Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EMBO J ; 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38719996

RESUMO

Extracellular vesicles (EVs) are important mediators of communication between cells. Here, we reveal a new mode of intercellular communication by melanosomes, large EVs secreted by melanocytes for melanin transport. Unlike small EVs, which are disintegrated within the receiver cell, melanosomes stay intact within them, gain a unique protein signature, and can then be further transferred to another cell as "second-hand" EVs. We show that melanoma-secreted melanosomes passaged through epidermal keratinocytes or dermal fibroblasts can be further engulfed by resident macrophages. This process leads to macrophage polarization into pro-tumor or pro-immune cell infiltration phenotypes. Melanosomes that are transferred through fibroblasts can carry AKT1, which induces VEGF secretion from macrophages in an mTOR-dependent manner, promoting angiogenesis and metastasis in vivo. In melanoma patients, macrophages that are co-localized with AKT1 are correlated with disease aggressiveness, and immunotherapy non-responders are enriched in macrophages containing melanosome markers. Our findings suggest that interactions mediated by second-hand extracellular vesicles contribute to the formation of the metastatic niche, and that blocking the melanosome cues of macrophage diversification could be helpful in halting melanoma progression.

2.
Elife ; 112022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36124553

RESUMO

Despite the remarkable successes of cancer immunotherapies, the majority of patients will experience only partial response followed by relapse of resistant tumors. While treatment resistance has frequently been attributed to clonal selection and immunoediting, comparisons of paired primary and relapsed tumors in melanoma and breast cancers indicate that they share the majority of clones. Here, we demonstrate in both mouse models and clinical human samples that tumor cells evade immunotherapy by generating unique transient cell-in-cell structures, which are resistant to killing by T cells and chemotherapies. While the outer cells in this cell-in-cell formation are often killed by reactive T cells, the inner cells remain intact and disseminate into single tumor cells once T cells are no longer present. This formation is mediated predominantly by IFNγ-activated T cells, which subsequently induce phosphorylation of the transcription factors signal transducer and activator of transcription 3 (STAT3) and early growth response-1 (EGR-1) in tumor cells. Indeed, inhibiting these factors prior to immunotherapy significantly improves its therapeutic efficacy. Overall, this work highlights a currently insurmountable limitation of immunotherapy and reveals a previously unknown resistance mechanism which enables tumor cells to survive immune-mediated killing without altering their immunogenicity.


Cancer immunotherapies use the body's own immune system to fight off cancer. But, despite some remarkable success stories, many patients only see a temporary improvement before the immunotherapy stops being effective and the tumours regrow. It is unclear why this occurs, but it may have to do with how the immune system attacks cancer cells. Immunotherapies aim to activate a special group of cells known as killer T-cells, which are responsible for the immune response to tumours. These cells can identify cancer cells and inject toxic granules through their membranes, killing them. However, killer T-cells are not always effective. This is because cancer cells are naturally good at avoiding detection, and during treatment, their genes can mutate, giving them new ways to evade the immune system. Interestingly, when scientists analysed the genes of tumour cells before and after immunotherapy, they found that many of the genes that code for proteins recognized by T-cells do not change significantly. This suggests that tumours' resistance to immune attack may be physical, rather than genetic. To investigate this hypothesis, Gutwillig et al. developed several mouse tumour models that stop responding to immunotherapy after initial treatment. Examining cells from these tumours revealed that when the immune system attacks, they reorganise by getting inside one another. This allows some cancer cells to hide under many layers of cell membrane. At this point killer T-cells can identify and inject the outer cell with toxic granules, but it cannot reach the cells inside. This ability of cancer cells to hide within one another relies on them recognising when the immune system is attacking. This happens because the cancer cells can detect certain signals released by the killer T-cells, allowing them to hide. Gutwillig et al. identified some of these signals, and showed that blocking them stopped cancer cells from hiding inside each other, making immunotherapy more effective. This new explanation for how cancer cells escape the immune system could guide future research and lead to new cancer treatments, or approaches to boost existing treatments. Understanding the process in more detail could allow scientists to prevent it from happening, by revealing which signals to block, and when, for best results.


Assuntos
Formação de Célula em Célula , Melanoma , Animais , Humanos , Fatores Imunológicos , Imunoterapia , Melanoma/terapia , Camundongos , Recidiva , Fator de Transcrição STAT3
3.
PLoS Biol ; 20(8): e3001756, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35969606

RESUMO

Mitotic spindle orientation (SO) is a conserved mechanism that governs cell fate and tissue morphogenesis. In the developing epidermis, a balance between self-renewing symmetric divisions and differentiative asymmetric divisions is necessary for normal development. While the cellular machinery that executes SO is well characterized, the extrinsic cues that guide it are poorly understood. Here, we identified the basal cell adhesion molecule (BCAM), a ß1 integrin coreceptor, as a novel regulator of epidermal morphogenesis. In utero RNAi-mediated depletion of Bcam in the mouse embryo did not hinder ß1 integrin distribution or cell adhesion and polarity. However, Bcam depletion promoted apoptosis, thinning of the epidermis, and symmetric cell division, and the defects were reversed by concomitant overexpression of the apoptosis inhibitor Xiap. Moreover, in mosaic epidermis, depletion of Bcam or Xiap induced symmetric divisions in neighboring wild-type cells. These results identify apoptosis and epidermal architecture as extrinsic cues that guide SO in the developing epidermis.


Assuntos
Integrina beta1 , Fuso Acromático , Animais , Apoptose , Divisão Celular , Polaridade Celular , Epiderme , Integrina beta1/metabolismo , Camundongos , Fuso Acromático/metabolismo
4.
Nat Cell Biol ; 24(7): 1049-1063, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35798842

RESUMO

Anchored cells of the basal epidermis constantly undergo proliferation in an overcrowded environment. An important regulator of epidermal proliferation is YAP, which can be controlled by both cell-matrix and cell-cell interactions. Here, we report that THY1, a GPI-anchored protein, inhibits epidermal YAP activity through converging molecular mechanisms. THY1 deficiency leads to increased adhesion by activating the integrin-ß1-SRC module. Notably, regardless of high cellular densities, the absence of THY1 leads to the dissociation of an adherens junction complex that enables the release and translocation of YAP. Due to increased YAP-dependent proliferation, Thy1-/- mice display enhanced wound repair and hair follicle regeneration. Taken together, our work reveals THY1 as a crucial regulator of cell-matrix and cell-cell interactions that controls YAP activity in skin homeostasis and regeneration.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Ciclo Celular , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Epiderme/metabolismo , Homeostase , Camundongos , Pele/metabolismo
5.
BMC Biol ; 20(1): 145, 2022 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-35710398

RESUMO

BACKGROUND: The establishment of tissue architecture requires coordination between distinct processes including basement membrane assembly, cell adhesion, and polarity; however, the underlying mechanisms remain poorly understood. The actin cytoskeleton is ideally situated to orchestrate tissue morphogenesis due to its roles in mechanical, structural, and regulatory processes. However, the function of many pivotal actin-binding proteins in mammalian development is poorly understood. RESULTS: Here, we identify a crucial role for anillin (ANLN), an actin-binding protein, in orchestrating epidermal morphogenesis. In utero RNAi-mediated silencing of Anln in mouse embryos disrupted epidermal architecture marked by adhesion, polarity, and basement membrane defects. Unexpectedly, these defects cannot explain the profoundly perturbed epidermis of Anln-depleted embryos. Indeed, even before these defects emerge, Anln-depleted epidermis exhibits abnormalities in mitotic rounding and its associated processes: chromosome segregation, spindle orientation, and mitotic progression, though not in cytokinesis that was disrupted only in Anln-depleted cultured keratinocytes. We further show that ANLN localizes to the cell cortex during mitotic rounding, where it regulates the distribution of active RhoA and the levels, activity, and structural organization of the cortical actomyosin proteins. CONCLUSIONS: Our results demonstrate that ANLN is a major regulator of epidermal morphogenesis and identify a novel role for ANLN in mitotic rounding, a near-universal process that governs cell shape, fate, and tissue morphogenesis.


Assuntos
Proteínas Contráteis , Proteínas dos Microfilamentos , Citoesqueleto de Actina/metabolismo , Animais , Proteínas Contráteis/metabolismo , Citocinese/fisiologia , Mamíferos , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo
6.
Development ; 147(23)2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33310787

RESUMO

Planar cell polarity (PCP) is essential for tissue morphogenesis and homeostasis; however, the mechanisms that orchestrate the cell shape and packing dynamics required to establish PCP are poorly understood. Here, we identified a major role for the globular (G)-actin-binding protein thymosin-ß4 (TMSB4X) in PCP establishment and cell adhesion in the developing epidermis. Depletion of Tmsb4x in mouse embryos hindered eyelid closure and hair-follicle angling owing to PCP defects. Tmsb4x depletion did not preclude epidermal cell adhesion in vivo or in vitro; however, it resulted in abnormal structural organization and stability of adherens junction (AJ) due to defects in filamentous (F)-actin and G-actin distribution. In cultured keratinocytes, TMSB4X depletion increased the perijunctional G/F-actin ratio and decreased G-actin incorporation into junctional actin networks, but it did not change the overall actin expression level or cellular F-actin content. A pharmacological treatment that increased the G/F-actin ratio and decreased actin polymerization mimicked the effects of Tmsb4x depletion on both AJs and PCP. Our results provide insights into the regulation of the actin pool and its involvement in AJ function and PCP establishment.


Assuntos
Polaridade Celular/genética , Desenvolvimento Embrionário/genética , Morfogênese/genética , Timosina/genética , Citoesqueleto de Actina/genética , Actinas/genética , Junções Aderentes/genética , Animais , Adesão Celular/genética , Forma Celular/genética , Células Epidérmicas/metabolismo , Epiderme/crescimento & desenvolvimento , Homeostase/genética , Queratinócitos/metabolismo , Camundongos , Proteínas dos Microfilamentos/genética
7.
J Vis Exp ; (165)2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-33191932

RESUMO

Genetically modified mouse models (GEMM) have been instrumental in assessing gene function, modeling human diseases, and serving as preclinical model to assess therapeutic avenues. However, their time-, labor- and cost-intensive nature limits their utility for systematic analysis of gene function. Recent advances in genome-editing technologies overcome those limitations and allow for the rapid generation of specific gene perturbations directly within specific mouse organs in a multiplexed and rapid manner. Here, we describe a CRISPR/Cas9-based method (Clustered Regularly Interspaced Short Palindromic Repeats) to generate thousands of gene knock-out clones within the epithelium of the skin and oral cavity of mice, and provide a protocol detailing the steps necessary to perform a direct in vivo CRISPR screen for tumor suppressor genes. This approach can be applied to other organs or other CRISPR/Cas9 technologies such as CRISPR-activation or CRISPR-inactivation to study the biological function of genes during tissue homeostasis or in various disease settings.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Testes Genéticos , Boca/metabolismo , Pele/metabolismo , Animais , Sequência de Bases , Proteína 9 Associada à CRISPR/genética , Epitélio/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Integrases/metabolismo , Camundongos , Microinjeções , Fenótipo , Ultrassom
8.
Front Cell Dev Biol ; 8: 615, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32766247

RESUMO

Striatin, a subunit of the serine/threonine phosphatase PP2A, is a core member of the conserved striatin-interacting phosphatase and kinase (STRIPAK) complexes. The protein is expressed in the cell junctions between epithelial cells, which play a role in maintaining cell-cell adhesion. Since the cell junctions are crucial for the function of the mammalian inner ear, we examined the localization and function of striatin in the mouse cochlea. Our results show that in neonatal mice, striatin is specifically expressed in the cell-cell junctions of the inner hair cells, the receptor cells in the mammalian cochlea. Auditory brainstem response measurements of striatin-deficient mice indicated a progressive, high-frequency hearing loss, suggesting that striatin is essential for normal hearing. Moreover, scanning electron micrographs of the organ of Corti revealed a moderate degeneration of the outer hair cells in the middle and basal regions, concordant with the high-frequency hearing loss. Additionally, striatin-deficient mice show aberrant ribbon synapse maturation. Loss of the outer hair cells, combined with the aberrant ribbon synapse distribution, may lead to the observed auditory impairment. Together, these results suggest a novel function for striatin in the mammalian auditory system.

9.
Nat Commun ; 11(1): 2711, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32483135

RESUMO

p16INK4a (CDKN2A) is a central tumor suppressor, which induces cell-cycle arrest and senescence. Cells expressing p16INK4a accumulate in aging tissues and appear in premalignant lesions, yet their physiologic effects are poorly understood. We found that prolonged expression of transgenic p16INK4a in the mouse epidermis induces hyperplasia and dysplasia, involving high proliferation rates of keratinocytes not expressing the transgene. Continuous p16INK4a expression increases the number of epidermal papillomas formed after carcinogen treatment. Wnt-pathway ligands and targets are activated upon prolonged p16INK4a expression, and Wnt inhibition suppresses p16INK4a-induced hyperplasia. Senolytic treatment reduces p16INK4a-expressing cell numbers, and inhibits Wnt activation and hyperplasia. In human actinic keratosis, a precursor of squamous cell carcinoma, p16INK4a-expressing cells are found adjacent to dividing cells, consistent with paracrine interaction. These findings reveal that chronic p16INK4a expression is sufficient to induce hyperplasia through Wnt-mediated paracrine stimulation, and suggest that this tumor suppressor can promote early premalignant epidermal lesion formation.


Assuntos
Transformação Celular Neoplásica/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Epiderme/metabolismo , Via de Sinalização Wnt/genética , Animais , Proliferação de Células/genética , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Humanos , Hiperplasia/genética , Hiperplasia/metabolismo , Queratinócitos/metabolismo , Ceratose/genética , Ceratose/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Papiloma/genética , Papiloma/metabolismo , Papiloma/patologia
10.
Mol Cell Oncol ; 6(5): e1609877, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31528690

RESUMO

The WASP-family verprolin-homologous (Wave) complex enhances actin-related protein 2/3 (Arp2/3)-mediated actin polymerization. Recently, we have identified a novel Wave complex-actin-Wnt/ß-catenin-Sox9 pathway that regulates epidermal morphogenesis and proliferation. These findings highlight a mechanism by which actin polymerization impacts crucial signaling events in vivo.

11.
Cell Rep ; 28(7): 1785-1798.e6, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31412247

RESUMO

Melanoma is the deadliest skin cancer due to its high rate of metastasis, frequently to the brain. Brain metastases are incurable; therefore, understanding melanoma brain metastasis is of great clinical importance. We used a mouse model of spontaneous melanoma brain metastasis to study the interactions of melanomas with the brain microenvironment. We find that CXCL10 is upregulated in metastasis-associated astrocytes in mice and humans and is functionally important for the chemoattraction of melanoma cells. Moreover, CXCR3, the receptor for CXCL10, is upregulated in brain-tropic melanoma cells. Targeting melanoma expression of CXCR3 by nanoparticle-mediated siRNA delivery or by shRNA transduction inhibits melanoma cell migration and attenuates brain metastasis in vivo. These findings suggest that the instigation of pro-inflammatory signaling in astrocytes is hijacked by brain-metastasizing tumor cells to promote their metastatic capacity and that the CXCL10-CXCR3 axis may be a potential therapeutic target for the prevention of melanoma brain metastasis.


Assuntos
Astrócitos/patologia , Neoplasias Encefálicas/secundário , Quimiocina CXCL10/metabolismo , Modelos Animais de Doenças , Inflamação/imunologia , Melanoma/patologia , Receptores CXCR3/metabolismo , Animais , Astrócitos/imunologia , Astrócitos/metabolismo , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Movimento Celular , Quimiocina CXCL10/genética , Humanos , Inflamação/metabolismo , Inflamação/patologia , Masculino , Melanoma/imunologia , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Receptores CXCR3/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia , Microambiente Tumoral
12.
J Cell Biol ; 218(4): 1390-1406, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30867227

RESUMO

Development of the skin epidermis requires tight spatiotemporal control over the activity of several signaling pathways; however, the mechanisms that orchestrate these events remain poorly understood. Here, we identify a key role for the Wave complex proteins ABI1 and Wave2 in regulating signals that control epidermal shape and growth. In utero RNAi-mediated silencing of Abi1 or Wasf2 induced cellular hyperproliferation and defects in architecture of the interfollicular epidermis (IFE) and delayed hair follicle growth. Unexpectedly, SOX9, a hair follicle growth regulator, was aberrantly expressed throughout the IFE of the mutant embryos, and its forced overexpression mimicked the Wave complex loss-of-function phenotype. Moreover, Wnt signaling, which regulates SOX9+ cell specification, was up-regulated in Wave complex loss-of-function IFE. Importantly, we show that the Wave complex regulates filamentous actin content and that a decrease in actin levels is sufficient to elevate Wnt/ß-catenin signaling. Our results identify a novel role for Wave complex- and actin-regulated signaling via Wnt and SOX9 in skin development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proliferação de Células , Proteínas do Citoesqueleto/metabolismo , Epiderme/metabolismo , Queratinócitos/metabolismo , Fatores de Transcrição SOX9/metabolismo , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , Via de Sinalização Wnt , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células Cultivadas , Proteínas do Citoesqueleto/genética , Epiderme/anormalidades , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Folículo Piloso/anormalidades , Folículo Piloso/metabolismo , Camundongos , Morfogênese , Fosforilação , Fatores de Transcrição SOX9/genética , Família de Proteínas da Síndrome de Wiskott-Aldrich/genética , beta Catenina/metabolismo
13.
Exp Cell Res ; 378(2): 232-237, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30872138

RESUMO

Animal cells exhibit a wide range of shapes that reflect their diverse functions. Cell shape is determined by a balance between internal and external forces and therefore involves the cytoskeleton and its associated adhesion structures. Cell shape dynamics during development and homeostasis are tightly regulated and closely coordinated with cell fate determination. Defects in cell shape are a hallmark of many pathological conditions including cancer and skin diseases. This review highlights the links between cell shape and cell fate in the epidermis, which have been studied for over 40 years both in vitro and in vivo. Briefly discussing seminal experiments showing the strong coupling between keratinocyte cell shape and their fate we primarily focus on recent studies uncovering novel cellular and molecular mechanisms linking epidermal cell shape with cell growth, differentiation, asymmetric division, and delamination.


Assuntos
Diferenciação Celular , Forma Celular , Epiderme/fisiologia , Animais , Divisão Celular , Humanos , Queratinócitos/fisiologia , Mitose , Pele/citologia
14.
Stem Cells ; 37(3): 417-429, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30548157

RESUMO

Mutations in key transcription factors SOX2 and P63 were linked with developmental defects and postnatal abnormalities such as corneal opacification, neovascularization, and blindness. The latter phenotypes suggest that SOX2 and P63 may be involved in corneal epithelial regeneration. Although P63 has been shown to be a key regulator of limbal stem cells, the expression pattern and function of SOX2 in the adult cornea remained unclear. Here, we show that SOX2 regulates P63 to control corneal epithelial stem/progenitor cell function. SOX2 and P63 were co-expressed in the stem/progenitor cell compartments of the murine cornea in vivo and in undifferentiated human limbal epithelial stem/progenitor cells in vitro. In line, a new consensus site that allows SOX2-mediated regulation of P63 enhancer was identified while repression of SOX2 reduced P63 expression, suggesting that SOX2 is upstream to P63. Importantly, knockdown of SOX2 significantly attenuated cell proliferation, long-term colony-forming potential of stem/progenitor cells, and induced robust cell differentiation. However, this effect was reverted by forced expression of P63, suggesting that SOX2 acts, at least in part, through P63. Finally, miR-450b was identified as a direct repressor of SOX2 that was required for SOX2/P63 downregulation and cell differentiation. Altogether, we propose that SOX2/P63 pathway is an essential regulator of corneal stem/progenitor cells while mutations in SOX2 or P63 may disrupt epithelial regeneration, leading to loss of corneal transparency and blindness. Stem Cells 2019;37:417-429.


Assuntos
Diferenciação Celular , Proliferação de Células , Epitélio Corneano/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Alcaloides , Animais , Camundongos , Células NIH 3T3 , Piperidinas , Fatores de Transcrição SOXB1/genética , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética
15.
Sci Signal ; 10(481)2017 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-28559444

RESUMO

The establishment of epithelial architecture is a complex process involving cross-talk between cells and the basement membrane. Basement membrane assembly requires integrin activity but the role of the associated actomyosin cytoskeleton is poorly understood. Here, we identify the actin-bundling protein T-plastin (Pls3) as a regulator of basement membrane assembly and epidermal morphogenesis. In utero depletion of Pls3 transcripts in mouse embryos caused basement membrane and polarity defects in the epidermis but had little effect on cell adhesion and differentiation. Loss-of-function experiments demonstrated that the apicobasal polarity defects were secondary to the disruption of the basement membrane. However, the basement membrane itself was profoundly sensitive to subtle perturbations in the actin cytoskeleton. We further show that Pls3 localized to the cell cortex, where it was essential for the localization and activation of myosin II. Inhibition of myosin II motor activity disrupted basement membrane organization. Our results provide insights into the regulation of cortical actomyosin and its importance for basement membrane assembly and skin morphogenesis.


Assuntos
Membrana Basal/fisiologia , Embrião de Mamíferos/citologia , Epiderme/crescimento & desenvolvimento , Glicoproteínas de Membrana/fisiologia , Proteínas dos Microfilamentos/fisiologia , Morfogênese , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Animais , Adesão Celular/fisiologia , Diferenciação Celular , Polaridade Celular , Embrião de Mamíferos/metabolismo , Epiderme/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos ICR , Microtúbulos/metabolismo , Miosina Tipo II/metabolismo
16.
Handb Exp Pharmacol ; 235: 263-284, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27807694

RESUMO

The ability of cells to generate, maintain, and repair tissues with complex architecture, in which distinct cells function as coherent units, relies on polarity cues. Polarity can be described as an asymmetry along a defined axis, manifested at the molecular, structural, and functional levels. Several types of cell and tissue polarities were described in the literature, including front-back, apical-basal, anterior-posterior, and left-right polarity. Extensive research provided insights into the specific regulators of each polarization process, as well as into generic elements that affect all types of polarities. The actin cytoskeleton and the associated adhesion structures are major regulators of most, if not all, known forms of polarity. Actin filaments exhibit intrinsic polarity and their ability to bind many proteins including the mechanosensitive adhesion and motor proteins, such as myosins, play key roles in cell polarization. The actin cytoskeleton can generate mechanical forces and together with the associated adhesions, probe the mechanical, structural, and chemical properties of the environment, and transmit signals that impact numerous biological processes, including cell polarity. In this article we highlight novel mechanisms whereby the mechanical forces and actin-adhesion complexes regulate cell and tissue polarity in a variety of natural and experimental systems.


Assuntos
Citoesqueleto de Actina/fisiologia , Polaridade Celular , Animais , Adesão Celular , Humanos
17.
PLoS Genet ; 12(10): e1006369, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27736875

RESUMO

Despite recent advances in our understanding of the pathogenesis of ectodermal dysplasias (EDs), the molecular basis of many of these disorders remains unknown. In the present study, we aimed at elucidating the genetic basis of a new form of ED featuring facial dysmorphism, scalp hypotrichosis and hypodontia. Using whole exome sequencing, we identified 2 frameshift and 2 missense mutations in TSPEAR segregating with the disease phenotype in 3 families. TSPEAR encodes the thrombospondin-type laminin G domain and EAR repeats (TSPEAR) protein, whose function is poorly understood. TSPEAR knock-down resulted in altered expression of genes known to be regulated by NOTCH and to be involved in murine hair and tooth development. Pathway analysis confirmed that down-regulation of TSPEAR in keratinocytes is likely to affect Notch signaling. Accordingly, using a luciferase-based reporter assay, we showed that TSPEAR knock-down is associated with decreased Notch signaling. In addition, NOTCH1 protein expression was reduced in patient scalp skin. Moreover, TSPEAR silencing in mouse hair follicle organ cultures was found to induce apoptosis in follicular epithelial cells, resulting in decreased hair bulb diameter. Collectively, these observations indicate that TSPEAR plays a critical, previously unrecognized role in human tooth and hair follicle morphogenesis through regulation of the Notch signaling pathway.


Assuntos
Displasia Ectodérmica/genética , Morfogênese/genética , Proteínas/genética , Receptor Notch1/biossíntese , Animais , Diferenciação Celular/genética , Análise Mutacional de DNA , Displasia Ectodérmica/patologia , Mutação da Fase de Leitura/genética , Regulação da Expressão Gênica no Desenvolvimento , Folículo Piloso/crescimento & desenvolvimento , Humanos , Camundongos , Linhagem , Receptor Notch1/genética , Transdução de Sinais/genética , Dente/crescimento & desenvolvimento , Dente/metabolismo
18.
J Cell Sci ; 128(12): 2209-17, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-26021351

RESUMO

Actomyosin contractility is a highly regulated process that affects many fundamental biological processes in each and every cell in our body. In this Cell Science at a Glance article and the accompanying poster, we mined the literature and databases to map the contractome of non-muscle cells. Actomyosin contractility is involved in at least 49 distinct cellular functions that range from providing cell architecture to signal transduction and nuclear activity. Containing over 100 scaffolding and regulatory proteins, the contractome forms a highly complex network with more than 230 direct interactions between its components, 86 of them involving phosphorylation. Mapping these interactions, we identify the key regulatory pathways involved in the assembly of actomyosin structures and in activating myosin to produce contractile forces within non-muscle cells at the exact time and place necessary for cellular function.


Assuntos
Citoesqueleto de Actina/fisiologia , Actomiosina/metabolismo , Movimento Celular/fisiologia , Redes Reguladoras de Genes , Mapas de Interação de Proteínas , Actomiosina/genética , Humanos , Fosforilação , Transdução de Sinais
19.
Nat Cell Biol ; 17(5): 592-604, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25915128

RESUMO

During mouse development, core planar cell polarity (PCP) proteins become polarized in the epidermal plane to guide angling/morphogenesis of hair follicles. How PCP is established is poorly understood. Here, we identify a key role for Wdr1 (also known as Aip1), an F-actin-binding protein that enhances cofilin/destrin-mediated F-actin disassembly. We show that cofilin and destrin function redundantly in developing epidermis, but their combined depletion perturbs cell adhesion, cytokinesis, apicobasal polarity and PCP. Although Wdr1 depletion accentuates single-loss-of-cofilin/destrin phenotypes, alone it resembles core PCP mutations. Seeking a mechanism, we find that Wdr1 and cofilin/destrin-mediated actomyosin remodelling are essential for generating or maintaining cortical tension within the developing epidermal sheet and driving the cell shape and planar orientation changes that accompany establishment of PCP in mammalian epidermis. Our findings suggest intriguing evolutionary parallels but mechanistic modifications to the distal wing hinge-mediated mechanical forces that drive cell shape change and orient PCP in the Drosophila wing disc.


Assuntos
Polaridade Celular , Forma Celular , Epiderme/metabolismo , Queratinócitos/metabolismo , Mecanotransdução Celular , Proteínas dos Microfilamentos/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Actomiosina/metabolismo , Animais , Adesão Celular , Linhagem Celular , Citocinese , Citoesqueleto/metabolismo , Destrina/deficiência , Destrina/genética , Células Epidérmicas , Evolução Molecular , Genótipo , Terapia a Laser , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Microscopia de Vídeo , Fenótipo , Transporte Proteico , Interferência de RNA , Estresse Mecânico , Fatores de Tempo , Transfecção
20.
J Cell Sci ; 125(Pt 7): 1666-72, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22328507

RESUMO

Podosomes, which are formed by different monocyte derivatives, are small adhesion structures whose coordinated dynamics and cytoskeletal reorganization drive their motile and invasive features. Using live-cell microscopy, we explored the temporal molecular steps of the de novo assembly and disassembly of podosomes in cultured osteoclasts. We demonstrate here that the earliest visible step in podosome assembly is the local accumulation of the plaque protein paxillin, along with cortactin, which stabilizes actin networks, followed by robust polymerization of actin filaments and their association with α-actinin. Only then is a local increase in integrin ß3 levels apparent in the podosome ring domain. Thus, local actin polymerization in cortactin- and paxillin-rich locations nucleates podosome assembly before the local accumulation of ß3 integrin. We further show that actin polymerization is also important for the recruitment and maintenance of plaque proteins in the mature podosome ring domain. Our model implies that core bundle dynamics play a central role in regulating podosome stability.


Assuntos
Actinas/metabolismo , Extensões da Superfície Celular/metabolismo , Animais , Células Cultivadas , Cortactina/metabolismo , Camundongos , Miosina Tipo II/metabolismo , Paxilina/metabolismo , Polimerização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...