Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cancer Res ; 83(12): 2016-2033, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37078793

RESUMO

Cellular components of the tumor microenvironment, including myeloid cells, play important roles in the progression of lung adenocarcinoma (LUAD) and its response to therapy. Here, we characterize the function of the ubiquitin ligases Siah1a/2 in regulating the differentiation and activity of alveolar macrophages (AM) and assess the implication of Siah1a/2 control of AMs for carcinogen-induced LUAD. Macrophage-specific genetic ablation of Siah1a/2 promoted accumulation of AMs with an immature phenotype and increased expression of protumorigenic and pro-inflammatory Stat3 and ß-catenin gene signatures. Administration of urethane to wild-type mice promoted enrichment of immature-like AMs and lung tumor development, which was enhanced by macrophage-specific Siah1a/2 ablation. The profibrotic gene signature seen in Siah1a/2-ablated immature-like macrophages was associated with increased tumor infiltration of CD14+ myeloid cells and poorer survival of patients with LUAD. Single-cell RNA-seq confirmed the presence of a cluster of immature-like AMs expressing a profibrotic signature in lungs of patients with LUAD, a signature enhanced in smokers. These findings identify Siah1a/2 in AMs as gatekeepers of lung cancer development. SIGNIFICANCE: The ubiquitin ligases Siah1a/2 control proinflammatory signaling, differentiation, and profibrotic phenotypes of alveolar macrophages to suppress lung carcinogenesis.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Macrófagos Alveolares , Ubiquitina-Proteína Ligases , Animais , Camundongos , Macrófagos Alveolares/enzimologia , Macrófagos Alveolares/imunologia , Adenocarcinoma de Pulmão/induzido quimicamente , Neoplasias Pulmonares/induzido quimicamente , Microambiente Tumoral , Ubiquitina-Proteína Ligases/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
Mol Cancer Res ; 18(10): 1560-1573, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32571981

RESUMO

Mechanisms regulating nuclear organization control fundamental cellular processes, including the cell and chromatin organization. Their disorganization, including aberrant nuclear architecture, has been often implicated in cellular transformation. Here, we identify Lamin A, among proteins essential for nuclear architecture, as SPANX (sperm protein associated with the nucleus on the X chromosome), a cancer testis antigen previously linked to invasive tumor phenotypes, interacting protein in melanoma. SPANX interaction with Lamin A was mapped to the immunoglobulin fold-like domain, a region critical for Lamin A function, which is often mutated in laminopathies. SPANX downregulation in melanoma cell lines perturbed nuclear organization, decreased cell viability, and promoted senescence-associated phenotypes. Moreover, SPANX knockdown (KD) in melanoma cells promoted proliferation arrest, a phenotype mediated in part by IRF3/IL1A signaling. SPANX KD in melanoma cells also prompted the secretion of IL1A, which attenuated the proliferation of naïve melanoma cells. Identification of SPANX as a nuclear architecture complex component provides an unexpected insight into the regulation of Lamin A and its importance in melanoma. IMPLICATIONS: SPANX, a testis protein, interacts with LMNA and controls nuclear architecture and melanoma growth.


Assuntos
Lamina Tipo A/metabolismo , Laminas/metabolismo , Melanoma/genética , Proteínas Nucleares/genética , Humanos , Melanoma/patologia , Transfecção
3.
Nat Commun ; 11(1): 99, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31911617

RESUMO

Understanding the mechanisms underlying anti-tumor immunity is pivotal for improving immune-based cancer therapies. Here, we report that growth of BRAF-mutant melanoma cells is inhibited, up to complete rejection, in Siah2-/- mice. Growth-inhibited tumors exhibit increased numbers of intra-tumoral activated T cells and decreased expression of Ccl17, Ccl22, and Foxp3. Marked reduction in Treg proliferation and tumor infiltration coincide with G1 arrest in tumor infiltrated Siah2-/- Tregs in vivo or following T cell stimulation in culture, attributed to elevated expression of the cyclin-dependent kinase inhibitor p27, a Siah2 substrate. Growth of anti-PD-1 therapy resistant melanoma is effectively inhibited in Siah2-/- mice subjected to PD-1 blockade, indicating synergy between PD-1 blockade and Siah2 loss. Low SIAH2 and FOXP3 expression is identified in immune responsive human melanoma tumors. Overall, Siah2 regulation of Treg recruitment and cell cycle progression effectively controls melanoma development and Siah2 loss in the host sensitizes melanoma to anti-PD-1 therapy.


