fbpx
Wikipedia

Stem-cell niche

Stem-cell niche refers to a microenvironment, within the specific anatomic location where stem cells are found, which interacts with stem cells to regulate cell fate.[1] The word 'niche' can be in reference to the in vivo or in vitro stem-cell microenvironment. During embryonic development, various niche factors act on embryonic stem cells to alter gene expression, and induce their proliferation or differentiation for the development of the fetus. Within the human body, stem-cell niches maintain adult stem cells in a quiescent state, but after tissue injury, the surrounding micro-environment actively signals to stem cells to promote either self-renewal or differentiation to form new tissues. Several factors are important to regulate stem-cell characteristics within the niche: cell–cell interactions between stem cells, as well as interactions between stem cells and neighbouring differentiated cells, interactions between stem cells and adhesion molecules, extracellular matrix components, the oxygen tension, growth factors, cytokines, and the physicochemical nature of the environment including the pH, ionic strength (e.g. Ca2+ concentration) and metabolites, like ATP, are also important.[2] The stem cells and niche may induce each other during development and reciprocally signal to maintain each other during adulthood.

Scientists are studying the various components of the niche and trying to replicate the in vivo niche conditions in vitro.[2] This is because for regenerative therapies, cell proliferation and differentiation must be controlled in flasks or plates, so that sufficient quantity of the proper cell type are produced prior to being introduced back into the patient for therapy.

Human embryonic stem cells are often grown in fibrotastic growth factor-2 containing, fetal bovine serum supplemented media. They are grown on a feeder layer of cells, which is believed to be supportive in maintaining the pluripotent characteristics of embryonic stem cells. However, even these conditions may not truly mimic in vivo niche conditions.

Adult stem cells remain in an undifferentiated state throughout adult life. However, when they are cultured in vitro, they often undergo an 'aging' process in which their morphology is changed and their proliferative capacity is decreased. It is believed that correct culturing conditions of adult stem cells needs to be improved so that adult stem cells can maintain their stemness over time.[citation needed]

A Nature Insight review defines niche as follows:

"Stem-cell populations are established in 'niches' — specific anatomic locations that regulate how they participate in tissue generation, maintenance and repair. The niche saves stem cells from depletion, while protecting the host from over-exuberant stem-cell proliferation. It constitutes a basic unit of tissue physiology, integrating signals that mediate the balanced response of stem cells to the needs of organisms. Yet the niche may also induce pathologies by imposing aberrant function on stem cells or other targets. The interplay between stem cells and their niche creates the dynamic system necessary for sustaining tissues, and for the ultimate design of stem-cell therapeutics ... The simple location of stem cells is not sufficient to define a niche. The niche must have both anatomic and functional dimensions."[3]

History edit

Though the concept of stem cell niche was prevailing in vertebrates, the first characterization of stem cell niche in vivo was worked out in Drosophila germinal development.[citation needed]

The architecture of the stem-cell niche edit

By continuous intravital imaging in mice, researchers were able to explore the structure of the stem cell niche and to obtain the fate of individual stem cells (SCs) and their progeny over time in vivo. In particular in intestinal crypt,[4] two distinct groups of SCs have been identified: the "border stem cells" located in the upper part of the niche at the interface with transit amplifying cells (TAs), and "central stem cells" located at the crypt base. The proliferative potential of the two groups was unequal and correlated with the cells' location (central or border). It was also shown that the two SC compartments acted in accord to maintain a constant cell population and a steady cellular turnover. A similar dependence of self-renewal potential on proximity to the niche border was reported in the context of hair follicle, in an in vivo live-imaging study.[5]

This bi-compartmental structure of stem cell niche has been mathematically modeled to obtain the optimal architecture that leads to the maximum delay in double-hit mutant production.[6] They found that the bi-compartmental SC architecture minimizes the rate of two-hit mutant production compared to the single SC compartment model. Moreover, the minimum probability of double-hit mutant generation corresponds to purely symmetric division of SCs with a large proliferation rate of border stem cells along with a small, but non-zero, proliferation rate of central stem cells.[citation needed]

Stem cell niches harboring continuously dividing cells, such as those located at the base of the intestinal gland, are maintained at small population size. This presents a challenge to the maintenance of multicellular tissues, as small populations of asexually dividing individuals will accumulate deleterious mutations through genetic drift and succumb to mutational meltdown.[7] Mathematical modeling of the intestinal gland reveals that the small population size within the stem cell niche minimizes the probability of carcinogenesis occurring anywhere, at the expense of gradually accumulated deleterious mutations throughout organismal lifetime—a process that contributes to tissue degradation and aging.[8] Therefore, the population size of the stem cell niche represents an evolutionary trade-off between the probability of cancer formation and the rate of aging.

Examples edit

Germline edit

Germline stem cells (GSCs) are found in organisms that continuously produce sperm and eggs until they are sterile. These specialized stem cells reside in the GSC niche, the initial site for gamete production, which is composed of the GSCs, somatic stem cells, and other somatic cells. In particular, the GSC niche is well studied in the genetic model organism Drosophila melanogaster and has provided an extensive understanding of the molecular basis of stem cell regulation.[citation needed]

 
GSC niche in"Drosophila melanogaster" germarium

GSC niche in Drosophila ovaries edit

In Drosophila melanogaster, the GSC niche resides in the anterior-most region of each ovariole, known as the germarium. The GSC niche consists of necessary somatic cells-terminal filament cells, cap cells, escort cells, and other stem cells which function to maintain the GSCs.[9] The GSC niche holds on average 2–3 GSCs, which are directly attached to somatic cap cells and Escort stem cells, which send maintenance signals directly to the GSCs.[10] GSCs are easily identified through histological staining against vasa protein (to identify germ cells) and 1B1 protein (to outline cell structures and a germline specific fusome structure). Their physical attachment to the cap cells is necessary for their maintenance and activity.[10] A GSC will divide asymmetrically to produce one daughter cystoblast, which then undergoes 4 rounds of incomplete mitosis as it progresses down the ovariole (through the process of oogenesis) eventually emerging as a mature egg chamber; the fusome found in the GSCs functions in cyst formation and may regulate asymmetrical cell divisions of the GSCs.[11] Because of the abundant genetic tools available for use in Drosophila melanogaster and the ease of detecting GSCs through histological stainings, researchers have uncovered several molecular pathways controlling GSC maintenance and activity.[12] [13]

Molecular mechanisms of GSC maintenance and activity edit

Local signals edit

The Bone Morphogenetic Protein (BMP) ligands Decapentaplegic (Dpp) and Glass-bottom-boat (Gbb) ligand are directly signalled to the GSCs, and are essential for GSC maintenance and self-renewal.[14] BMP signalling in the niche functions to directly repress expression of Bag-of-marbles (Bam) in GSCs, which is up-regulated in developing cystoblast cells.[15] Loss of function of dpp in the niche results in de-repression of Bam in GSCs, resulting in rapid differentiation of the GSCs.[10] Along with BMP signalling, cap cells also signal other molecules to GSCs: Yb and Piwi. Both of these molecules are required non-autonomously to the GSCs for proliferation-piwi is also required autonomously in the GSCs for proliferation.[16] In the germarium, BMP signaling has a short-range effect, therefore the physical attachment of GSCs to cap cells is important for maintenance and activity.[citation needed]

Physical attachment of GSCs to cap cells edit

The GSCs are physically attached to the cap cells by Drosophila E-cadherin (DE-cadherin) adherens junctions and if this physical attachment is lost GSCs will differentiate and lose their identity as a stem cell.[10] The gene encoding DE-cadherin, shotgun (shg), and a gene encoding Beta-catenin ortholog, armadillo, control this physical attachment.[17] A GTPase molecule, rab11, is involved in cell trafficking of DE-cadherins. Knocking out rab11 in GSCs results in detachment of GSCs from the cap cells and premature differentiation of GSCs.[18] Additionally, zero population growth (zpg), encoding a germline-specific gap junction is required for germ cell differentiation.[19]

Systemic signals regulating GSCs edit

Both diet and insulin-like signaling directly control GSC proliferation in Drosophila melanogaster. Increasing levels of Drosophila insulin-like peptide (DILP) through diet results in increased GSC proliferation.[20] Up-regulation of DILPs in aged GSCs and their niche results in increased maintenance and proliferation.[21] It has also been shown that DILPs regulate cap cell quantities and regulate the physical attachment of GSCs to cap cells.[21]

Renewal mechanisms edit

There are two possible mechanisms for stem cell renewal, symmetrical GSC division or de-differentiation of cystoblasts. Normally, GSCs will divide asymmetrically to produce one daughter cystoblast, but it has been proposed that symmetrical division could result in the two daughter cells remaining GSCs.[22][23] If GSCs are ablated to create an empty niche and the cap cells are still present and sending maintenance signals, differentiated cystoblasts can be recruited to the niche and de-differentiate into functional GSCs.[24]

Stem cell aging edit

As the Drosophila female ages, the stem cell niche undergoes age-dependent loss of GSC presence and activity. These losses are thought to be caused in part by degradation of the important signaling factors from the niche that maintains GSCs and their activity. Progressive decline in GSC activity contributes to the observed reduction in fecundity of Drosophila melanogaster at old age; this decline in GSC activity can be partially attributed to a reduction of signaling pathway activity in the GSC niche.[25][26] It has been found that there is a reduction in Dpp and Gbb signaling through aging. In addition to a reduction in niche signaling pathway activity, GSCs age cell-autonomously. In addition to studying the decline of signals coming from the niche, GSCs age intrinsically; there is age-dependent reduction of adhesion of GSCs to the cap cells and there is accumulation of reactive oxygen species (ROS) resulting in cellular damage which contributes to GSC aging. There is an observed reduction in the number of cap cells and the physical attachment of GSCs to cap cells through aging. Shg is expressed at significantly lower levels in an old GSC niche in comparison to a young one.[26]

GSC niche in Drosophila testes edit

Males of Drosophila melanogaster each have two testes – long, tubular, coiled structures – and at the anterior most tip of each lies the GSC niche. The testis GSC niche is built around a population of non-mitotic hub cells (a.k.a. niche cells), to which two populations of stem cells adhere: the GSCs and the somatic stem cells (SSCs, a.k.a. somatic cyst stem cells/cyst stem cells). Each GSC is enclosed by a pair of SSCs, though each stem cell type is still in contact with the hub cells. In this way, the stem cell niche consists of these three cell types, as not only do the hub cells regulate GSC and SSC behaviour, but the stem cells also regulate the activity of each other. The Drosophila testis GSC niche has proven a valuable model system for examining a wide range of cellular processes and signalling pathways.[27]

Outside the testis GSC niche edit

The process of spermatogenesis begins when the GSCs divide asymmetrically, producing a GSC that maintains hub contact, and a gonialblast that exits the niche. The SSCs divide with their GSC partner, and their non-mitotic progeny, the somatic cyst cells (SCCs, a.k.a. cyst cells) will enclose the gonialblast. The gonialblast then undergoes four rounds of synchronous, transit-amplifying divisions with incomplete cytokinesis to produce a sixteen-cell spermatogonial cyst. This spermatogonial cyst then differentiates and grows into a spermatocyte, which will eventually undergo meiosis and produce sperm.[27]

Molecular signalling in the testis GSC niche edit

The two main molecular signalling pathways regulating stem cell behaviour in the testis GSC niche are the Jak-STAT and BMP signalling pathways. Jak-STAT signalling originates in the hub cells, where the ligand Upd is secreted to the GSCs and SSCs.[28][29] This leads to activation of the Drosophila STAT, Stat92E, a transcription factor which effects GSC adhesion to the hub cells,[30] and SSC self-renewal via Zfh-1.[31] Jak-STAT signalling also influences the activation of BMP signalling, via the ligands Dpp and Gbb. These ligands are secreted into the GSCs from the SSCs and hub cells, activate BMP signalling, and suppress the expression of Bam, a differentiation factor.[32] Outside of the niche, gonialblasts no longer receive BMP ligands, and are free to begin their differentiation program. Other important signalling pathways include the MAPK and Hedgehog, which regulate germline enclosure [33] and somatic cell self-renewal,[34] respectively.

