits the effectiveness of HSC transplantation, delaying the resumption of blood cell formationincreasing the risk of infection or bleedingor even causing the transplant to fail completely. HSC death is a particular problem if the number of donor stem cells is low to begin with. Umbilical cord blood, for example, generally contains insufficient numbers of stem cells for it to be used as a source of HSCs for transplantation into adult patients.

HSCs die through a process called apoptosis, driven by two proteins called BIM and BMF. Permanently inhibiting these two proteins prevents HSCs from dying and improves the efficiency of HSC transplantation in mice. But mice receiving these apoptosis-resistant stem cells soon develop autoimmune disease and/or lymphomas because the HSCs, and the blood cells they produce, do not die when they are supposed to.

"Thus, inhibiting apoptosis transiently during the stressful period of transplantation could be an attractive strategy to improve transplantation outcome without increasing the risk of long-term adverse effects," says Dr. Miriam Erlacher of the University Medical Center of Freiburg.

Erlacher and colleagues isolated HSCs from mice and infected them with a genetically engineered adenovirus that transiently produces a human protein called BCL-XL that inhibits BIM and BMF. These virally infected HSCs were resistant to apoptosis for the 7-9 days that BCL-XL was expressed, and, upon transplantation into recipient mice, their ability to establish themselves in the bone marrow and produce new blood cells was greatly enhanced. Moreover, because the transplanted HSCs only expressed BCL-XL for a few days, they didn't promote the formation of lymphomas in recipient animals.

Adenoviral infection is slightly toxic to HSCs, however, so Erlacher and colleagues developed an alternative approach in which purified BCL-XL could be introduced directly into isolated HSCs. This second method also provided temporary protection from apoptosis and improved the cells' ability to undergo transplantation.

"Our findings suggest that transiently inhibiting apoptosis by manipulating donor HSCs increases the fitness of these cells without elevating the risk of adverse pathology," Erlacher says. "Transient apoptosis inhibition is therefore a promising approach to reduce the risk of graft failure and improve HSC transplantation outcomes."

Explore further: Dose of transplanted blood-forming stem cells affects their behavior

More information: Kollek et al., 2017. J. Exp. Med. jem.rupress.org/cgi/doi/10.1084/jem.20161721

Please sign in to add a comment. Registration is free, and takes less than a minute. Read more

Excerpt from:
Researchers point way to improved stem cell transplantation therapies - Medical Xpress

Clarkson professor awarded $420000 grant to study development of intestinal stem cells using zebrafish vertebrate … – North Country Now

Clarkson University Associate Professor of Biology Kenneth Wallace showcases his subjects of study -- zebrafish -- a common aquarium fish who share more than 70 percent of their genes with humans.

POTSDAM -- Kenneth Wallace, associate professor of biology at Clarkson University, has been awarded a $420,000 grant from the Eunice Kennedy Shriver National Institute of Child Health & Human Development at the National Institutes of Health to investigate development of intestinal stem cells using the zebrafish vertebrate model system.

While much has been discovered about how stem cells are controlled during the adult phase, much less is known about the origins of these stem cell compartments. Little is known about when the stem cells form and how they are regulated. To uncover more about how stem cells are regulated during development of the intestine, Wallace will use zebrafish, which have become a widely-used vertebrate model system.

Zebrafish are a common aquarium fish, which are small easy to care for and have embryos that develop rapidly in an external environment. They also share more than 70 percent of their genes with humans, making them an excellent system to study both development and the origins of disease. Understanding of the genes and mechanisms involved in formation and regulation of the fish intestinal stem cells will provide information about how human intestinal stem cells are regulated.

Aside from the main research component, a secondary goal of the grant and project is to provide resources for undergraduate Clarkson University students to perform independent research on the molecular and cellular basis of embryonic development under Wallaces supervision. This will give them first-hand knowledge of developmental biology research practices and perhaps pique future interest in the field and research.

Read more here:
Clarkson professor awarded $420000 grant to study development of intestinal stem cells using zebrafish vertebrate ... - North Country Now

Stem Cell Therapy: A Lethal Cure – Medical News Bulletin

Stem cell therapy is a two-step process. First, the patients blood cells are destroyed by chemotherapy, radiation therapy or immunosuppression. This conditioning process also eradicates any cancer cells that survived first-line treatment. Second, the patient receives stem cells harvested from a donors bone marrow or peripheral blood (circulating blood). While this can be an effective cure, it can cause graft-versus-host disease (GVHD) in up to 50% of patients. GVHD is more likely to develop in patients who have received a peripheral blood transplant and can kill 15%-20% of patients.

Two types of GVHD can develop, acute and chronic, and patients may develop either one, both or neither type. GVHD is less likely to occur and symptoms are milder if the donor cells closely match those of the patient. Acute GVHD can develop within 100 days of a transplant. The first step of stem cell therapy can cause tissue damage, and bacteria from the gut can escape into the bloodstream. This primes the patients antigen-presenting cells (cells that activate the immune response), which subsequently encourage donor T cells to proliferate and attack the patients tissues. Symptoms include vomiting, diarrhea, skin rashes, nausea, vomiting and liver problems. This can be resolved relatively quickly in one third of patients using immunosuppressive treatments, but some patients can progress to chronic GVHD.

The biological mechanisms responsible for chronic GVHD are not completely understood, but scientists believe that other immune system cells from the donor (B cells and macrophages) are stimulated and damage the patients tissues. Symptoms include dry eyes, mouth sores, muscle weakness, fatigue and joint problems.

Unfortunately, development of effective treatments for GVHD is not keeping up with the increasing number of GVHD patients or with advances in understanding this disease. At present, standard treatments include corticosteroids and drugs that reduce IL-2, an immune system chemical that helps T-cells multiply and diversify. These treatments have various side effects including suppressing the patients immune system, thereby increasing risk of infection.

One challenge stalling drug research is that a small degree of graft-versus-host response must occur for successful stem cell therapy: donor cells will destroy any cancer cells that remain after the first stage of therapy. This challenge is discussed in a recent article in Science Health.Although several treatments have been trialed, success is variable and often targets only acute GVHD or chronic GVHD. Biomarkers have also been detected that may help identify individuals at risk of developing severe GVHD, information that may aid the development of personalized treatment strategies. Drugs that have been approved for other diseases, but not for GVHD, show promise and include ibrutinib for chronic GVHD (approved for specific blood cancers) and ruxolitinib for acute GVHD (approved for bone marrow disorders).

The impact of stem cell therapy must not be underestimated: up to 50% of recipients will develop GVHD. Unfortunately, some individuals will develop chronic GVHD, a condition that is just as difficult to survive as cancer. This highlights the importance of developing continued care strategies for individuals receiving stem cell therapy as a final defence against cancer.

Written byNatasha Tetlow, PhD

Reference: Cohen J. A stem cell transplant helped beat back a young doctors cancer. Now, its assaulting his body. Science Health. 2017. Available at: DOI: 10.1126/science.aan7079

Continue reading here:
Stem Cell Therapy: A Lethal Cure - Medical News Bulletin