Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS Genet ; 19(8): e1010873, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37566613

RESUMO

Aberrantly up-regulated activity of the type II transmembrane protease Matriptase-1 has been associated with the development and progression of a range of epithelial-derived carcinomas, and a variety of signaling pathways can mediate Matriptase-dependent tumorigenic events. During mammalian carcinogenesis, gain of Matriptase activity often results from imbalanced ratios between Matriptase and its cognate transmembrane inhibitor Hai1. Similarly, in zebrafish, unrestrained Matriptase activity due to loss of hai1a results in epidermal pre-neoplasms already during embryogenesis. Here, based on our former findings of a similar tumor-suppressive role for the Na+/K+-pump beta subunit ATP1b1a, we identify epithelial polarity defects and systemic hypotonic stress as another mode of aberrant Matriptase activation in the embryonic zebrafish epidermis in vivo. In this case, however, a different oncogenic pathway is activated which contains PI3K, AKT and NFkB, rather than EGFR and PLD (as in hai1a mutants). Strikingly, epidermal pre-neoplasm is only induced when epithelial polarity defects in keratinocytes (leading to disturbed Matriptase subcellular localization) occur in combination with systemic hypotonic stress (leading to increased proteolytic activity of Matriptase). A similar combinatorial effect of hypotonicity and loss of epithelial polarity was also obtained for the activity levels of Matriptase-1 in human MCF-10A epithelial breast cells. Together, this is in line with the multi-factor concept of carcinogenesis, with the notion that such factors can even branch off from one and the same initiator (here ATP1a1b) and can converge again at the level of one and the same mediator (here Matriptase). In sum, our data point to tonicity and epithelial cell polarity as evolutionarily conserved regulators of Matriptase activity that upon de-regulation can constitute an alternative mode of Matriptase-dependent carcinogenesis in vivo.


Assuntos
Epiderme , Peixe-Zebra , Animais , Humanos , Peixe-Zebra/genética , Pressão Osmótica , Carcinogênese , Proteínas Secretadas Inibidoras de Proteinases/genética , Mamíferos
2.
J Physiol ; 600(23): 5027-5054, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36226443

RESUMO

Cytotoxic T lymphocytes (CTL) and natural killer (NK) cells recognize and eliminate cancer cells. However, immune evasion, downregulation of immune function by the tumour microenvironment and resistance of cancer cells are major problems. Although CTL and NK cells are both important to eliminate cancer, most studies address them individually. We quantified sequential primary human CTL and NK cell cytotoxicity against the melanoma cell line SK-Mel-5. At high effector-to-target ratios, NK cells or melan-A (MART-1)-specific CTL eliminated all SK-Mel-5 cells within 24 h, indicating that SK-Mel-5 cells are not resistant initially. However, at lower effector-to-target ratios, which resemble numbers of the immune contexture in human cancer, a substantial number of SK-Mel-5 cells survived. Pre-exposure to CTL induced resistance in surviving SK-Mel-5 cells to subsequent CTL or NK cell cytotoxicity, and pre-exposure to NK cells induced resistance in surviving SK-Mel-5 cells to NK cells. Higher human leucocyte antigen class I expression or interleukin-6 levels were correlated with resistance to NK cells, whereas reduction in MART-1 antigen expression was correlated with reduced CTL cytotoxicity. The CTL cytotoxicity was rescued beyond control levels by exogenous MART-1 antigen. In contrast to the other three combinations, CTL cytotoxicity against SK-Mel-5 cells was enhanced following NK cell pre-exposure. Our assay allows quantification of sequential CTL and NK cell cytotoxicity and might guide strategies for efficient CTL-NK cell anti-melanoma therapies. KEY POINTS: Cytotoxic T lymphocytes (CTL) and natural killer (NK) cells eliminate cancer cells. Both CTL and NK cells attack the same targets, but most studies address them individually. In a sequential cytotoxicity model, the interdependence of antigen-specific CTL and NK cell cytotoxicity against melanoma is quantified. High numbers of antigen-specific CTL and NK cells eliminate all melanoma cells. However, lower numbers induce resistance if secondary CTL or NK cell exposure follows initial CTL exposure or if secondary NK cell exposure follows initial NK cell exposure. On the contrary, if secondary CTL exposure follows initial NK cell exposure, cytotoxicity is enhanced. Alterations in human leucocyte antigen class I expression and interleukin-6 levels are correlated with resistance to NK cells, whereas a reduction in antigen expression is correlated with reduced CTL cytotoxicity; CTL cytotoxicity is rescued beyond control levels by exogenous antigen. This assay and the results on interdependencies will help us to understand and optimize immune therapies against cancer.


Assuntos
Melanoma , Linfócitos T Citotóxicos , Humanos , Antígeno MART-1 , Interleucina-6 , Células Matadoras Naturais , Microambiente Tumoral
3.
Bio Protoc ; 12(8): e4383, 2022 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-35800101

RESUMO

CD4+ T cells are essential players in orchestrating the specific immune response against intracellular pathogens, and in inhibiting tumor development in an early stage. The activation of T cells is triggered by engagement of T cell receptors (TCRs). Here, CD3 and CD28 molecules are key factors, (co)stimulating signaling pathways essential for activation and proliferation of CD4+ T cells. T cell activation induces the formation of a tight mechanical bond between T cell and target cell, the so-called immunological synapse (IS). Due to this, mechanical cell properties, including stiffness, play a significant role in modulating cell functions. In the past, many approaches were made to investigate mechanical properties of immune cells, including micropipette aspiration, microplate-based rheometry, techniques based on deformation during cytometry, or the use of optical tweezers. However, the stiffness of T lymphocytes at a subcellular level at the IS still remains largely elusive. With this protocol, we introduce a method based on atomic force microscopy (AFM), to investigate the local cellular stiffness of T cells on functionalized glass/Polydimethylsiloxan (PDMS) surfaces, which mimicks focal stimulation of target cells inducing IS formation by T cells. By applying the peak force nanomechanical mapping (QNM) technique, cellular surface structures and the local stiffness are determined simultaneously, with a resolution of approximately 60 nm. This protocol can be easily adapted to investigate the mechanical impact of numerous factors influencing IS formation and T cell activation. Graphical abstract: Overview of the experimental workflow. Individual experimental steps are shown on the left, hands on and incubation times for each step are shown right.

4.
EMBO J ; 40(12): e106787, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33998017

RESUMO

Eukaryotic development relies on dynamic cell shape changes and segregation of fate determinants to achieve coordinated compartmentalization at larger scale. Studies in invertebrates have identified polarity programmes essential for morphogenesis; however, less is known about their contribution to adult tissue maintenance. While polarity-dependent fate decisions in mammals utilize molecular machineries similar to invertebrates, the hierarchies and effectors can differ widely. Recent studies in epithelial systems disclosed an intriguing interplay of polarity proteins, adhesion molecules and mechanochemical pathways in tissue organization. Based on major advances in biophysics, genome editing, high-resolution imaging and mathematical modelling, the cell polarity field has evolved to a remarkably multidisciplinary ground. Here, we review emerging concepts how polarity and cell fate are coupled, with emphasis on tissue-scale mechanisms, mechanobiology and mammalian models. Recent findings on the role of polarity signalling for tissue mechanics, micro-environmental functions and fate choices in health and disease will be summarized.


Assuntos
Polaridade Celular , Animais , Fenômenos Biomecânicos , Homeostase , Humanos , Neoplasias , Regeneração
5.
J Invest Dermatol ; 141(4): 713-716, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33752808

RESUMO

The epidermal integrin α3ß1 promotes skin tumorigenesis in experimental models; yet, the underlying molecular mechanisms remain mostly unclear. In their article, Ramovs et al. (2020a) identify two spatially separated α3ß1-dependent signaling branches fostering skin tumor outgrowth. In basal keratinocytes, α3ß1/laminin (LN)-332 drives FAK/Src activation, whereas in suprabasal layers, junctional α3ß1 and the tetraspanin CD151 mediate signal transducer and protein kinase B (Akt)‒dependent survival that is independent of LN-332 binding.


Assuntos
Integrina alfa3 , Laminina , Adesão Celular , Integrina alfa3beta1 , Tetraspanina 24
6.
Curr Opin Cell Biol ; 67: 99-108, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33099084

RESUMO

Melanocytes are neuroectoderm-derived pigment-producing cells with highly polarized dendritic morphology. They protect the skin against ultraviolet radiation by providing melanin to neighbouring keratinocytes. However, the mechanisms underlying melanocyte polarization and its relevance for diseases remain mostly elusive. Numerous studies have instead revealed roles for polarity regulators in other neuroectoderm-derived lineages including different neuronal cell types. Considering the shared ontogeny and morphological similarities, these lineages may be used as reference models for the exploration of melanocyte polarity, for example, regarding dendrite formation, spine morphogenesis and polarized organelle transport. In this review, we summarize and compare the latest progress in understanding polarity regulation in neuronal cells and melanocytes and project key open questions for future work.


Assuntos
Diferenciação Celular , Linhagem da Célula , Polaridade Celular , Melanócitos/citologia , Placa Neural/citologia , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Melanócitos/metabolismo , Melanossomas/metabolismo
7.
Nat Commun ; 10(1): 3362, 2019 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-31358743

RESUMO

Epithelial homeostasis requires balanced progenitor cell proliferation and differentiation, whereas disrupting this equilibrium fosters degeneration or cancer. Here we studied how cell polarity signaling orchestrates epidermal self-renewal and differentiation. Using genetic ablation, quantitative imaging, mechanochemical reconstitution and atomic force microscopy, we find that mammalian Par3 couples genome integrity and epidermal fate through shaping keratinocyte mechanics, rather than mitotic spindle orientation. Par3 inactivation impairs RhoA activity, actomyosin contractility and viscoelasticity, eliciting mitotic failures that trigger aneuploidy, mitosis-dependent DNA damage responses, p53 stabilization and premature differentiation. Importantly, reconstituting myosin activity is sufficient to restore mitotic fidelity, genome integrity, and balanced differentiation and stratification. Collectively, this study deciphers a mechanical signaling network in which Par3 acts upstream of Rho/actomyosin contractility to promote intrinsic force generation, thereby maintaining mitotic accuracy and cellular fitness at the genomic level. Disturbing this network may compromise not only epidermal homeostasis but potentially also that of other self-renewing epithelia.


Assuntos
Polaridade Celular/fisiologia , Epiderme/metabolismo , Genômica/métodos , Homeostase , Queratinócitos/metabolismo , Transdução de Sinais/fisiologia , Actomiosina/genética , Actomiosina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Animais Recém-Nascidos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Polaridade Celular/genética , Células Cultivadas , Queratinócitos/citologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitose/genética , Transdução de Sinais/genética
8.
Oncogene ; 37(37): 5136-5146, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29789715

RESUMO

The polarity proteins Par3 and aPKC are key regulators of processes altered in cancer. Par3/aPKC are thought to dynamically interact with Par6 but increasing evidence suggests that aPKC and Par3 also exert complex-independent functions. Whereas aPKCλ serves as tumor promotor, Par3 can either promote or suppress tumorigenesis. Here we asked whether and how Par3 and aPKCλ genetically interact to control two-stage skin carcinogenesis. Epidermal loss of Par3, aPKCλ, or both, strongly reduced tumor multiplicity and increased latency but inhibited invasion to similar extents, indicating that Par3 and aPKCλ function as a complex to promote tumorigenesis. Molecularly, Par3/aPKCλ cooperate to promote Akt, ERK and NF-κB signaling during tumor initiation to sustain growth, whereas aPKCλ dominates in promoting survival. In the inflammatory tumorigenesis phase Par3/aPKCλ cooperate to drive Stat3 activation and hyperproliferation. Unexpectedly, the reduced inflammatory signaling did not alter carcinogen-induced immune cell numbers but reduced IL-4 Receptor-positive stromal macrophage numbers in all mutant mice, suggesting that epidermal aPKCλ and Par3 promote a tumor-permissive environment. Importantly, aPKCλ also serves a distinct, carcinogen-independent role in controlling skin immune cell homeostasis. Collectively, our data demonstrates that Par3 and aPKCλ cooperate to promote skin tumor initiation and progression, likely through sustaining growth, survival, and inflammatory signaling.


Assuntos
Carcinogênese/genética , Moléculas de Adesão Celular/genética , Proteína Quinase C/genética , Neoplasias Cutâneas/genética , Pele/patologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Carcinogênese/patologia , Proteínas de Ciclo Celular , Polaridade Celular/genética , Proliferação de Células/genética , Modelos Animais de Doenças , Inflamação/patologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , NF-kappa B/genética , Receptores de Interleucina-4/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/genética , Neoplasias Cutâneas/patologia
9.
J Exp Med ; 214(2): 339-358, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28096290

RESUMO

Melanoma, an aggressive skin malignancy with increasing lifetime risk, originates from melanocytes (MCs) that are in close contact with surrounding epidermal keratinocytes (KCs). How the epidermal microenvironment controls melanomagenesis remains poorly understood. In this study, we identify an unexpected non-cell autonomous role of epidermal polarity proteins, molecular determinants of cytoarchitecture, in malignant melanoma. Epidermal Par3 inactivation in mice promotes MC dedifferentiation, motility, and hyperplasia and, in an autochthonous melanoma model, results in increased tumor formation and lung metastasis. KC-specific Par3 loss up-regulates surface P-cadherin that is essential to promote MC proliferation and phenotypic switch toward dedifferentiation. In agreement, low epidermal PAR3 and high P-cadherin expression correlate with human melanoma progression, whereas elevated P-cadherin levels are associated with reduced survival of melanoma patients, implying that this mechanism also drives human disease. Collectively, our data show that reduced KC Par3 function fosters a permissive P-cadherin-dependent niche for MC transformation, invasion, and metastasis. This reveals a previously unrecognized extrinsic tumor-suppressive mechanism, whereby epithelial polarity proteins dictate the cytoarchitecture and fate of other tissue-resident cells to suppress their malignant outgrowth.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Epiderme/química , Melanoma/prevenção & controle , Proteínas de Membrana/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Caderinas/análise , Comunicação Celular , Movimento Celular , Proliferação de Células , Humanos , Melanócitos/patologia , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica
10.
Invest Ophthalmol Vis Sci ; 56(10): 5965-73, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26377082

RESUMO

PURPOSE: Conjunctival melanoma (CM) is an ocular surface tumor that can lead to fatal metastases. Patients developing, tumor-associated lymphangiogenesis have a significantly increased risk of metastatic disease, because tumor spread primarily occurs via lymphatic vessels to the draining lymph node. Here, we describe a novel immune-competent mouse model of CM that displays tumor-associated lymphangiogenesis with development of metastatic tumors. METHODS: C57BL/6N mice received C57BL/6N-derived dermal melanoma cells (hepatocyte growth factor [HGF] cyclin dependent kinase-4 [Cdk4]+) or B16F10 via subconjunctival injection. A clinical score quantified primary tumor growth and metastases were identified by macroscopic examination of the draining lymph nodes, lung, and spleen. Confirmation of tumors and metastases was achieved by immunohistochemical staining for markers of pigmented cells (tyrosinase related protein-2 [TRP2]) and S-100, and of cell proliferation (Ki67). The intra- and peritumoral CD31+ blood and lymphatic vessel endothelium hyaluronan receptor-1 (LYVE-1)+ lymphatic vessels were quantified immunohistochemically. RESULTS: All mice rapidly developed aggressive TRP2+, S100+, and Ki67+ CM. Metastatic tumors were found in the lymph node (9%) and lung (6%) of HGF-Cdk4(R24C)-treated mice and in the spleen (8%) and lung (17%) of B16F10-treated mice. The amount of peri- and intratumoral blood vessels was significantly increased compared with lymphatic vessels. CONCLUSIONS: This CM model in immune-competent animals offers new possibilities to study the pathobiology of tumor growth, invasion, and mechanisms of metastatic tumor spread, and provides a robust model to explore new immune-based and antilymphangiogenic treatment modalities of this malignancy.