GSC niche in mouse testes edit

The murine GSC niche in males, also called spermatogonial stem cell (SSC) niche, is located in the basal region of seminiferous tubules in the testes. The seminiferous epithelium is composed of sertoli cells that are in contact with the basement membrane of the tubules, which separates the sertoli cells from the interstitial tissue below. This interstitial tissue comprises Leydig cells, macrophages, mesenchymal cells, capillary networks, and nerves.[35]

During development, primordial germ cells migrate into the seminiferous tubules and downward towards the basement membrane whilst remaining attached to the sertoli cells where they will subsequently differentiate into SSCs, also referred to as Asingle spermatogonia.[35][36] These SSCs can either self-renew or commit to differentiating into spermatozoa upon the proliferation of Asingle into Apaired spermatogonia. The 2 cells of Apaired spermatogonia remain attached by intercellular bridges and subsequently divide into Aaligned spermatogonia, which is made up of 4–16 connected cells. Aaligned spermatogonia then undergo meiosis I to form spermatocytes and meiosis II to form spermatids which will mature into spermatozoa.[37][38] This differentiation occurs along the longitudinal axis of sertoli cells, from the basement membrane to the apical lumen of the seminiferous tubules. However, sertoli cells form tight junctions that separate SSCs and spermatogonia in contact with the basement membrane from the spermatocytes and spermatids to create a basal and an adluminal compartment, whereby differentiating spermatocytes must traverse the tight junctions.[35][39] These tight junctions form the blood testis barrier (BTB) and have been suggested to play a role in isolating differentiated cells in the adluminal compartment from secreted factors by the interstitial tissue and vasculature neighboring the basal compartment.[35]

Molecular mechanisms of SSC maintenance and activity edit

Physical cues edit

The basement membrane of the seminiferous tubule is a modified form of extracellular matrix composed of fibronectin, collagens, and laminin.[35] β1- integrin is expressed on the surface of SSCs and is involved in their adhesion to the laminin component of the basement membrane although other adhesion molecules are likely also implicated in the attachment of SSCs to the basement membrane.[40] E cadherin expression on SSCs in mice, unlike in Drosophila, have been shown to be dispensable as the transplantation of cultured SSCs lacking E-cadherin are able to colonize host seminiferous tubules and undergo spermatogenesis.[41] In addition the blood testis barrier provides architectural support and is composed of tight junction components such as occludins, claudins and zonula occludens (ZOs) which show dynamic expression during spermatogenesis.[42] For example, claudin 11 has been shown to be a necessary component of these tight junctions as mice lacking this gene have a defective blood testis barrier and do not produce mature spermatozoa.[40]

Molecular signals regulating SSC renewal edit

GDNF (Glial cell-derived neurotrophic factor) is known to stimulate self-renewal of SSCs and is secreted by the sertoli cells under the influence of gonadotropin FSH. GDNF is a related member of the TGFβ superfamily of growth factors and when overexpressed in mice, an increase in undifferentiated spermatogonia was observed which led to the formation of germ tumours.[35][40] In corroboration for its role as a renewal factor, heterozygous knockout male mice for GDNF show decreased spermatogenesis that eventually leads to infertility.[40] In addition the supplementation of GDNF has been shown to extend the expansion of mouse SSCs in culture. However, the GDNF receptor c-RET and co-receptor GFRa1 are not solely expressed on the SSCs but also on Apaired and Aaligned, therefore showing that GDNF is a renewal factor for Asingle to Aaligned in general rather than being specific to the Asingle SSC population. FGF2 (Fibroblast growth factor −2), secreted by sertoli cells, has also been shown to influence the renewal of SSCs and undifferentiated spermatogonia in a similar manner to GDNF.[35]

Although sertoli cells appear to play a major role in renewal, it expresses receptors for testosterone that is secreted by Leydig cells whereas germ cells do not contain this receptor- thus alluding to an important role of Leydig cells upstream in mediating renewal. Leydig cells also produce CSF 1 (Colony stimulating factor −1) for which SSCs strongly express the receptor CSF1R.[37] When CSF 1 was added in culture with GDNF and FGF2 no further increase in proliferation was observed, however, the longer the germ cells remained in culture with CSF-1 the greater the SSC density observed when these germ cells were transplanted into host seminiferous tubules. This showed CSF 1 to be a specific renewal factor that tilts the SSCs towards renewal over differentiation, rather than affecting proliferation of SSCs and spermatogonia. GDNF, FGF 2 and CSF 1 have also been shown to influence self-renewal of stem cells in other mammalian tissues.[35]

Plzf (Promyelocytic leukaemia zinc finger) has also been implicated in regulating SSC self-renewal and is expressed by Asingle, Apaired and Aaligned spermatogonia. Plzf directly inhibits the transcription of a receptor, c-kit, in these early spermatogonia. However, its absence in late spermatogonia permits c-kit expression, which is subsequently activated by its ligand SCF (stem cell factor) secreted by sertoli cells, resulting in further differentiation. Also, the addition of BMP4 and Activin-A have shown to reduce self-renewal of SSCs in culture and increase stem cell differentiation, with BMP4 shown to increase the expression of c-kit.[37]

Aging of the SSC niche edit

Prolonged spermatogenesis relies on the maintenance of SSCs, however, this maintenance declines with age and leads to infertility. Mice between 12 and 14 months of age show decreased testis weight, reduced spermatogenesis and SSC content. Although stem cells are regarded as having the potential to infinitely replicate in vitro, factors provided by the niche are crucial in vivo. Indeed, serial transplantation of SSCs from male mice of different ages into young mice 3 months of age, whose endogenous spermatogenesis had been ablated, was used to estimate stem cell content given that each stem cell would generate a colony of spermatogenesis.[35][43] The results of this experiment showed that transplanted SSCs could be maintained far longer than their replicative lifespan for their age. In addition, a study also showed that SSCs from young fertile mice could not be maintained nor undergo spermatogenesis when transplanted into testes of old, infertile mice. Together, these results points towards a deterioration of the SSC niche itself with aging rather than the loss of intrinsic factors in the SSC.[43]

Vertebrate adult stem cell niches edit

Hematopoietic stem cell niche edit

Vertebrate hematopoietic stem cells niche in the bone marrow is formed by cells subendosteal osteoblasts, sinusoidal endothelial cells and bone marrow stromal (also sometimes called reticular) cells which includes a mix of fibroblastoid, monocytic and adipocytic cells (which comprise marrow adipose tissue).[1]

Hair follicle stem cell niche edit

The hair follicle stem cell niche is one of the more closely studied niches thanks to its relative accessibility and role in important diseases such as melanoma. The bulge area at the junction of arrector pili muscle to the hair follicle sheath has been shown to host the skin stem cells which can contribute to all epithelial skin layers. There cells are maintained by signaling in concert with niche cells – signals include paracrine (e.g. sonic hedgehog), autocrine and juxtacrine signals.[44] The bulge region of the hair follicle relies on these signals to maintain the stemness of the cells. Fate mapping or cell lineage tracing has shown that Keratin 15 positive stem cells' progeny participate in all epithelial lineages.[45] The follicle undergoes cyclic regeneration in which these stem cells migrate to various regions and differentiate into the appropriate epithelial cell type. Some important signals in the hair follicle stem cell niche produced by the mesenchymal dermal papilla or the bulge include BMP, TGF-β and Fibroblast growth factor (FGF) ligands and Wnt inhibitors.[46] While, Wnt signaling pathways and β-catenin are important for stem cell maintenance,[47] over-expression of β-catenin in hair follicles induces improper hair growth. Therefore, these signals such as Wnt inhibitors produced by surrounding cells are important to maintain and facilitate the stem cell niche.[48]

Intestinal stem cell niche edit

Intestinal organoids have been used to study intestinal stem cell niches. An intestinal organoid culture can be used to indirectly assess the effect of the manipulation on the stem cells through assessing the organoid's survival and growth. Research using intestinal organoids have demonstrated that the survival of intestinal stem cells is improved by the presence of neurons and fibroblasts,[49] and through the administration of IL-22.[50]

Cardiovascular stem cell niche edit

Cardiovascular stem cell niches can be found within the right ventricular free wall, atria and outflow tracks of the heart. They are composed of Isl1+/Flk1+ cardiac progenitor cells (CPCs) that are localized into discrete clusters within a ColIV and laminin extracellular matrix (ECM). ColI and fibronectin are predominantly found outside the CPC clusters within the myocardium. Immunohistochemical staining has been used to demonstrate that differentiating CPCs, which migrate away from the progenitor clusters and into the ColI and fibronectin ECM surrounding the niche, down-regulate Isl1 while up-regulating mature cardiac markers such as troponin C.[51] There is a current controversy over the role of Isl1+ cells in the cardiovascular system. While major publications have identified these cells as CPC's and have found a very large number in the murine and human heart, recent publications have found very few Isl1+ cells in the murine fetal heart and attribute their localization to the sinoatrial node,[52] which is known as an area that contributes to heart pacemaking. The role of these cells and their niche are under intense research and debate.[citation needed]

Neural stem cell niche edit

Neural stem cell niches are divided in two : the Subependymal zone (SEZ) and the Subgranular zone (SGZ).

The SEZ is a thin area beneath the ependymal cell layer that contains three types of neural stem cells : infrequently dividing neural stem cells (NSCs), rapidly dividing transit amplifying precursors (TaPs) and neuroblasts (NBs). The SEZ extracellular matrix (ECM) has significant differences in composition compared to surrounding tissues. Recently, it was described that progenitor cells, NSCs, TaPs and NBs were attached to ECM structures called Fractones.[53] These structures are rich in laminin, collagen and heparan sulfate proteoglycans.[54] Other ECM molecules, such as tenascin-C, MMPs and different proteoglycans are also implicated in the neural stem cell niche.[55]

Cancer stem cell niche edit

Cancer tissue is morphologically heterogenous, not only due to the variety of cell types present, endothelial, fibroblast and various immune cells, but cancer cells themselves are not a homogenous population either.[citation needed]

In accordance with the hierarchy model of tumours, the cancer stem cells (CSC) are maintained by biochemical and physical contextual signals emanating from the microenvironment, called the cancer stem cell niche.[56] The CSC niche is very similar to normal stem cells niche (embryonic stem cell (ESC), Adult Stem Cell ASC) in function (maintaining of self-renewal, undifferentiated state and ability to differentiate) and in signalling pathways (Activin/Noda, Akt/PTEN, JAK/STAT, PI3-K, TGF-β, Wnt and BMP).[57] It is hypothesized that CSCs arise form aberrant signalling of the microenvironment and participates not only in providing survival signals to CSCs but also in metastasis by induction of epithelial-mesenchymal transition (EMT).[citation needed]

Hypoxia edit

Hypoxic condition in stem cell niches (ESC, ASC or CSC) is necessary for maintaining stem cells in an undifferentiated state and also for minimizing DNA damage via oxidation. The maintaining of the hypoxic state is under control of Hypoxia-Inducible transcription Factors (HIFs).[58] HIFs contribute to tumour progression, cell survival and metastasis by regulation of target genes as VEGF, GLUT-1, ADAM-1, Oct4 and Notch.[57]

Hypoxia in the CSC niche edit

Hypoxia plays an important role in the regulation of cancer stem cell niches and EMT through the promotion of HIFs.[59] These HIFs help maintain cancer stem cell niches by regulating important stemness genes such as Oct4, Nanog, SOX2, Klf4, and cMyc.[60][61] HIFs also regulate important tumor suppressor genes such as p53 and genes that promote metastasis.[62][63] Although HIFs increase the survival of cells by decreasing the effects of oxidative stress, they have also been shown to decrease factors such as RAD51 and H2AX that maintain genomic stability.[64] In the hypoxic condition there is an increase of intracellular Reactive Oxygen Species (ROS) which also promote CSCs survival via stress response.[65][66] ROS stabilizes HIF-1α which promotes the Met proto-oncogene, which drives metastasis or motogenic escape in melanoma cells.[67] All of these factors contribute to a cancer stem cell phenotype which is why it is often referred to as a hypoxic stem cell niche. Hypoxic environments are often found in tumors where the cells are dividing faster that angiogenesis can occur. It is important to study hypoxia as an aspect of cancer because hypoxic environments have been shown to be resistant to radiation therapy.[68] Radiation has been shown to increase the amounts of HIF-1.[69] EMT induction by hypoxia though interactions between HIF-1α and ROS is crucial for metastasis in cancers such as melanoma. It has been found that many genes associated with melanoma are regulated by hypoxia such as MXI1, FN1, and NME1.[70]

Epithelial–mesenchymal transition edit

Epithelial–mesenchymal transition is a morphogenetic process, normally occurs in embryogenesis that is "hijacked" by cancer stem cells by detaching from their primary place and migrating to another one. The dissemination is followed by reverse transition so-called Epithelial-Mesenchymal Transition (EMT). This process is regulated by CSCs microenvironment via the same signalling pathways as in embryogenesis using the growth factors (TGF-β, PDGF, EGF), cytokine IL-8 and extracellular matrix components. These growth factors' interactions through intracellular signal transducers like β-catenin has been shown to induce metastatic potential.[71][72] A characteristic of EMT is loss of the epithelial markers (E-cadherin, cytokeratins, claudin, occluding, desmoglein, desmocolin) and gain of mesenchymal markers (N-cadherin, vimentin, fibronectin).[73]

There is also certain degree of similarity in homing-mobilization of normal stem cells and metastasis-invasion of cancer stem cells. There is an important role of Matrix MetalloProteinases (MMP), the principal extracellular matrix degrading enzymes, thus for example matrix metalloproteinase-2 and −9 are induced to expression and secretion by stromal cells during metastasis of colon cancer via direct contact or paracrine regulation. The next sharing molecule is Stromal cell-Derived Factor-1 (SDF-1).[73][74]

Inflammation edit

The EMT and cancer progression can be triggered also by chronic inflammation. The main roles have molecules (IL-6, IL-8, TNF-α, NFκB, TGF-β, HIF-1α) which can regulate both processes through regulation of downstream signalling that overlapping between EMT and inflammation.[57] The downstream pathways involving in regulation of CSCs are Wnt, SHH, Notch, TGF-β, RTKs-EGF, FGF, IGF, HGF.

NFκB regulates the EMT, migration and invasion of CSCs through Slug, Snail and Twist. The activation of NFκB leads to increase not only in production of IL-6, TNF-α and SDF-1 but also in delivery of growth factors.

The source of the cytokine production are lymphocytes (TNF-α), Mesenchymal Stem Cells (SDF-1, IL-6, IL8).

Interleukin 6 mediates activation of STAT3. The high level of STAT3 was described in isolated CSCs from liver, bone, cervical and brain cancer. The inhibition of STAT3 results in dramatic reduction in their formation. Generally IL-6 contributes a survival advantage to local stem cells and thus facilitates tumorigenesis.[57]

SDF-1α secreted from Mesenchymal Stem Cells (MSCs) has important role in homing and maintenance of Hematopoietic Stem Cell (HSC) in bone marrow niche but also in homing and dissemination of CSC.[74]

Angiogenesis edit

Hypoxia is a main stimulant for angiogenesis, with HIF-1α being the primary mediator. Angiogenesis induced by hypoxic conditions is called an "Angiogenic switch". HIF-1 promotes expression of several angiogenic factors: Vascular Endothelial Growth Factor (VEGF), basic Fibroblast Growth Factor (bFGF), Placenta-Like Growth Factor (PLGF), Platelet-Derived Growth Factor (PDGF) and Epidermal Growth Factor. But there is evidence that the expression of angiogenic agens by cancer cells can also be HIF-1 independent. It seems that there is an important role of Ras protein, and that intracellular levels of calcium regulate the expression of angiogenic genes in response to hypoxia.[73]

The angiogenic switch downregulates angiogenesis suppressor proteins, such as thrombospondin, angiostatin, endostatin and tumstatin. Angiogenesis is necessary for the primary tumour growth.[citation needed]

Injury-induced edit

During injury, support cells are able to activate a program for repair, recapitulating aspects of development in the area of damage. These areas become permissive for stem cell renewal, migration and differentiation. For instance in the CNS, injury is able to activate a developmental program in astrocytes that allow them to express molecules that support stem cells such as chemokines i.e. SDF-1[75] and morphogens such as sonic hedgehog.[76]

Extracellular Matrix Mimicking Strategies For Stem Cell Niche edit

It is evident that biophysio-chemical characteristics of ECM such as composition, shape, topography, stiffness, and mechanical strength can control the stem cell behavior. These ECM factors are equally important when stem cells are grown in vitro. Given a choice between niche cell-stem cell interaction and ECM-stem cell interaction, mimicking ECM is preferred as that can be precisely controlled by scaffold fabrication techniques, processing parameters or post-fabrication modifications. In order to mimic, it is essential to understand natural properties of ECM and their role in stem cell fate processes. Various studies involving different types of scaffolds that regulate stem cells fate by mimicking these ECM properties have been done.[2])

[77]

References edit

  1. ^ a b Birbrair A, Frenette PS (April 2016). "Niche heterogeneity in the bone marrow". Annals of the New York Academy of Sciences. 1370 (1): 82–96. Bibcode:2016NYASA1370...82B. doi:10.1111/nyas.13016. PMC 4938003. PMID 27015419.
  2. ^ a b c Jhala D (2015). "A review on extracellular matrix mimicking strategies for an artificial stem cell niche". Polymer Reviews. 55 (4): 561–595. doi:10.1080/15583724.2015.1040552. S2CID 94588894.
  3. ^ Scadden DT (June 2006). "The stem-cell niche as an entity of action". Nature. 441 (7097): 1075–1079. Bibcode:2006Natur.441.1075S. doi:10.1038/nature04957. PMID 16810242. S2CID 4418385.
  4. ^ Ritsma L, Ellenbroek SI, Zomer A, Snippert HJ, de Sauvage FJ, Simons BD, et al. (March 2014). "Intestinal crypt homeostasis revealed at single-stem-cell level by in vivo live imaging". Nature. 507 (7492): 362–365. Bibcode:2014Natur.507..362R. doi:10.1038/nature12972. PMC 3964820. PMID 24531760.
  5. ^ Rompolas P, Mesa KR, Greco V (October 2013). "Spatial organization within a niche as a determinant of stem-cell fate". Nature. 502 (7472): 513–518. Bibcode:2013Natur.502..513R. doi:10.1038/nature12602. PMC 3895444. PMID 24097351.
  6. ^ Shahriyari L, Komarova NL (July 2015). "The role of the bi-compartmental stem cell niche in delaying cancer". Physical Biology. 12 (5): 055001. Bibcode:2015PhBio..12e5001S. doi:10.1088/1478-3975/12/5/055001. PMID 26228740. S2CID 7171931.
  7. ^ Cannataro VL, McKinley SA, St Mary CM (April 2016). "The implications of small stem cell niche sizes and the distribution of fitness effects of new mutations in aging and tumorigenesis". Evolutionary Applications. 9 (4): 565–582. doi:10.1111/eva.12361. PMC 4831459. PMID 27099622.
  8. ^ Cannataro VL, McKinley SA, St Mary CM (July 2017). "The evolutionary trade-off between stem cell niche size, aging, and tumorigenesis". Evolutionary Applications. 10 (6): 590–602. doi:10.1111/eva.12476. PMC 5469181. PMID 28616066.
  9. ^ Li L, Xie T (2005). "Stem cell niche: structure and function". Annual Review of Cell and Developmental Biology. 21: 605–631. doi:10.1146/annurev.cellbio.21.012704.131525. PMID 16212509.
  10. ^ a b c d Xie T, Spradling AC (October 2000). "A niche maintaining germ line stem cells in the Drosophila ovary". Science. 290 (5490): 328–330. Bibcode:2000Sci...290..328X. doi:10.1126/science.290.5490.328. PMID 11030649.
  11. ^ Lin H, Yue L, Spradling AC (April 1994). "The Drosophila fusome, a germline-specific organelle, contains membrane skeletal proteins and functions in cyst formation". Development. 120 (4): 947–956. doi:10.1242/dev.120.4.947. PMID 7600970.
  12. ^ Ting, X., 2013. Control of germline stem cell self‐renewal and differentiation in the Drosophila ovary: concerted actions of niche signals and intrinsic factors. Wiley Interdisciplinary Reviews: Developmental Biology, 2(2), pp.261-273.
  13. ^ Zhang, H. and Cai, Y., 2020. Signal transduction pathways regulating Drosophila ovarian germline stem cells. Current opinion in insect science, 37, pp.1-7.
  14. ^ Song X, Wong MD, Kawase E, Xi R, Ding BC, McCarthy JJ, Xie T (March 2004). "Bmp signals from niche cells directly repress transcription of a differentiation-promoting gene, bag of marbles, in germline stem cells in the Drosophila ovary". Development. 131 (6): 1353–1364. doi:10.1242/dev.01026. PMID 14973291.
  15. ^ Chen D, McKearin D (October 2003). "Dpp signaling silences bam transcription directly to establish asymmetric divisions of germline stem cells". Current Biology. 13 (20): 1786–1791. doi:10.1016/j.cub.2003.09.033. PMID 14561403.
  16. ^ Cox DN, Chao A, Lin H (February 2000). "piwi encodes a nucleoplasmic factor whose activity modulates the number and division rate of germline stem cells". Development. 127 (3): 503–514. doi:10.1242/dev.127.3.503. PMID 10631171.
  17. ^ Song X, Zhu CH, Doan C, Xie T (June 2002). "Germline stem cells anchored by adherens junctions in the Drosophila ovary niches". Science. 296 (5574): 1855–1857. Bibcode:2002Sci...296.1855S. doi:10.1126/science.1069871. PMID 12052957. S2CID 25830121.
  18. ^ Bogard N, Lan L, Xu J, Cohen RS (October 2007). "Rab11 maintains connections between germline stem cells and niche cells in the Drosophila ovary". Development. 134 (19): 3413–3418. doi:10.1242/dev.008466. PMID 17715175.
  19. ^ Gilboa L, Forbes A, Tazuke SI, Fuller MT, Lehmann R (December 2003). "Germ line stem cell differentiation in Drosophila requires gap junctions and proceeds via an intermediate state". Development. 130 (26): 6625–6634. doi:10.1242/dev.00853. PMID 14660550.
  20. ^ Drummond-Barbosa D, Spradling AC (March 2001). "Stem cells and their progeny respond to nutritional changes during Drosophila oogenesis". Developmental Biology. 231 (1): 265–278. doi:10.1006/dbio.2000.0135. PMID 11180967.
  21. ^ a b Hsu HJ, Drummond-Barbosa D (January 2009). "Insulin levels control female germline stem cell maintenance via the niche in Drosophila". Proceedings of the National Academy of Sciences of the United States of America. 106 (4): 1117–1121. Bibcode:2009PNAS..106.1117H. doi:10.1073/pnas.0809144106. PMC 2633547. PMID 19136634.
  22. ^ Margolis J, Spradling A (November 1995). "Identification and behavior of epithelial stem cells in the Drosophila ovary". Development. 121 (11): 3797–3807. doi:10.1242/dev.121.11.3797. PMID 8582289.
  23. ^ Xie T, Spradling AC (July 1998). "decapentaplegic is essential for the maintenance and division of germline stem cells in the Drosophila ovary". Cell. 94 (2): 251–260. doi:10.1016/s0092-8674(00)81424-5. PMID 9695953.
  24. ^ Kai T, Spradling A (April 2003). "An empty Drosophila stem cell niche reactivates the proliferation of ectopic cells". Proceedings of the National Academy of Sciences of the United States of America. 100 (8): 4633–4638. Bibcode:2003PNAS..100.4633K. doi:10.1073/pnas.0830856100. PMC 153607. PMID 12676994.
  25. ^ Zhao R, Xuan Y, Li X, Xi R (June 2008). "Age-related changes of germline stem cell activity, niche signaling activity and egg production in Drosophila". Aging Cell. 7 (3): 344–354. doi:10.1111/j.1474-9726.2008.00379.x. PMID 18267001.
  26. ^ a b Pan L, Chen S, Weng C, Call G, Zhu D, Tang H, et al. (October 2007). "Stem cell aging is controlled both intrinsically and extrinsically in the Drosophila ovary". Cell Stem Cell. 1 (4): 458–469. doi:10.1016/j.stem.2007.09.010. PMID 18371381.
  27. ^ a b La Marca JE, Somers WG (2014). "The Drosophila gonads: models for stem cell proliferation, self-renewal, and differentiation". AIMS Genetics. 1 (1): 55–80. doi:10.3934/genet.2014.1.55.
  28. ^ Kiger AA, Jones DL, Schulz C, Rogers MB, Fuller MT (December 2001). "Stem cell self-renewal specified by JAK-STAT activation in response to a support cell cue". Science. 294 (5551): 2542–2545. Bibcode:2001Sci...294.2542K. doi:10.1126/science.1066707. PMID 11752574. S2CID 206506814.
  29. ^ Tulina N, Matunis E (December 2001). "Control of stem cell self-renewal in Drosophila spermatogenesis by JAK-STAT signaling". Science. 294 (5551): 2546–2549. Bibcode:2001Sci...294.2546T. doi:10.1126/science.1066700. PMID 11752575. S2CID 43266825.
  30. ^ Leatherman JL, Dinardo S (August 2010). "Germline self-renewal requires cyst stem cells and stat regulates niche adhesion in Drosophila testes". Nature Cell Biology. 12 (8): 806–811. doi:10.1038/ncb2086. PMC 2917891. PMID 20622868.
  31. ^ Leatherman JL, Dinardo S (July 2008). "Zfh-1 controls somatic stem cell self-renewal in the Drosophila testis and nonautonomously influences germline stem cell self-renewal". Cell Stem Cell. 3 (1): 44–54. doi:10.1016/j.stem.2008.05.001. PMC 2601693. PMID 18593558.
  32. ^ Kawase E, Wong MD, Ding BC, Xie T (March 2004). "Gbb/Bmp signaling is essential for maintaining germline stem cells and for repressing bam transcription in the Drosophila testis". Development. 131 (6): 1365–1375. doi:10.1242/dev.01025. PMID 14973292.
  33. ^ Sarkar A, Parikh N, Hearn SA, Fuller MT, Tazuke SI, Schulz C (July 2007). "Antagonistic roles of Rac and Rho in organizing the germ cell microenvironment". Current Biology. 17 (14): 1253–1258. doi:10.1016/j.cub.2007.06.048. PMID 17629483.
  34. ^ Michel M, Kupinski AP, Raabe I, Bökel C (August 2012). "Hh signalling is essential for somatic stem cell maintenance in the Drosophila testis niche". Development. 139 (15): 2663–2669. doi:10.1242/dev.075242. PMID 22745310.
  35. ^ a b c d e f g h i Oatley JM, Brinster RL (April 2012). "The germline stem cell niche unit in mammalian testes". Physiological Reviews. 92 (2): 577–595. doi:10.1152/physrev.00025.2011. PMC 3970841. PMID 22535892.
  36. ^ Griswold MD, Oatley JM (January 2013). "Concise review: Defining characteristics of mammalian spermatogenic stem cells". Stem Cells. 31 (1): 8–11. doi:10.1002/stem.1253. PMC 5312674. PMID 23074087.
  37. ^ a b c de Rooij DG (August 2009). "The spermatogonial stem cell niche". Microscopy Research and Technique. 72 (8): 580–585. doi:10.1002/jemt.20699. PMID 19263493.
  38. ^ Bowles J, Koopman P (October 2007). "Retinoic acid, meiosis and germ cell fate in mammals". Development. 134 (19): 3401–3411. doi:10.1242/dev.001107. PMID 17715177.
  39. ^ Hess RA, de Franca LR (2009). "Spermatogenesis and Cycle of the Seminiferous Epithelium". In Cheng CY (ed.). Molecular Mechanisms in Spermatogenesis. Advances in Experimental Medicine and Biology. Vol. 636. pp. 1–15. doi:10.1007/978-0-387-09597-4_1. ISBN 978-0-387-09597-4. PMID 19856159.
  40. ^ a b c d Kanatsu-Shinohara M, Shinohara T (2013). "Spermatogonial stem cell self-renewal and development". Annual Review of Cell and Developmental Biology. 29: 163–187. doi:10.1146/annurev-cellbio-101512-122353. PMID 24099084.
  41. ^ Yoshida S (2011). "Stem Cell Niche System in Mouse Spermatogenesis". Male Germline Stem Cells: Developmental and Regenerative Potential. Vol. 2011. pp. 159–175. doi:10.1007/978-1-61737-973-4_8. ISBN 978-1-61737-972-7. {{cite book}}: |journal= ignored (help)
  42. ^ Chihara M, Otsuka S, Ichii O, Hashimoto Y, Kon Y (July 2010). "Molecular dynamics of the blood-testis barrier components during murine spermatogenesis". Molecular Reproduction and Development. 77 (7): 630–639. doi:10.1002/mrd.21200. PMID 20578065. S2CID 21630147.
  43. ^ a b Ryu BY, Orwig KE, Oatley JM, Avarbock MR, Brinster RL (June 2006). "Effects of aging and niche microenvironment on spermatogonial stem cell self-renewal". Stem Cells. 24 (6): 1505–1511. doi:10.1634/stemcells.2005-0580. PMC 5501308. PMID 16456131.
  44. ^ Aloni-Grinstein R, Shetzer Y, Kaufman T, Rotter V (August 2014). "p53: the barrier to cancer stem cell formation". FEBS Letters. 588 (16): 2580–2589. doi:10.1016/j.febslet.2014.02.011. PMID 24560790.
  45. ^ Morris RJ, Liu Y, Marles L, Yang Z, Trempus C, Li S, et al. (April 2004). "Capturing and profiling adult hair follicle stem cells". Nature Biotechnology. 22 (4): 411–417. doi:10.1038/nbt950. PMID 15024388. S2CID 9257482.
  46. ^ Rompolas P, Greco V (2014). "Stem cell dynamics in the hair follicle niche". Seminars in Cell & Developmental Biology. 25–26: 34–42. doi:10.1016/j.semcdb.2013.12.005. PMC 3988239. PMID 24361866.
  47. ^ Hosseini V, Kalantary-Charvadeh A, Hajikarami M, Fayyazpour P, Rahbarghazi R, Totonchi M, Darabi M (October 2021). "A small molecule modulating monounsaturated fatty acids and Wnt signaling confers maintenance to induced pluripotent stem cells against endodermal differentiation". Stem Cell Research & Therapy. 12 (1): 550. doi:10.1186/s13287-021-02617-x. PMC 8532309. PMID 34674740.
  48. ^ Deschene ER, Myung P, Rompolas P, Zito G, Sun TY, Taketo MM, et al. (March 2014). "β-Catenin activation regulates tissue growth non-cell autonomously in the hair stem cell niche". Science. 343 (6177): 1353–1356. Bibcode:2014Sci...343.1353D. doi:10.1126/science.1248373. PMC 4096864. PMID 24653033.
  49. ^ Pastuła A, Middelhoff M, Brandtner A, Tobiasch M, Höhl B, Nuber AH, et al. (2016). "Three-Dimensional Gastrointestinal Organoid Culture in Combination with Nerves or Fibroblasts: A Method to Characterize the Gastrointestinal Stem Cell Niche". Stem Cells International. 2016: 3710836. doi:10.1155/2016/3710836. PMC 4677245. PMID 26697073.
  50. ^ Lindemans C, Mertelsmann A, Dudakov JA, Velardi E, Hua G, O'Connor M, et al. (2014). "IL-22 Administration Protects Intestinal Stem Cells from Gvhd". Biology of Blood and Marrow Transplantation. 20 (2): S53–S54. doi:10.1016/j.bbmt.2013.12.056.
  51. ^ Schenke-Layland K, Nsair A, Van Handel B, Angelis E, Gluck JM, Votteler M, et al. (April 2011). "Recapitulation of the embryonic cardiovascular progenitor cell niche". Biomaterials. 32 (11): 2748–2756. doi:10.1016/j.biomaterials.2010.12.046. PMC 3414535. PMID 21257198.
  52. ^ Weinberger F, Mehrkens D, Friedrich FW, Stubbendorff M, Hua X, Müller JC, et al. (May 2012). "Localization of Islet-1-positive cells in the healthy and infarcted adult murine heart". Circulation Research. 110 (10): 1303–1310. doi:10.1161/CIRCRESAHA.111.259630. PMC 5559221. PMID 22427341.
  53. ^ Luo, Pifu; Moritani, Masayuki; Dessem, Dean (2001-07-02). "Jaw-muscle spindle afferent pathways to the trigeminal motor nucleus in the rat". The Journal of Comparative Neurology. 435 (3): 341–353. doi:10.1002/cne.1034. ISSN 0021-9967. PMID 11406816. S2CID 36398505.
  54. ^ Mercier, Frederic (2016). "Fractones: extracellular matrix niche controlling stem cell fate and growth factor activity in the brain in health and disease". Cellular and Molecular Life Sciences. 73 (24): 4661–4674. doi:10.1007/s00018-016-2314-y. ISSN 1420-682X. PMID 27475964. S2CID 28119663.
  55. ^ "The neural stem cell microenvironment | StemBook". www.stembook.org. Retrieved 2022-04-29.
  56. ^ van de Stolpe A (2013). "On the origin and destination of cancer stem cells: a conceptual evaluation". American Journal of Cancer Research. 3 (1): 107–116. PMC 3555199. PMID 23359140.
  57. ^ a b c d Cabarcas SM, Mathews LA, Farrar WL (November 2011). "The cancer stem cell niche--there goes the neighborhood?". International Journal of Cancer. 129 (10): 2315–2327. doi:10.1002/ijc.26312. PMC 6953416. PMID 21792897.
  58. ^ Borovski T, De Sousa E, Melo F, Vermeulen L, Medema JP (February 2011). "Cancer stem cell niche: the place to be". Cancer Research. 71 (3): 634–639. doi:10.1158/0008-5472.CAN-10-3220. PMID 21266356.
  59. ^ Peitzsch C, Perrin R, Hill RP, Dubrovska A, Kurth I (August 2014). "Hypoxia as a biomarker for radioresistant cancer stem cells". International Journal of Radiation Biology. 90 (8): 636–652. doi:10.3109/09553002.2014.916841. PMID 24844374. S2CID 25813277.
  60. ^ Covello KL, Kehler J, Yu H, Gordan JD, Arsham AM, Hu CJ, et al. (March 2006). "HIF-2alpha regulates Oct-4: effects of hypoxia on stem cell function, embryonic development, and tumor growth". Genes & Development. 20 (5): 557–570. doi:10.1101/gad.1399906. PMC 1410808. PMID 16510872.
  61. ^ Keith B, Simon MC (May 2007). "Hypoxia-inducible factors, stem cells, and cancer". Cell. 129 (3): 465–472. doi:10.1016/j.cell.2007.04.019. PMC 3150586. PMID 17482542.
  62. ^ Bertout JA, Majmundar AJ, Gordan JD, Lam JC, Ditsworth D, Keith B, et al. (August 2009). "HIF2alpha inhibition promotes p53 pathway activity, tumor cell death, and radiation responses". Proceedings of the National Academy of Sciences of the United States of America. 106 (34): 14391–14396. Bibcode:2009PNAS..10614391B. doi:10.1073/pnas.0907357106. PMC 2726037. PMID 19706526.
  63. ^ Liu L, Zhu XD, Wang WQ, Shen Y, Qin Y, Ren ZG, et al. (May 2010). "Activation of beta-catenin by hypoxia in hepatocellular carcinoma contributes to enhanced metastatic potential and poor prognosis". Clinical Cancer Research. 16 (10): 2740–2750. doi:10.1158/1078-0432.CCR-09-2610. PMID 20460486.
  64. ^ Bindra RS, Schaffer PJ, Meng A, Woo J, Måseide K, Roth ME, et al. (October 2004). "Down-regulation of Rad51 and decreased homologous recombination in hypoxic cancer cells". Molecular and Cellular Biology. 24 (19): 8504–8518. doi:10.1128/MCB.24.19.8504-8518.2004. PMC 516750. PMID 15367671.
  65. ^ Singh S, Brocker C, Koppaka V, Chen Y, Jackson BC, Matsumoto A, et al. (March 2013). "Aldehyde dehydrogenases in cellular responses to oxidative/electrophilic stress". Free Radical Biology & Medicine. 56: 89–101. doi:10.1016/j.freeradbiomed.2012.11.010. PMC 3631350. PMID 23195683.
  66. ^ Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, et al. (April 2009). "Association of reactive oxygen species levels and radioresistance in cancer stem cells". Nature. 458 (7239): 780–783. Bibcode:2009Natur.458..780D. doi:10.1038/nature07733. PMC 2778612. PMID 19194462.
  67. ^ Comito G, Calvani M, Giannoni E, Bianchini F, Calorini L, Torre E, et al. (August 2011). "HIF-1α stabilization by mitochondrial ROS promotes Met-dependent invasive growth and vasculogenic mimicry in melanoma cells". Free Radical Biology & Medicine. 51 (4): 893–904. doi:10.1016/j.freeradbiomed.2011.05.042. hdl:2158/496457. PMID 21703345.
  68. ^ Brown JM (2007). "Tumor Hypoxia in Cancer Therapy". Oxygen Biology and Hypoxia. Methods in Enzymology. Vol. 435. pp. 297–321. doi:10.1016/S0076-6879(07)35015-5. ISBN 9780123739704. PMID 17998060.
  69. ^ Moeller BJ, Cao Y, Li CY, Dewhirst MW (May 2004). "Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules". Cancer Cell. 5 (5): 429–441. doi:10.1016/s1535-6108(04)00115-1. PMID 15144951.
  70. ^ Olbryt M, Habryka A, Tyszkiewicz T, Rusin A, Cichoń T, Jarząb M, Krawczyk Z (October 2011). "Melanoma-associated genes, MXI1, FN1, and NME1, are hypoxia responsive in murine and human melanoma cells". Melanoma Research. 21 (5): 417–425. doi:10.1097/CMR.0b013e328348db2f. PMID 21912348. S2CID 33171556.
  71. ^ Moustakas A, Heldin CH (October 2007). "Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression". Cancer Science. 98 (10): 1512–1520. doi:10.1111/j.1349-7006.2007.00550.x. PMID 17645776. S2CID 23032956.
  72. ^ Zhou B, Liu Y, Kahn M, Ann DK, Han A, Wang H, et al. (March 2012). "Interactions between β-catenin and transforming growth factor-β signaling pathways mediate epithelial-mesenchymal transition and are dependent on the transcriptional co-activator cAMP-response element-binding protein (CREB)-binding protein (CBP)". The Journal of Biological Chemistry. 287 (10): 7026–7038. doi:10.1074/jbc.M111.276311. PMC 3293544. PMID 22241478.
  73. ^ a b c Gout S, Huot J (December 2008). "Role of cancer microenvironment in metastasis: focus on colon cancer". Cancer Microenvironment. 1 (1): 69–83. doi:10.1007/s12307-008-0007-2. PMC 2654352. PMID 19308686.
  74. ^ a b Li L, Neaves WB (May 2006). "Normal stem cells and cancer stem cells: the niche matters". Cancer Research. 66 (9): 4553–4557. doi:10.1158/0008-5472.CAN-05-3986. PMID 16651403.
  75. ^ Imitola J, Raddassi K, Park KI, Mueller FJ, Nieto M, Teng YD, et al. (December 2004). "Directed migration of neural stem cells to sites of CNS injury by the stromal cell-derived factor 1alpha/CXC chemokine receptor 4 pathway". Proceedings of the National Academy of Sciences of the United States of America. 101 (52): 18117–18122. Bibcode:2004PNAS..10118117I. doi:10.1073/pnas.0408258102. PMC 536055. PMID 15608062.
  76. ^ Wang Y, Imitola J, Rasmussen S, O'Connor KC, Khoury SJ (October 2008). "Paradoxical dysregulation of the neural stem cell pathway sonic hedgehog-Gli1 in autoimmune encephalomyelitis and multiple sclerosis". Annals of Neurology. 64 (4): 417–427. doi:10.1002/ana.21457. PMC 2757750. PMID 18991353.
  77. ^ Vishwakarma A (2017-04-01). Biology and Engineering of Stem Cell Niches. Academic Press, 2017. ISBN 9780128027561.

