Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EMBO Mol Med ; 13(11): e13714, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34661368

RESUMO

Risk stratification of COVID-19 patients is essential for pandemic management. Changes in the cell fitness marker, hFwe-Lose, can precede the host immune response to infection, potentially making such a biomarker an earlier triage tool. Here, we evaluate whether hFwe-Lose gene expression can outperform conventional methods in predicting outcomes (e.g., death and hospitalization) in COVID-19 patients. We performed a post-mortem examination of infected lung tissue in deceased COVID-19 patients to determine hFwe-Lose's biological role in acute lung injury. We then performed an observational study (n = 283) to evaluate whether hFwe-Lose expression (in nasopharyngeal samples) could accurately predict hospitalization or death in COVID-19 patients. In COVID-19 patients with acute lung injury, hFwe-Lose is highly expressed in the lower respiratory tract and is co-localized to areas of cell death. In patients presenting in the early phase of COVID-19 illness, hFwe-Lose expression accurately predicts subsequent hospitalization or death with positive predictive values of 87.8-100% and a negative predictive value of 64.1-93.2%. hFwe-Lose outperforms conventional inflammatory biomarkers and patient age and comorbidities, with an area under the receiver operating characteristic curve (AUROC) 0.93-0.97 in predicting hospitalization/death. Specifically, this is significantly higher than the prognostic value of combining biomarkers (serum ferritin, D-dimer, C-reactive protein, and neutrophil-lymphocyte ratio), patient age and comorbidities (AUROC of 0.67-0.92). The cell fitness marker, hFwe-Lose, accurately predicts outcomes in COVID-19 patients. This finding demonstrates how tissue fitness pathways dictate the response to infection and disease and their utility in managing the current COVID-19 pandemic.


Assuntos
COVID-19 , Biomarcadores , Flores , Humanos , Pandemias , Curva ROC , Estudos Retrospectivos , SARS-CoV-2 , Índice de Gravidade de Doença
2.
Adv Cancer Res ; 148: 171-199, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32723563

RESUMO

Cancer is a complex disease with high incidence and mortality rates. The important role played by the tumor microenvironment in regulating oncogenesis, tumor growth, and metastasis is by now well accepted in the scientific community. SPARC is known to participate in tumor-stromal interactions and impact cancer growth in ambiguous ways, which either enhance or suppress cancer aggressiveness, in a context-dependent manner. p53 transcription factor, a well-established tumor suppressor, has been reported to promote tumor growth in certain situations, such as hypoxia, thus displaying a duality in its action. Although both proteins are being tested in clinical trials, the synergistic relation between them is yet to be explored in clinical practice. In this review, we address the controversial roles of SPARC and p53 as double agents in cancer, briefly summarizing the interaction found between these two molecules and its importance in cancer.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Osteonectina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Humanos , Neoplasias/genética , Osteonectina/genética , Proteína Supressora de Tumor p53/genética
3.
Semin Cancer Biol ; 63: 11-18, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31323289

RESUMO

In many cases, cancers are difficult to eliminate because they develop resistance to a primary chemotherapy or targeted therapy. Tumors grow into diverse cell subpopulations, increasing the ability to resist elimination. The phenomenon of 'cell competition' describes our body's natural surveillance system to optimize tissue fitness by forcing viable but aberrant cells to undergo cell death. Cell competition is not simply comparison of cell division potential. Competition factors signal for 'loser' cell elimination and 'winner' cell dominance. New evidence demonstrates it is possible to restrict cancer growth by strengthening the cell fitness of surrounding healthy tissue via anti-apoptotic pathways. Hence, cell competition provides strong conceptual explanation for oncogenesis, tumor growth and suppression. Tumor heterogeneity is a hallmark of many cancers and establishes gradients in which competitive interactions are able to occur among tumor cell subpopulations as well as neighboring stromal tissue. Here we review cellular/molecular competition pathways in the context of tumor evolution, heterogeneity and response to interventions. We propose strategies to exploit these mediators and design novel broad-spectrum therapeutic approaches that eliminate cancer and enhance fitness of neighboring tissue to improve patient outcomes.


Assuntos
Neoplasias/genética , Neoplasias/patologia , Animais , Comunicação Celular/fisiologia , Evolução Clonal , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Heterogeneidade Genética , Humanos , Neoplasias/etiologia , Neoplasias/metabolismo
4.
Nucleic Acids Res ; 47(19): 10212-10234, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31538203

RESUMO

Chronic hypoxia is associated with a variety of physiological conditions such as rheumatoid arthritis, ischemia/reperfusion injury, stroke, diabetic vasculopathy, epilepsy and cancer. At the molecular level, hypoxia manifests its effects via activation of HIF-dependent transcription. On the other hand, an important transcription factor p53, which controls a myriad of biological functions, is rendered transcriptionally inactive under hypoxic conditions. p53 and HIF-1α are known to share a mysterious relationship and play an ambiguous role in the regulation of hypoxia-induced cellular changes. Here we demonstrate a novel pathway where HIF-1α transcriptionally upregulates both WT and MT p53 by binding to five response elements in p53 promoter. In hypoxic cells, this HIF-1α-induced p53 is transcriptionally inefficient but is abundantly available for protein-protein interactions. Further, both WT and MT p53 proteins bind and chaperone HIF-1α to stabilize its binding at its downstream DNA response elements. This p53-induced chaperoning of HIF-1α increases synthesis of HIF-regulated genes and thus the efficiency of hypoxia-induced molecular changes. This basic biology finding has important implications not only in the design of anti-cancer strategies but also for other physiological conditions where hypoxia results in disease manifestation.


Assuntos
Hipóxia Celular/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Mapas de Interação de Proteínas/genética , Proteína Supressora de Tumor p53/genética , Regulação da Expressão Gênica , Humanos , Chaperonas Moleculares/genética , Regiões Promotoras Genéticas/genética , Elementos de Resposta/genética , Transdução de Sinais/genética
5.
Nature ; 572(7768): 260-264, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31341286

RESUMO

In humans, the adaptive immune system uses the exchange of information between cells to detect and eliminate foreign or damaged cells; however, the removal of unwanted cells does not always require an adaptive immune system1,2. For example, cell selection in Drosophila uses a cell selection mechanism based on 'fitness fingerprints', which allow it to delay ageing3, prevent developmental malformations3,4 and replace old tissues during regeneration5. At the molecular level, these fitness fingerprints consist of combinations of Flower membrane proteins3,4,6. Proteins that indicate reduced fitness are called Flower-Lose, because they are expressed in cells marked to be eliminated6. However, the presence of Flower-Lose isoforms at a cell's membrane does not always lead to elimination, because if neighbouring cells have similar levels of Lose proteins, the cell will not be killed4,6,7. Humans could benefit from the capability to recognize unfit cells, because accumulation of damaged but viable cells during development and ageing causes organ dysfunction and disease8-17. However, in Drosophila this mechanism is hijacked by premalignant cells to gain a competitive growth advantage18. This would be undesirable for humans because it might make tumours more aggressive19-21. It is unknown whether a similar mechanism of cell-fitness comparison is present in humans. Here we show that two human Flower isoforms (hFWE1 and hFWE3) behave as Flower-Lose proteins, whereas the other two isoforms (hFWE2 and hFWE4) behave as Flower-Win proteins. The latter give cells a competitive advantage over cells expressing Lose isoforms, but Lose-expressing cells are not eliminated if their neighbours express similar levels of Lose isoforms; these proteins therefore act as fitness fingerprints. Moreover, human cancer cells show increased Win isoform expression and proliferate in the presence of Lose-expressing stroma, which confers a competitive growth advantage on the cancer cells. Inhibition of the expression of Flower proteins reduces tumour growth and metastasis, and induces sensitivity to chemotherapy. Our results show that ancient mechanisms of cell recognition and selection are active in humans and affect oncogenic growth.


Assuntos
Canais de Cálcio/metabolismo , Proliferação de Células , Proteínas de Drosophila/metabolismo , Neoplasias/patologia , Isoformas de Proteínas/metabolismo , Animais , Canais de Cálcio/genética , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Drosophila melanogaster , Feminino , Técnicas de Silenciamento de Genes , Humanos , Masculino , Metástase Neoplásica , Neoplasias/tratamento farmacológico , Isoformas de Proteínas/genética
6.
J Biol Chem ; 293(12): 4262-4276, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29382728

RESUMO

p53 is an important tumor-suppressor protein that is mutated in more than 50% of cancers. Strategies for restoring normal p53 function are complicated by the oncogenic properties of mutant p53 and have not met with clinical success. To counteract mutant p53 activity, a variety of drugs with the potential to reconvert mutant p53 to an active wildtype form have been developed. However, these drugs are associated with various negative effects such as cellular toxicity, nonspecific binding to other proteins, and inability to induce a wildtype p53 response in cancer tissue. Here, we report on the effects of a curcumin analog, HO-3867, on p53 activity in cancer cells from different origins. We found that HO-3867 covalently binds to mutant p53, initiates a wildtype p53-like anticancer genetic response, is exclusively cytotoxic toward cancer cells, and exhibits high anticancer efficacy in tumor models. In conclusion, HO-3867 is a p53 mutant-reactivating drug with high clinical anticancer potential.


Assuntos
Apoptose/efeitos dos fármacos , Curcumina/análogos & derivados , Proteínas Mutantes/genética , Mutação , Neoplasias/patologia , Piperidonas/farmacologia , Proteína Supressora de Tumor p53/genética , Animais , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Curcumina/farmacologia , Feminino , Humanos , Camundongos , Camundongos Nus , Proteínas Mutantes/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Mol Cell Cardiol ; 98: 1-10, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27374117

RESUMO

Vitamin D deficiency in human subjects is associated with hypertension, metabolic syndrome and related risk factors of cardiovascular diseases. Serum 25-hydroxyvitamin D levels correlate inversely with adiposity in obese and lean individuals. Bioactive vitamin D, or calcitriol, exerts anti-inflammatory effects on adipocytes, preadipocytes and macrophages in vitro. We tested the hypothesis that vitamin D deficiency alters the phenotype of perivascular adipose tissue (PVAT) leading to impaired function in resistance artery. To examine the effects of vitamin D and PVAT on vascular reactivity, myograph experiments were performed on arteries, with or without intact PVAT, from mice maintained on vitamin D-deficient, vitamin D-sufficient or vitamin D-supplemented diet. Systolic blood pressure was significantly increased in mice on vitamin D-deficient diet. Importantly, vitamin D deficiency enhanced angiotensin II-induced vasoconstriction and impaired the normal ability of PVAT to suppress contractile responses of the underlying mesenteric resistance artery to angiotensin II and serotonin. Furthermore, vitamin D deficiency caused upregulation of the mRNA expression of tumor necrosis factor-α, hypoxia-inducible factor-1α and its downstream target lysyl oxidase in mesenteric PVAT. Incubation of mesenteric arteries under hypoxic conditions impaired the anti-contractile effects of intact PVAT on those arteries from mice on vitamin D-sufficient diet. Vitamin D supplementation protected arteries against hypoxia-induced impairment of PVAT function. The protective effects of vitamin D against vascular dysfunction, hypertension and cardiovascular diseases may be mediated, at least in part, through regulation of inflammatory and hypoxia signaling pathways in PVAT.


Assuntos
Tecido Adiposo/metabolismo , Artérias/fisiologia , Hipóxia/metabolismo , Inflamação/metabolismo , Resistência Vascular , Vitamina D/metabolismo , Ração Animal , Animais , Pressão Sanguínea , Feminino , Perfilação da Expressão Gênica , Masculino , Artérias Mesentéricas/fisiologia , Camundongos , Contração Muscular , Fenótipo , Vitamina D/administração & dosagem
8.
Stem Cells Int ; 2015: 498328, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26106428

RESUMO

Coronary artery stenting or angioplasty procedures frequently result in long-term endothelial dysfunction or loss and complications including arterial thrombosis and myocardial infarction. Stem cell-based therapies have been proposed to support endothelial regeneration. Mesenchymal stem cells (MSCs) differentiate into endothelial cells (ECs) in the presence of VEGF-A in vitro. Application of VEGF-A and MSC-derived ECs at the interventional site is a complex clinical challenge. In this study, we examined the effect of atherogenic cytokines (IL-6, TNFα, and Ang II) on EC differentiation and function. MSCs (CD44(+), CD73(+), CD90(+), CD14(-), and CD45(-)) were isolated from the bone marrow of Yucatan microswine. Naïve MSCs cultured in differentiation media containing VEGF-A (50 ng/mL) demonstrated increased expression of EC-specific markers (vWF, PECAM-1, and VE-cadherin), VEGFR-2 and Sox18, and enhanced endothelial tube formation. IL-6 or TNFα caused a dose-dependent attenuation of EC marker expression in VEGF-A-stimulated MSCs. In contrast, Ang II enhanced EC marker expression in VEGF-A-stimulated MSCs. Addition of Ang II to VEGF-A and IL-6 or TNFα was sufficient to rescue the EC phenotype. Thus, Ang II promotes but IL-6 and TNFα inhibit VEGF-A-induced differentiation of MSCs into ECs. These findings have important clinical implications for therapies intended to increase cardiac vascularity and reendothelialize coronary arteries following intervention.

9.
Differentiation ; 89(3-4): 87-96, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25913202

RESUMO

OBJECTIVE: Mesenchymal stem cells (MSC) have shown therapeutic potential to engraft and either differentiate into or support differentiation of vascular endothelial cells (EC), smooth muscle cells and cardiomyocytes in animal models of ischemic heart disease. Following intracoronary or transendocardial delivery of MSCs, however, only a small fraction of cells engraft and the majority of those persist as an immature cell phenotype. The goal of the current study was to decipher the molecular pathways and mechanisms that control MSC differentiation into ECs. Vascular endothelial growth factor (VEGF-165) treatment is known to enhance in vitro differentiation of MSCs into ECs. We tested the possible involvement of the Sry-type HMG box (Sox) family of transcription factors in this process. METHOD AND RESULTS: MSCs were isolated from the bone marrow of Yucatan microswine and underwent a 10 day differentiation protocol. VEGF-165 (50ng/ml) treatment of MSCs in vitro induced a significant increase in the protein expression of VEGFR-2, Sox9 and Sox18, in addition to the EC markers PECAM-1, VE-cadherin and vWF, as determined by Western blot or flow cytometry. siRNA-mediated knockdown of Sox18, as opposed to Sox9, in MSCs prevented VEGF-165-mediated induction of EC markers and capillary tube formation. Inhibition of VEGFR-2 signaling (SC-202850) reduced Sox18 and reduced VEGF-165-induced differentiation of MSCs to ECs. CONCLUSION: Here we demonstrate that VEGF-165 mediates MSC differentiation into ECs via VEGFR-2-dependent induction of Sox18, which ultimately coordinates the transcriptional upregulation of specific markers of the EC phenotype.


Assuntos
Diferenciação Celular/genética , Células-Tronco Mesenquimais/citologia , Fatores de Transcrição SOXF/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Fatores de Transcrição SOXF/genética , Transdução de Sinais , Suínos , Porco Miniatura , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
10.
Stem Cell Res Ther ; 6: 4, 2015 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-25563650

RESUMO

INTRODUCTION: Increased levels of angiotensin II (Ang II) and activity of Ang II receptor type 1 (AT1R) elicit detrimental effects in cardiovascular disease. However, the role of Ang II receptor type 2 (AT2R) remains poorly defined. Mesenchymal stem cells (MSCs) replenish and repair endothelial cells in the cardiovascular system. Herein, we investigated a novel role of angiotensin signaling in enhancing vascular endothelial growth factor (VEGF)-A-mediated differentiation of MSCs into endothelial cells (ECs). METHODS: Bone marrow was aspirated from the femurs of Yucatan microswine. MSCs were extracted via ficoll density centrifugation technique and were strongly immunopositive for MSC markers, CD44, CD90, and CD105, but negative for hematopoietic markers, CD14 and CD45. Subsequently, naïve MSCs were differentiated for 10 days in varying concentrations and combinations of VEGF-A, Ang II, and AT1R or AT2R antagonists. Markers specific to ECs were determined by FACS analysis. RESULTS: AT1R and AT2R expression and cellular localization was demonstrated in MSCs stimulated with VEGF-A and Ang II via quantitative RT-PCR and immunofluorescence, respectively. Differentiation of naïve MSCs in media containing Ang II (2 ng/ml) plus low-dose VEGF-A (2 ng/ml) produced a significantly higher percentage of cells that were positive for expression of EC markers (for example, platelet endothelial cell adhesion molecule, vascular endothelial Cadherin and von Willebrand factor) compared to VEGF-A alone. Ang II alone failed to induce EC marker expression. MSCs differentiated with the combination of Ang II and VEGF-A were capable of forming capillary tubes using an in vitro angiogenesis assay. Induction of EC marker expression was greatly attenuated by co-treatment of Ang II/VEGF-A with the AT2R antagonist PD123319, but not the AT1R antagonist telmisartan. CONCLUSIONS: We report the presence of functional AT2R receptor on porcine bone marrow-derived MSCs, where it positively regulates EC differentiation. These findings have significant implications toward therapeutic approaches based on activation of AT2R, which could be a means to stimulate regeneration of damaged endothelium and prevent vascular thrombosis.


Assuntos
Angiotensina II/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Endoteliais/citologia , Células-Tronco Mesenquimais/citologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Bloqueadores do Receptor Tipo 2 de Angiotensina II/farmacologia , Animais , Antígenos CD/metabolismo , Benzimidazóis/farmacologia , Benzoatos/farmacologia , Células da Medula Óssea/citologia , Caderinas/metabolismo , Sinergismo Farmacológico , Células Endoteliais/metabolismo , Fêmur/citologia , Imidazóis/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Microscopia de Fluorescência , Piridinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/genética , Receptor Tipo 2 de Angiotensina/metabolismo , Suínos , Telmisartan , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética , Fator de von Willebrand/metabolismo
11.
Physiol Rep ; 3(1)2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25602015

RESUMO

Preeclampsia is a cardiovascular disorder of late pregnancy that is, commonly characterized by hypertension, renal structural damage and dysfunction, and fetal growth restriction. Prevailing etiologic models of this disorder include T-cell dysfunction as an initiating cause of preeclampsia. Indoleamine 2,3-dioxygenase (IDO), an enzyme that mediates the conversion of tryptophan to kynurenine, has been linked to preeclampsia in humans, and is known to regulate T-cell activity and an endothelial-derived relaxing factor. To test the hypothesis that IDO is causally involved in the pathogenesis of preeclampsia, mice deficient for IDO (IDO-KO) were generated on a C57BL/6 background. IDO-KO and wild-type C57BL/6 mice were bred, and preeclampsia phenotypes were evaluated during pregnancy. Pregnant IDO-KO mice exhibited pathognomonic renal glomerular endotheliosis, proteinuria, pregnancy-specific endothelial dysfunction, intrauterine growth restriction, and mildly elevated blood pressure compared to wild-type mice. Together these findings highlight an important role for IDO in the generation of phenotypes typical of preeclampsia. Loss of IDO function may represent a risk factor for the development of preeclampsia. By extension, increased IDO activity, reductions in IDO reactants, or increases in IDO products may represent novel therapeutic approaches for this disorder.

12.
Hypertension ; 65(2): 345-51, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25385762

RESUMO

Myogenic responses by resistance vessels are a key component of autoregulation in brain, thus playing a crucial role in regulating cerebral blood flow and protecting the blood-brain barrier against potentially detrimental elevations in blood pressure. Although cerebrovascular disease is often accompanied by alterations in myogenic responses, mechanisms that control these changes are poorly understood. Peroxisome proliferator-activated receptor γ has emerged as a regulator of vascular tone. We hypothesized that interference with peroxisome proliferator-activated receptor γ in smooth muscle would augment myogenic responses in cerebral arteries. We studied transgenic mice expressing a dominant-negative mutation in peroxisome proliferator-activated receptor γ selectively in smooth muscle (S-P467L) and nontransgenic littermates. Myogenic tone in middle cerebral arteries from S-P467L was elevated 3-fold when compared with nontransgenic littermates. Rho kinase is thought to play a major role in cerebrovascular disease. The Rho kinase inhibitor, Y-27632, abolished augmented myogenic tone in middle cerebral arteries from S-P467L mice. CN-03, which modifies RhoA making it constitutively active, elevated myogenic tone to ≈60% in both strains, via a Y-27632-dependent mechanism. Large conductance Ca(2+)-activated K(+) channels (BKCa) modulate myogenic tone. Inhibitors of BKCa caused greater constriction in middle cerebral arteries from nontransgenic littermates when compared with S-P467L. Expression of RhoA or Rho kinase-I/II protein was similar in cerebral arteries from S-P467L mice. Overall, the data suggest that peroxisome proliferator-activated receptor γ in smooth muscle normally inhibits Rho kinase and promotes BKCa function, thus influencing myogenic tone in resistance arteries in brain. These findings have implications for mechanisms that underlie large- and small-vessel disease in brain, as well as regulation of cerebral blood flow.


Assuntos
Circulação Cerebrovascular/fisiologia , Artéria Cerebral Média/fisiologia , Músculo Liso Vascular/fisiologia , PPAR gama/deficiência , Vasoconstrição/fisiologia , Animais , Circulação Cerebrovascular/efeitos dos fármacos , Acetato de Desoxicorticosterona/toxicidade , Indução Enzimática , Perfilação da Expressão Gênica , Genes Dominantes , Hipertensão/induzido quimicamente , Hipertensão/genética , Hipertensão/fisiopatologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/efeitos dos fármacos , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/fisiologia , Camundongos , Camundongos Knockout , Artéria Cerebral Média/efeitos dos fármacos , Artéria Cerebral Média/enzimologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/enzimologia , PPAR gama/efeitos dos fármacos , PPAR gama/genética , PPAR gama/fisiologia , Cloreto de Sódio/toxicidade , Tetraetilamônio/farmacologia , Proteínas rho de Ligação ao GTP/biossíntese , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/fisiologia , Quinases Associadas a rho/biossíntese , Quinases Associadas a rho/genética , Quinases Associadas a rho/fisiologia , Proteína rhoA de Ligação ao GTP
13.
Expert Opin Ther Pat ; 24(12): 1383-95, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25363248

RESUMO

INTRODUCTION: Triggering receptor expressed on myeloid cells (TREM) receptors and TREM-like transcript (TLT; or TREML) receptors of the immunoglobulin superfamily are known as key modulators of host immune responses. TREM-1 (CD354) and TREM-2 share the transmembrane adaptor DNAX-activation protein of 12 kDa (DAP12), but they possess separate stimulatory and inhibitory functional roles, especially in myeloid cells. AREAS COVERED: This review covers findings related to TREMs and TLTs published in patent applications from their discovery in 2000 to the present. New roles for TREM-1, TREM-2, TLT-1 and TLT-2 in maladies ranging from acute and chronic inflammatory disorders to cardiovascular diseases and cancers are discussed. Putative endogenous ligands and novel synthetic peptide blockers are also discussed. EXPERT OPINION: So far, therapeutic use of activators/blockers specific for TREMs and TLTs has been limited to preclinical animal models. TREM-1 is an immediate therapeutic target for acute and chronic inflammatory conditions, especially sepsis. Certain mutations in DAP12 and TREM-2 manifest into a disorder named polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy, and newly identified TREM-2 variants confer a significant increase in risk of developing Alzheimer's disease. This makes TREM-2 an attractive therapeutic target for neurodegenerative diseases.


Assuntos
Glicoproteínas de Membrana/efeitos dos fármacos , Patentes como Assunto , Receptores Imunológicos/efeitos dos fármacos , Animais , Humanos , Glicoproteínas de Membrana/fisiologia , Receptores Imunológicos/fisiologia , Receptor Gatilho 1 Expresso em Células Mieloides
14.
Hypertension ; 64(5): 1088-93, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25185134

RESUMO

Although peroxisome proliferator-activated receptor-γ (PPARγ) is thought to play a protective role in the vasculature, its cell-specific effect, particularly in resistance vessels, is poorly defined. Nitric oxide (NO) plays a major role in vascular biology in the brain. We examined the hypothesis that selective interference with PPARγ in vascular muscle would impair NO-dependent responses and augment vasoconstrictor responses in the cerebral circulation. We studied mice expressing a dominant negative mutation in human PPARγ (P467L) under the control of the smooth muscle myosin heavy chain promoter (S-P467L). In S-P467L mice, dilator responses to exogenously applied or endogenously produced NO were greatly impaired in cerebral arteries in vitro and in small cerebral arterioles in vivo. Select NO-independent responses, including vasodilation to low concentrations of potassium, were also impaired in S-P467L mice. In contrast, increased expression of wild-type PPARγ in smooth muscle had little effect on vasomotor responses. Mechanisms underlying impairment of both NO-dependent and NO-independent vasodilator responses after interference with PPARγ involved Rho kinase with no apparent contribution by oxidative stress-related mechanisms. These findings support the concept that via effects on Rho kinase-dependent signaling, PPARγ in vascular muscle is a major determinant of vascular tone in resistance vessels and, in particular, NO-mediated signaling in cerebral arteries and brain microvessels. Considering the importance of NO and Rho kinase, these findings have implications for regulation of cerebral blood flow and the pathogenesis of large and small vessel disease in brain.


Assuntos
Circulação Cerebrovascular/fisiologia , Músculo Liso Vascular/fisiologia , PPAR gama/fisiologia , Animais , Arteríolas/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Óxido Nítrico/metabolismo , PPAR gama/genética , Transdução de Sinais/fisiologia , Vasodilatação/fisiologia , Quinases Associadas a rho/fisiologia
15.
Expert Rev Clin Immunol ; 10(2): 243-56, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24325404

RESUMO

Innate immune receptors represent important therapeutic targets for inflammatory disorders. In particular, the Toll-like receptor (TLR) family has emerged as a promoter of chronic inflammation that contributes to obesity, insulin resistance and atherosclerosis. Importantly, triggering receptor expressed on myeloid cells-1 (TREM-1) has been characterized as an 'amplifier' of TLR2 and TLR4 signaling. TREM-1- and TREM-2-dependent signaling, as opposed to TREM-like transcript-1 (TLT-1 or TREML1), are mediated through association with the transmembrane adaptor DNAX activation protein of 12 kDa (DAP12). Recessive inheritance of rare mutations in DAP12 or TREM-2 results in a disorder called polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy, and surprisingly these subjects are not immunocompromised. Recent progress into the roles of TREM/DAP12 signaling is critically reviewed here with a focus on metabolic, cardiovascular and inflammatory diseases. The expanding repertoire of putative ligands for TREM receptors is also discussed.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doenças do Sistema Imunitário/imunologia , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Predisposição Genética para Doença , Humanos , Imunidade Inata/genética , Inflamação/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Proteínas de Membrana/genética , Polimorfismo Genético , Receptor Cross-Talk , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Transdução de Sinais , Receptor Gatilho 1 Expresso em Células Mieloides
16.
Biochim Biophys Acta ; 1829(12): 1257-1265, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24184271

RESUMO

Hereditary hemochromatosis (HH) is a common autosomal recessive disorder of iron overload among Caucasians of northern European descent. Over 85% of all cases with HH are due to mutations in the hemochromatosis protein (HFE) involved in iron metabolism. Although the importance in iron homeostasis is well recognized, the mechanism of sensing and regulating iron absorption by HFE, especially in the absence of iron response element in its gene, is not fully understood. In this report, we have identified an inverted repeat sequence (ATGGTcttACCTA) within 1700bp (-1675/+35) of the HFE promoter capable to form cruciform structure that binds PARP1 and strongly represses HFE promoter. Knockdown of PARP1 increases HFE mRNA and protein. Similarly, hemin or FeCl3 treatments resulted in increase in HFE expression by reducing nuclear PARP1 pool via its apoptosis induced cleavage, leading to upregulation of the iron regulatory hormone hepcidin mRNA. Thus, PARP1 binding to the inverted repeat sequence on the HFE promoter may serve as a novel iron sensing mechanism as increased iron level can trigger PARP1 cleavage and relief of HFE transcriptional repression.


Assuntos
Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/genética , Sequências Repetidas Invertidas , Proteínas de Membrana/genética , Poli(ADP-Ribose) Polimerases/fisiologia , Regiões Promotoras Genéticas/fisiologia , Transcrição Gênica , Western Blotting , Cloretos/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética , Compostos Férricos/farmacologia , Células HCT116 , Células HEK293 , Células HeLa , Proteína da Hemocromatose , Células Hep G2 , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Luciferases/metabolismo , Proteínas de Membrana/metabolismo , Noxas/farmacologia , Fragmentos de Peptídeos/metabolismo , Poli(ADP-Ribose) Polimerase-1 , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Am J Physiol Regul Integr Comp Physiol ; 304(9): R690-701, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23447133

RESUMO

Agonists of the nuclear hormone receptor peroxisome proliferator-activated receptor γ (PPARγ) have potent insulin-sensitizing effects and inhibit atherosclerosis progression in patients with Type II diabetes. Conversely, missense mutations in the ligand-binding domain of PPARγ that render the transcription factor dominant negative (DN) cause early-onset hypertension and Type II diabetes. We tested the hypothesis that DN PPARγ-mediated interference of endogenous wild-type PPARγ in the endothelium and vascular smooth muscle exacerbates atherosclerosis in apolipoprotein E-deficient (ApoE(-/-)) mice. Endothelium-specific expression of DN PPARγ on the ApoE(-/-) background unmasked significant impairment of endothelium-dependent relaxation in aortic rings, increased systolic blood pressure, altered expression of atherogenic markers (e.g., Cd36, Mcp1, Catalase), and enhanced diet-induced atherosclerotic lesion formation in aorta. Smooth muscle-specific expression of DN PPARγ, which induces aortic dysfunction and increased systolic blood pressure at baseline, also resulted in enhanced diet-induced atherosclerotic lesion formation in aorta on the ApoE(-/-) background that was associated with altered expression of a shared, yet distinct, set of atherogenic markers (e.g., Cd36, Mcp1, Osteopontin, Vcam1). In particular, induction of Osteopontin expression by smooth muscle-specific DN PPARγ correlated with increased plaque calcification. These data demonstrate that inhibition of PPARγ function specifically in the vascular endothelium or smooth muscle may contribute to cardiovascular disease.


Assuntos
Aterosclerose/genética , Aterosclerose/fisiopatologia , Endotélio Vascular/fisiopatologia , Hipertensão/genética , Hipertensão/fisiopatologia , Músculo Liso Vascular/fisiopatologia , PPAR gama/genética , PPAR gama/fisiologia , Doenças Vasculares/genética , Doenças Vasculares/fisiopatologia , Acetilcolina/farmacologia , Animais , Aorta Torácica/patologia , Aorta Torácica/fisiopatologia , Apolipoproteínas E/genética , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Dieta , Progressão da Doença , Genes Dominantes/genética , Genes Dominantes/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miografia , Nitroprussiato/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Vasodilatadores/farmacologia
18.
Am J Physiol Regul Integr Comp Physiol ; 304(10): R818-28, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23535460

RESUMO

An indispensable role for the brain renin-angiotensin system (RAS) has been documented in most experimental animal models of hypertension. To identify the specific efferent pathway activated by the brain RAS that mediates hypertension, we examined the hypothesis that elevated arginine vasopressin (AVP) release is necessary for hypertension in a double-transgenic model of brain-specific RAS hyperactivity (the "sRA" mouse model). sRA mice experience elevated brain RAS activity due to human angiotensinogen expression plus neuron-specific human renin expression. Total daily loss of the 4-kDa AVP prosegment (copeptin) into urine was grossly elevated (≥8-fold). Immunohistochemical staining for AVP was increased in the supraoptic nucleus of sRA mice (~2-fold), but no quantitative difference in the paraventricular nucleus was observed. Chronic subcutaneous infusion of a nonselective AVP receptor antagonist conivaptan (YM-087, Vaprisol, 22 ng/h) or the V(2)-selective antagonist tolvaptan (OPC-41061, 22 ng/h) resulted in normalization of the baseline (~15 mmHg) hypertension in sRA mice. Abdominal aortas and second-order mesenteric arteries displayed AVP-specific desensitization, with minor or no changes in responses to phenylephrine and endothelin-1. Mesenteric arteries exhibited substantial reductions in V(1A) receptor mRNA, but no significant changes in V(2) receptor expression in kidney were observed. Chronic tolvaptan infusion also normalized the (5 mmol/l) hyponatremia of sRA mice. Together, these data support a major role for vasopressin in the hypertension of mice with brain-specific hyperactivity of the RAS and suggest a primary role of V(2) receptors.


Assuntos
Pressão Sanguínea/fisiologia , Encéfalo/metabolismo , Hipertensão/metabolismo , Sistema Renina-Angiotensina/fisiologia , Vasopressinas/metabolismo , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos , Benzazepinas/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Hipertensão/genética , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Camundongos , Camundongos Transgênicos , Receptores de Vasopressinas/genética , Receptores de Vasopressinas/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Tolvaptan , Vasopressinas/genética
19.
Cell Metab ; 16(4): 462-72, 2012 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-23040068

RESUMO

Dominant-negative (DN) mutations in the nuclear hormone receptor peroxisome proliferator-activated receptor-γ (PPARγ) cause hypertension by an unknown mechanism. Hypertension and vascular dysfunction are recapitulated by expression of DN PPARγ specifically in vascular smooth muscle of transgenic mice. DN PPARγ increases RhoA and Rho-kinase activity, and inhibition of Rho-kinase restores normal reactivity and reduces arterial pressure. RhoBTB1, a component of the Cullin-3 RING E3 ubiquitin ligase complex, is a PPARγ target gene. Decreased RhoBTB1, Cullin-3, and neddylated Cullin-3 correlated with increased levels of the Cullin-3 substrate RhoA. Knockdown of Cullin-3 or inhibition of cullin-RING ligase activity in aortic smooth muscle cells increased RhoA. Cullin-RING ligase inhibition enhanced agonist-mediated contraction in aortic rings from normal mice by a Rho-kinase-dependent mechanism, and it increased arterial pressure in vivo. We conclude that Cullin-3 regulates vascular function and arterial pressure, thus providing a mechanistic link between mutations in Cullin-3 and hypertension in humans.


Assuntos
Proteínas Culina/metabolismo , Músculo Liso Vascular/metabolismo , PPAR gama/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Pressão Sanguínea/fisiologia , Proteínas Culina/antagonistas & inibidores , Proteínas Culina/genética , Células HEK293 , Humanos , Camundongos , Camundongos Transgênicos , PPAR gama/antagonistas & inibidores , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transfecção
20.
Circ Res ; 111(11): 1446-58, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22962432

RESUMO

RATIONALE: Activation of peroxisome proliferator-activated receptor-γ (PPARγ) by thiazolidinediones lowers blood pressure, whereas PPARγ mutations cause hypertension. Previous studies suggest these effects may be mediated through the vasculature, but the underlying mechanisms remain unclear. OBJECTIVE: To identify PPARγ mechanisms and transcriptional targets in vascular smooth muscle and their role in regulating resistance artery tone. METHODS AND RESULTS: We studied mesenteric artery (MA) from transgenic mice expressing dominant-negative (DN) mutant PPARγ driven by a smooth muscle cell-specific promoter. MA from transgenic mice exhibited a robust increase in myogenic tone. Patch clamp analysis revealed a reduced large conductance Ca(2+)-activated K(+) (BKCa) current in freshly dissociated smooth muscle cell from transgenic MA. Inhibition of protein kinase C corrected both enhanced myogenic constriction and impaired the large conductance Ca(2+)-activated K(+) channel function. Gene expression profiling revealed a marked loss of the regulator of G protein signaling 5 (RGS5) mRNA in transgenic MA, which was accompanied by a substantial increase in angiotensin II-induced constriction in MA. Small interfering RNA targeting RGS5 caused augmented myogenic tone in intact mesenteric arteries and increased activation of protein kinase C in smooth muscle cell cultures. PPARγ and PPARδ each bind to a PPAR response element close to the RGS5 promoter. RGS5 expression in nontransgenic MA was induced after activation of either PPARγ or PPARδ, an effect that was markedly blunted by DN PPARγ. CONCLUSIONS: We conclude that RGS5 in smooth muscle is a PPARγ and PPARδ target, which when activated blunts angiotensin II-mediated activation of protein kinase C, and preserves the large conductance Ca(2+)-activated K(+) channel activity, thus providing tight control of myogenic tone in the microcirculation.


Assuntos
Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/fisiologia , Artérias Mesentéricas/fisiologia , PPAR gama/fisiologia , Proteína Quinase C/metabolismo , Proteínas RGS/metabolismo , Angiotensina II/farmacologia , Animais , Western Blotting , Feminino , Perfilação da Expressão Gênica , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Masculino , Artérias Mesentéricas/citologia , Artérias Mesentéricas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Mutação , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Naftalenos/farmacologia , PPAR gama/genética , PPAR gama/metabolismo , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteínas RGS/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tetraetilamônio/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...