Assuntos
Neoplasias da Túnica Conjuntiva/patologia , Modelos Animais de Doenças , Imunocompetência , Linfangiogênese , Metástase Linfática/patologia , Melanoma/patologia , Animais , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Neoplasias da Túnica Conjuntiva/metabolismo , Feminino , Imuno-Histoquímica , Masculino , Melanoma/metabolismo , Melanoma/secundário , Camundongos , Camundongos Endogâmicos C57BL , Prognóstico , Células Tumorais Cultivadas/metabolismo
11.
Invest Ophthalmol Vis Sci ; 56(9): 5691-7, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26313304

RESUMO

PURPOSE: Malignant melanomas of the ocular surface (conjunctival melanoma [CM]) and within the eye (uveal melanoma [UM]) show different types of metastatic behavior. While CM has a propensity to spread first to regional lymph nodes, UM metastasizes almost exclusively via the hematogenic route to the liver. We investigated whether these different metastatic patterns might be attributable to differential hem- and lymphangiogenic characteristics of CM and UM cells. METHODS: Human CM (CM2005.1, CRMM1, CRMM2) and UM (Mel270, Mel290, OM431) cell lines were analyzed for VEGF-A, -C, and -D expression by RT-PCR and ELISA. The influence of CM- or UM-conditioned medium on blood (BEC) and lymphatic (LEC) endothelial cell proliferation and migration was measured using 3-[4,5-dimethylthiazol-2yl]-2,5-diphenyl-tetrazolium bromide (MTT) and scratch assays, respectively. RESULTS: Vascular endothelial growth factor-A, -C and -D mRNA, and VEGF-A and -D protein were expressed by all CM and UM cell lines, while VEGF-C protein was only expressed by UM cell lines. The CM- and UM-conditioned medium did neither differentially affect BEC (P = 0.86) and LEC (P = 0.90) proliferation, nor BEC (P = 0.56) and LEC (P = 0.90) migration. CONCLUSIONS: Conjunctival melanoma cell lines did not show a higher prolymphangiogenic potential, and UM cell lines did not show a higher prohemangiogenic potential. Accordingly, other mechanisms within the tumor microenvironment might account for the diverging metastatic patterns of conjunctival versus uveal melanomas.


Assuntos
Neoplasias da Túnica Conjuntiva/genética , Regulação Neoplásica da Expressão Gênica , Vasos Linfáticos/patologia , Melanoma/genética , RNA Mensageiro/genética , Neoplasias Uveais/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias da Túnica Conjuntiva/metabolismo , Neoplasias da Túnica Conjuntiva/patologia , Humanos , Vasos Linfáticos/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Neoplasias Uveais/metabolismo , Neoplasias Uveais/patologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator C de Crescimento do Endotélio Vascular/biossíntese
12.
Nat Commun ; 6: 8128, 2015 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-26306570

RESUMO

Planar spindle orientation in polarized epithelial cells depends on the precise localization of the dynein-dynactin motor protein complex at the lateral cortex. The contribution of cell adhesion molecules to the cortical localization of the dynein-dynactin complex is poorly understood. Here we find that junctional adhesion molecule-A (JAM-A) regulates the planar orientation of the mitotic spindle during epithelial morphogenesis. During mitosis, JAM-A triggers a transient activation of Cdc42 and PI(3)K, generates a gradient of PtdIns(3,4,5)P3 at the cortex and regulates the formation of the cortical actin cytoskeleton. In the absence of functional JAM-A, dynactin localization at the cortex is reduced, the mitotic spindle apparatus is misaligned and epithelial morphogenesis in three-dimensional culture is compromised. Our findings indicate that a PI(3)K- and cortical F-actin-dependent pathway of planar spindle orientation operates in polarized epithelial cells to regulate epithelial morphogenesis, and we identify JAM-A as a junctional regulator of this pathway.


Assuntos
Citoesqueleto de Actina/metabolismo , Dineínas/metabolismo , Molécula A de Adesão Juncional/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fuso Acromático/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Polaridade Celular , Cães , Complexo Dinactina , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Humanos , Molécula A de Adesão Juncional/metabolismo , Células Madin Darby de Rim Canino , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose/genética
13.
Biomed Res Int ; 2014: 639058, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25254213

RESUMO

Lymphangiogenesis is a very early step in lymphatic metastasis. It is regulated and promoted not only by the tumor cells themselves, but also by cells of the tumor microenvironment, including cancer associated fibroblasts, mesenchymal stem cells, dendritic cells, or macrophages. Even the extracellular matrix as well as cytokines and growth factors are involved in the process of lymphangiogenesis and metastasis. The cellular and noncellular components influence each other and can be influenced by the tumor cells. The knowledge about mechanisms behind lymphangiogenesis in the tumor microenvironmental crosstalk is growing and offers starting points for new therapeutic approaches.


Assuntos
Linfangiogênese/genética , Metástase Linfática/genética , Neoplasias/metabolismo , Microambiente Tumoral/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Humanos , Metástase Linfática/patologia , Vasos Linfáticos/patologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Transdução de Sinais
14.
J Cell Sci ; 125(Pt 15): 3501-10, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22935653

RESUMO

The establishment and maintenance of cell and tissue polarity is crucial for a range of biological processes, such as oriented division, migration, adhesion and barrier function. The molecular pathways that regulate cell and tissue polarity have been extensively studied in lower organisms as well as in mammalian cell culture. By contrast, relatively little is still known about how polarization regulates the in vivo formation and homeostasis of mammalian tissues. Several recent papers have identified crucial roles for mammalian polarity proteins in a range of in vivo processes, including stem cell behavior, cell fate determination, junction formation and maintenance and organ development. Using the epidermis of the skin as a model system, this Commentary aims to discuss the in vivo significance of cell and tissue polarity in the regulation of mammalian tissue morphogenesis, homeostasis and disease. Specifically, we discuss the mechanisms by which the molecular players previously identified to determine polarity in vitro and/or in lower organisms regulate epidermal stratification; orient cell division to drive cell fate determination within the epidermal lineage; and orient hair follicles. We also describe how altered polarity signaling contributes to skin cancer.


Assuntos
Polaridade Celular/fisiologia , Citoesqueleto/fisiologia , Epiderme/fisiologia , Animais , Diferenciação Celular/fisiologia , Células Epidérmicas , Humanos , Transdução de Sinais
15.
Cancer Cell ; 22(3): 389-403, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22975380

RESUMO

Cell polarization is crucial during development and tissue homeostasis and is regulated by conserved proteins of the Scribble, Crumbs, and Par complexes. In mouse skin tumorigenesis, Par3 deficiency results in reduced papilloma formation and growth. Par3 mediates its tumor-promoting activity through regulation of growth and survival, since Par3 deletion increases apoptosis and reduces growth in vivo and in vitro. In contrast, Par3-deficient mice are predisposed to formation of keratoacanthomas, cutaneous tumors thought to originate from different cellular origin and frequently observed in humans. Par3 expression is reduced in both mouse and human keratoacanthomas, indicating tumor-suppressive properties of Par3. Our results identify a dual function of Par3 in skin cancer, with both pro-oncogenic and tumor-suppressive activity depending on the tumor type.


Assuntos
Moléculas de Adesão Celular/metabolismo , Transformação Celular Neoplásica , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/genética , Proteínas de Ciclo Celular , Polaridade Celular , Proliferação de Células , Células Cultivadas , Queratinócitos/metabolismo , Ceratoacantoma/genética , Ceratoacantoma/metabolismo , Ceratoacantoma/patologia , Camundongos , Camundongos Transgênicos , Proteína Quinase C/metabolismo , Pele/metabolismo , Neoplasias Cutâneas/genética
16.
Small GTPases ; 3(1): 4-14, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22710731

RESUMO

Polarized cell migration is a crucial process in the development and repair of tissues, as well as in pathological conditions, including cancer. Recent studies have elucidated important roles for Rho GTPases in the establishment and maintenance of polarity prior to and during cell migration. Here, we show that Tiam1, a specific activator of the small GTPase Rac, is required for the polarized outgrowth of protrusions in primary astrocytes during the initial phase of cell polarization after scratch-wounding monolayers of cells. Tiam1 deficiency delays closure of wounds in confluent monolayers. Lack of Tiam1 impairs adoption of an asymmetrical cell shape as well as microtubule organization within protrusions. Positioning of the centrosome and Golgi apparatus, however, are independent of Tiam1-Rac signaling. We speculate that the function of Tiam1 in polarized outgrowth of astrocyte protrusions involves regulation of microtubule organization, possibly by stabilizing the microtubule cytoskeleton. Our results add Tiam1 as a player to the growing list of proteins involved in polarized outgrowth of protrusions and further elucidate the signaling pathways leading to cell polarization.


Assuntos
Astrócitos/citologia , Astrócitos/metabolismo , Polaridade Celular , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Microtúbulos/metabolismo , Animais , Células Cultivadas , Centrossomo/metabolismo , Centrossomo/ultraestrutura , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnicas de Inativação de Genes , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Fatores de Troca do Nucleotídeo Guanina/genética , Camundongos , Camundongos Knockout , Microtúbulos/ultraestrutura , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T
17.
Semin Cancer Biol ; 22(3): 208-15, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22465739

RESUMO

Cell polarity is essential in many biological processes and required for development as well as maintenance of tissue integrity. Loss of polarity is considered both a hallmark and precondition for human cancer. Three conserved polarity protein complexes regulate different modes of polarity that are conserved throughout numerous cell types and species. These complexes are the Crumbs, Par and Scribble complex. Given the importance of cell polarity for normal tissue homeostasis, aberrant polarity signaling is suggested to contribute to the multistep processes of human cancer. Most human cancers are formed from epithelial cells. Evidence confirming the roles for polarity proteins in different phases of the oncogenic trajectory comes from functional studies using mammalian cells as well as Drosophila and zebrafish models. Furthermore, several reports have revealed aberrant expression and localization of polarity proteins in different human tumors. In this review we will give an overview on the current data available that couple polarity signaling to tumorigenesis, particularly in epithelial cells.


Assuntos
Polaridade Celular/fisiologia , Transformação Celular Neoplásica/patologia , Células Epiteliais/fisiologia , Proteínas de Membrana/fisiologia , Metástase Neoplásica/fisiopatologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Transformação Celular Neoplásica/metabolismo , Expressão Gênica , Humanos , Transdução de Sinais/fisiologia
18.
J Cell Biol ; 196(5): 623-39, 2012 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-22371556

RESUMO

The PAR-3-atypical protein kinase C (aPKC)-PAR-6 complex has been implicated in the development of apicobasal polarity and the formation of tight junctions (TJs) in vertebrate epithelial cells. It is recruited by junctional adhesion molecule A (JAM-A) to primordial junctions where aPKC is activated by Rho family small guanosine triphosphatases. In this paper, we show that aPKC can interact directly with JAM-A in a PAR-3-independent manner. Upon recruitment to primordial junctions, aPKC phosphorylates JAM-A at S285 to promote the maturation of immature cell-cell contacts. In fully polarized cells, S285-phosphorylated JAM-A is localized exclusively at the TJs, and S285 phosphorylation of JAM-A is required for the development of a functional epithelial barrier. Protein phosphatase 2A dephosphorylates JAM-A at S285, suggesting that it antagonizes the activity of aPKC. Expression of nonphosphorylatable JAM-A/S285A interferes with single lumen specification during cyst development in three-dimensional culture. Our data suggest that aPKC phosphorylates JAM-A at S285 to regulate cell-cell contact maturation, TJ formation, and single lumen specification.


Assuntos
Moléculas de Adesão Celular/metabolismo , Proteína Quinase C/metabolismo , Receptores de Superfície Celular/metabolismo , Serina/metabolismo , Junções Íntimas/fisiologia , Sequência de Aminoácidos , Animais , Moléculas de Adesão Celular/genética , Linhagem Celular , Polaridade Celular/fisiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Camundongos , Mitose/fisiologia , Dados de Sequência Molecular , Morfogênese/fisiologia , Fosforilação , Proteína Quinase C/genética , Proteína Fosfatase 2/metabolismo , Interferência de RNA , Receptores de Superfície Celular/genética
19.
Nat Rev Mol Cell Biol ; 9(11): 846-59, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18946474

RESUMO

Cell polarization is crucial for the development of multicellular organisms, and aberrant cell polarization contributes to various diseases, including cancer. How cell polarity is established and how it is maintained remain fascinating questions. Conserved proteins of the partitioning defective (PAR), Scribble and Crumbs complexes guide the establishment of cell polarity in various organisms. Moreover, GTPases that regulate actin cytoskeletal dynamics have been implicated in cell polarization. Recent findings provide insights into polarization mechanisms and show intriguing crosstalk between small GTPases and members of polarity complexes in regulating cell polarization in different cellular contexts and cell types.


Assuntos
Polaridade Celular , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteínas/metabolismo , Animais , Movimento Celular , Citoesqueleto/química , Humanos
20.
Front Biosci ; 13: 6520-36, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18508678

RESUMO

The organization of tissues depends on intercellular junctions that connect individual cells to each other. In sheets of epithelial cells the junctions contain different components like adherens junctions or tight junctions in an asymmetric distribution along the cell-cell contacts. Tight junctions are located at the most apical region of cell junctions, act as a regulatable barrier for small solutes, and separate the apical membrane domain from the basolateral membrane domain. For a long time, the mechanisms that underly the formation of tight junctions and the development of apico-basal membrane polarity in epithelial cells have been poorly understood. Recently, strong evidence has been provided which implicates a conserved set of cell polarity proteins--the PAR proteins--in this process. Here we discuss the mechanisms by which PAR proteins regulate the formation of cell junctions with a special emphasis on vertebrate epithelial cells.


Assuntos
Polaridade Celular/fisiologia , Células Endoteliais/fisiologia , Células Epiteliais/fisiologia , Proteínas de Membrana/fisiologia , Proteínas de Neoplasias/fisiologia , Animais , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/fisiologia , Humanos , Junções Intercelulares/fisiologia , Masculino , Mamíferos , Glicoproteínas de Membrana/fisiologia , Núcleosídeo-Fosfato Quinase/fisiologia , Proteína Quinase C/metabolismo , Células de Sertoli/citologia , Células de Sertoli/fisiologia , Espermátides/citologia , Espermátides/fisiologia , Proteínas de Junções Íntimas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA