Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Res ; 73(3): 1168-79, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23243018

RESUMO

The cell surface glycoprotein Trask/CDCP1 is phosphorylated during anchorage loss in epithelial cells in which it inhibits integrin clustering, outside-in signaling, and cell adhesion. Its role in cancer has been difficult to understand, because of the lack of a discernible pattern in its various alterations in cancer cells. To address this issue, we generated mice lacking Trask function. Mammary tumors driven by the PyMT oncogene and skin tumors driven by the SmoM2 oncogene arose with accelerated kinetics in Trask-deficient mice, establishing a tumor suppressing function for this gene. Mechanistic investigations in mammary tumor cell lines derived from wild-type or Trask-deficient mice revealed a derepression of integrin signaling and an enhancement of integrin-growth factor receptor cross-talk, specifically in unanchored cell states. A similar restrictive link between anchorage and growth in untransformed epithelial cells was observed and disrupted by elimination of Trask. Together our results establish a tumor-suppressing function in Trask that restricts epithelial cell growth to the anchored state.


Assuntos
Antígenos de Neoplasias/fisiologia , Integrinas/fisiologia , Neoplasias Mamárias Experimentais/patologia , Glicoproteínas de Membrana/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Animais , Moléculas de Adesão Celular , Feminino , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Tamoxifeno/farmacologia
2.
Spermatogenesis ; 1(1): 73-86, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21866278

RESUMO

During spermatogenesis, step 1 spermatids (round spermatids) derive from spermatocytes following meiosis I and II at stage XIV of the epithelial cycle begin a series of morphological transformation and differentiation via 19 steps in rats to form spermatozoa. This process is known as spermiogenesis, which is marked by condensation of the genetic material in the spermatid head, formation of the acrosome and elongation of the tail. Since developing spermatids are lacking the robust protein synthesis and transcriptional activity, the cellular, molecular and morphological changes associated with spermiogenesis rely on the Sertoli cell in the seminiferous epithelium via desmosome and gap junction between Sertoli cells and step 1-7 spermatids. Interestingly, a unique anchoring junction type arises at the interface of step 8 spermatid and Sertoli cell known as apical ectoplasmic specialization (apical ES). Once it appears, apical ES is the only anchoring device restricted to the interface of step 8-19 spermatids and Sertoli cells to confer spermatid polarity, adhesion, signal communication and structural support, and to provide nutritional support during spermiogenesis, replacing desmosome and gap junction. While the adhesion protein complexes that constitute the apical ES are known, the signaling protein complexes that regulate apical ES dynamics, however, remain largely unknown. Herein we report the presence of a FAK (focal adhesion kinase)-p130Cas (p130 Crk-associated substrate)-DOCK180 (Dedicator of cytokinesis 180)-RhoA (Ras homolog gene family, member A)-vinculin signaling protein complex at the apical ES, which is also an integrated component of the ß1-integrin-based adhesion protein complex based on co-immunoprecipitation experiment. It was also shown that besides p-FAK-Tyr(397) and p-FAK-Tyr(576), ß1-integrin, p130Cas, RhoA and vinculin displayed stage-specific expression in the seminiferous epithelium during the epithelial cycle with predominant localization at the apical ES as demonstrated by immunohistochemistry. Based on these findings, functional studies can now be performed to assess the role of this ß1-integrin-p-FAK-p130Cas-DOCK180-RhoA-vinculin protein complex in apical ES dynamics during spermiogenesis.

3.
PLoS One ; 6(4): e19154, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21559459

RESUMO

Trask/CDCP1 is a transmembrane protein with a large extracellular and small intracellular domains. The intracellular domain (ICD) undergoes tyrosine phosphorylation by Src kinases during anchorage loss and, when phosphorylated, Trask functions to inhibit cell adhesion. The extracellular domain (ECD) undergoes proteolytic cleavage by serine proteases, although the functional significance of this remains unknown. There is conflicting evidence regarding whether it functions to signal the phosphorylation of the ICD. To better define the structural determinants that mediate the anti-adhesive functions of Trask, we generated a series of deletion mutants of Trask and expressed them in tet-inducible cell models to define the structural elements involved in cell adhesion signaling. We find that the ECD is dispensable for the phosphorylation of the ICD or for the inhibition of cell adhesion. The anti-adhesive functions of Trask are entirely embodied within its ICD and are specifically due to tyrosine phosphorylation of the ICD as this function is completely lost in a phosphorylation-defective tyrosine-phenylalanine mutant. Both full length and cleaved ECDs are fully capable of phosphorylation and undergo phosphorylation during anchorage loss and cleavage is not an upstream signal for ICD phosphorylation. These data establish that the anti-adhesive functions of Trask are mediated entirely through its tyrosine phosphorylation. It remains to be defined what role, if any, the Trask ECD plays in its adhesion functions.