Assuntos
Melanoma/imunologia , Proteínas Nucleares/imunologia , Linfócitos T Reguladores/imunologia , Ubiquitina-Proteína Ligases/imunologia , Animais , Quimiocina CCL17/genética , Quimiocina CCL17/imunologia , Quimiocina CCL22/genética , Quimiocina CCL22/imunologia , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/imunologia , Humanos , Melanoma/genética , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Ubiquitina-Proteína Ligases/genética
4.
JCI Insight ; 2(23)2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-29212953

RESUMO

Primary and secondary hypertension are major risk factors for cardiovascular disease, the leading cause of death worldwide. Elevated secretion of aldosterone resulting from primary aldosteronism (PA) is a key driver of secondary hypertension. Here, we report an unexpected role for the ubiquitin ligase Siah1 in adrenal gland development and PA. Siah1a-/- mice exhibit altered adrenal gland morphology, as reflected by a diminished X-zone, enlarged medulla, and dysregulated zonation of the glomerulosa as well as increased aldosterone levels and aldosterone target gene expression and reduced plasma potassium levels. Genes involved in catecholamine biosynthesis and cAMP signaling are upregulated in the adrenal glands of Siah1a-/- mice, while genes related to retinoic acid signaling and cholesterol biosynthesis are downregulated. Loss of Siah1 leads to increased expression of the Siah1 substrate PIAS1, an E3 SUMO protein ligase implicated in the suppression of LXR, a key regulator of cholesterol levels in the adrenal gland. In addition, SIAH1 sequence variants were identified in patients with PA; such variants impaired SIAH1 ubiquitin ligase activity, resulting in elevated PIAS1 expression. These data identify a role for the Siah1-PIAS1 axis in adrenal gland organization and function and point to possible therapeutic targets for hyperaldosteronism.


Assuntos
Glândulas Suprarrenais/metabolismo , Aldosterona/metabolismo , Proteínas/fisiologia , Glândulas Suprarrenais/patologia , Medula Suprarrenal/patologia , Adulto , Animais , Colesterol/biossíntese , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Hiperaldosteronismo/genética , Hiperaldosteronismo/metabolismo , Rim/metabolismo , Masculino , Camundongos Knockout , Pessoa de Meia-Idade , Mutação , Proteínas Nucleares/genética , Potássio/sangue , Proteínas Inibidoras de STAT Ativados/biossíntese , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas/genética , Transdução de Sinais/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Tretinoína/fisiologia , Ubiquitina-Proteína Ligases/genética , Zona Glomerulosa/metabolismo , Zona Glomerulosa/patologia
5.
Cell Rep ; 15(9): 1884-92, 2016 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-27210757

RESUMO

Melanoma is one of the most lethal cutaneous malignancies, characterized by chemoresistance and a striking propensity to metastasize. The transcription factor ATF2 elicits oncogenic activities in melanoma, and its inhibition attenuates melanoma development. Here, we show that expression of a transcriptionally inactive form of Atf2 (Atf2(Δ8,9)) promotes development of melanoma in mouse models. Atf2(Δ8,9)-driven tumors show enhanced pigmentation, immune infiltration, and metastatic propensity. Similar to mouse Atf2(Δ8,9), we have identified a transcriptionally inactive human ATF2 splice variant 5 (ATF2(SV5)) that enhances the growth and migration capacity of cultured melanoma cells and immortalized melanocytes. ATF2(SV5) expression is elevated in human melanoma specimens and is associated with poor prognosis. These findings point to an oncogenic function for ATF2 in melanoma development that appears to be independent of its transcriptional activity.