stem, cell, niche, this, article, needs, additional, citations, verification, please, help, improve, this, article, adding, citations, reliable, sources, unsourced, material, challenged, removed, find, sources, news, newspapers, books, scholar, jstor, october,. This article needs additional citations for verification Please help improve this article by adding citations to reliable sources Unsourced material may be challenged and removed Find sources Stem cell niche news newspapers books scholar JSTOR October 2015 Learn how and when to remove this message Stem cell niche refers to a microenvironment within the specific anatomic location where stem cells are found which interacts with stem cells to regulate cell fate 1 The word niche can be in reference to the in vivo or in vitro stem cell microenvironment During embryonic development various niche factors act on embryonic stem cells to alter gene expression and induce their proliferation or differentiation for the development of the fetus Within the human body stem cell niches maintain adult stem cells in a quiescent state but after tissue injury the surrounding micro environment actively signals to stem cells to promote either self renewal or differentiation to form new tissues Several factors are important to regulate stem cell characteristics within the niche cell cell interactions between stem cells as well as interactions between stem cells and neighbouring differentiated cells interactions between stem cells and adhesion molecules extracellular matrix components the oxygen tension growth factors cytokines and the physicochemical nature of the environment including the pH ionic strength e g Ca2 concentration and metabolites like ATP are also important 2 The stem cells and niche may induce each other during development and reciprocally signal to maintain each other during adulthood Scientists are studying the various components of the niche and trying to replicate the in vivo niche conditions in vitro 2 This is because for regenerative therapies cell proliferation and differentiation must be controlled in flasks or plates so that sufficient quantity of the proper cell type are produced prior to being introduced back into the patient for therapy Human embryonic stem cells are often grown in fibrotastic growth factor 2 containing fetal bovine serum supplemented media They are grown on a feeder layer of cells which is believed to be supportive in maintaining the pluripotent characteristics of embryonic stem cells However even these conditions may not truly mimic in vivo niche conditions Adult stem cells remain in an undifferentiated state throughout adult life However when they are cultured in vitro they often undergo an aging process in which their morphology is changed and their proliferative capacity is decreased It is believed that correct culturing conditions of adult stem cells needs to be improved so that adult stem cells can maintain their stemness over time citation needed A Nature Insight review defines niche as follows Stem cell populations are established in niches specific anatomic locations that regulate how they participate in tissue generation maintenance and repair The niche saves stem cells from depletion while protecting the host from over exuberant stem cell proliferation It constitutes a basic unit of tissue physiology integrating signals that mediate the balanced response of stem cells to the needs of organisms Yet the niche may also induce pathologies by imposing aberrant function on stem cells or other targets The interplay between stem cells and their niche creates the dynamic system necessary for sustaining tissues and for the ultimate design of stem cell therapeutics The simple location of stem cells is not sufficient to define a niche The niche must have both anatomic and functional dimensions 3 Contents 1 History 2 The architecture of the stem cell niche 3 Examples 3 1 Germline 3 2 GSC niche in Drosophila ovaries 3 2 1 Molecular mechanisms of GSC maintenance and activity 3 2 1 1 Local signals 3 2 1 2 Physical attachment of GSCs to cap cells 3 2 1 3 Systemic signals regulating GSCs 3 2 1 4 Renewal mechanisms 3 2 2 Stem cell aging 3 3 GSC niche in Drosophila testes 3 3 1 Outside the testis GSC niche 3 3 2 Molecular signalling in the testis GSC niche 3 4 GSC niche in mouse testes 3 4 1 Molecular mechanisms of SSC maintenance and activity 3 4 1 1 Physical cues 3 4 1 2 Molecular signals regulating SSC renewal 3 4 1 3 Aging of the SSC niche 3 5 Vertebrate adult stem cell niches 3 5 1 Hematopoietic stem cell niche 3 5 2 Hair follicle stem cell niche 3 5 3 Intestinal stem cell niche 3 5 4 Cardiovascular stem cell niche 3 5 5 Neural stem cell niche 4 Cancer stem cell niche 4 1 Hypoxia 4 1 1 Hypoxia in the CSC niche 4 2 Epithelial mesenchymal transition 4 3 Inflammation 4 4 Angiogenesis 4 5 Injury induced 5 Extracellular Matrix Mimicking Strategies For Stem Cell Niche 6 ReferencesHistory editThough the concept of stem cell niche was prevailing in vertebrates the first characterization of stem cell niche in vivo was worked out in Drosophila germinal development citation needed The architecture of the stem cell niche editBy continuous intravital imaging in mice researchers were able to explore the structure of the stem cell niche and to obtain the fate of individual stem cells SCs and their progeny over time in vivo In particular in intestinal crypt 4 two distinct groups of SCs have been identified the border stem cells located in the upper part of the niche at the interface with transit amplifying cells TAs and central stem cells located at the crypt base The proliferative potential of the two groups was unequal and correlated with the cells location central or border It was also shown that the two SC compartments acted in accord to maintain a constant cell population and a steady cellular turnover A similar dependence of self renewal potential on proximity to the niche border was reported in the context of hair follicle in an in vivo live imaging study 5 This bi compartmental structure of stem cell niche has been mathematically modeled to obtain the optimal architecture that leads to the maximum delay in double hit mutant production 6 They found that the bi compartmental SC architecture minimizes the rate of two hit mutant production compared to the single SC compartment model Moreover the minimum probability of double hit mutant generation corresponds to purely symmetric division of SCs with a large proliferation rate of border stem cells along with a small but non zero proliferation rate of central stem cells citation needed Stem cell niches harboring continuously dividing cells such as those located at the base of the intestinal gland are maintained at small population size This presents a challenge to the maintenance of multicellular tissues as small populations of asexually dividing individuals will accumulate deleterious mutations through genetic drift and succumb to mutational meltdown 7 Mathematical modeling of the intestinal gland reveals that the small population size within the stem cell niche minimizes the probability of carcinogenesis occurring anywhere at the expense of gradually accumulated deleterious mutations throughout organismal lifetime a process that contributes to tissue degradation and aging 8 Therefore the population size of the stem cell niche represents an evolutionary trade off between the probability of cancer formation and the rate of aging Examples editGermline edit Germline stem cells GSCs are found in organisms that continuously produce sperm and eggs until they are sterile These specialized stem cells reside in the GSC niche the initial site for gamete production which is composed of the GSCs somatic stem cells and other somatic cells In particular the GSC niche is well studied in the genetic model organism Drosophila melanogaster and has provided an extensive understanding of the molecular basis of stem cell regulation citation needed nbsp GSC niche in Drosophila melanogaster germarium GSC niche in Drosophila ovaries edit In Drosophila melanogaster the GSC niche resides in the anterior most region of each ovariole known as the germarium The GSC niche consists of necessary somatic cells terminal filament cells cap cells escort cells and other stem cells which function to maintain the GSCs 9 The GSC niche holds on average 2 3 GSCs which are directly attached to somatic cap cells and Escort stem cells which send maintenance signals directly to the GSCs 10 GSCs are easily identified through histological staining against vasa protein to identify germ cells and 1B1 protein to outline cell structures and a germline specific fusome structure Their physical attachment to the cap cells is necessary for their maintenance and activity 10 A GSC will divide asymmetrically to produce one daughter cystoblast which then undergoes 4 rounds of incomplete mitosis as it progresses down the ovariole through the process of oogenesis eventually emerging as a mature egg chamber the fusome found in the GSCs functions in cyst formation and may regulate asymmetrical cell divisions of the GSCs 11 Because of the abundant genetic tools available for use in Drosophila melanogaster and the ease of detecting GSCs through histological stainings researchers have uncovered several molecular pathways controlling GSC maintenance and activity 12 13 Molecular mechanisms of GSC maintenance and activity edit Local signals edit The Bone Morphogenetic Protein BMP ligands Decapentaplegic Dpp and Glass bottom boat Gbb ligand are directly signalled to the GSCs and are essential for GSC maintenance and self renewal 14 BMP signalling in the niche functions to directly repress expression of Bag of marbles Bam in GSCs which is up regulated in developing cystoblast cells 15 Loss of function of dpp in the niche results in de repression of Bam in GSCs resulting in rapid differentiation of the GSCs 10 Along with BMP signalling cap cells also signal other molecules to GSCs Yb and Piwi Both of these molecules are required non autonomously to the GSCs for proliferation piwi is also required autonomously in the GSCs for proliferation 16 In the germarium BMP signaling has a short range effect therefore the physical attachment of GSCs to cap cells is important for maintenance and activity citation needed Physical attachment of GSCs to cap cells edit The GSCs are physically attached to the cap cells by Drosophila E cadherin DE cadherin adherens junctions and if this physical attachment is lost GSCs will differentiate and lose their identity as a stem cell 10 The gene encoding DE cadherin shotgun shg and a gene encoding Beta catenin ortholog armadillo control this physical attachment 17 A GTPase molecule rab11 is involved in cell trafficking of DE cadherins Knocking out rab11 in GSCs results in detachment of GSCs from the cap cells and premature differentiation of GSCs 18 Additionally zero population growth zpg encoding a germline specific gap junction is required for germ cell differentiation 19 Systemic signals regulating GSCs edit Both diet and insulin like signaling directly control GSC proliferation in Drosophila melanogaster Increasing levels of Drosophila insulin like peptide DILP through diet results in increased GSC proliferation 20 Up regulation of DILPs in aged GSCs and their niche results in increased maintenance and proliferation 21 It has also been shown that DILPs regulate cap cell quantities and regulate the physical attachment of GSCs to cap cells 21 Renewal mechanisms edit There are two possible mechanisms for stem cell renewal symmetrical GSC division or de differentiation of cystoblasts Normally GSCs will divide asymmetrically to produce one daughter cystoblast but it has been proposed that symmetrical division could result in the two daughter cells remaining GSCs 22 23 If GSCs are ablated to create an empty niche and the cap cells are still present and sending maintenance signals differentiated cystoblasts can be recruited to the niche and de differentiate into functional GSCs 24 Stem cell aging edit As the Drosophila female ages the stem cell niche undergoes age dependent loss of GSC presence and activity These losses are thought to be caused in part by degradation of the important signaling factors from the niche that maintains GSCs and their activity Progressive decline in GSC activity contributes to the observed reduction in fecundity of Drosophila melanogaster at old age this decline in GSC activity can be partially attributed to a reduction of signaling pathway activity in the GSC niche 25 26 It has been found that there is a reduction in Dpp and Gbb signaling through aging In addition to a reduction in niche signaling pathway activity GSCs age cell autonomously In addition to studying the decline of signals coming from the niche GSCs age intrinsically there is age dependent reduction of adhesion of GSCs to the cap cells and there is accumulation of reactive oxygen species ROS resulting in cellular damage which contributes to GSC aging There is an observed reduction in the number of cap cells and the physical attachment of GSCs to cap cells through aging Shg is expressed at significantly lower levels in an old GSC niche in comparison to a young one 26 GSC niche in Drosophila testes edit Males of Drosophila melanogaster each have two testes long tubular coiled structures and at the anterior most tip of each lies the GSC niche The testis GSC niche is built around a population of non mitotic hub cells a k a niche cells to which two populations of stem cells adhere the GSCs and the somatic stem cells SSCs a k a somatic cyst stem cells cyst stem cells Each GSC is enclosed by a pair of SSCs though each stem cell type is still in contact with the hub cells In this way the stem cell niche consists of these three cell types as not only do the hub cells regulate GSC and SSC behaviour but the stem cells also regulate the activity of each other The Drosophila testis GSC niche has proven a valuable model system for examining a wide range of cellular processes and signalling pathways 27 Outside the testis GSC niche edit The process of spermatogenesis begins when the GSCs divide asymmetrically producing a GSC that maintains hub contact and a gonialblast that exits the niche The SSCs divide with their GSC partner and their non mitotic progeny the somatic cyst cells SCCs a k a cyst cells will enclose the gonialblast The gonialblast then undergoes four rounds of synchronous transit amplifying divisions with incomplete cytokinesis to produce a sixteen cell spermatogonial cyst This spermatogonial cyst then differentiates and grows into a spermatocyte which will eventually undergo meiosis and produce sperm 27 Molecular signalling in the testis GSC niche edit The two main molecular signalling pathways regulating stem cell behaviour in the testis GSC niche are the Jak STAT and BMP signalling pathways Jak STAT signalling originates in the hub cells where the ligand Upd is secreted to the GSCs and SSCs 28 29 This leads to activation of the Drosophila STAT Stat92E a transcription factor which effects GSC adhesion to the hub cells 30 and SSC self renewal via Zfh 1 31 Jak STAT signalling also influences the activation of BMP signalling via the ligands Dpp and Gbb These ligands are secreted into the GSCs from the SSCs and hub cells activate BMP signalling and suppress the expression of Bam a differentiation factor 32 Outside of the niche gonialblasts no longer receive BMP ligands and are free to begin their differentiation program Other important signalling pathways include the MAPK and Hedgehog which regulate germline enclosure 33 and somatic cell self renewal 34 respectively GSC niche in mouse testes edit The murine GSC niche in males also called spermatogonial stem cell SSC niche is located in the basal region of seminiferous tubules in the testes The seminiferous epithelium is composed of sertoli cells that are in contact with the basement membrane of the tubules which separates the sertoli cells from the interstitial tissue below This interstitial tissue comprises Leydig cells macrophages mesenchymal cells capillary networks and nerves 35 During development primordial germ cells migrate into the seminiferous tubules and downward towards the basement membrane whilst remaining attached to the sertoli cells where they will subsequently differentiate into SSCs also referred to as Asingle spermatogonia 35 36 These SSCs can either self renew or commit to differentiating into spermatozoa upon the proliferation of Asingle into Apaired spermatogonia The 2 cells of Apaired spermatogonia remain attached by intercellular bridges and subsequently divide into Aaligned spermatogonia which is made up of 4 16 connected cells Aaligned spermatogonia then undergo meiosis I to form spermatocytes and meiosis II to form spermatids which will mature into spermatozoa 37 38 This differentiation occurs along the longitudinal axis of sertoli cells from the basement membrane to the apical lumen of the seminiferous tubules However sertoli cells form tight junctions that separate SSCs and spermatogonia in contact with the basement membrane from the spermatocytes and spermatids to create a basal and an adluminal compartment whereby differentiating spermatocytes must traverse the tight junctions 35 39 These tight junctions form the blood testis barrier BTB and have been suggested to play a role in isolating differentiated cells in the adluminal compartment from secreted factors by the interstitial tissue and vasculature neighboring the basal compartment 35 Molecular mechanisms of SSC maintenance and activity edit Physical cues edit The basement membrane of the seminiferous tubule is a modified form of extracellular matrix composed of fibronectin collagens and laminin 35 b1 integrin is expressed on the surface of SSCs and is involved in their adhesion to the laminin component of the basement membrane although other adhesion molecules are likely also implicated in the attachment of SSCs to the basement membrane 40 E cadherin expression on SSCs in mice unlike in Drosophila have been shown to be dispensable as the transplantation of cultured SSCs lacking E cadherin are able to colonize host seminiferous tubules and undergo spermatogenesis 41 In addition the blood testis barrier provides architectural support and is composed of tight junction components such as occludins claudins and zonula occludens ZOs which show dynamic expression during spermatogenesis 42 For example claudin 11 has been shown to be a necessary component of these tight junctions as mice lacking this gene have a defective blood testis barrier and do not produce mature spermatozoa 40 Molecular signals regulating SSC renewal edit GDNF Glial cell derived neurotrophic factor is known to stimulate self renewal of SSCs and is secreted by the sertoli cells under the influence of gonadotropin FSH GDNF is a related member of the TGFb superfamily of growth factors and when overexpressed in mice an increase in undifferentiated spermatogonia was observed which led to the formation of germ tumours 35 40 In corroboration for its role as a renewal factor heterozygous knockout male mice for GDNF show decreased spermatogenesis that eventually leads to infertility 40 In addition the supplementation of GDNF has been shown to extend the expansion of mouse SSCs in culture However the GDNF receptor c RET and co receptor GFRa1 are not solely expressed on the SSCs but also on Apaired and Aaligned therefore showing that GDNF is a renewal factor for Asingle to Aaligned in general rather than being specific to the Asingle SSC population FGF2 Fibroblast growth factor 2 secreted by sertoli cells has also been shown to influence the renewal of SSCs and undifferentiated spermatogonia in a similar manner to GDNF 35 Although sertoli cells appear to play a major role in renewal it expresses receptors for testosterone that is secreted by Leydig cells whereas germ cells do not contain this receptor thus alluding to an important role of Leydig cells upstream in mediating renewal Leydig cells also produce CSF 1 Colony stimulating factor 1 for which SSCs strongly express the receptor CSF1R 37 When CSF 1 was added in culture with GDNF and FGF2 no further increase in proliferation was observed however the longer the germ cells remained in culture with CSF 1 the greater the SSC density observed when these germ cells were transplanted into host seminiferous tubules This showed CSF 1 to be a specific renewal factor that tilts the SSCs towards renewal over differentiation rather than affecting proliferation of SSCs and spermatogonia GDNF FGF 2 and CSF 1 have also been shown to influence self renewal of stem cells in other mammalian tissues 35 Plzf Promyelocytic leukaemia zinc finger has also been implicated in regulating SSC self renewal and is expressed by Asingle Apaired and Aaligned spermatogonia Plzf directly inhibits the transcription of a receptor c kit in these early spermatogonia However its absence in late spermatogonia permits c kit expression which is subsequently activated by its ligand SCF stem cell factor secreted by sertoli cells resulting in further differentiation Also the addition of BMP4 and Activin A have shown to reduce self renewal of SSCs in culture and increase stem cell differentiation with BMP4 shown to increase the expression of c kit 37 Aging of the SSC niche edit Prolonged spermatogenesis relies on the maintenance of SSCs however this maintenance declines with age and leads to infertility Mice between 12 and 14 months of age show decreased testis weight reduced spermatogenesis and SSC content Although stem cells are regarded as having the potential to infinitely replicate in vitro factors provided by the niche are crucial in vivo Indeed serial transplantation of SSCs from male mice of different ages into young mice 3 months of age whose endogenous spermatogenesis had been ablated was used to estimate stem cell content given that each stem cell would generate a colony of spermatogenesis 35 43 The results of this experiment showed that transplanted SSCs could be maintained far longer than their replicative lifespan for their age In addition a study also showed that SSCs from young fertile mice could not be maintained nor undergo spermatogenesis when transplanted into testes of old infertile mice Together these results points towards a deterioration of the SSC niche itself with aging rather than the loss of intrinsic factors in the SSC 43 Vertebrate adult stem cell niches edit Hematopoietic stem cell niche edit Further information Hematopoietic stem cell niche Vertebrate hematopoietic stem cells niche in the bone marrow is formed by cells subendosteal osteoblasts sinusoidal endothelial cells and bone marrow stromal also sometimes called reticular cells which includes a mix of fibroblastoid monocytic and adipocytic cells which comprise marrow adipose tissue 1 Hair follicle stem cell niche edit The hair follicle stem cell niche is one of the more closely studied niches thanks to its relative accessibility and role in important diseases such as melanoma The bulge area at the junction of arrector pili muscle to the hair follicle sheath has been shown to host the skin stem cells which can contribute to all epithelial skin layers There cells are maintained by signaling in concert with niche cells signals include paracrine e g sonic hedgehog autocrine and juxtacrine signals 44 The bulge region of the hair follicle relies on these signals to maintain the stemness of the cells Fate mapping or cell lineage tracing has shown that Keratin 15 positive stem cells progeny participate in all epithelial lineages 45 The follicle undergoes cyclic regeneration in which these stem cells migrate to various regions and differentiate into the appropriate epithelial cell type Some important signals in the hair follicle stem cell niche produced by the mesenchymal dermal papilla or the bulge include BMP TGF b and Fibroblast growth factor FGF ligands and Wnt inhibitors 46 While Wnt signaling pathways and b catenin are important for stem cell maintenance 47 over expression of b catenin in hair follicles induces improper hair growth Therefore these signals such as Wnt inhibitors produced by surrounding cells are important to maintain and facilitate the stem cell niche 48 Intestinal stem cell niche edit Intestinal organoids have been used to study intestinal stem cell niches An intestinal organoid culture can be used to indirectly assess the effect of the manipulation on the stem cells through assessing the organoid s survival and growth Research using intestinal organoids have demonstrated that the survival of intestinal stem cells is improved by the presence of neurons and fibroblasts 49 and through the administration of IL 22 50 Cardiovascular stem cell niche edit Cardiovascular stem cell niches can be found within the right ventricular free wall atria and outflow tracks of the heart They are composed of Isl1 Flk1 cardiac progenitor cells CPCs that are localized into discrete clusters within a ColIV and laminin extracellular matrix ECM ColI and fibronectin are predominantly found outside the CPC clusters within the myocardium Immunohistochemical staining has been used to demonstrate that differentiating CPCs which migrate away from the progenitor clusters and into the ColI and fibronectin ECM surrounding the niche down regulate Isl1 while up regulating mature cardiac markers such as troponin C 51 There is a current controversy over the role of Isl1 cells in the cardiovascular system While major publications have identified these cells as CPC s and have found a very large number in the murine and human heart recent publications have found very few Isl1 cells in the murine fetal heart and attribute their localization to the sinoatrial node 52 which is known as an area that contributes to heart pacemaking The role of these cells and their niche are under intense research and debate citation needed Neural stem cell niche edit Neural stem cell niches are divided in two the Subependymal zone SEZ and the Subgranular zone SGZ The SEZ is a thin area beneath the ependymal cell layer that contains three types of neural stem cells infrequently dividing neural stem cells NSCs rapidly dividing transit amplifying precursors TaPs and neuroblasts NBs The SEZ extracellular matrix ECM has significant differences in composition compared to surrounding tissues Recently it was described that progenitor cells NSCs TaPs and NBs were attached to ECM structures called Fractones 53 These structures are rich in laminin collagen and heparan sulfate proteoglycans 54 Other ECM molecules such as tenascin C MMPs and different proteoglycans are also implicated in the neural stem cell niche 55 Cancer stem cell niche editCancer tissue is morphologically heterogenous not only due to the variety of cell types present endothelial fibroblast and various immune cells but cancer cells themselves are not a homogenous population either citation needed In accordance with the hierarchy model of tumours the cancer stem cells CSC are maintained by biochemical and physical contextual signals emanating from the microenvironment called the cancer stem cell niche 56 The CSC niche is very similar to normal stem cells niche embryonic stem cell ESC Adult Stem Cell ASC in function maintaining of self renewal undifferentiated state and ability to differentiate and in signalling pathways Activin Noda Akt PTEN JAK STAT PI3 K TGF b Wnt and BMP 57 It is hypothesized that CSCs arise form aberrant signalling of the microenvironment and participates not only in providing survival signals to CSCs but also in metastasis by induction of epithelial mesenchymal transition EMT citation needed Hypoxia edit Hypoxic condition in stem cell niches ESC ASC or CSC is necessary for maintaining stem cells in an undifferentiated state and also for minimizing DNA damage via oxidation The maintaining of the hypoxic state is under control of Hypoxia Inducible transcription Factors HIFs 58 HIFs contribute to tumour progression cell survival and metastasis by regulation of target genes as VEGF GLUT 1 ADAM 1 Oct4 and Notch 57 Hypoxia in the CSC niche edit Hypoxia plays an important role in the regulation of cancer stem cell niches and EMT through the promotion of HIFs 59 These HIFs help maintain cancer stem cell niches by regulating important stemness genes such as Oct4 Nanog SOX2 Klf4 and cMyc 60 61 HIFs also regulate important tumor suppressor genes such as p53 and genes that promote metastasis 62 63 Although HIFs increase the survival of cells by decreasing the effects of oxidative stress they have also been shown to decrease factors such as RAD51 and H2AX that maintain genomic stability 64 In the hypoxic condition there is an increase of intracellular Reactive Oxygen Species ROS which also promote CSCs survival via stress response 65 66 ROS stabilizes HIF 1a which promotes the Met proto oncogene which drives metastasis or motogenic escape in melanoma cells 67 All of these factors contribute to a cancer stem cell phenotype which is why it is often referred to as a hypoxic stem cell niche Hypoxic environments are often found in tumors where the cells are dividing faster that angiogenesis can occur It is important to study hypoxia as an aspect of cancer because hypoxic environments have been shown to be resistant to radiation therapy 68 Radiation has been shown to increase the amounts of HIF 1 69 EMT induction by hypoxia though interactions between HIF 1a and ROS is crucial for metastasis in cancers such as melanoma It has been found that many genes associated with melanoma are regulated by hypoxia such as MXI1 FN1 and NME1 70 Epithelial mesenchymal transition edit Epithelial mesenchymal transition is a morphogenetic process normally occurs in embryogenesis that is hijacked by cancer stem cells by detaching from their primary place and migrating to another one The dissemination is followed by reverse transition so called Epithelial Mesenchymal Transition EMT This process is regulated by CSCs microenvironment via the same signalling pathways as in embryogenesis using the growth factors TGF b PDGF EGF cytokine IL 8 and extracellular matrix components These growth factors interactions through intracellular signal transducers like b catenin has been shown to induce metastatic potential 71 72 A characteristic of EMT is loss of the epithelial markers E cadherin cytokeratins claudin occluding desmoglein desmocolin and gain of mesenchymal markers N cadherin vimentin fibronectin 73 There is also certain degree of similarity in homing mobilization of normal stem cells and metastasis invasion of cancer stem cells There is an important role of Matrix MetalloProteinases MMP the principal extracellular matrix degrading enzymes thus for example matrix metalloproteinase 2 and 9 are induced to expression and secretion by stromal cells during metastasis of colon cancer via direct contact or paracrine regulation The next sharing molecule is Stromal cell Derived Factor 1 SDF 1 73 74 Inflammation edit The EMT and cancer progression can be triggered also by chronic inflammation The main roles have molecules IL 6 IL 8 TNF a NFkB TGF b HIF 1a which can regulate both processes through regulation of downstream signalling that overlapping between EMT and inflammation 57 The downstream pathways involving in regulation of CSCs are Wnt SHH Notch TGF b RTKs EGF FGF IGF HGF NFkB regulates the EMT migration and invasion of CSCs through Slug Snail and Twist The activation of NFkB leads to increase not only in production of IL 6 TNF a and SDF 1 but also in delivery of growth factors The source of the cytokine production are lymphocytes TNF a Mesenchymal Stem Cells SDF 1 IL 6 IL8 Interleukin 6 mediates activation of STAT3 The high level of STAT3 was described in isolated CSCs from liver bone cervical and brain cancer The inhibition of STAT3 results in dramatic reduction in their formation Generally IL 6 contributes a survival advantage to local stem cells and thus facilitates tumorigenesis 57 SDF 1a secreted from Mesenchymal Stem Cells MSCs has important role in homing and maintenance of Hematopoietic Stem Cell HSC in bone marrow niche but also in homing and dissemination of CSC 74 Angiogenesis edit Hypoxia is a main stimulant for angiogenesis with HIF 1a being the primary mediator Angiogenesis induced by hypoxic conditions is called an Angiogenic switch HIF 1 promotes expression of several angiogenic factors Vascular Endothelial Growth Factor VEGF basic Fibroblast Growth Factor bFGF Placenta Like Growth Factor PLGF Platelet Derived Growth Factor PDGF and Epidermal Growth Factor But there is evidence that the expression of angiogenic agens by cancer cells can also be HIF 1 independent It seems that there is an important role of Ras protein and that intracellular levels of calcium regulate the expression of angiogenic genes in response to hypoxia 73 The angiogenic switch downregulates angiogenesis suppressor proteins such as thrombospondin angiostatin endostatin and tumstatin Angiogenesis is necessary for the primary tumour growth citation needed Injury induced edit Main article Injury induced stem cell niches During injury support cells are able to activate a program for repair recapitulating aspects of development in the area of damage These areas become permissive for stem cell renewal migration and differentiation For instance in the CNS injury is able to activate a developmental program in astrocytes that allow them to express molecules that support stem cells such as chemokines i e SDF 1 75 and morphogens such as sonic hedgehog 76 Extracellular Matrix Mimicking Strategies For Stem Cell Niche editIt is evident that biophysio chemical characteristics of ECM such as composition shape topography stiffness and mechanical strength can control the stem cell behavior These ECM factors are equally important when stem cells are grown in vitro Given a choice between niche cell stem cell interaction and ECM stem cell interaction mimicking ECM is preferred as that can be precisely controlled by scaffold fabrication techniques processing parameters or post fabrication modifications In order to mimic it is essential to understand natural properties of ECM and their role in stem cell fate processes Various studies involving different types of scaffolds that regulate stem cells fate by mimicking these ECM properties have been done 2 77 References edit a b Birbrair A Frenette PS April 2016 Niche heterogeneity in the bone marrow Annals of the New York Academy of Sciences 1370 1 82 96 Bibcode 2016NYASA1370 82B doi 10 1111 nyas 13016 PMC 4938003 PMID 27015419 a b c Jhala D 2015 A review on extracellular matrix mimicking strategies for an artificial stem cell niche Polymer Reviews 55 4 561 595 doi 10 1080 15583724 2015 1040552 S2CID 94588894 Scadden DT June 2006 The stem cell niche as an entity of action Nature 441 7097 1075 1079 Bibcode 2006Natur 441 1075S doi 10 1038 nature04957 PMID 16810242 S2CID 4418385 Ritsma L Ellenbroek SI Zomer A Snippert HJ de Sauvage FJ Simons BD et al March 2014 Intestinal crypt homeostasis revealed at single stem cell level by in vivo live imaging Nature 507 7492 362 365 Bibcode 2014Natur 507 362R doi 10 1038 nature12972 PMC 3964820 PMID 24531760 Rompolas P Mesa KR Greco V October 2013 Spatial organization within a niche as a determinant of stem cell fate Nature 502 7472 513 518 Bibcode 2013Natur 502 513R doi 10 1038 nature12602 PMC 3895444 PMID 24097351 Shahriyari L Komarova NL July 2015 The role of the bi compartmental stem cell niche in delaying cancer Physical Biology 12 5 055001 Bibcode 2015PhBio 12e5001S doi 10 1088 1478 3975 12 5 055001 PMID 26228740 S2CID 7171931 Cannataro VL McKinley SA St Mary CM April 2016 The implications of small stem cell niche sizes and the distribution of fitness effects of new mutations in aging and tumorigenesis Evolutionary Applications 9 4 565 582 doi 10 1111 eva 12361 PMC 4831459 PMID 27099622 Cannataro VL McKinley SA St Mary CM July 2017 The evolutionary trade off between stem cell niche size aging and tumorigenesis Evolutionary Applications 10 6 590 602 doi 10 1111 eva 12476 PMC 5469181 PMID 28616066 Li L Xie T 2005 Stem cell niche structure and function Annual Review of Cell and Developmental Biology 21 605 631 doi 10 1146 annurev cellbio 21 012704 131525 PMID 16212509 a b c d Xie T Spradling AC October 2000 A niche maintaining germ line stem cells in the Drosophila ovary Science 290 5490 328 330 Bibcode 2000Sci 290 328X doi 10 1126 science 290 5490 328 PMID 11030649 Lin H Yue L Spradling AC April 1994 The Drosophila fusome a germline specific organelle contains membrane skeletal proteins and functions in cyst formation Development 120 4 947 956 doi 10 1242 dev 120 4 947 PMID 7600970 Ting X 2013 Control of germline stem cell self renewal and differentiation in the Drosophila ovary concerted actions of niche signals and intrinsic factors Wiley Interdisciplinary Reviews Developmental Biology 2 2 pp 261 273 Zhang H and Cai Y 2020 Signal transduction pathways regulating Drosophila ovarian germline stem cells Current opinion in insect science 37 pp 1 7 Song X Wong MD Kawase E Xi R Ding BC McCarthy JJ Xie T March 2004 Bmp signals from niche cells directly repress transcription of a differentiation promoting gene bag of marbles in germline stem cells in the Drosophila ovary Development 131 6 1353 1364 doi 10 1242 dev 01026 PMID 14973291 Chen D McKearin D October 2003 Dpp signaling silences bam transcription directly to establish asymmetric divisions of germline stem cells Current Biology 13 20 1786 1791 doi 10 1016 j cub 2003 09 033 PMID 14561403 Cox DN Chao A Lin H February 2000 piwi encodes a nucleoplasmic factor whose activity modulates the number and division rate of germline stem cells Development 127 3 503 514 doi 10 1242 dev 127 3 503 PMID 10631171 Song X Zhu CH Doan C Xie T June 2002 Germline stem cells anchored by adherens junctions in the Drosophila ovary niches Science 296 5574 1855 1857 Bibcode 2002Sci 296 1855S doi 10 1126 science 1069871 PMID 12052957 S2CID 25830121 Bogard N Lan L Xu J Cohen RS October 2007 Rab11 maintains connections between germline stem cells and niche cells in the Drosophila ovary Development 134 19 3413 3418 doi 10 1242 dev 008466 PMID 17715175 Gilboa L Forbes A Tazuke SI Fuller MT Lehmann R December 2003 Germ line stem cell differentiation in Drosophila requires gap junctions and proceeds via an intermediate state Development 130 26 6625 6634 doi 10 1242 dev 00853 PMID 14660550 Drummond Barbosa D Spradling AC March 2001 Stem cells and their progeny respond to nutritional changes during Drosophila oogenesis Developmental Biology 231 1 265 278 doi 10 1006 dbio 2000 0135 PMID 11180967 a b Hsu HJ Drummond Barbosa D January 2009 Insulin levels control female germline stem cell maintenance via the niche in Drosophila Proceedings of the National Academy of Sciences of the United States of America 106 4 1117 1121 Bibcode 2009PNAS 106 1117H doi 10 1073 pnas 0809144106 PMC 2633547 PMID 19136634 Margolis J Spradling A November 1995 Identification and behavior of epithelial stem cells in the Drosophila ovary Development 121 11 3797 3807 doi 10 1242 dev 121 11 3797 PMID 8582289 Xie T Spradling AC July 1998 decapentaplegic is essential for the maintenance and division of germline stem cells in the Drosophila ovary Cell 94 2 251 260 doi 10 1016 s0092 8674 00 81424 5 PMID 9695953 Kai T Spradling A April 2003 An empty Drosophila stem cell niche reactivates the proliferation of ectopic cells Proceedings of the National Academy of Sciences of the United States of America 100 8 4633 4638 Bibcode 2003PNAS 100 4633K doi 10 1073 pnas 0830856100 PMC 153607 PMID 12676994 Zhao R Xuan Y Li X Xi R June 2008 Age related changes of germline stem cell activity niche signaling activity and egg production in Drosophila Aging Cell 7 3 344 354 doi 10 1111 j 1474 9726 2008 00379 x PMID 18267001 a b Pan L Chen S Weng C Call G Zhu D Tang H et al October 2007 Stem cell aging is controlled both intrinsically and extrinsically in the Drosophila ovary Cell Stem Cell 1 4 458 469 doi 10 1016 j stem 2007 09 010 PMID 18371381 a b La Marca JE Somers WG 2014 The Drosophila gonads models for stem cell proliferation self renewal and differentiation AIMS Genetics 1 1 55 80 doi 10 3934 genet 2014 1 55 Kiger AA Jones DL Schulz C Rogers MB Fuller MT December 2001 Stem cell self renewal specified by JAK STAT activation in response to a support cell cue Science 294 5551 2542 2545 Bibcode 2001Sci 294 2542K doi 10 1126 science 1066707 PMID 11752574 S2CID 206506814 Tulina N Matunis E December 2001 Control of stem cell self renewal in Drosophila spermatogenesis by JAK STAT signaling Science 294 5551 2546 2549 Bibcode 2001Sci 294 2546T doi 10 1126 science 1066700 PMID 11752575 S2CID 43266825 Leatherman JL Dinardo S August 2010 Germline self renewal requires cyst stem cells and stat regulates niche adhesion in Drosophila testes Nature Cell Biology 12 8 806 811 doi 10 1038 ncb2086 PMC 2917891 PMID 20622868 Leatherman JL Dinardo S July 2008 Zfh 1 controls somatic stem cell self renewal in the Drosophila testis and nonautonomously influences germline stem cell self renewal Cell Stem Cell 3 1 44 54 doi 10 1016 j stem 2008 05 001 PMC 2601693 PMID 18593558 Kawase E Wong MD Ding BC Xie T March 2004 Gbb Bmp signaling is essential for maintaining germline stem cells and for repressing bam transcription in the Drosophila testis Development 131 6 1365 1375 doi 10 1242 dev 01025 PMID 14973292 Sarkar A Parikh N Hearn SA Fuller MT Tazuke SI Schulz C July 2007 Antagonistic roles of Rac and Rho in organizing the germ cell microenvironment Current Biology 17 14 1253 1258 doi 10 1016 j cub 2007 06 048 PMID 17629483 Michel M Kupinski AP Raabe I Bokel C August 2012 Hh signalling is essential for somatic stem cell maintenance in the Drosophila testis niche Development 139 15 2663 2669 doi 10 1242 dev 075242 PMID 22745310 a b c d e f g h i Oatley JM Brinster RL April 2012 The germline stem cell niche unit in mammalian testes Physiological Reviews 92 2 577 595 doi 10 1152 physrev 00025 2011 PMC 3970841 PMID 22535892 Griswold MD Oatley JM January 2013 Concise review Defining characteristics of mammalian spermatogenic stem cells Stem Cells 31 1 8 11 doi 10 1002 stem 1253 PMC 5312674 PMID 23074087 a b c de Rooij DG August 2009 The spermatogonial stem cell niche Microscopy Research and Technique 72 8 580 585 doi 10 1002 jemt 20699 PMID 19263493 Bowles J Koopman P October 2007 Retinoic acid meiosis and germ cell fate in mammals Development 134 19 3401 3411 doi 10 1242 dev 001107 PMID 17715177 Hess RA de Franca LR 2009 Spermatogenesis and Cycle of the Seminiferous Epithelium In Cheng CY ed Molecular Mechanisms in Spermatogenesis Advances in Experimental Medicine and Biology Vol 636 pp 1 15 doi 10 1007 978 0 387 09597 4 1 ISBN 978 0 387 09597 4 PMID 19856159 a b c d Kanatsu Shinohara M Shinohara T 2013 Spermatogonial stem cell self renewal and development Annual Review of Cell and Developmental Biology 29 163 187 doi 10 1146 annurev cellbio 101512 122353 PMID 24099084 Yoshida S 2011 Stem Cell Niche System in Mouse Spermatogenesis Male Germline Stem Cells Developmental and Regenerative Potential Vol 2011 pp 159 175 doi 10 1007 978 1 61737 973 4 8 ISBN 978 1 61737 972 7 a href Template Cite book html title Template Cite book cite book a journal ignored help Chihara M Otsuka S Ichii O Hashimoto Y Kon Y July 2010 Molecular dynamics of the blood testis barrier components during murine spermatogenesis Molecular Reproduction and Development 77 7 630 639 doi 10 1002 mrd 21200 PMID 20578065 S2CID 21630147 a b Ryu BY Orwig KE Oatley JM Avarbock MR Brinster RL June 2006 Effects of aging and niche microenvironment on spermatogonial stem cell self renewal Stem Cells 24 6 1505 1511 doi 10 1634 stemcells 2005 0580 PMC 5501308 PMID 16456131 Aloni Grinstein R Shetzer Y Kaufman T Rotter V August 2014 p53 the barrier to cancer stem cell formation FEBS Letters 588 16 2580 2589 doi 10 1016 j febslet 2014 02 011 PMID 24560790 Morris RJ Liu Y Marles L Yang Z Trempus C Li S et al April 2004 Capturing and profiling adult hair follicle stem cells Nature Biotechnology 22 4 411 417 doi 10 1038 nbt950 PMID 15024388 S2CID 9257482 Rompolas P Greco V 2014 Stem cell dynamics in the hair follicle niche Seminars in Cell amp Developmental Biology 25 26 34 42 doi 10 1016 j semcdb 2013 12 005 PMC 3988239 PMID 24361866 Hosseini V Kalantary Charvadeh A Hajikarami M Fayyazpour P Rahbarghazi R Totonchi M Darabi M October 2021 A small molecule modulating monounsaturated fatty acids and Wnt signaling confers maintenance to induced pluripotent stem cells against endodermal differentiation Stem Cell Research amp Therapy 12 1 550 doi 10 1186 s13287 021 02617 x PMC 8532309 PMID 34674740 Deschene ER Myung P Rompolas P Zito G Sun TY Taketo MM et al March 2014 b Catenin activation regulates tissue growth non cell autonomously in the hair stem cell niche Science 343 6177 1353 1356 Bibcode 2014Sci 343 1353D doi 10 1126 science 1248373 PMC 4096864 PMID 24653033 Pastula A Middelhoff M Brandtner A Tobiasch M Hohl B Nuber AH et al 2016 Three Dimensional Gastrointestinal Organoid Culture in Combination with Nerves or Fibroblasts A Method to Characterize the Gastrointestinal Stem Cell Niche Stem Cells International 2016 3710836 doi 10 1155 2016 3710836 PMC 4677245 PMID 26697073 Lindemans C Mertelsmann A Dudakov JA Velardi E Hua G O Connor M et al 2014 IL 22 Administration Protects Intestinal Stem Cells from Gvhd Biology of Blood and Marrow Transplantation 20 2 S53 S54 doi 10 1016 j bbmt 2013 12 056 Schenke Layland K Nsair A Van Handel B Angelis E Gluck JM Votteler M et al April 2011 Recapitulation of the embryonic cardiovascular progenitor cell niche Biomaterials 32 11 2748 2756 doi 10 1016 j biomaterials 2010 12 046 PMC 3414535 PMID 21257198 Weinberger F Mehrkens D Friedrich FW Stubbendorff M Hua X Muller JC et al May 2012 Localization of Islet 1 positive cells in the healthy and infarcted adult murine heart Circulation Research 110 10 1303 1310 doi 10 1161 CIRCRESAHA 111 259630 PMC 5559221 PMID 22427341 Luo Pifu Moritani Masayuki Dessem Dean 2001 07 02 Jaw muscle spindle afferent pathways to the trigeminal motor nucleus in the rat The Journal of Comparative Neurology 435 3 341 353 doi 10 1002 cne 1034 ISSN 0021 9967 PMID 11406816 S2CID 36398505 Mercier Frederic 2016 Fractones extracellular matrix niche controlling stem cell fate and growth factor activity in the brain in health and disease Cellular and Molecular Life Sciences 73 24 4661 4674 doi 10 1007 s00018 016 2314 y ISSN 1420 682X PMID 27475964 S2CID 28119663 The neural stem cell microenvironment StemBook www stembook org Retrieved 2022 04 29 van de Stolpe A 2013 On the origin and destination of cancer stem cells a conceptual evaluation American Journal of Cancer Research 3 1 107 116 PMC 3555199 PMID 23359140 a b c d Cabarcas SM Mathews LA Farrar WL November 2011 The cancer stem cell niche there goes the neighborhood International Journal of Cancer 129 10 2315 2327 doi 10 1002 ijc 26312 PMC 6953416 PMID 21792897 Borovski T De Sousa E Melo F Vermeulen L Medema JP February 2011 Cancer stem cell niche the place to be Cancer Research 71 3 634 639 doi 10 1158 0008 5472 CAN 10 3220 PMID 21266356 Peitzsch C Perrin R Hill RP Dubrovska A Kurth I August 2014 Hypoxia as a biomarker for radioresistant cancer stem cells International Journal of Radiation Biology 90 8 636 652 doi 10 3109 09553002 2014 916841 PMID 24844374 S2CID 25813277 Covello KL Kehler J Yu H Gordan JD Arsham AM Hu CJ et al March 2006 HIF 2alpha regulates Oct 4 effects of hypoxia on stem cell function embryonic development and tumor growth Genes amp Development 20 5 557 570 doi 10 1101 gad 1399906 PMC 1410808 PMID 16510872 Keith B Simon MC May 2007 Hypoxia inducible factors stem cells and cancer Cell 129 3 465 472 doi 10 1016 j cell 2007 04 019 PMC 3150586 PMID 17482542 Bertout JA Majmundar AJ Gordan JD Lam JC Ditsworth D Keith B et al August 2009 HIF2alpha inhibition promotes p53 pathway activity tumor cell death and radiation responses Proceedings of the National Academy of Sciences of the United States of America 106 34 14391 14396 Bibcode 2009PNAS 10614391B doi 10 1073 pnas 0907357106 PMC 2726037 PMID 19706526 Liu L Zhu XD Wang WQ Shen Y Qin Y Ren ZG et al May 2010 Activation of beta catenin by hypoxia in hepatocellular carcinoma contributes to enhanced metastatic potential and poor prognosis Clinical Cancer Research 16 10 2740 2750 doi 10 1158 1078 0432 CCR 09 2610 PMID 20460486 Bindra RS Schaffer PJ Meng A Woo J Maseide K Roth ME et al October 2004 Down regulation of Rad51 and decreased homologous recombination in hypoxic cancer cells Molecular and Cellular Biology 24 19 8504 8518 doi 10 1128 MCB 24 19 8504 8518 2004 PMC 516750 PMID 15367671 Singh S Brocker C Koppaka V Chen Y Jackson BC Matsumoto A et al March 2013 Aldehyde dehydrogenases in cellular responses to oxidative electrophilic stress Free Radical Biology amp Medicine 56 89 101 doi 10 1016 j freeradbiomed 2012 11 010 PMC 3631350 PMID 23195683 Diehn M Cho RW Lobo NA Kalisky T Dorie MJ Kulp AN et al April 2009 Association of reactive oxygen species levels and radioresistance in cancer stem cells Nature 458 7239 780 783 Bibcode 2009Natur 458 780D doi 10 1038 nature07733 PMC 2778612 PMID 19194462 Comito G Calvani M Giannoni E Bianchini F Calorini L Torre E et al August 2011 HIF 1a stabilization by mitochondrial ROS promotes Met dependent invasive growth and vasculogenic mimicry in melanoma cells Free Radical Biology amp Medicine 51 4 893 904 doi 10 1016 j freeradbiomed 2011 05 042 hdl 2158 496457 PMID 21703345 Brown JM 2007 Tumor Hypoxia in Cancer Therapy Oxygen Biology and Hypoxia Methods in Enzymology Vol 435 pp 297 321 doi 10 1016 S0076 6879 07 35015 5 ISBN 9780123739704 PMID 17998060 Moeller BJ Cao Y Li CY Dewhirst MW May 2004 Radiation activates HIF 1 to regulate vascular radiosensitivity in tumors role of reoxygenation free radicals and stress granules Cancer Cell 5 5 429 441 doi 10 1016 s1535 6108 04 00115 1 PMID 15144951 Olbryt M Habryka A Tyszkiewicz T Rusin A Cichon T Jarzab M Krawczyk Z October 2011 Melanoma associated genes MXI1 FN1 and NME1 are hypoxia responsive in murine and human melanoma cells Melanoma Research 21 5 417 425 doi 10 1097 CMR 0b013e328348db2f PMID 21912348 S2CID 33171556 Moustakas A Heldin CH October 2007 Signaling networks guiding epithelial mesenchymal transitions during embryogenesis and cancer progression Cancer Science 98 10 1512 1520 doi 10 1111 j 1349 7006 2007 00550 x PMID 17645776 S2CID 23032956 Zhou B Liu Y Kahn M Ann DK Han A Wang H et al March 2012 Interactions between b catenin and transforming growth factor b signaling pathways mediate epithelial mesenchymal transition and are dependent on the transcriptional co activator cAMP response element binding protein CREB binding protein CBP The Journal of Biological Chemistry 287 10 7026 7038 doi 10 1074 jbc M111 276311 PMC 3293544 PMID 22241478 a b c Gout S Huot J December 2008 Role of cancer microenvironment in metastasis focus on colon cancer Cancer Microenvironment 1 1 69 83 doi 10 1007 s12307 008 0007 2 PMC 2654352 PMID 19308686 a b Li L Neaves WB May 2006 Normal stem cells and cancer stem cells the niche matters Cancer Research 66 9 4553 4557 doi 10 1158 0008 5472 CAN 05 3986 PMID 16651403 Imitola J Raddassi K Park KI Mueller FJ Nieto M Teng YD et al December 2004 Directed migration of neural stem cells to sites of CNS injury by the stromal cell derived factor 1alpha CXC chemokine receptor 4 pathway Proceedings of the National Academy of Sciences of the United States of America 101 52 18117 18122 Bibcode 2004PNAS 10118117I doi 10 1073 pnas 0408258102 PMC 536055 PMID 15608062 Wang Y Imitola J Rasmussen S O Connor KC Khoury SJ October 2008 Paradoxical dysregulation of the neural stem cell pathway sonic hedgehog Gli1 in autoimmune encephalomyelitis and multiple sclerosis Annals of Neurology 64 4 417 427 doi 10 1002 ana 21457 PMC 2757750 PMID 18991353 Vishwakarma A 2017 04 01 Biology and Engineering of Stem Cell Niches Academic Press 2017 ISBN 9780128027561 Retrieved from https en wikipedia org w index php title Stem cell niche amp oldid 1221971472, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.