Assuntos
Antígenos CD/química , Antígenos CD/fisiologia , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/fisiologia , Proteínas de Neoplasias/química , Proteínas de Neoplasias/fisiologia , Antígenos de Neoplasias , Adesão Celular , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Microscopia de Fluorescência/métodos , Microscopia de Contraste de Fase/métodos , Mutação , Fases de Leitura Aberta , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Transfecção , Tirosina/química
4.
Mol Cell Biol ; 31(4): 766-82, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21189288

RESUMO

Trask is a recently described transmembrane substrate of Src kinases whose expression and phosphorylation has been correlated with the biology of some cancers. Little is known about the molecular functions of Trask, although its phosphorylation has been associated with cell adhesion. We have studied the effects of Trask phosphorylation on cell adhesion, integrin activation, clustering, and focal adhesion signaling. The small hairpin RNA (shRNA) knockdown of Trask results in increased cell adhesiveness and a failure to properly inactivate focal adhesion signaling, even in the unanchored state. On the contrary, the experimentally induced phosphorylation of Trask results in the inhibition of cell adhesion and inhibition of focal adhesion signaling. This is mediated through the inhibition of integrin clustering without affecting integrin affinity state or ligand binding activity. Furthermore, Trask signaling and focal adhesion signaling inactivate each other and signal in exclusion with each other, constituting a switch that underlies cell anchorage state. These data provide considerable insight into how Trask functions to regulate cell adhesion and reveal a novel pathway through which Src kinases can oppose integrin-mediated cell adhesion.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Adesões Focais/metabolismo , Integrinas/metabolismo , Proteínas de Neoplasias/metabolismo , Quinases da Família src/metabolismo , Antígenos CD/genética , Antígenos de Neoplasias , Sequência de Bases , Adesão Celular/fisiologia , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Integrinas/química , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Fosforilação , RNA Interferente Pequeno/genética , Transdução de Sinais/fisiologia , Especificidade por Substrato
5.
Am J Pathol ; 174(5): 1756-65, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19349359

RESUMO

The roles of epithelial cells encompass both cellular- and tissue-level functions that involve numerous cell-cell and cell-matrix interactions, which ultimately mediate the highly structured arrangement of cells on a basement membrane. Although maintaining this basic structure is critical for preserving tissue integrity, plasticity in epithelial cell behavior is also critical for processes such as cell migration during development or wound repair, mitotic cell detachment, and physiological shedding. The mechanisms that mediate epithelial cell plasticity are only beginning to be understood. We previously identified Trask, a transmembrane protein that is phosphorylated by src kinases during mitosis. In this study, we report that the phosphorylation of Trask is associated with anchorage loss in epithelial cells. Phosphorylation of Trask is seen during the cell-detachment phase of mitosis, in experimentally induced interphase detachment, and during cell migration in experimental epithelial models. An analysis of human tissues shows that Trask is widely expressed in many epithelial tissues but not in most tissues of mesenchymal origin, except for a subset of early hematopoietic cells. Trask is not phosphorylated in epithelial tissues in vivo; however, its phosphorylation is seen in epithelial cells undergoing mitosis or physiological shedding. Trask is a novel epithelial membrane protein that is phosphorylated by src kinases when epithelial cells disengage from their tissue framework, identifying an important new regulator of epithelial tissue dynamics.


Assuntos
Antígenos CD/metabolismo , Neoplasias da Mama/metabolismo , Moléculas de Adesão Celular/metabolismo , Adesão Celular/fisiologia , Células Epiteliais/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Antígenos CD/genética , Antígenos de Neoplasias , Mama/citologia , Mama/metabolismo , Neoplasias da Mama/patologia , Moléculas de Adesão Celular/genética , Movimento Celular/fisiologia , Citometria de Fluxo , Imunofluorescência , Humanos , Immunoblotting , Técnicas Imunoenzimáticas , Interfase/fisiologia , Mesoderma/citologia , Mesoderma/metabolismo , Mitose/fisiologia , Proteínas de Neoplasias/genética , Fosforilação , Transdução de Sinais
6.
Clin Cancer Res ; 15(7): 2311-22, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19318475

RESUMO

PURPOSE: The frequently elevated activities of the c-src and c-yes products in human epithelial tumors suggest that these activated tyrosine kinases have tumorigenic functions analogous to the v-src and v-yes oncogene products. Studies of v-src-transformed fibroblasts have identified many of the effectors of this potent oncogene; however, because c-src and c-yes lack the mutational and promiscuous activities of their retroviral oncogene homologues, their presumptive tumorigenic functions in human epithelial tumors are more subtle, less well-defined, and await identification of possible effectors more directly relevant to epithelial cells. EXPERIMENTAL DESIGN: We recently identified a transmembrane glycoprotein named Trask that is expressed in epithelial tissues but not fibroblasts and is phosphorylated by SRC kinases in mitotic epithelial cells. In this study, we have surveyed the expression and phosphorylation of Trask in many human epithelial cancer cell lines and surgical tissues and tumors. RESULTS: Trask is widely expressed in human epithelial tissues, but its phosphorylation is tightly regulated and restricted to detached mitotic cells or cells undergoing physiologic shedding. However, abberant Trask phosphorylation is seen in many epithelial tumors from all stages including preinvasive, invasive, and metastatic tumors. Trask phosphorylation requires SRC kinases, and is also aberrantly hyperphosphorylated in the SRC-activated PyMT mouse epithelial tumors and dephosphorylated by the SRC inhibitor treatment of these tumors. CONCLUSIONS: The widespread phosphorylation of Trask in many human epithlelial cancers identifies a new potential effector of SRC kinases in human epithelial tumorigenesis.


Assuntos
Antígenos CD/metabolismo , Carcinoma/enzimologia , Moléculas de Adesão Celular/metabolismo , Proteínas de Neoplasias/metabolismo , Antígenos de Neoplasias , Neoplasias da Mama/enzimologia , Carcinoma/metabolismo , Carcinoma/patologia , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Colo/enzimologia , Células Epiteliais/enzimologia , Feminino , Humanos , Fosforilação , Quinases da Família src/metabolismo
7.
J Magn Reson Imaging ; 26(6): 1618-25, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17968965

RESUMO

PURPOSE: To determine if inhibitors of the human growth factor receptor (HER) family can be used to enhance tumor vascular permeability and perfusion and optimize the efficacy of cytotoxic chemotherapeutics. Poor tumor vascular function limits the delivery and efficacy of cancer chemotherapeutics and HER family tyrosine kinases mediate tumor-endothelial signaling in both of these compartments. MATERIALS AND METHODS: BT474 human breast cancer tumors were established in mice and the biologic effects of the HER tyrosine kinase inhibitor (TKI) gefitinib on tumor vascular function was determined by dynamic contrast-enhanced MRI (DCE-MRI), and on tumor vascular architecture and perfusion by immunofluorescence microscopy. RESULTS: A brief dose of gefitinib enhances the antitumor activity of paclitaxel in vivo but not in cell culture, suggesting that its chemoenhancing activity involves the in vivo microenvironment. A brief high dose of gefitinib induces a decrease in endothelial transfer constant (Kps) and a concomitant increase in tumor fractional plasma volume (fPV). These changes are accompanied by a rapid reduction in tumor volume, likely due to decreased tumor edema, and modestly improved tumor vascular architecture and perfusion on microscopy. CONCLUSION: These data suggest that HER family TKIs have the potential to optimize the tumor microenvironment for delivery of cytotoxic chemotherapeutics.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Permeabilidade Capilar/efeitos dos fármacos , Imageamento por Ressonância Magnética/métodos , Neovascularização Patológica/patologia , Quinazolinas/farmacologia , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Neoplasias da Mama/irrigação sanguínea , Meios de Contraste , Gadolínio DTPA , Gefitinibe , Camundongos , Camundongos Nus
8.
FASEB J ; 21(2): 438-48, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17167075

RESUMO

The blood-testis barrier (BTB) is one of the tightest blood-tissue barriers in mammals. As such, it poses a challenge to deliver any drugs to the seminiferous epithelium of the testis, such as a nonhormonal male contraceptive. To circumvent this problem, a genetically engineered follicle-stimulating hormone (FSH) mutant protein was produced in Spodoptera furgiperda (Sf)-9 insect cells to serve as a testis-specific carrier. Subsequently, a 22-amino acid peptide corresponding to the second extracellular loop of occludin, which was known to disrupt BTB integrity in vivo, was inserted to the FSH mutant by polymerase chain reaction (PCR), as well as chemical cross-linking. This molecule was found to have negligible hormonal activity but was still capable of binding to FSH receptors, which are restricted to Sertoli cells in mammals. When this FSH mutant-occludin peptide conjugate was administered to adult rats at 40 microg/adult rat (approximately 300 gm b.w.) via intraperitoneally (i.p.) injection, it induced transient and reversible disruption of the BTB, while at 150 microg/rat, it induced partial germ cell loss from the testis, particularly elongating/elongate spermatids. Most importantly, this effect was limited to the BTB without compromising the TJ-barrier integrity or cell adhesion in epithelia of other organs, such as kidney, liver, and small intestine. In summary, the use of an FSH mutant-occludin peptide conjugate is a feasible nanodevice to transiently compromise the BTB.


Assuntos
Barreira Hematotesticular/efeitos dos fármacos , Hormônio Foliculoestimulante/farmacologia , Proteínas de Membrana/farmacologia , Proteínas Mutantes/farmacologia , Animais , Anticorpos/sangue , Barreira Hematotesticular/metabolismo , Adesão Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Hormônio Foliculoestimulante/química , Hormônio Foliculoestimulante/metabolismo , Células Germinativas/citologia , Células Germinativas/efeitos dos fármacos , Células Germinativas/imunologia , Immunoblotting , Injeções Intraperitoneais , Masculino , Proteínas de Membrana/química , Proteínas de Membrana/genética , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Estrutura Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Ocludina , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Receptores do FSH/metabolismo , Espermatogênese , Testículo/citologia , Testículo/metabolismo , Testículo/ultraestrutura
9.
J Endocrinol ; 191(3): 571-86, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17170215

RESUMO

In adult rat testes, blood-testis barrier (BTB) restructuring facilitates the migration of preleptotene spermatocytes from the basal to the adluminal compartment that occurs at stage VIII of the epithelial cycle. Structural proteins at the BTB must utilize an efficient mechanism (e.g. endocytosis) to facilitate its transient 'opening'. Dynamin II, a large GTPase known to be involved in endocytosis, was shown to be a product of Sertoli and germ cells in the testis. It was also localized to the BTB, as well as the apical ectoplasmic specialization (apical ES), during virtually all stages of the epithelial cycle. By co-immunoprecipitation, dynamin II was shown to associate with occludin, N-cadherin, zonula occludens-1 (ZO-1), beta-catenin, junctional adhesion molecule-A, and p130Cas, but not nectin-3. An in vivo model in rats previously characterized for studying adherens junction (AJ) dynamics in the testes by adjudin (formerly called AF-2364, 1-(2,4-dichlorobenzyl)-1H-indazole-3-carbohydrazide) treatment was used in our studies. At the time of germ cell loss from the seminiferous epithelium as a result of adjudin-induced AJ restructuring without disrupting the BTB integrity, a significant decline in the steady-state dynamin II protein level was detected. This change was associated with a concomitant increase in the levels of two protein complexes at the BTB, namely occludin/ZO-1 and N-cadherin/beta-catenin. Interestingly, these changes were also accompanied by a significant increase in the structural interaction of dynamin II with beta-catenin and ZO-1. Beta-catenin and ZO-1 are adaptors that structurally link the cadherin- and occludin-based protein complexes together at the BTB in an 'engaged'state to reinforce the barrier function in normal testes. However, beta-catenin and ZO-1 were 'disengaged' from each other but bound to dynamin II during adjudin-induced AJ restructuring in the testis. The data reported herein suggest that dynamin II may assist the 'disengagement' of beta-catenin from ZO-1 during BTB restructuring. Thus, this may permit the occludin/ZO-1 complexes to maintain the BTB integrity when the cadherin/catenin complexes are dissociated to facilitate germ cell movement.


Assuntos
Barreira Hematotesticular , Caderinas/metabolismo , Dinamina II/metabolismo , Proteínas de Membrana/metabolismo , Testículo/fisiologia , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/metabolismo , Animais , Caderinas/análise , Moléculas de Adesão Celular/análise , Moléculas de Adesão Celular/metabolismo , Movimento Celular/fisiologia , Células Cultivadas , Dinamina II/análise , Hidrazinas/análise , Hidrazinas/metabolismo , Hidrazinas/farmacologia , Imuno-Histoquímica/métodos , Indazóis/análise , Indazóis/metabolismo , Indazóis/farmacologia , Masculino , Proteínas de Membrana/análise , Microscopia de Fluorescência , Nectinas , Ocludina , Fosfoproteínas/análise , Fosfoproteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Espermatozoides/fisiologia , Testículo/metabolismo , Proteína da Zônula de Oclusão-1 , beta Catenina/análise , beta Catenina/metabolismo
10.
Nat Med ; 12(11): 1323-8, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17072312

RESUMO

Throughout spermatogenesis, developing germ cells remain attached to Sertoli cells via testis-specific anchoring junctions. If adhesion between these cell types is compromised, germ cells detach from the seminiferous epithelium and infertility often results. Previously, we reported that Adjudin is capable of inducing germ cell loss from the epithelium. In a small subset of animals, however, oral administration of Adjudin (50 mg per kg body weight (b.w.) for 29 d) resulted in adverse effects such as liver inflammation and muscle atrophy. Here, we report a novel approach in which Adjudin is specifically targeted to the testis by conjugating Adjudin to a recombinant follicle-stimulating hormone (FSH) mutant, which serves as its 'carrier'. Using this approach, infertility was induced in adult rats when 0.5 microg Adjudin per kg b.w. was administered intraperitoneally, which was similar to results when 50 mg per kg b.w. was given orally. This represents a substantial increase in Adjudin's selectivity and efficacy as a male contraceptive.


Assuntos
Adesão Celular , Anticoncepcionais Masculinos , Células Germinativas/citologia , Animais , Hormônio Foliculoestimulante/administração & dosagem , Hormônio Foliculoestimulante/química , Hidrazinas/administração & dosagem , Hidrazinas/química , Hidrazinas/farmacocinética , Hidrazinas/farmacologia , Indazóis/administração & dosagem , Indazóis/química , Indazóis/farmacocinética , Indazóis/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Testículo/efeitos dos fármacos
11.
Curr Top Dev Biol ; 71: 263-96, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16344108

RESUMO

The blood-testis barrier (BTB) in mammals, such as rats, is composed of the tight junction (TJ), the basal ectoplasmic specialization (basal ES), the basal tubulobulbar complex (basal TBC) (both are testis-specific actin-based adherens junction [AJ] types), and the desmosome-like junction that are present side-by-side in the seminiferous epithelium. The BTB physically divides the seminiferous epithelium into basal and apical (or adluminal) compartments, and is pivotal to spermatogenesis. Besides its function as an immunological barrier to segregate the postmeiotic germ-cell antigens from the systemic circulation, it creates a unique microenvironment for germ-cell development and confers cell polarity. During spermatogenesis, the BTB in rodents must physically disassemble to permit the passage of preleptotene and leptotene spermatocytes. This occurs at late stage VII through early stage VIII of the epithelial cycle. Studies have shown that this dynamic BTB restructuring to facilitate germ-cell migration is regulated by two cytokines, namely transforming growth factor-beta3 (TGF-beta3) and tumor necrosis factor-alpha (TNFalpha), via downstream mitogen-activated protein kinases. These cytokines determine the homeostasis of TJ- and basal ES-structural proteins, proteases, protease inhibitors, and other extracellular matrix (ECM) proteins (e.g., collagen) in the seminiferous epithelium. Some of these molecules are known regulators of focal contacts between the ECM and other actively migrating cells, such as macrophages, fibroblasts, or malignant cells. These findings also illustrate that cell-cell junction restructuring at the BTB is regulated by mechanisms involved in the junction turnover at the cell-matrix interface. This review critically discusses these latest findings in the field in light of their significance in the biology and regulation of the BTB pertinent to spermatogenesis.


Assuntos
Barreira Hematotesticular/fisiologia , Espermatogênese/fisiologia , Animais , Barreira Hematotesticular/ultraestrutura , Masculino , Modelos Biológicos
12.
Contraception ; 72(4): 251-61, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16181968

RESUMO

Earlier studies have shown that 1-(2,4-dichlorobenzyl)-1H-indazole-3-carbohydrazide (AF-2364) is a potential male contraceptive when administered orally to adult Sprague-Dawley rats. This compound induces reversible germ cell loss from the seminiferous epithelium by disrupting cell adhesion function between Sertoli and germ cells, in particular, elongating/elongate/round spermatids and spermatocytes but not spermatogonia. Thus, this event is accompanied by a transient loss of fertility in treated rats. Once the drug is metabolically cleared, the remaining spermatogonia can begin repopulating the epithelium, and fertility bounces back. In this review, we summarize recent findings regarding the possible use of this drug for male contraception and its mechanism of action in the rat testis. We also provide an update on the efficacy results of using different treatment regimens in adult rats where AF-2364 was administered by gavage vs. intraperitoneal and intramuscular administration. These results have clearly indicated that AF-2364 is indeed a reversible male contraceptive. Furthermore, the tissue distribution in multiple organs and biological fluids using [3H]-AF-2364 is also reviewed. These data have clearly illustrated the low bioavailability of AF-2364 in rats and that this compound is not specifically taken up by any organs including the testis or the epididymis. These summaries are helpful to investigators in the field who seek to understand the molecular mechanism of action of AF-2364 in the rat testis and to explore its possible use for male contraception.


Assuntos
Anticoncepcionais Masculinos , Hidrazinas , Indazóis , Animais , Disponibilidade Biológica , Adesão Celular/efeitos dos fármacos , Fertilidade/efeitos dos fármacos , Hidrazinas/administração & dosagem , Hidrazinas/farmacocinética , Hidrazinas/farmacologia , Indazóis/administração & dosagem , Indazóis/farmacocinética , Indazóis/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/fisiologia , Espermatozoides/efeitos dos fármacos , Espermatozoides/fisiologia , Distribuição Tecidual
13.
Dev Biol ; 286(1): 1-15, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16153630

RESUMO

Mitogen-activated protein kinases (MAPKs) are important regulators of many cellular processes. In mammalian testes, these kinases are involved in controlling cell division, differentiation, survival and death, and are therefore critical to spermatogenesis. Recent studies have also illustrated their involvement in junction restructuring in the seminiferous epithelium, especially at the ectoplasmic specialization (ES), a testis-specific adherens junction (AJ) type. ES contributes to the adhesion between Sertoli cells at the blood-testis barrier, as well as between Sertoli and developing spermatids (step 9 and beyond) at the adluminal compartment. MAPKs regulate AJ dynamics in the testis via their effects on the turnover of junction-associated protein complexes, the production of proteases and protease inhibitors, and the cytoskeleton structure. In this review, roles of the three major MAPK members, namely extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 MAPK, in ES dynamics are critically discussed. An integrated model of how these three MAPKs regulate adhesion function in the seminiferous epithelium is also presented. This model will serve as the framework for future investigation in the field.


Assuntos
Junções Aderentes/enzimologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Espermatogênese/fisiologia , Animais , Barreira Hematotesticular , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Modelos Biológicos , Testículo/enzimologia
14.
J Biol Chem ; 280(26): 25029-47, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15870075

RESUMO

When Sertoli and germ cells were co-cultured in vitro in serum-free chemically defined medium, functional anchoring junctions such as cell-cell intermediate filament-based desmosome-like junctions and cell-cell actin-based adherens junctions (e.g. ectoplasmic specialization (ES)) were formed within 1-2 days. This event was marked by the induction of several protein kinases such as phosphatidylinositol 3-kinase (PI3K), phosphorylated protein kinase B (PKB; also known as Akt), p21-activated kinase-2 (PAK-2), and their downstream effector (ERK) as well as an increase in PKB intrinsic activity. PI3K, phospho (p)-PKB, and PAK were co-localized to the site of apical ES in the seminiferous epithelium of the rat testis in immunohistochemistry studies. Furthermore, PI3K also co-localized with p-PKB to the same site in the epithelium as determined by fluorescence microscopy, consistent with their localization at the ES. These kinases were shown to associate with ES-associated proteins such as beta1-integrin, phosphorylated focal adhesion kinase, and c-Src by co-immunoprecipitation, suggesting that the integrin.laminin protein complex at the apical ES likely utilizes these protein kinases as regulatory proteins to modulate Sertoli-germ cell adherens junction dynamics via the ERK signaling pathway. To validate this hypothesis further, an in vivo model using AF-2364 (1-(2,4-dichlorobenzyl)-1H-indazole-3-carbohydrazide) to perturb Sertoli-germ cell anchoring junction function, inducing germ cell loss from the epithelium in adult rats, was used in conjunction with specific inhibitors. Interestingly, the event of germ cell loss induced by AF-2364 in vivo was also associated with induction of PI3K, p-PKB, PAK-2, and p-ERK as well as a surge in intrinsic PKB activity. Perhaps the most important of all, pretreatment of rats with wortmannin (a PI3K inhibitor) or anti-beta1-integrin antibody via intratesticular injection indeed delayed AF-2364-induced spermatid loss from the epithelium. In summary, these results illustrate that Sertoli-germ cell anchoring junction dynamics in the testis are regulated, at least in part, via the beta1-integrin/PI3K/PKB/ERK signaling pathway.


Assuntos
Células Germinativas/metabolismo , Metabolismo dos Lipídeos , Proteínas Quinases/química , Células de Sertoli/citologia , Espermatogênese , Testículo/metabolismo , Androstadienos/farmacologia , Animais , Comunicação Celular , Técnicas de Cultura de Células , Quelantes/farmacologia , Técnicas de Cocultura , Meios de Cultura/farmacologia , Meios de Cultura Livres de Soro/metabolismo , Meios de Cultura Livres de Soro/farmacologia , Citoesqueleto/metabolismo , Ácido Edético/farmacologia , Ácido Egtázico/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Integrina beta1/metabolismo , Lipídeos/química , Masculino , Microscopia Eletrônica , Microscopia de Fluorescência , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Fatores de Tempo , Tirosina/química , Wortmanina
15.
J Cell Physiol ; 205(1): 141-57, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15880438

RESUMO

During spermatogenesis, both adherens junctions (AJ) (such as ectoplasmic specialization (ES), a testis-specific AJ type at the Sertoli cell-spermatid interface (apical ES) or Sertoli-Sertoli cell interface (basal ES) in the apical compartment and BTB, respectively) and tight junctions (TJ) undergo extensive restructuring to permit germ cells to move across the blood-testis barrier (BTB) as well as the seminiferous epithelium from the basal compartment to the luminal edge to permit fully developed spermatids (spermatozoa) to be sloughed at spermiation. However, the integrity of the BTB cannot be compromised throughout spermatogenesis so that postmeiotic germ cell-specific antigens can be sequestered from the systemic circulation at all times. We thus hypothesize that AJ disruption in the seminiferous epithelium unlike other epithelia, can occur without compromising the BTB-barrier, even though these junctions, namely TJ and basal ES, co-exist side-by-side in the BTB. Using an intratesticular androgen suppression-induced germ cell loss model, we have shown that the disruption of AJs indeed was limited to the Sertoli-germ cell interface without perturbing the BTB. The testis apparently is using a unique physiological mechanism to induce the production of both TJ- and AJ-integral membrane proteins and their associated adaptors to maintain BTB integrity yet permitting a transient loss of cell adhesion function by dissociating N-cadherin from beta-catenin at the apical and basal ES. The enhanced production of TJ proteins, such as occludin and ZO-1, at the BTB site can supersede the transient loss of cadherin-catenin function at the basal ES. This thus allows germ cell depletion from the epithelium without compromising BTB integrity. It is plausible that the testis is using this novel mechanism to facilitate the movement of preleptotene and leptotene spermatocytes across the BTB at late stage VIII through early stage IX of the epithelial cycle in the rat while maintaining the BTB immunological barrier function.


Assuntos
Junções Aderentes/metabolismo , Androgênios/metabolismo , Barreira Hematotesticular/fisiologia , Epitélio Seminífero/citologia , Células de Sertoli/citologia , Espermatozoides/citologia , Animais , Peso Corporal , Caderinas/metabolismo , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Modelos Animais , Ocludina , Tamanho do Órgão , Fosfoproteínas/metabolismo , Fosfotirosina/metabolismo , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Epitélio Seminífero/metabolismo , Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Transativadores/metabolismo , Regulação para Cima , Proteína da Zônula de Oclusão-1 , beta Catenina
16.
Biol Reprod ; 73(3): 458-71, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15858215

RESUMO

During spermatogenesis, extensive restructuring of cell junctions takes place in the seminiferous epithelium to facilitate germ cell movement. However, the mechanism that regulates this event remains largely unknown. Recent studies have shown that nitric oxide (NO) likely regulates tight junction (TJ) dynamics in the testis via the cGMP/protein kinase G (cGMP-dependent protein kinase, PRKG) signaling pathway. Due to the proximity of TJ and adherens junctions (AJ) in the testis, in particular at the blood-testis barrier, it is of interest to investigate if NO can affect AJ dynamics. Studies using Sertoli-germ cell cocultures in vitro have shown that the levels of NOS (nitric oxide synthase), cGMP, and PRKG were induced when anchoring junctions were being established. Using an in vivo model in which adult rats were treated with adjudin [a molecule that induces adherens junction disruption, formerly called AF-2364, 1-(2,4-dichlorobenzyl)-IH-indazole-3-carbohydrazide], the event of AJ disruption was also associated with a transient iNOS (inducible nitric oxide synthase, NOS2) induction. Immunohistochemistry has illustrated that NOS2 was intensely accumulated in Sertoli and germ cells in the epithelium during adjudin-induced germ cell loss, with a concomitant accumulation of intracellular cGMP and an induction of PRKG but not cAMP or protein kinase A (cAMP-dependent protein kinase, PRKA). To identify the NOS-mediated downstream signaling partners, coimmunoprecipitation was used to demonstrate that NOS2 and eNOS (endothelial nitric oxide synthase, NOS3) were structurally associated with the N-cadherin (CDH2)/beta-catenin (CATNB)/actin complex but not the nectin-3 (poliovirus receptor-related 3, PVRL 3)/afadin (myeloid/lymphoid or mixed lineage-leukemia tranlocation to 4 homolog, MLLT4) nor the integrin beta1 (ITB1)-mediated protein complexes, illustrating the spatial vicinity of NOS with selected AJ-protein complexes. Interestingly, CDH2 and CATNB were shown to dissociate from NOS during the adjudin-mediated AJ disruption, implicating the CDH2/CATNB protein complex is the likely downstream target of the NO signaling. Furthermore, PRKG, the downstream signaling protein of NOS, was shown to interact with CATNB in the rat testis. Perhaps the most important of all, pretreatment of testes with KT5823, a specific PRKG inhibitor, can indeed delay the adjudin-induced germ cell loss, further validating NOS/NO regulates Sertoli-germ cell AJ dynamics via the cGMP/PRKG pathway. These results illustrate that the CDH2/CATNB-mediated adhesion function in the testis is regulated, at least in part, via the NOS/cGMP/PRKG/CATNB pathway.


Assuntos
Junções Aderentes/fisiologia , Células de Sertoli/fisiologia , Transdução de Sinais/fisiologia , Espermatozoides/fisiologia , Animais , Carbazóis/farmacologia , Células Cultivadas , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Proteínas do Citoesqueleto/metabolismo , Regulação da Expressão Gênica/fisiologia , Hidrazinas/farmacologia , Indazóis/farmacologia , Indóis/farmacologia , Masculino , Óxido Nítrico Sintase/biossíntese , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transativadores/metabolismo , beta Catenina
17.
Endocrinology ; 146(3): 1268-84, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15591133

RESUMO

Using a well characterized model of cell-cell actin-based adherens junction (AJ) disruption by suppressing the intratesticular testosterone level in adult rats with testosterone-estradiol implants, we have confirmed earlier findings that Sertoli-germ cell AJ dynamics are regulated by the activation of kinases via putative signaling pathways but with some unexpected findings as follows. First, the loss of germ cells from the seminiferous epithelium during androgen suppression was associated with a surge in myotubularin-related protein 2 (MTMR2, a lipid phosphatase, in which adult MTMR2-/- mice were recently shown to be azoospermic because of the loss of cell adhesion function between germ and Sertoli cells); kinases: phosphatidylinositol 3-kinase, c-Src, and C-terminal Src kinase; adaptors: alpha-actinin, vinculin, afadin, and p130 Crk-associated protein; and AJ-integral membrane proteins at the ectoplasmic specialization (ES, a testis-specific cell-cell actin-based AJ type) site: N-cadherin, beta-catenin, integrin beta1, and nectin 3. Second, MTMR2, instead of structurally interacting with phosphatidylinositol 3-kinase, a protein and lipid kinase, was shown to associate only with c-Src, a nonreceptor protein tyrosine kinase, as demonstrated by both coimmunoprecipitation and fluorescent microscopy at the site of apical ES, but none of the kinases, adaptors, and AJ-integral proteins that were examined. Collectively, these results suggest that the MTMR2/c-Src is an important phosphatase/kinase protein pair in AJ dynamics in the testis. Because c-Src is known to associate with the cadherin/catenin protein complex at the ES in the testis, we next sought to investigate any changes in the protein-protein interactions of this protein complex during androgen suppression-induced germ cell loss. Indeed, there was a loss of N-cadherin and beta-catenin association, accompanied by a surge in Tyr phosphorylation of beta-catenin, during germ cell loss from the epithelium. Third, and perhaps the most important of all, during natural recovery of the epithelium after removal of testosterone-estradiol implants when spermatids were reattaching to Sertoli cells, an increase in N-cadherin and beta-catenin association was detected with a concomitant loss in the increased Tyr phosphorylation in beta-catenin. In summary, these results illustrate that the cadherin/catenin is a crucial cell adhesion complex that regulates AJ dynamics in the testis, and its functionality is likely modulated by the MTMR2/c-Src protein complex.


Assuntos
Junções Aderentes/fisiologia , Regulação da Expressão Gênica , Células Germinativas/citologia , Células de Sertoli/citologia , Animais , Caderinas/metabolismo , Técnicas de Cocultura , Acetato de Ciproterona/farmacologia , Proteínas do Citoesqueleto/metabolismo , Células Germinativas/metabolismo , Immunoblotting , Imuno-Histoquímica , Masculino , Microscopia de Fluorescência , Modelos Biológicos , Fosforilação , Ligação Proteica , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Fosfatases não Receptoras , Ratos , Ratos Sprague-Dawley , Túbulos Seminíferos/metabolismo , Testículo/metabolismo , Fatores de Tempo , Transativadores/metabolismo , Tirosina/metabolismo , beta Catenina
18.
Endocrinology ; 146(3): 1192-204, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15591141

RESUMO

Apical ectoplasmic specialization (ES) is a unique testis-specific cell-cell actin-based adherens junction type restricted to the Sertoli-round/elongating/elongate spermatid interface in the seminiferous epithelium. An endogenous testosterone (T) suppression model was used to study the regulation of apical ES dynamics in the testis. By providing sustained releases of T and estradiol using subdermal implants in rats, this treatment reduced endogenous testicular T level. This in turn led to sloughing of spermatids (step 8 and beyond) from the seminiferous epithelium, which can be reversed by removing the implants, or replacing them with a higher dose of T implants. This model thus allows us to study the restructuring events at the apical ES. It was shown that apical ES restructuring involved proteins that were usually restricted to the cell-matrix focal adhesion site in other epithelia. For instance, the protein levels of beta1-integrin, Tyr-phosphorylated focal adhesion kinase (p-FAK), and c-Src were induced during the T suppression-induced germ cell loss and recovery, implicating that these proteins are putative regulators of ES dynamics. Indeed, the formation of p-FAK/c-Src protein complex, but not their association with beta1-integrin, was stimulated during T suppression-induced germ cell loss. ERK, a MAPK known to regulate focal adhesion turnover, was also activated during the androgen suppression-induced spermatid loss and the early phase of the recovery when germ cells began to repopulate the epithelium. Collectively, these data suggest that the apical ES is a cell-cell adherens junction type with the characteristics of cell-matrix focal contacts. In addition to its role in conferring cell adhesion formation, the p-FAK/c-Src protein complex apparently also regulates apical ES disruption via the ERK signaling pathway.


Assuntos
Proteínas Tirosina Quinases/metabolismo , Epitélio Seminífero/metabolismo , Testículo/metabolismo , Testosterona/farmacologia , Junções Aderentes/metabolismo , Androgênios/metabolismo , Animais , Proteína Tirosina Quinase CSK , Adesão Celular , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Adesões Focais/metabolismo , Células Germinativas/metabolismo , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Integrina beta1/metabolismo , Masculino , Microscopia de Fluorescência , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Espermátides/metabolismo , Espermatogênese , Testosterona/metabolismo , Fatores de Tempo , Quinases da Família src
19.
Endocrinology ; 146(4): 1893-908, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15618353

RESUMO

The blood-testis barrier (BTB), in contrast to the blood-brain and blood-retina barriers, is composed of coexisting tight junctions, gap junctions, and basal ectoplasmic specializations, a testis-specific type of adherens junction. Recent studies showed that BTB restructuring that facilitates germ cell migration during spermatogenesis involves proteolysis, an event that is usually restricted to the cell-matrix interface in other epithelia. For instance, a surge in alpha(2)-macroglobulin (alpha(2)-MG), a protease inhibitor produced by Sertoli cells, was detected at the Sertoli-Sertoli and Sertoli-germ cell interface in the epithelium during cadmium chloride-induced BTB disruption in adult rats. It is thus proposed that the increase in alpha(2)-MG is crucial for protecting the epithelium from unwanted proteolysis as well as regulating the availability of cytokines that affect junction turnover. Although both tight junction and adherens junction dynamics at the BTB are regulated via the p38 MAPK signaling pathway, the mechanism(s) that regulates alpha(2)-MG is entirely unknown. In this study, we have shown that by administering dimethylaminopurine, a c-Jun N-terminal protein kinase (JNK) inhibitor, to the testis, JNK activity was blocked specifically and alpha(2)-MG production was inhibited, worsening the cadmium chloride-induced damage to the epithelium. Studies coupled with inhibitors, immunoblottings, and immunofluorescent and electron microscopy have unequivocally demonstrated that the JNK signaling pathway is a putative regulatory pathway for alpha(2)-MG production in the testis. This finding illustrates for the first time that a cell-matrix restructuring event occurs in normal cell physiology at the cell-cell interface in the testis, highlighting the significance of alpha(2)-MG in the regulation of BTB function.


Assuntos
Adenina/análogos & derivados , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Transdução de Sinais/fisiologia , Testículo/metabolismo , alfa-Macroglobulinas/fisiologia , Adenina/farmacologia , Animais , Cloreto de Cádmio/toxicidade , Masculino , Ratos , Ratos Sprague-Dawley , Epitélio Seminífero/citologia , Epitélio Seminífero/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
20.
J Cell Sci ; 117(Pt 5): 783-98, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14734653

RESUMO

An in vivo model was used to investigate the regulation of tight junction (TJ) dynamics in the testis when adult rats were treated with CdCl(2). It was shown that the CdCl(2)-induced disruption of the blood-testis barrier (BTB) associated with a transient induction in testicular TGF-beta2 and TGF-beta3 (but not TGF-beta1) and the phosphorylated p38 mitogen activated protein (MAP) kinase, concomitant with a loss of occludin and zonula occludens-1 (ZO-1) from the BTB site in the seminiferous epithelium. These results suggest that BTB dynamics in vivo are regulated by TGF-beta2/-beta3 via the p38 MAP kinase pathway. Indeed, SB202190, a specific p38 MAP kinase inhibitor, blocked the CdCl(2)-induced occludin and ZO-1 loss from the BTB. This result clearly illustrates that CdCl(2) mediates its BTB disruptive effects via the TGF-beta3/p38 MAP kinase signaling pathway. Besides, this CdCl(2)-induced occludin and ZO-1 loss from the BTB also associated with a significant loss of the cadherin/catenin and the nectin/afadin protein complexes at the site of cell-cell actin-based adherens junctions (AJs). An induction of alpha(2)-macroglobulin (a non-specific protease inhibitor) was also observed during BTB damage and when the seminiferous epithelium was being depleted of germ cells. These data illustrate that a primary disruption of the BTB can lead to a secondary loss of cell adhesion function at the site of AJs, concomitant with an induction in protease inhibitor, which apparently is used to protect the epithelium from unwanted proteolysis. alpha(2)-Macroglobulin was also shown to associate physically with TGF-beta3, afadin and nectin 3, but not occludin, E-cadherin or N-cadherin, indicating its possible role in junction restructuring in vivo. Additionally, the use of SB202190 to block the TGF-beta3/p-38 MAP kinase pathway also prevented the CdCl(2)-induced loss of cadherin/catenin and nectin/afadin protein complexes from the AJ sites, yet it had no apparent effect on alpha(2)-macroglobulin. These results demonstrate for the first time that the TGF-beta3/p38 MAP kinase signaling pathway is being used to regulate both TJ and AJ dynamics in the testis, mediated by the effects of TGF-beta3 on TJ- and AJ-integral membrane proteins and adaptors, but not protease inhibitors.


Assuntos
Barreira Hematotesticular/fisiologia , Testículo/metabolismo , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/metabolismo , Animais , Barreira Hematotesticular/efeitos dos fármacos , Barreira Hematotesticular/ultraestrutura , Cloreto de Cádmio/farmacologia , Proteínas do Citoesqueleto/metabolismo , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Proteínas de Membrana/metabolismo , Ocludina , Peptídeo Hidrolases/metabolismo , Fosfoproteínas/metabolismo , Inibidores de Proteases/metabolismo , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Epitélio Seminífero/efeitos dos fármacos , Epitélio Seminífero/metabolismo , Testículo/efeitos dos fármacos , Junções Íntimas/metabolismo , Junções Íntimas/ultraestrutura , Fator de Crescimento Transformador beta/metabolismo , Proteína da Zônula de Oclusão-1 , alfa-Macroglobulinas/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...