Assuntos
Fator 2 Ativador da Transcrição/genética , Carcinogênese/genética , Melanoma/genética , Melanoma/patologia , Mutação/genética , Neoplasias Cutâneas/genética , Transcrição Gênica , Processamento Alternativo/genética , Animais , Carcinogênese/patologia , Modelos Animais de Doenças , Humanos , Melanócitos/metabolismo , Melanócitos/patologia , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Nevo/genética , Nevo/patologia , Oncogenes , Proteínas Proto-Oncogênicas B-raf/deficiência , Proteínas Proto-Oncogênicas B-raf/metabolismo , Neoplasias Cutâneas/patologia
6.
Cancer Res ; 75(7): 1399-412, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25712345

RESUMO

Melanoma development involves members of the AGC kinase family, including AKT, PKC, and, most recently, PDK1, as elucidated recently in studies of Braf::Pten mutant melanomas. Here, we report that PDK1 contributes functionally to skin pigmentation and to the development of melanomas harboring a wild-type PTEN genotype, which occurs in about 70% of human melanomas. The PDK1 substrate SGK3 was determined to be an important mediator of PDK1 activities in melanoma cells. Genetic or pharmacologic inhibition of PDK1 and SGK3 attenuated melanoma growth by inducing G1 phase cell-cycle arrest. In a synthetic lethal screen, pan-PI3K inhibition synergized with PDK1 inhibition to suppress melanoma growth, suggesting that focused blockade of PDK1/PI3K signaling might offer a new therapeutic modality for wild-type PTEN tumors. We also noted that responsiveness to PDK1 inhibition associated with decreased expression of pigmentation genes and increased expression of cytokines and inflammatory genes, suggesting a method to stratify patients with melanoma for PDK1-based therapies. Overall, our work highlights the potential significance of PDK1 as a therapeutic target to improve melanoma treatment.


Assuntos
Melanoma/enzimologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias Cutâneas/enzimologia , Animais , Benzoatos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Pontos de Checagem da Fase G1 do Ciclo Celular , Humanos , Proteínas Imediatamente Precoces/metabolismo , Indazóis/farmacologia , Metástase Linfática , Melanoma/tratamento farmacológico , Melanoma/secundário , Camundongos Knockout , Terapia de Alvo Molecular , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Pirimidinas/farmacologia , Piruvato Desidrogenase Quinase de Transferência de Acetil , Pele/enzimologia , Pele/patologia , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia
7.
PLoS Genet ; 10(5): e1004348, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24809345

RESUMO

The endoplasmic reticulum (ER) responds to changes in intracellular homeostasis through activation of the unfolded protein response (UPR). Yet, it is not known how UPR-signaling coordinates adaptation versus cell death. Previous studies suggested that signaling through PERK/ATF4 is required for cell death. We show that high levels of ER stress (i.e., ischemia-like conditions) induce transcription of the ubiquitin ligases Siah1/2 through the UPR transducers PERK/ATF4 and IRE1/sXBP1. In turn, Siah1/2 attenuates proline hydroxylation of ATF4, resulting in its stabilization, thereby augmenting ER stress output. Conversely, ATF4 activation is reduced upon Siah1/2 KD in cultured cells, which attenuates ER stress-induced cell death. Notably, Siah1a(+/-)::Siah2(-/-) mice subjected to neuronal ischemia exhibited smaller infarct volume and were protected from ischemia-induced death, compared with the wild type (WT) mice. In all, Siah1/2 constitutes an obligatory fine-tuning mechanism that predisposes cells to death under severe ER stress conditions.


Assuntos
Isoenzimas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Resposta a Proteínas não Dobradas , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Retículo Endoplasmático/metabolismo , Ativação Enzimática , Humanos , Camundongos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Estresse Fisiológico , Transcrição Gênica , Ubiquitina-Proteína Ligases/genética
8.
Cell Biochem Biophys ; 67(1): 15-24, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23700162

RESUMO

The Siah ubiquitin ligases are members of the RING finger E3 ligases. The Siah E3s are conserved from fly to mammals. Primarily implicated in cellular stress responses, Siah ligases play a key role in hypoxia, through the regulation of HIF-1α transcription stability and activity. Concomitantly, physiological conditions associated with varying oxygen tension often highlight the importance of Siah, as seen in cancer and neurodegenerative disorders. Notably, recent studies also point to the role of these ligases in fundamental processes including DNA damage response, cellular organization and polarity. This review summarizes the current understanding of upstream regulators and downstream effectors of Siah.


Assuntos
Ubiquitina-Proteína Ligases/metabolismo , Animais , Dano ao DNA , Humanos , Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo
9.
Pigment Cell Melanoma Res ; 26(1): 136-42, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23035722

RESUMO

To date, there are no effective therapies for tumors bearing NRAS mutations, which are present in 15-20% of human melanomas. Here we extend our earlier studies where we demonstrated that the small molecule BI-69A11 inhibits the growth of melanoma cell lines. Gene expression analysis revealed the induction of interferon- and cell death-related genes that were associated with responsiveness of melanoma cell lines to BI-69A11. Strikingly, the administration of BI-69A11 inhibited melanoma development in genetically modified mice bearing an inducible form of activated Nras and a deletion of the Ink4a gene (Nras((Q61K)) ::Ink4a(-/-) ). Biweekly administration of BI-69A11 starting at 10 weeks or as late as 24 weeks after the induction of mutant Nras expression inhibited melanoma development (100 and 36%, respectively). BI-69A11 treatment did not inhibit the development of histiocytic sarcomas, which constitute about 50% of the tumors in this model. BI-69A11-resistant Nras((Q61K)) ::Ink4a(-/-) tumors exhibited increased CD45 expression, reflective of immune cell infiltration and upregulation of gene networks associated with the cytoskeleton, DNA damage response, and small molecule transport. The ability to attenuate the development of NRAS mutant melanomas supports further development of BI-69A11 for clinical assessment.


Assuntos
Substituição de Aminoácidos/genética , Benzimidazóis/uso terapêutico , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Melanoma/tratamento farmacológico , Lesões Pré-Cancerosas/tratamento farmacológico , Quinolonas/uso terapêutico , Neoplasias Cutâneas/tratamento farmacológico , Proteínas ras/genética , Animais , Benzimidazóis/administração & dosagem , Benzimidazóis/farmacologia , Proliferação de Células/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Injeções Intraperitoneais , Antígeno Ki-67/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Melanoma/genética , Melanoma/patologia , Camundongos , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Proteínas Proto-Oncogênicas B-raf/genética , Quinolonas/administração & dosagem , Quinolonas/farmacologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Biol Chem ; 286(31): 27333-41, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21659512

RESUMO

The RING finger E3 ubiquitin ligase Siah2 is implicated in control of diverse cellular biological events, including MAPK signaling and hypoxia. Here we demonstrate that Siah2 is subject to regulation by the deubiquitinating enzyme USP13. Overexpression of USP13 increases Siah2 stability by attenuating its autodegradation. Consequently, the ability of Siah2 to target its substrates prolyl hydroxylase 3 and Spry2 (Sprouty2) for ubiquitin-mediated proteasomal degradation is attenuated. Conversely, inhibition of USP13 expression with corresponding shRNA decreases the stability of both Siah2 and its substrate Spry2. Thus, USP13 limits Siah2 autodegradation and its ubiquitin ligase activity against its target substrates. Strikingly, the effect of USP13 on Siah2 is not mediated by its isopeptidase activity: mutations in its ubiquitin-binding sequences positioned within the ubiquitin-specific processing protease and ubiquitin-binding domains, but not within putative catalytic sites, abolish USP13 binding to and effect on Siah2 autodegradation and targeted ubiquitination. Notably, USP13 expression is attenuated in melanoma cells maintained under hypoxia, thereby relieving Siah2 inhibition and increasing its activity under low oxygen levels. Significantly, on melanoma tissue microarray, high nuclear expression of USP13 coincided with high nuclear expression of Siah2. Overall, this study identifies a new layer of Siah2 regulation mediated by USP13 binding to ubiquitinated Siah2 protein with a concomitant inhibitory effect on its activity under normoxia.


Assuntos
Endopeptidases/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Sítios de Ligação , Western Blotting , Linhagem Celular , Endopeptidases/genética , Estabilidade Enzimática , Humanos , Hidrólise , Imunoprecipitação , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase , Proteases Específicas de Ubiquitina
11.
Genes Cancer ; 1(4): 316-330, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20740050

RESUMO

The transcription factor ATF2 was previously shown to be an ATM substrate. Upon phosphorylation by ATM, ATF2 exhibits a transcription-independent function in the DNA damage response through localization to DNA repair foci and control of cell cycle arrest. To assess the physiological significance of this phosphorylation, we generated ATF2 mutant mice in which the ATM phosphoacceptor sites (S472/S480) were mutated (ATF2(KI)). ATF2(KI) mice are more sensitive to ionizing radiation (IR) than wild-type (ATF2 (WT)) mice: following IR, ATF2(KI) mice exhibited higher levels of apoptosis in the intestinal crypt cells and impaired hepatic steatosis. Molecular analysis identified impaired activation of the cell cycle regulatory protein p21(Cip/Waf1) in cells and tissues of IR-treated ATF2(KI) mice, which was p53 independent. Analysis of tumor development in p53(KO) crossed with ATF2(KI) mice indicated a marked decrease in amount of time required for tumor development. Further, when subjected to two-stage skin carcinogenesis process, ATF2(KI) mice developed skin tumors faster and with higher incidence, which also progressed to the more malignant carcinomas, compared with the control mice. Using 3 mouse models, we establish the importance of ATF2 phosphorylation by ATM in the acute cellular response to DNA damage and maintenance of genomic stability.

12.
Cancer Res ; 69(6): 2638-46, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19276359

RESUMO

Hypoxia-inducible factor-1 (HIF-1) is a known cancer progression factor, promoting growth, spread, and metastasis. However, in selected contexts, HIF-1 is a tumor suppressor coordinating hypoxic cell cycle suppression and apoptosis. Prior studies focused on HIF-1 function in established malignancy; however, little is known about its role during the entire process of carcinogenesis from neoplasia induction to malignancy. Here, we tested HIF-1 gain of function during multistage murine skin chemical carcinogenesis in K14-HIF-1alpha(Pro402A564G) (K14-HIF-1alphaDPM) transgenic mice. Transgenic papillomas appeared earlier and were more numerous (6 +/- 3 transgenic versus 2 +/- 1.5 nontransgenic papillomas per mouse), yet they were more differentiated, their proliferation was lower, and their malignant conversion was profoundly inhibited (7% in transgenic versus 40% in nontransgenic mice). Moreover, transgenic cancers maintained squamous differentiation whereas epithelial-mesenchymal transformation was frequent in nontransgenic malignancies. Transgenic basal keratinocytes up-regulated the HIF-1 target N-myc downstream regulated gene-1, a known tumor suppressor gene in human malignancy, and its expression was maintained in transgenic papillomas and cancer. We also discovered a novel HIF-1 target gene, selenium binding protein-1 (Selenbp1), a gene of unknown function whose expression is lost in human cancer. Thus, HIF-1 can function as a tumor suppressor through transactivation of genes that are themselves targets for negative selection in human cancers.


Assuntos
Carcinoma de Células Escamosas/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Cutâneas/genética , 9,10-Dimetil-1,2-benzantraceno , Animais , Carcinógenos , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Progressão da Doença , Células Epiteliais/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas de Ligação a Selênio/biossíntese , Proteínas de Ligação a Selênio/genética , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Acetato de Tetradecanoilforbol , Transfecção
13.
Blood ; 111(7): 3343-54, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18199827

RESUMO

Hypoxia inducible factor-1 (HIF-1) is a master regulatory transcription factor controlling multiple cell-autonomous and non-cell-autonomous processes, such as metabolism, angiogenesis, matrix invasion, and cancer metastasis. Here we used a new line of transgenic mice with constitutive gain of HIF-1 function in basal keratinocytes and demonstrated a signaling pathway from HIF-1 to nuclear factor kappa B (NFkappaB) activation to enhanced epithelial chemokine and cytokine elaboration. This pathway was responsible for a phenotypically silent accumulation of stromal inflammatory cells and a marked inflammatory hypersensitivity to a single 12-O-tetradecanoylphorbol-13-acetate (TPA) challenge. HIF-1-induced NFkappaB activation was composed of 2 elements, IkappaB hyperphosphorylation and phosphorylation of Ser276 on p65, enhancing p65 nuclear localization and transcriptional activity, respectively. NFkappaB transcriptional targets macrophage inflammatory protein-2 (MIP-2/CXCL2/3), keratinocyte chemokine (KC/CXCL1), and tumor necrosis factor [alfa] (TNFalpha) were constitutively up-regulated and further increased after TPA challenge both in cultured keratinocytes and in transgenic mice. Whole animal KC, MIP-2, or TNFalpha immunodepletion each abrogated TPA-induced inflammation, whereas blockade of either VEGF or placenta growth factor (PlGF) signaling did not affect transgenic inflammatory hyper-responsiveness. Thus, epithelial HIF-1 gain of function remodels the local environment by cell-autonomous NFkappaB-mediated chemokine and cytokine secretion, which may be another mechanism by which HIF-1 facilitates either inflammatory diseases or malignant progression.


Assuntos
Núcleo Celular/metabolismo , Hipersensibilidade a Drogas/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Queratinócitos/metabolismo , Comunicação Parácrina , Fator de Transcrição RelA/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/genética , Animais , Carcinógenos/toxicidade , Núcleo Celular/genética , Núcleo Celular/patologia , Citocinas/genética , Citocinas/metabolismo , Hipersensibilidade a Drogas/genética , Hipersensibilidade a Drogas/patologia , Epitélio/metabolismo , Epitélio/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Queratinócitos/patologia , Camundongos , Camundongos Transgênicos , Comunicação Parácrina/efeitos dos fármacos , Comunicação Parácrina/genética , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Células Estromais/metabolismo , Células Estromais/patologia , Acetato de Tetradecanoilforbol/toxicidade , Fator de Transcrição RelA/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
14.
Invest Ophthalmol Vis Sci ; 46(3): 1010-6, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15728559

RESUMO

PURPOSE: To characterize ocular disease in HIF-2alpha-null mice. METHODS: Histologic, electroretinographic (ERG), and molecular studies were performed on samples obtained from age- and gender-matched HIF-2alpha-null (HIF-2alpha-KO), HIF-2alpha-heterozygous (HIF-2alpha-HET), and wild-type (WT) littermate mice. RESULTS: HIF-2alpha-KO mice exhibited marked thinning of the retina and abnormal retinal vasculature. The pathologic changes in HIF-2alpha-KO mice were associated with a virtual absence of postreceptor function. The expression of a surrogate marker for HIF-2alpha mRNA localized to vascular endothelial, amacrine, and retinal pigment epithelial (RPE) cells. Several HIF-2alpha target genes involved in angiogenesis, retinal protection, and stress responses have altered expression patterns in HIF-2alpha-KO retinas. CONCLUSIONS: HIF-2alpha-KO mice exhibit marked retinopathy consistent with complete loss of vision by 1 month of age. Impaired HIF-2alpha signaling in HIF-2alpha-KO mice likely produces functional deficits in cell types in which HIF-2alpha normally is expressed, ultimately resulting in retinopathy. Future studies will address whether the molecular abnormalities described in this study are directly responsible for the retinal disease in HIF-2alpha-KO mice.


Assuntos
Doenças Retinianas/etiologia , Doenças Retinianas/fisiopatologia , Transativadores/deficiência , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Eletrorretinografia , Feminino , Deleção de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora , Retina/fisiologia , Doenças Retinianas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/genética , Transtornos da Visão/etiologia , Transtornos da Visão/fisiopatologia
15.
Blood ; 105(8): 3133-40, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15626745

RESUMO

Erythropoiesis in the adult mammal depends critically on erythropoietin, an inducible cytokine with pluripotent effects. Erythropoietin gene expression increases under conditions associated with lowered oxygen content such as anemia and hypoxia. HIF-1alpha, the founding member of the hypoxia-inducible factor (HIF) alpha class, was identified by its ability to bind and activate the hypoxia-responsive enhancer in the erythropoietin regulatory region in vitro. The existence of multiple HIF alpha members raises the question of which HIF alpha member or members regulates erythropoietin expression in vivo. We previously reported that mice lacking wild-type HIF-2alpha, encoded by the EPAS1 gene, exhibit pancytopenia. In this study, we have characterized the etiology of this hematopoietic phenotype. Molecular studies of EPAS1-null kidneys reveal dramatically decreased erythropoietin gene expression. EPAS1-null as well as heterozygous mice have impaired renal erythropoietin induction in response to hypoxia. Treatment of EPAS1-null mice with exogenous erythropoietin reverses the hematopoietic and other defects. We propose that HIF-2alpha is an essential regulator of murine erythropoietin production. Impairments in HIF signaling, involving either HIF-1alpha or HIF-2alpha, may play a prominent role in conditions involving altered hematopoietic or erythropoietin homeostasis.


Assuntos
Eritropoetina/genética , Hematopoese/fisiologia , Pancitopenia/fisiopatologia , Transativadores/genética , Transativadores/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Feminino , Expressão Gênica , Hipóxia/fisiopatologia , Rim/fisiologia , Masculino , Camundongos , Camundongos Mutantes , Pancitopenia/metabolismo , Fenótipo , RNA Mensageiro/análise
16.
Nat Genet ; 35(4): 331-40, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14608355

RESUMO

Hypoxia-inducible factor (HIF) transcription factors respond to multiple environmental stressors, including hypoxia and hypoglycemia. We report that mice lacking the HIF family member HIF-2alpha (encoded by Epas1) have a syndrome of multiple-organ pathology, biochemical abnormalities and altered gene expression patterns. Histological and ultrastructural analyses showed retinopathy, hepatic steatosis, cardiac hypertrophy, skeletal myopathy, hypocellular bone marrow, azoospermia and mitochondrial abnormalities in these mice. Serum and urine metabolite studies showed hypoglycemia, lactic acidosis, altered Krebs cycle function and dysregulated fatty acid oxidation. Biochemical assays showed enhanced generation of reactive oxygen species (ROS), whereas molecular analyses indicated reduced expression of genes encoding the primary antioxidant enzymes (AOEs). Transfection analyses showed that HIF-2alpha could efficiently transactivate the promoters of the primary AOEs. Prenatal or postnatal treatment of Epas1-/- mice with a superoxide dismutase (SOD) mimetic reversed several aspects of the null phenotype. We propose a rheostat role for HIF-2alpha that allows for the maintenance of ROS as well as mitochondrial homeostasis.


Assuntos
Anormalidades Múltiplas , Homeostase/fisiologia , Proteínas de Neoplasias , Espécies Reativas de Oxigênio , Transativadores/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Hipóxia Celular , Complexo IV da Cadeia de Transporte de Elétrons , Regulação da Expressão Gênica , Coração/fisiologia , Homozigoto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mimetismo Molecular , Músculo Esquelético/ultraestrutura , Estresse Oxidativo , Peroxidases , Peroxirredoxina III , Peroxirredoxinas , Superóxido Dismutase , Superóxidos , Taxa de Sobrevida , Transativadores/deficiência , Transativadores/genética , Transfecção
17.
Blood ; 102(5): 1634-40, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12750163

RESUMO

Hypoxic stress plays a role in pathophysiologic states such as myocardial infarction and cerebral vascular events as well as in normal physiologic conditions including development and hematopoiesis. Members of the hypoxia inducible factor (HIF) family function as transcriptional regulators of genes involved in the hypoxic response. After generating adult mice that globally lack endothelial PAS domain protein 1 (EPAS1, also known as HIF-2alpha/HRF/HLF/MOP3), the second member of the HIF family, characterization of the hematopoietic cell population indicated that the loss of EPAS1/HIF-2alpha resulted in pancytopenia. Using bone marrow reconstitution experiments of lethally irradiated hosts, we have defined the extent and site of hematopoietic impairment in the EPAS1/HIF-2alpha null mice. These data suggest a critical role for EPAS1/HIF-2alpha in maintaining a functional microenvironment in the bone marrow for effective hematopoiesis.


Assuntos
Hematopoese/fisiologia , Transativadores/genética , Transativadores/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Medula Óssea/fisiologia , Transplante de Medula Óssea , Citometria de Fluxo , Hematócrito , Hipóxia/fisiopatologia , Camundongos , Camundongos Endogâmicos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA