Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Cell Biol ; 154(4): 799-814, 2001 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-11514592

RESUMO

We show here that the alpha, beta, and gamma isotypes of peroxisome proliferator-activated receptor (PPAR) are expressed in the mouse epidermis during fetal development and that they disappear progressively from the interfollicular epithelium after birth. Interestingly, PPARalpha and beta expression is reactivated in the adult epidermis after various stimuli, resulting in keratinocyte proliferation and differentiation such as tetradecanoylphorbol acetate topical application, hair plucking, or skin wound healing. Using PPARalpha, beta, and gamma mutant mice, we demonstrate that PPARalpha and beta are important for the rapid epithelialization of a skin wound and that each of them plays a specific role in this process. PPARalpha is mainly involved in the early inflammation phase of the healing, whereas PPARbeta is implicated in the control of keratinocyte proliferation. In addition and very interestingly, PPARbeta mutant primary keratinocytes show impaired adhesion and migration properties. Thus, the findings presented here reveal unpredicted roles for PPARalpha and beta in adult mouse epidermal repair.


Assuntos
Epiderme/fisiologia , Queratinócitos/fisiologia , Peroxissomos/fisiologia , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , Cicatrização/genética , Animais , Adesão Celular , Divisão Celular , Movimento Celular , Colágeno/metabolismo , Elastina/metabolismo , Células Epidérmicas , Folículo Piloso/lesões , Queratinócitos/citologia , Macrófagos/citologia , Camundongos , Camundongos Mutantes , Neutrófilos/citologia , Pele/lesões , Acetato de Tetradecanoilforbol/farmacologia , Regulação para Cima
2.
Oncogene ; 37(2): 160-173, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-28892046

RESUMO

Most anticancer therapies to date focus on druggable features of tumor epithelia. Despite the increasing repertoire of treatment options, patient responses remain varied. Moreover, tumor resistance and relapse remain persistent clinical challenges. These observations imply an incomplete understanding of tumor heterogeneity. The tumor microenvironment is a major determinant of disease progression and therapy outcome. Cancer-associated fibroblasts (CAFs) are the dominant cell type within the reactive stroma of tumors. They orchestrate paracrine pro-tumorigenic signaling with adjacent tumor cells, thus exacerbating the hallmarks of cancer and accelerating tumor malignancy. Although CAF-derived soluble factors have been investigated for tumor stroma-directed therapy, the underlying transcriptional programs that enable the oncogenic functions of CAFs remain poorly understood. Nuclear receptors (NRs), a large family of ligand-responsive transcription factors, are pharmacologically viable targets for the suppression of CAF-facilitated oncogenesis. In this study, we defined the expression profiles of NRs in CAFs from clinical cutaneous squamous cell carcinoma (SCC) biopsies. We further identified a cluster of driver NRs in CAFs as important modifiers of CAF function with profound influence on cancer cell invasiveness, proliferation, drug resistance, energy metabolism and oxidative stress status. Importantly, guided by the NR profile of CAFs, retinoic acid receptor ß and androgen receptor antagonists were identified for concurrent therapy with cisplatin, resulting in the inhibition of chemoresistance in recurred SCC:CAF xenografts. Our work demonstrates that treatments targeting both the tumor epithelia and the surrounding CAFs can extend the efficacy of conventional chemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Fibroblastos Associados a Câncer/efeitos dos fármacos , Carcinoma de Células Escamosas/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Neoplasias Cutâneas/patologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biópsia , Fibroblastos Associados a Câncer/patologia , Carcinogênese/efeitos dos fármacos , Carcinogênese/patologia , Carcinoma de Células Escamosas/tratamento farmacológico , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Terapia Combinada/métodos , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Cultura Primária de Células , Receptores Citoplasmáticos e Nucleares/metabolismo , Neoplasias Cutâneas/tratamento farmacológico , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Biomater Sci ; 6(1): 168-178, 2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29167844

RESUMO

Adipose-derived stem cells (ASCs) are found in a location within the adipose tissue known as the stem cell niche. The ASCs in the niche are maintained in the quiescent state, and upon exposure to various microenvironmental triggers are prompted to undergo proliferation or differentiation. These microenvironmental triggers also modulate the extracellular matrix (ECM), which interacts with the cells through the cytoskeleton and induces downstream events inside the cells that bring about a change in cell behaviour. In response to these changes, the cells remodel the ECM, which will differ according to the type of tissue being formed by the cells. As the ECM itself plays an important role in the regulation of cellular differentiation, this study aims to explore the role of the cell-secreted ECM at various stages of differentiation of stem cells in triggering the differentiation of ASCs. To this end, the ASCs cultured in proliferation, osteogenic and adipogenic media were decellularized and the secreted ECM was characterized. Overall, it was found that osteo-differentiated ASCs produced higher amounts of collagen and glycosaminoglycans (GAG) compared to the undifferentiated and adipo-differentiated ASCs. The two types of differentiated ECMs were subsequently shown to trigger initial but not terminal differentiation of ASCs into osteo- and adipo-lineages respectively, as indicated by the upregulation of lineage specific markers. In addition, integrin subunits alpha (α) 6 and integrin beta (ß) 1 were found to be produced by ASCs cultured on cell-secreted ECM-coated substrates, suggesting that the integrins α6 and ß1 play an instrumental role in cell-ECM interactions. Taken together, this study demonstrates the importance of the ECM in cellular fate decisions and how ECM-coated substrates can potentially be used for various tissue engineering applications.


Assuntos
Tecido Adiposo/citologia , Adipócitos/citologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Células Cultivadas , Matriz Extracelular/química , Humanos , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , Engenharia Tecidual/métodos
4.
Oncogene ; 36(46): 6408-6419, 2017 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-28745316

RESUMO

Metastatic cancer cells acquire energy-intensive processes including increased invasiveness and chemoresistance. However, how the energy demand is met and the molecular drivers that coordinate an increase in cellular metabolic activity to drive epithelial-mesenchymal transition (EMT), the first step of metastasis, remain unclear. Using different in vitro and in vivo EMT models with clinical patient's samples, we showed that EMT is an energy-demanding process fueled by glucose metabolism-derived adenosine triphosphate (ATP). We identified angiopoietin-like 4 (ANGPTL4) as a key player that coordinates an increase in cellular energy flux crucial for EMT via an ANGPTL4/14-3-3γ signaling axis. This augmented cellular metabolic activity enhanced metastasis. ANGPTL4 knockdown suppresses an adenylate energy charge elevation, delaying EMT. Using an in vivo dual-inducible EMT model, we found that ANGPTL4 deficiency reduces cancer metastasis to the lung and liver. Unbiased kinase inhibitor screens and Ingenuity Pathway Analysis revealed that ANGPTL4 regulates the expression of 14-3-3γ adaptor protein via the phosphatidylinositol-3-kinase/AKT and mitogen-activated protein kinase signaling pathways that culminate to activation of transcription factors, CREB, cFOS and STAT3. Using a different mode of action, as compared with protein kinases, the ANGPTL4/14-3-3γ signaling axis consolidated cellular bioenergetics and stabilized critical EMT proteins to coordinate energy demand and enhanced EMT competency and metastasis, through interaction with specific phosphorylation signals on target proteins.


Assuntos
Proteínas 14-3-3/metabolismo , Proteína 4 Semelhante a Angiopoietina/metabolismo , Transição Epitelial-Mesenquimal , Transdução de Sinais , Proteínas 14-3-3/genética , Trifosfato de Adenosina/metabolismo , Proteína 4 Semelhante a Angiopoietina/genética , Animais , Linhagem Celular Tumoral , Células HEK293 , Células Hep G2 , Humanos , Immunoblotting , Células MCF-7 , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Microscopia Confocal , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Interferência de RNA , Fator de Transcrição STAT3/metabolismo , Transplante Heterólogo
5.
Biochim Biophys Acta ; 1452(2): 103-20, 1999 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-10559464

RESUMO

The full length estrogen receptor from Oreochromis aureus (OaER) was cloned and expressed in vitro and in vivo as a functional transcription factor. Amino acid residues involved in the thermal stability of the receptor are located at/near subzones beta1 and beta3, which are highly conserved in other non-piscine species but not in OaER. Hormone binding studies, however, indicate that OaER is thermally stable but exhibited a approximately 3-fold reduced affinity for estrogen at elevated temperatures. Transfection of OaER into various cell lines cultured at different temperatures displayed a significant estrogen dose-response shift compared with that of chicken ER (cER). At 37 degrees C, OaER requires approximately 80-fold more estrogen to achieve half-maximal stimulation of CAT. Lowering of the incubation temperature from 37 degrees C to 25 degrees C or 20 degrees C resulted in a 4-fold increase in its affinity for estrogen. The thermally deficient transactivation of OaER at temperatures above 25 degrees C was fully prevented by high levels of estrogen. Thus, compared to cER, the OaER exhibits reduced affinity for estrogen at elevated temperature as reflected in its deficient transactivation capability. Amino acid replacements of OaER beta3 subzones with corresponding amino acids from cER could partially rescue this temperature sensitivity. The three-dimensional structure of the OaER ligand binding domain (LBD) was modelled based on conformational similarity and sequence homology with human RXRalpha apo, RARgamma holo and ERalpha LBDs. Unliganded and 17beta-estradiol-liganded OaER LBD retained the overall folding pattern of the nuclear receptor LBDs. The residues at/near the subzone beta3 of the LBD constitute the central core of OaER structure. Thus, amino acid alteration at this region potentially alters the structure and consequently its temperature-dependent ligand binding properties.


Assuntos
Estrogênios/metabolismo , Receptores de Estrogênio/biossíntese , Tilápia/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Clonagem Molecular , DNA Complementar/biossíntese , DNA Complementar/química , Estrogênios/química , Expressão Gênica , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Receptores de Estrogênio/química , Receptores de Estrogênio/genética , Alinhamento de Sequência , Temperatura , Transcrição Gênica , Ativação Transcricional
6.
FEBS Lett ; 459(1): 57-63, 1999 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-10508917

RESUMO

The Oreochromis aureus vitellogenin (OaVtg) gene contains three imperfect oestrogen response elements (EREs) and GATA and VBP (vitellogenin binding protein) binding sites. An analysis of the promoter indicates that the 5'-flanking region up to position -625 is sufficient to mediate E(2) control. Furthermore, transfection of deletion and mutagenised promoters indicates that both GATA and VBP synergise with ER, and thus contribute to the regulation of the endogenous OaVtg gene. These findings support the notion that the interplay of promoter elements mediates proper hormone-dependent and tissue-specific expression of the OaVtg gene, regardless of non-consensus sequence context of EREs and VBP.


Assuntos
Proteínas Aviárias , Proteínas de Transporte/fisiologia , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica , Receptores de Estrogênio/fisiologia , Fatores de Transcrição/fisiologia , Vitelogeninas/genética , Animais , Fatores de Transcrição de Zíper de Leucina Básica , Células COS , Fator de Transcrição GATA6 , Perciformes , Regiões Promotoras Genéticas , Transcrição Gênica
7.
Mol Cell Endocrinol ; 120(2): 177-92, 1996 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-8832578

RESUMO

The O. aureus estrogen receptor (OaER) gene of 40.4 kb containing ten exons is the first complete piscine gene to be cloned. There are two extra introns: intron I that divides the 5' UTR into two exons, and intron V that intersperses D and E1 exons. Except for I and V, other introns have identical positions to those of human ER gene. All the donor and acceptor splice sites exhibit consensus sequences. The promoter lacks consensus TATA and CAAT boxes. This region exhibits several putative regulatory elements. A functional imperfect ERE deviating at two bases is located in the leader exon, thus suggesting that this gene is autoregulated. The OaER gene lacks an A region whereas its C and E domains are highly conserved. Within the ER subfamily, OaER exhibits the longest F domain of 77 amino acids. OaER has a long 3'UTR constituting >1/2 of its transcript. Using RT-PCR and SI nuclease mapping, we report for the first time the usage of both alternative transcriptional start sites and polyadenylation signals during estrogen-induced OaER expression. Thus, O. aureus may have four species of ER transcripts differing structurally in their transcriptional start sites and lengths of their 3' UTR.


Assuntos
Receptores de Estrogênio/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Peixes , Humanos , Dados de Sequência Molecular , Alinhamento de Sequência , Análise de Sequência , Transcrição Gênica
8.
Mol Cell Endocrinol ; 123(2): 149-61, 1996 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-8961252

RESUMO

The Oreochromis aureus estrogen receptor (OaER) 5' region is 62.2% AT-rich and does not display well-positioned consensus TATA and CAAT boxes. A functional imperfect 13 bp ERE, TGTTAtggTGACC, deviating at 2 bases is located in the leader exon. Transient transfection assays indicate that this ERE confers a 5-fold increase in SEAP reporter gene activity at 20 h post E2-induction. The continued elevation of transcription after the initial peak could be responsible for the "memory' effect of E2-induced vitellogenesis. The 4.4 kb OaER 3' region is AT-rich and has a high representation of 1/2 EREs and GREs. There are 10 copies of the destabilizing pentamer ATTTA which, in transient transfection experiments strongly suppressed SEAP activity. The 3' EREs are functional, and on their own, induce a bimodal increase in SEAP activity of 9-fold at 6 h and 11-fold at 18 h post-E2 induction. A recombinant construct of SEAP gene flanked by the 5' upstream and 3' flanking regions of the OaER gene allowed these regulatory signals to "cross-talk' to achieve a 5- and 25-fold increase for the first and second peaks, respectively. This bimodal response to E2-induction is attributed to both transcriptional and translational controls over the SEAP gene. This work represents a novel illustration of a synergistic interaction between the 5' and 3' regulatory elements of a steroid receptor in autoregulation.


Assuntos
Regulação da Expressão Gênica , Receptores de Estrogênio/genética , Transcrição Gênica , Animais , Peixes , Plasmídeos , Transfecção
9.
Mol Cell Endocrinol ; 146(1-2): 103-20, 1998 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-10022768

RESUMO

The Oreochromis aureus vitellogenin, OaVtg, gene spans 9 kb and contains 34 exons. Its transcription start site is located 15 bp upstream of the translational start codon. Although the OaVtg promoter has a nonconsensus TATA, transient transfection assay showed that this promoter is capable of driving basal transcription. Two imperfect estrogen response elements: EREp (proximal) and EREd (distal) are located in the promoter at - 532 and - 1352, respectively. In competition gel mobility-shift assays, only EREp exhibited specific binding of the recombinant estrogen receptor protein, GST-C/D OaER. Another imperfect ERE (EREexon2) was detected within exon 2 of the OaVtg gene. This is a novel finding for a vitellogenin (Vtg) gene. EREexon2 similarly showed specific recognition of GST-C/D OaER. Both EREp and EREexon2 showed comparable binding affinities as consensus ERE. In transient transfections, the OaVtg promoter, EREp and EREd elicited significant increase in estrogen-dependent synthesis of CAT protein. Hence, we propose that the non-consensus OaVtg EREs contribute to the estrogen-dependent regulation of the OaVtg gene in vivo.


Assuntos
Estrogênios/farmacologia , Regiões Promotoras Genéticas , Tilápia/genética , Vitelogeninas/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Cloranfenicol O-Acetiltransferase/genética , Escherichia coli/genética , Éxons , Feminino , Expressão Gênica , Íntrons , Dados de Sequência Molecular , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Proteínas Recombinantes/metabolismo , Sequências Reguladoras de Ácido Nucleico , Elementos de Resposta , Transcrição Gênica , Vitelogeninas/química
10.
J Reprod Immunol ; 23(2): 109-18, 1993 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8510075

RESUMO

In an effort to evaluate the relationships among pregnancy, cellular cytotoxicity and herpes simplex virus (HSV) infection, we conducted a series of experiments investigating: (1) the maternal cellular cytotoxic response to HSV infection as compared with non-pregnant hosts, (2) the influence of both cyclooxygenase and lipoxygenase products on cytotoxicity by selective inhibition of their metabolic pathways, and (3) the potential pregnancy-related differences in immune response to selective inhibition of eicosanoid metabolism. Indomethacin was used for cyclooxygenase blockade and nordihydroguaiaretic acid was used to evaluate lipoxygenase inhibition. In the non-infected animals no differences in cytotoxicity were observed between pregnant (1.5% +/- 0.7%) and non-pregnant (4.6% +/- 2.0%) groups. HSV infection increased cytotoxicity equally in both groups (pregnant: 10.6% +/- 2.0% vs. non-pregnant: 14.2% +/- 3.4%). Indomethacin did not significantly alter cytotoxicity in either the pregnant or the non-pregnant groups compared with controls (12.8% +/- 1.8% vs. 10.6% +/- 2.0% and 14.3% +/- 3.9% vs. 14.2% +/- 3.4%, respectively). In contrast, NDGA elicited a significant reduction in the cytotoxic response in both pregnant and non-pregnant hosts (6.2% +/- 1.1% vs. 10.6% +/- 2.0% and 5.7% +/- 1.1% vs. 14.2% +/- 3.4%, respectively). From our study we conclude that: (1) cytotoxicity is maintained at low levels in the absence of HSV infection, (2) HSV infection induces a significant augmentation in host cellular cytotoxicity, (3) pregnant and non-pregnant cytotoxic responses to HSV infection appear comparable, (4) indomethacin does not augment in vitro cytotoxicity to HSV infection and (5) NDGA suppresses cytotoxicity, providing evidence that lipoxygenase metabolites are essential to cytotoxic cell function.


Assuntos
Citotoxicidade Imunológica/fisiologia , Eicosanoides/imunologia , Herpes Simples/imunologia , Complicações Infecciosas na Gravidez/imunologia , Prenhez/imunologia , Animais , Testes Imunológicos de Citotoxicidade , Citotoxicidade Imunológica/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Eicosanoides/metabolismo , Feminino , Herpes Simples/metabolismo , Terapia de Imunossupressão/métodos , Indometacina/farmacologia , Lipoxigenase/imunologia , Masoprocol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Complicações Infecciosas na Gravidez/metabolismo , Prenhez/metabolismo , Prostaglandina-Endoperóxido Sintases/imunologia
11.
J Reprod Immunol ; 21(2): 139-48, 1992 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1548628

RESUMO

Fetal trophoblast is generally resistant to lysis by cytotoxic cells. We hypothesized that progesterone and estrogens secreted by the trophoblast act at the choriodecidual interface where they are present in high concentrations to provide a local, paracrine immunosuppressive effect on cellular cytotoxicity. Using peripheral blood mononuclear cells as effector cells in a cytotoxicity assay, we evaluated the effects of progesterone, estrone, estradiol and estriol, either alone or in combination, on cellular cytotoxicity. Both progesterone and estradiol suppressed cytotoxicity in a dose-dependent manner. Estrone, estriol, pregnenolone and cholesterol had no effect. A synergistic suppression of cytotoxicity was observed when estrone, estradiol, estriol and progesterone were combined. We speculate that trophoblast production of progesterone and estradiol may be an important local immunosuppressive mechanism contributing to fetal survival.


Assuntos
Citotoxicidade Imunológica/efeitos dos fármacos , Estradiol/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Progesterona/farmacologia , Células Cultivadas , Testes Imunológicos de Citotoxicidade , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Humanos , Leucócitos Mononucleares/imunologia , Mifepristona/farmacologia , Trofoblastos/fisiologia
12.
DNA Seq ; 5(6): 359-70, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-8777315

RESUMO

The nucleotide sequence of the estrogen receptor gene of Oreochromis aureus (OaER) indicates that the hormone-binding E domain is composed of 4 exons interspersed by short introns of only 0.18-1.3 kb each. All 4 E exons exhibit consensus sequences flanking the donor and acceptor splice sites. Analysis of introns revealed (i) numerous palindromic and half-palindromic steroid responsive elements including ERE, TRE and GRE, (ii) six alternative polyadenylation signals and (iii) putative control regions identified by the clustering of transcription factor binding sites. Of particular interest is the presence of a TATA and CAAT box in intron IV. The hydropathicity profile shows that the E exons are relatively hydrophobic. Two receptor dimerization regions have been observed: a conserved heptad repeat of hydrophobic residues (R168-M193) and a perfect leucine zipper (L36-L57). The presence of multiple sites for kinase activity in these regions suggests the importance of phosphorylation in the regulation of receptor functions and ligand-affinity.


Assuntos
Receptores de Estrogênio/genética , Tilápia/genética , Tilápia/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação/genética , Mapeamento Cromossômico , Clonagem Molecular , Sequência Consenso , Sequência Conservada , DNA/genética , DNA/metabolismo , Éxons , Íntrons , Dados de Sequência Molecular , Fosforilação , Conformação Proteica , Receptores de Estrogênio/química , Receptores de Estrogênio/metabolismo , Sequências Repetitivas de Ácido Nucleico
13.
DNA Seq ; 5(6): 371-9, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-8777316

RESUMO

Although the estrogen receptor gene of Oreochromis aureus (OaER) shows 85% homology to rainbow trout ER (rtER), the molecular organization of its exons and introns in the hormone-binding E domain is more closely related to the human ER gene. Comparison with other vertebrates yielded reduced homologies of 64-67%, probably due to evolutionary speciation. The E1 and E2 exons of OaER are interspersed by a short intron of 1.3 kb which is flanked by consensus splice sites. This is in sharp contrast to the 11 kb intron separating E1 and E2 exons of rtER which also displayed a rare GC donor junction. Three conserved cys at 83, 112 and 195, which are important for formation of 3-D ligand-binding pocket were found in OaER. However, the 4th conserved cys is replaced by a ser. This substitution which is the result of a single base mutation probably suggests different affinity for estrogen or transactivation of the OaER gene. Two overlapping steroid binding and receptor dimerization domains have been observed. The E domain of OaER and rtER has diversified significantly from that of other non-piscine vertebrates, such that they form a separate subgroup in the UPGMA tree of steroid receptors.


Assuntos
Receptores de Estrogênio/genética , Receptores de Esteroides/genética , Tilápia/genética , Tilápia/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação/genética , DNA/genética , DNA/metabolismo , Éxons , Humanos , Íntrons , Dados de Sequência Molecular , Filogenia , Mutação Puntual , Receptores de Estrogênio/química , Receptores de Estrogênio/metabolismo , Receptores de Esteroides/química , Receptores de Esteroides/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
14.
Cell Death Differ ; 20(3): 478-89, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23175186

RESUMO

Following acute-phase infection, activated T cells are terminated to achieve immune homeostasis, failure of which results in lymphoproliferative and autoimmune diseases. We report that sterile α- and heat armadillo-motif-containing protein (SARM), the most conserved Toll-like receptors adaptor, is proapoptotic during T-cell immune response. SARM expression is significantly reduced in natural killer (NK)/T lymphoma patients compared with healthy individuals, suggesting that decreased SARM supports NK/T-cell proliferation. T cells knocked down of SARM survived and proliferated more significantly compared with wild-type T cells following influenza infection in vivo. During activation of cytotoxic T cells, the SARM level fell before rising, correlating inversely with cell proliferation and subsequent T-cell clearance. SARM knockdown rescued T cells from both activation- and neglect-induced cell deaths. The mitochondria-localized SARM triggers intrinsic apoptosis by generating reactive oxygen species and depolarizing the mitochondrial potential. The proapoptotic function is attributable to the C-terminal sterile alpha motif and Toll/interleukin-1 receptor domains. Mechanistically, SARM mediates intrinsic apoptosis via B cell lymphoma-2 (Bcl-2) family members. SARM suppresses B cell lymphoma-extra large (Bcl-xL) and downregulates extracellular signal-regulated kinase phosphorylation, which are cell survival effectors. Overexpression of Bcl-xL and double knockout of Bcl-2 associated X protein and Bcl-2 homologous antagonist killer substantially reduced SARM-induced apoptosis. Collectively, we have shown how T-cell death following infection is mediated by SARM-induced intrinsic apoptosis, which is crucial for T-cell homeostasis.


Assuntos
Proteínas do Domínio Armadillo/metabolismo , Proteínas do Citoesqueleto/metabolismo , Mitocôndrias/metabolismo , Linfócitos T/metabolismo , Animais , Apoptose , Proteínas do Domínio Armadillo/antagonistas & inibidores , Proteínas do Domínio Armadillo/genética , Caspase 9/metabolismo , Células Cultivadas , Proteínas do Citoesqueleto/antagonistas & inibidores , Proteínas do Citoesqueleto/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Humanos , Ativação Linfocitária , Linfoma de Células T/metabolismo , Linfoma de Células T/patologia , Camundongos , Camundongos Transgênicos , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Linfócitos T/imunologia , Transfecção , Proteína X Associada a bcl-2/antagonistas & inibidores , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo , Proteína bcl-X/antagonistas & inibidores , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
15.
Cell Death Dis ; 4: e552, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23519121

RESUMO

Tumor metastasis is the main cause of death in cancer patients. Anoikis resistance is one critical malefactor of metastatic cancer cells to resist current clinical chemotherapeutic treatments. Although endoperoxide-containing compounds have long been suggested as anticancer drugs, few have been clinically employed due to their instability, complex synthesis procedure or low tumor cell selectivity. Herein, we describe a one-pot strategy to synthesize novel amino endoperoxides and their derivatives with good yields and stabilities. In vitro cell-based assays revealed that 4 out of the 14 amino endoperoxides selectively induce metastatic breast carcinoma cells but not normal breast cells to undergo apoptosis, in a dose-dependent manner. Mechanistic studies showed that the most potent amino endoperoxide, 4-Me, is selective for cancer cells expressing a high level of Nox4. The anticancer effects are further shown to be associated with reduced O2(-):H2O2 ratio and increased ·OH level in the cancerous cells. Animal study showed that 4-Me impairs orthotopic breast tumor growth as well as tumor cell metastasis to lymph nodes. Altogether, our study suggests that anticancer strategies that focus on redox-based apoptosis induction in tumors are clinically viable.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Carcinoma/tratamento farmacológico , NADPH Oxidases/genética , Peróxidos/farmacologia , Anoikis/efeitos dos fármacos , Antineoplásicos/síntese química , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , NADPH Oxidase 4 , NADPH Oxidases/metabolismo , Metástase Neoplásica , Oxirredução , Peróxidos/síntese química , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Cell Death Dis ; 4: e848, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-24113182

RESUMO

Epithelial-mesenchymal transition (EMT) is a crucial step in tumor progression, and the TGFß-SMAD signaling pathway as an inductor of EMT in many tumor types is well recognized. However, the role of non-canonical TGFß-TAK1 signaling in EMT remains unclear. Herein, we show that TAK1 deficiency drives metastatic skin squamous cell carcinoma earlier into EMT that is conditional on the elevated cellular ROS level. The expression of TAK1 is consistently reduced in invasive squamous cell carcinoma biopsies. Tumors derived from TAK1-deficient cells also exhibited pronounced invasive morphology. TAK1-deficient cancer cells adopt a more mesenchymal morphology characterized by higher number of focal adhesions, increase surface expression of integrin α5ß1 and active Rac1. Notably, these mutant cells exert an increased cell traction force, an early cellular response during TGFß1-induced EMT. The mRNA level of ZEB1 and SNAIL, transcription factors associated with mesenchymal phenotype is also upregulated in TAK1-deficient cancer cells compared with control cancer cells. We further show that TAK1 modulates Rac1 and RhoA GTPases activities via a redox-dependent downregulation of RhoA by Rac1, which involves the oxidative modification of low-molecular weight protein tyrosine phosphatase. Importantly, the treatment of TAK1-deficient cancer cells with Y27632, a selective inhibitor of Rho-associated protein kinase and antioxidant N-acetylcysteine augment and hinders EMT, respectively. Our findings suggest that a dysregulated balance in the activation of TGFß-TAK1 and TGFß-SMAD pathways is pivotal for TGFß1-induced EMT. Thus, TAK1 deficiency in metastatic cancer cells increases integrin:Rac-induced ROS, which negatively regulated Rho by LMW-PTP to accelerate EMT.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , MAP Quinase Quinase Quinases/metabolismo , Neoplasias de Células Escamosas/enzimologia , Neoplasias de Células Escamosas/patologia , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Cutâneas/patologia , Animais , Fenômenos Biomecânicos/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células HEK293 , Humanos , Integrina beta1/metabolismo , Integrina beta3/metabolismo , MAP Quinase Quinase Quinases/deficiência , Camundongos , Modelos Biológicos , Invasividade Neoplásica , Oxirredução/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Neoplasias Cutâneas/enzimologia , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
17.
Nat Commun ; 4: 1673, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23575677

RESUMO

The use of nanomaterials has raised safety concerns, as their small size facilitates accumulation in and interaction with biological tissues. Here we show that exposure of endothelial cells to TiO2 nanomaterials causes endothelial cell leakiness. This effect is caused by the physical interaction between TiO2 nanomaterials and endothelial cells' adherens junction protein VE-cadherin. As a result, VE-cadherin is phosphorylated at intracellular residues (Y658 and Y731), and the interaction between VE-cadherin and p120 as well as ß-catenin is lost. The resulting signalling cascade promotes actin remodelling, as well as internalization and degradation of VE-cadherin. We show that injections of TiO2 nanomaterials cause leakiness of subcutaneous blood vessels in mice and, in a melanoma-lung metastasis mouse model, increase the number of pulmonary metastases. Our findings uncover a novel non-receptor-mediated mechanism by which nanomaterials trigger intracellular signalling cascades via specific interaction with VE-cadherin, resulting in nanomaterial-induced endothelial cell leakiness.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Nanoestruturas , Titânio/farmacologia , Animais , Apoptose , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Estresse Oxidativo
18.
Nutr Diabetes ; 2: e29, 2012 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23449528

RESUMO

Recent findings on the role of transforming growth factor (TGF)-ß/Smad3 signaling in the pathogenesis of obesity and type 2 diabetes have underscored its importance in metabolism and adiposity. Indeed, elevated TGF-ß has been previously reported in human adipose tissue during morbid obesity and diabetic neuropathy. In this review, we discuss the pleiotropic effects of TGF-ß/Smad3 signaling on metabolism and energy homeostasis, all of which has an important part in the etiology and progression of obesity-linked diabetes; these include adipocyte differentiation, white to brown fat phenotypic transition, glucose and lipid metabolism, pancreatic function, insulin signaling, adipocytokine secretion, inflammation and reactive oxygen species production. We summarize the recent in vivo findings on the role of TGF-ß/Smad3 signaling in metabolism based on the studies using Smad3(-/-) mice. Based on the presence of a dual regulatory effect of Smad3 on peroxisome proliferator-activated receptor (PPAR)ß/δ and PPARγ2 promoters, we propose a unifying mechanism by which this signaling pathway contributes to obesity and its associated diabetes. We also discuss how the inhibition of this signaling pathway has been implicated in the amelioration of many facets of metabolic syndromes, thereby offering novel therapeutic avenues for these metabolic conditions.

19.
Cell Death Differ ; 18(7): 1120-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21233843

RESUMO

Dysregulated reactive oxygen species (ROS) generation contributes to many human pathologies, including cancer and diabetes. During normal wound repair, inflammation-induced ROS production must be tightly controlled, but the mechanisms reining their generation remain unclear. Herein, we show that transforming growth factor ß-activated kinase 1 (TAK1) directly regulates stem cell factor (SCF) expression, which activates the protein kinase B (PKB)α pro-survival pathway in a cell-autonomous manner to protect keratinocytes from ROS-mediated cell death. TAK1 is a pivotal inflammatory mediator whose expression was transiently elevated during wound healing, paralleling the ROS production profile. TAK1 deficiency in keratinocytes led to increased apoptosis in response to anoikis and TNF-α treatment and was associated with elevated ROS level as analyzed by FACS. Using organotypic skin co-culture and comparative growth factor array analysis, we revealed a cell-autonomous mechanism that involved the SCF/c-Kit/PKBα signaling cascade. Ectopic expression of TAK1 or treatment with exogenous recombinant SCF restored the increased ROS production and apoptotic cell death in TAK1-deficient keratinocytes. Conversely, normal keratinocytes treated with various inhibitors targeting the SCF/c-Kit/PKBα pathway exhibited increased ROS production and TNF-α- or anoikis-induced apoptosis. Our study reveals a novel anti-apoptotic role for SCF in keratinocytes and identifies TAK1 as a novel player uniting inflammation and ROS regulation in skin redox biology.


Assuntos
Apoptose , Queratinócitos/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Células-Tronco/metabolismo , Células Cultivadas , Técnicas de Cocultura , Humanos , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Fator de Células-Tronco/antagonistas & inibidores , Fator de Células-Tronco/genética , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA