dacapo


EVOLUTIONARY HOMOLOGS


Table of contents

CDK inhibitors, cell cycle arrest and differentiation (part 2/2)

Cell adhesion to substratum has been shown to regulate cyclin A expression as well as cyclin D- and E-dependent kinases, the latter via the up-regulation of cyclin D1 and the down-regulation of cyclin-Cdk inhibitors p21 and p27, respectively. This adhesion-dependent regulation of cell cycle is thought to be mediated by integrins. Stable transfection and overexpression of the integrin-linked kinase (ILK), which interacts with the beta1 and beta3 integrin cytoplasmic domains (See Drosophila Myospheroid), induces anchorage-independent cell cycle progression but not serum-independent growth of rat intestinal epithelial cells (IEC18). ILK overexpression results in increased expression of cyclin D1, activation of Cdk4 and cyclin E-associated kinases, and hyperphosphorylation of the retinoblastoma protein. In addition, ILK overexpression results in the expression of p21 and p27 Cdk inhibitors with altered electrophoretic mobilities, with the p27 from ILK-overexpressing cells having reduced inhibitory activity. The transfer of serum-exposed IEC18 cells from adherent cultures to suspension cultures results in a rapid down-regulation of expression of cyclin D1 and cyclin A proteins, as well as in retinoblastoma protein dephosphorylation. In marked contrast, transfer of ILK-overexpressing cells from adherent to suspension cultures results in continued high levels of expression of cyclin D1 and cyclin A proteins, and a substantial proportion of the retinoblastoma protein remains in a hyperphosphorylated state. These results indicate that, when overexpressed, ILK induces signaling pathways resulting in the stimulation of G1/S cyclin-Cdk activities, which are normally regulated by cell adhesion and integrin engagement (Radeva, 1997).

The extracellular matrix (ECM) plays an essential role in the regulation of cell proliferation during angiogenesis. Cell adhesion to ECM is mediated by binding of cell surface integrin receptors, which both activate intracellular signaling cascades and mediate tension-dependent changes in cell shape and cytoskeletal structure. Although the growth control field has focused on early integrin and growth factor signaling events, recent studies suggest that cell shape may play an equally critical role in control of cell cycle progression. Studies were carried out to determine when cell shape exerts its regulatory effects during the cell cycle and to analyze the molecular basis for shape-dependent growth control. The shape of human capillary endothelial cells was controlled by culturing cells on microfabricated substrates containing ECM-coated adhesive islands with defined shape and size on the micrometer scale or on plastic dishes coated with defined ECM molecular coating densities. Cells that are prevented from spreading in medium containing soluble growth factors exhibit normal activation of the mitogen-activated kinase (erk1/erk2) growth signaling pathway. However, in contrast to spread cells, these cells fail to progress through G1 and enter S phase. This shape-dependent block in cell cycle progression correlates with the failure of three activities: (1) an increase in cyclin D1 protein levels; (2) the down-regulation of the cell cycle inhibitor p27(Kip1), and (3) the phosphorylation of the retinoblastoma protein in late G1. A similar block in cell cycle progression is induced before this same shape-sensitive restriction point by disrupting the actin network using cytochalasin or by inhibiting cytoskeletal tension generation using an inhibitor of actomyosin interactions. In contrast, neither modifications of cell shape, cytoskeletal structure, nor mechanical tension have any effect on S phase entry when added at later times. These findings demonstrate that although early growth factor and integrin signaling events are required for growth, these events alone are not sufficient. Subsequent cell cycle progression and, hence, cell proliferation is controlled by tension-dependent changes in cell shape and cytoskeletal structure that act by subjugating the molecular machinery that regulates the G1/S transition (Huang, 1998).

p21Cip1 is a cyclin-dependent kinase (Cdk) inhibitor that is transcriptionally activated by p53 in response to DNA damage. p21 effectively inhibits Cdk2, Cdk3, Cdk4, and Cdk6 kinases, but is much less effective toward Cdc2/cyclin B and Cdk5/p35, and does not associate with Cdk7/cyclin H. Overexpression of P21 arrests cells in G1. Thus, p21 is not a universal inhibitor of Cdks but displays selectivity for G1/S Cdk/cyclin complexes. Association of p21 with Cdks is greatly enhanced by cyclin binding. This property is shared by the structurally related inhibitor p27, suggesting a common biochemical mechanism for inhibition. With respect to Cdk2 and Cdk4 complexes, p27 shares the inhibitory potency of p21 but has slightly different kinase specificities. In normal diploid fibroblasts, the vast majority of active Cdk2 is associated with p21, but this active kinase can be fully inhibited by addition of exogenous p21. Reconstruction experiments using purified components indicate that multiple molecules of p21 can associate with Cdk/cyclin complexes and inactive complexes contain more than one molecule of p21. Together, these data suggest a model whereby p21 functions as an inhibitory buffer whose levels determine the threshold kinase activity required for cell cycle progression (Harper, 1995).

During development of the central nervous system, oligodendrocyte progenitor cells (O-2A) undergo an orderly pattern of cell proliferation and differentiation, culminating in the ability of oligodendrocytes to myelinate axons. p27(Kip1), a cyclin-dependent kinase inhibitor, is an important component of the decision of O-2A cells to withdraw from the cell cycle. In vitro, accumulation of p27 correlates with differentiation of oligodendrocytes. Only a fraction of O-2A cells derived from p27-knockout mice differentiate successfully, as compared to controls. The inability to differentiate correlates with continued proliferation, suggesting that p27 is an important component of the machinery required for the G1/G0 transition in O-2A cells. In vivo, expansion of O-2A precursors before withdrawal leads, in part, to a greater number of oligodendrocytes. Together, these data indicate that in the oligodendrocyte lineage, there is a role for p27 during the decision to withdraw from the cell cycle (Casaccia-Bonnefil, 1997).

Strict control of cellular proliferation is required to shape the complex structures of the developing embryo. The organ of Corti, the auditory neuroepithelium of the inner ear in mammals, consists of two types of terminally differentiated mechanosensory hair cells and at least four types of supporting cells arrayed precisely along the length of the spiral cochlea. In mice, the progenitors of greater than 80% of both hair cells and supporting cells undergo their terminal division between embryonic day 13 (E13) and E14. As in humans, these cells persist in a non-proliferative state throughout the adult life of the animal. The correct timing of cell cycle withdrawal in the developing organ of Corti requires p27Kip1, a cyclin-dependent kinase inhibitor that functions as an inhibitor of cell cycle progression. p27Kip1 expression is induced in the primordial organ of Corti between E12 and E14, correlating with the cessation of cell division of the progenitors of the hair cells and supporting cells. In wild-type animals, p27Kip1 expression is downregulated during subsequent hair cell differentiation, but it persists at high levels in differentiated supporting cells of the mature organ of Corti. In mice with a targeted deletion of the p27Kip1 gene, proliferation of the sensory cell progenitors continues after E14, leading to the appearance of supernumerary hair cells and supporting cells. In the absence of p27Kip1, mitotically active cells are still observed in the organ of Corti of postnatal day 6 animals, suggesting that the persistence of p27Kip1 expression in mature supporting cells may contribute to the maintenance of quiescence in this tissue and, possibly, to its inability to regenerate. Homozygous mutant mice are severely hearing impaired. Thus, p27Kip1 provides a link between developmental control of cell proliferation and the morphological development of the inner ear (Chen, 1999).

Different members of the Raf family of protein kinases display differences in their abilities to promote the entry of quiescent NIH 3T3 cells into the S phase of the cell cycle. Although conditional activation of deltaA-Raf:ER promotes cell cycle progression, activation of deltaRaf-1:ER and deltaB-Raf:ER elicit a G1 arrest that is not overcome by exogenously added growth factors. Activation of all three deltaRaf:ER kinases leads to elevated expression of cyclin D1 and cyclin E and reduced expression of p27Kip1. However, activation of deltaB-Raf:ER and deltaRaf-1:ER induces the expression of p21Cip1, whereas activation of deltaA-Raf:ER does not. A catalytically potentiated form of deltaA-Raf:ER, generated by point mutation, strongly induces p21Cip1 expression and elicits cell cycle arrest similar to deltaB-Raf:ER and deltaRaf-1:ER. These data suggest that the strength and duration of signaling by Raf kinases might influence the biological outcome of activation of this pathway. By titration of deltaB-Raf:ER activity it was demonstrated that low levels of Raf activity lead to activation of cyclin D1-cdk4 and cyclin E-cdk2 complexes and to cell cycle progression, whereas higher Raf activity elicits cell cycle arrest correlating with p21Cip1 induction and inhibition of cyclin-cdk activity. Using green fluorescent protein-tagged forms of deltaRaf-1:ER in primary mouse embryo fibroblasts (MEFs) it was demonstrated that p21Cip1 is induced by Raf in a p53-independent manner, leading to cell cycle arrest. By contrast, activation of Raf in p21Cip1-/- MEFs leads to a robust mitogenic response that is similar to that observed in response to platelet-derived growth factor. These data indicate that, depending on the level of kinase activity, Raf can elicit either cell cycle progression or cell cycle arrest in mouse fibroblasts. The ability of Raf to elicit cell cycle arrest is strongly associated with its ability to induce the expression of the cyclin-dependent kinase inhibitor p21Cip1 in a manner that bears analogy to alpha-factor arrest in Saccharomyces cerevisiae. These data are consistent with a role for Raf kinases in both proliferation and differentiation of mammalian cells (Woods, 1997).

Skeletal muscle differentiation entails the coordination of muscle-specific gene expression and terminal withdrawal from the cell cycle. This cell cycle arrest in the G0 phase requires the retinoblastoma tumor suppressor protein (Rb). The function of Rb is negatively regulated by cyclin-dependent kinases (Cdks), which are controlled by Cdk inhibitors. Expression of MyoD, a skeletal muscle-specific transcriptional regulator, activates the expression of the Cdk inhibitor p21 during differentiation of murine myocytes and in nonmyogenic cells. MyoD-mediated induction of p21 does not require the tumor suppressor protein p53 and correlates with cell cycle withdrawal. Thus, MyoD may induce terminal cell cycle arrest during skeletal muscle differentiation by increasing the expression of p21 (Halevy, 1995).

Hearing loss is most often the result of hair-cell degeneration due to genetic abnormalities or ototoxic and traumatic insults. In the postembryonic and adult mammalian auditory sensory epithelium, the organ of Corti, no hair-cell regeneration has ever been observed. However, nonmammalian hair-cell epithelia are capable of regenerating sensory hair cells as a consequence of nonsensory supporting-cell proliferation. The supporting cells of the organ of Corti are highly specialized, terminally differentiated cell types that apparently are incapable of proliferation. At the molecular level terminally differentiated cells have been shown to express high levels of cell-cycle inhibitors, in particular, cyclin-dependent kinase inhibitors, which are thought to be responsible for preventing these cells from reentering the cell cycle. The cyclin-dependent kinase inhibitor p27(Kip1) is selectively expressed in the supporting-cell population of the organ of Corti. Effects of p27(Kip1)-gene disruption include ongoing cell proliferation in postnatal and adult mouse organ of Corti at time points well after mitosis normally has ceased during embryonic development. This suggests that release from p27(Kip1)-induced cell-cycle arrest is sufficient to allow supporting-cell proliferation to occur. This finding may provide an important pathway for inducing hair-cell regeneration in the mammalian hearing organ (Lowenheim, 1999).

The proliferating precursor cells that give rise to postmitotic oligodendrocytes, the cells that make myelin in the central nervous system, are subject to regulation by the cyclin-dependent kinase inhibitor p27/Kip1. Two components of the cell cycle control system, cyclin D1 and the Cdc2 kinase, are present in the proliferating precursor cells but not in differentiated oligodendrocytes, suggesting that the control system is dismantled in the oligodendrocytes.The cyclin-dependent kinase (Cdk) inhibitor p27 progressively accumulates in the precursor cells as they proliferate and is present at high levels in oligodendrocytes. These findings are consistent with the possibility that the accumulation of p27 is part of both the intrinsic counting mechanism that determines when precursor cell proliferation stops and differentiation begins, and the effector mechanism that arrests the cell cycle when the counting mechanism indicates it is time. Others have recently found that p27-deficient mice have an increased number of cells in all of the organs examined; this suggests that this function of p27 is not restricted to the oligodendrocyte cell lineage (Durand, 1997).

gax is a relatively divergent Antp class homeobox gene with a homeodomain nearly identical to that of Mox-1, a recently described homeobox gene restricted to mesoderm and mesodermally derived tissue. Gax is less homologous to either proboscipedia (66% identity in the homeodomain) or to Deformed (61% identity in the homeodomain. gax is expressed in vascular smooth muscle cells (VSMCs) and is down-regulated in vitro by mitogen stimulation and in vivo in response to vascular injury that leads to cellular proliferation. Recombinant Gax protein microinjected into VSMCs and fibroblasts inhibited the mitogen-induced entry into S-phase when introduced either during quiescence or early stages of G1. Overexpression of gax with a replication-defective adenovirus vector results in G0/G1 cell cycle arrest of VSMCs and fibroblasts. The gax-induced growth inhibition correlates with a p53-independent up-regulation of the cyclin-dependent kinase inhibitor p21. Gax overexpression also leads to an association of p21 with cdk2 complexes and a decrease in cdk2 activity. Fibroblasts deficient in p21 are not susceptible to a reduction in cdk2 activity or growth inhibition by gax overexpression. Localized delivery of the virus to denuded rat carotid arteries significantly reduces neointima formation and luminal narrowing. These data indicate that gax overexpression can inhibit cell proliferation in a p21-dependent manner and can modulate injury-induced changes in vessel wall morphology that result from excessive cellular proliferation (Smith, 1997).

Much of the predisposition to hereditary breast and ovarian cancer has been attributed to inherited defects in the BRCA1 tumour-suppressor gene. The nuclear protein BRCA1 has the properties of a transcription factor, and can interact with the recombination and repair protein RAD51. Young women with germline alterations in BRCA1 develop breast cancer at rates 100-fold higher than the general population, and BRCA1-null mice die before day 8 of development. However, the mechanisms of BRCA1-mediated growth regulation and tumour suppression remain unknown. BRCA1 transactivates expression of the cyclin-dependent kinase inhibitor p21WAF1/CIP1 in a p53-independent manner, and BRCA1 inhibits cell-cycle progression into the S-phase following its transfection into human cancer cells. BRCA1 does not inhibit S-phase progression in p21-/- cells, unlike p21+/+ cells. Tumour-associated, transactivation-deficient mutants of BRCA1 are defective in both transactivation of p21 and cell-cycle inhibition. These data suggest that one mechanism by which BRCA1 contributes to cell-cycle arrest and growth suppression is through the induction of p21 (Somasundaram, 1997).

The cyclin-dependent kinase inhibitor p21(Cip1/WAF1) has been implicated as an inducer of differentiation. However, although expression of p21 is increased in postmitotic cells immediately adjacent to the proliferative compartment, its expression is decreased in cells further along the differentiation program. Expression of the p21 protein is decreased in terminally differentiated primary keratinocytes of mice, and this occurs by a proteasome-dependent pathway. Forced expression of p21 in these cells inhibits the expression of markers of terminal differentiation at both the protein and messenger RNA levels. These inhibitory effects on differentiation are not observed with a carboxyl-terminal truncation mutant or with the unrelated cyclin-dependent kinase inhibitor p16(INK4a), although all these proteins exert similar inhibition of cell growth. These findings reveal an inhibitory role for p21 in the late stages of differentiation that does not result from the effects of p21 on the cell cycle. An attractive model for future studies is that p21 may function as a specific bridge between signaling complexes (such as cyclin-CDK complexes and SAPKs) and other multiprotein apparatuses, such as the transcription machinery involved in differentation (Di Cunto, 1998).

Transient transfection of vectors expressing neuroD2, MASH1, ngn1 or related neural bHLH proteins, with their putative dimerization partner E12, can convert mouse P19 embryonal carcinoma cells into differentiated neurons. Transfected cells express numerous neuron-specific proteins, adopt a neuronal morphology and are electrically excitable. Pan-neuronal markers such as neurofilament-M, the HuC/D RNA-binding proteins, M6 and synapsin I are all present in most or all cells with neuronal morphology. Moreover, subsets of the transfected cells are immunoreactive for the neurotransmitters GABA and glutamate, the GABA synthetic enzyme glutamatic acid decarboxylase (GAD), the neuropeptide substance P, NMDA receptor 1, and the transcription factors Islet-1 or LIM1. At present, it is not known if the subsets of cells that express these proteins overlap. The same constellation of neurotransmitters, receptors and other markers of mature neuronal subtypes have been observed regardless of which neural bHLH vector is transfected. No expression of glial fibrillary acidic protein (GFAP) or markers of radial glia have been observed in any transfected cells. Thus, the expression of neural bHLH proteins is sufficient to confer a neuronal fate on uncommitted mammalian cells. Neuronal differentiation of transfected cells is preceded by elevated expression of the cyclin-dependent kinase inhibitor p27 Kip1 and cell cycle withdrawal. This demonstrates that the bHLH proteins can link neuronal differentiation to withdrawal from the cell cycle, possibly by activating the expression of p27 Kip1. The ability to generate mammalian neurons by transient expression of neural bHLH proteins should create new opportunities for studying neurogenesis and devising neural repair strategies (Farah, 2000).

Relative quiescence is a defining characteristic of hematopoietic stem cells, while their progeny have dramatic proliferative ability and inexorably move toward terminal differentiation. The quiescence of stem cells has been conjectured to be of critical biologic importance in protecting the stem cell compartment; this has been directly assessed using mice engineered to be deficient in the G1 checkpoint regulator, cyclin-dependent kinase inhibitor, p21cip1/waf1 (p21). In the absence of p21, hematopoietic stem cell proliferation and absolute number are increased under normal homeostatic conditions. Exposing the animals to cell cycle-specific myelotoxic injury results in premature death due to hematopoietic cell depletion. Further, self-renewal of primitive cells is impaired in serially transplanted bone marrow from p21-/- mice, leading to hematopoietic failure. Therefore, p21 is the molecular switch governing the entry of stem cells into the cell cycle, and in its absence, increased cell cycling leads to stem cell exhaustion. Under conditions of stress, restricted cell cycling is crucial to prevent premature stem cell depletion and hematopoietic death (Cheng, 2000).

A precise balance between proliferation and differentiation must be maintained during retinal development to obtain the correct proportion of each of the seven cell types found in the adult tissue. Cyclin kinase inhibitors can regulate cell cycle exit coincident with induction of differentiation programs during development. The p57 Kip2 cyclin kinase inhibitor is upregulated during G1/G0 in a subset of retinal progenitor cells exiting the cell cycle between embryonic day 14.5 and 16.5 of mouse development. Retroviral mediated overexpression of p57 Kip2 in embryonic retinal progenitor cells leads to premature cell cycle exit. Retinae from mice lacking p57 Kip2 exhibit inappropriate S-phase entry and apoptotic nuclei were found in the region where p57 Kip2 is normally expressed. Apoptosis precisely compensates for the inappropriate proliferation in the p57 Kip2-deficient retinae to preserve the correct proportion of the major retinal cell types. Postnatally, p57 Kip2 is expressed in a novel subpopulation of amacrine interneurons. At this stage, p57 Kip2 does not regulate proliferation. However, perhaps reflecting its role during this late stage of development, animals lacking p57 Kip2 show an alteration in amacrine subpopulations. p57 Kip2 is the first gene to be implicated as a regulator of amacrine subtype/subpopulation development. Consequently, it is proposed that p57 Kip2 has two roles during retinal development, acting first as a cyclin kinase inhibitor in mitotic progenitor cells, and then playing a distinct role in neuronal differentiation (Dyer, 2000).

Several lines of evidence suggest that Cip/Kip family members in general, and p57 Kip2 in particular, may be involved in developmental processes beyond their prescribed roles as cyclin kinase inhibitors. The Cip/Kip family members are promiscuous kinase inhibitors and may regulate kinases required for cell fate specification and/or differentiation during development. In addition to their shared cyclin kinase binding domains, the Cip/Kip proteins have distinct, structurally complex domains that may prove to contain novel biochemical properties. This is especially true for p57 Kip2 , which is the most structurally complex family member. Several of the abnormalities observed in the p57 Kip2 knockout mice (including defects in muscle, kidney, palate and chondrocyte development) appear to not be linked to proliferation defects. It has therefore been proposed that this protein directly influences cell fate specification and/or differentiation in these tissues. The Xenopus cyclin kinase inhibitor p27 Xic1, which is a member of the Cip/Kip family and is believed to be related to the mammalian protein p27 Kip1, has been shown to induce the M¨ller glial cell fate during retinal development. Consistent with the idea that cyclin kinase inhibitors may be bifunctional molecules, the Müller-inducing portion of the p27 Xic1 is found to be separable from the portion of protein that induces cell cycle exit. Cyclin kinase inhibitors are not the only molecules that may regulate both cell cycle progression and cell fate specification. A novel protein cloned from Xenopus called geminin was found to contain a neuralizing domain that was separable from a domain that has been shown to be involved in the regulation of DNA replication. Taken together, these reports indicate that cell cycle exit and cell fate specification are not only coordinated temporally during development but that individual molecules can play roles in both processes through distinct protein domains (Dyer, 2000 and references therein).

The molecular basis of the antagonism between cellular proliferation and differentiation is poorly understood. The role of the cyclin-dependent kinase inhibitor p27Xic1 has been investigated in the co-ordination of cell cycle exit and differentiation during early myogenesis in vivo using Xenopus embryos. p27Xic1 is highly expressed in the developing myotome; ablation of p27Xic1 protein prevents muscle differentiation, and that p27Xic1 synergizes with the transcription factor MyoD to promote muscle differentiation. Furthermore, the ability of p27Xic1 to promote myogenesis resides in an N-terminal domain and is separable from its cell cycle regulation function. This data demonstrates that a single cyclin-dependent kinase inhibitor, p27Xic1, controls in vivo muscle differentiation in Xenopus and that regulation of this process by p27Xic1 requires activities beyond cell cycle inhibition (Vernon, 2003a).

The role of the cyclin-dependent kinase inhibitor, p27Xic1, has been investigated in the coordination of cell cycle exit and differentiation during early neurogenesis. p27Xic1 is highly expressed in cells destined to become primary neurons and is essential for an early stage of neurogenesis. Ablation of p27Xic1 protein prevents differentiation of primary neurons, while overexpressing p27Xic1 promotes their formation. p27Xic1 may enhance neurogenesis by stabilizing the bHLH protein, neurogenin, although the molecular mechanism of this stabilization is unknown. Moreover, the ability of p27Xic1 to stabilize neurogenin and enhance neurogenesis localizes to an N-terminal domain of the molecule and is separable from its ability to inhibit the cell cycle (Vernon, 2003b).

Precursors of cochlear and vestibular hair cells of the inner ear exit the cell cycle at midgestation. Hair cells are mitotically quiescent during late-embryonic differentiation stages and postnatally. The retinoblastoma gene Rb and the encoded protein pRb are expressed in differentiating and mature hair cells. In addition to Rb, the cyclin dependent kinase inhibitor (CKI) p21 is expressed in developing hair cells, suggesting that p21 is an upstream effector of pRb activity. p21 apparently cooperates with other CKIs, since p21-null mice exhibit an unaltered inner ear phenotype. By contrast, Rb inactivation leads to aberrant hair cell proliferation, as analysed at birth in a loss-of-function/transgenic mouse model. Supernumerary hair cells express various cell type-specific differentiation markers, including components of stereocilia. The extent of alterations in stereociliary bundle morphology ranges from near-normal to severe disorganization. Apoptosis contributes to the mutant phenotype, but does not compensate for the production of supernumerary hair cells, resulting in hyperplastic sensory epithelia. The Rb-null-mediated proliferation leads to a distinct pathological phenotype, including multinucleated and enlarged hair cells, and infiltration of hair cells into the mesenchyme. These findings demonstrate that the pRb pathway is required for hair cell quiescence and that manipulation of the cell cycle machinery disrupts the coordinated development within the inner ear sensory epithelia (Mantela, 2005).

These data show that the CKI p21 is expressed in the differentiating cochlear and vestibular HCs, and that the expression is induced at the initiation of HC differentiation. In the auditory sensory epithelium, p21 expression is initiated at E14.5, at the stage when Math1 expression is first detected. It is possible that p21 induction in HCs is regulated by Math1, by analogy to the positive role of bHLH proteins such as Myod1 and myogenin in skeletal myogenesis. Thereafter, p21 together with other CKI(s) might have an active role in keeping pRb in a hypophosphorylated form. Thus, negative regulation at the level of both pRb and CKIs seems to be responsible for the maintenance of HC quiescence. No phenotypic alterations or aberrant mitoses were found in the inner ears of developing or adult p21-/- mice. Interestingly, in addition to p21, another CKI, p19, has been shown to be expressed in the late-embryonic organ of Corti, but its inactivation does not result in developmental abnormalities. Thus, functional redundancy may exist between p21 and p19 in developing cochlear HCs. In addition, developing vestibular HCs express p21, but do not show phenotypic changes following targeted gene disruption, most probably owing to functional compensation. The identity of the CKI that may cooperate with p21 in vestibular HCs remains to be identified, since p19 expression and the consequences of p19 inactivation have not been reported in vestibular organs (Mantela, 2005).

Integrin-extracellular matrix interactions play important roles in the coordinated integration of external and internal cues that are essential for proper development. To study the role of beta1 integrin in the mammary gland, Itgbeta1flox/flox mice were crossed with WAPiCre transgenic mice, which led to specific ablation of beta1 integrin in luminal alveolar epithelial cells. In the beta1 integrin mutant mammary gland, individual alveoli were disorganized resulting from alterations in cell-basement membrane associations. Activity of focal adhesion kinase (FAK) was also decreased in mutant mammary glands. Luminal cell proliferation was strongly inhibited in beta1 integrin mutant glands, which correlated with a specific increase of p21 Cip1 expression. In a p21 Cip1 null background, there was a partial rescue of BrdU incorporation, providing in vivo evidence linking p21 Cip1 to the proliferative defect observed in beta1 integrin mutant glands. A connection between p21 Cip1 and beta1 integrin as well as FAK was also established in primary mammary cells. These results point to the essential role of beta1 integrin signaling in mammary epithelial cell proliferation (Li, 2005).

Antagonistic regulation of p57kip2 by Hes/Hey downstream of Notch signaling and muscle regulatory factors regulates skeletal muscle growth arrest

A central question in development is to define how the equilibrium between cell proliferation and differentiation is temporally and spatially regulated during tissue formation. This study addresses how interactions between cyclin-dependent kinase inhibitors essential for myogenic growth arrest (p21cip1 and p57kip2), the Notch pathway and myogenic regulatory factors (MRFs) orchestrate the proliferation, specification and differentiation of muscle progenitor cells. It was first shown that cell cycle exit and myogenic differentiation can be uncoupled. In addition, it was establish that skeletal muscle progenitor cells require Notch signaling to maintain their cycling status. Using several mouse models combined with ex vivo studies, it was demonstrated that Notch signaling is required to repress p21cip1 and p57kip2 expression in muscle progenitor cells. Finally, a muscle-specific regulatory element of p57kip2 directly activated by MRFs was identified in myoblasts but was found to be repressed by the Notch targets Hes1/Hey1 in progenitor cells. A molecular mechanism is proposed whereby information provided by Hes/Hey downstream of Notch as well as MRF activities are integrated at the level of the p57kip2 enhancer to regulate the decision between progenitor cell maintenance and muscle differentiation (Zalc, 2014).

back to CDK inhibitors, cell cycle arrest and differentiation part 1/2


Table of contents


dacapo: Biological Overview | Regulation | Developmental Biology | Effects of Mutation | References

Home page: The Interactive Fly © 1995, 1996 Thomas B. Brody, Ph.D.

The Interactive Fly resides on the
Society for Developmental Biology's Web server.