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











Base de dados
Intervalo de ano de publicação
1.
Cytokine ; 176: 156514, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38277928

RESUMO

BACKGROUND: Inflammation is linked to coronavirus disease 2019 (COVID-19)-related heart failure (HF), but the specific mechanisms are unclear. This study aimed to assess the relationship between specific inflammatory factors, such as interleukin (IL)-1ß, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, IL-17, interferon (IFN)-α, and IFN-γ, and COVID-19-related HF. METHODS: We retrospectively identified 212 adult patients with COVID-19 who were hospitalized at Shanghai Public Health Center from March 1 to May 30, 2022 (including 80 patients with HF and 132 without HF). High-sensitivity C-reactive protein (hs-CRP), procalcitonin (PCT), and inflammatory factors, including IL-1ß, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, IL-17, IFN-α, and IFN-γ, were compared between patients with COVID-19 with and without HF. RESULTS: Patients with COVID-19 having and not having HF differed with regard to sex, age, hs-CRP, PCT, and IL-6 levels (p < 0.05). Logistic regression analysis indicated a significant positive association between IL and 6 and HF (odds ratio = 1.055; 95 % confidence interval: 1.019-1.093, p < 0.005). Sex, age, and hs-CRP were also associated with HF. Women had a greater risk of HF than men. Older age, higher levels of hs-CRP, and IL-6 were associated with a greater risk of HF. CONCLUSIONS: In patients with COVID-19, increased IL-6 levels are significantly associated with COVID-19-related HF.


Assuntos
COVID-19 , Insuficiência Cardíaca , Adulto , Feminino , Humanos , Masculino , Proteína C-Reativa/metabolismo , China , COVID-19/complicações , Interleucina-10 , Interleucina-12 , Interleucina-17 , Interleucina-2 , Interleucina-4 , Interleucina-5 , Interleucina-6 , Interleucina-8 , Estudos Retrospectivos
2.
Nat Cancer ; 5(4): 572-589, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38291304

RESUMO

Acquired drug resistance is a major challenge for cancer therapy and is the leading cause of cancer mortality; however, the mechanisms of drug resistance are diverse and the strategy to specifically target drug-resistant cancer cells remains an unmet clinical issue. Here, we established a colorectal cancer-derived organoid biobank and induced acquired drug resistance by repeated low-level exposures of chemo-agents. Chemosensitivity profiling and transcriptomic analysis studies revealed that chemoresistant cancer-derived organoids exhibited elevated expression of LGR4 and activation of the Wnt signaling pathway. Further, we generated a monoclonal antibody (LGR4-mAb) that potently inhibited LGR4-Wnt signaling and found that treatment with LGR4-mAb notably sensitized drug-induced ferroptosis. Mechanistically, LGR4-dependent Wnt signaling transcriptionally upregulated SLC7A11, a key inhibitor of ferroptosis, to confer acquired drug resistance. Our findings reveal that targeting of Wnt signaling by LGR4-mAb augments ferroptosis when co-administrated with chemotherapeutic agents, demonstrating a potential opportunity to fight refractory and recurrent cancers.


Assuntos
Neoplasias Colorretais , Resistencia a Medicamentos Antineoplásicos , Ferroptose , Receptores Acoplados a Proteínas G , Animais , Humanos , Camundongos , Sistema y+ de Transporte de Aminoácidos/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ferroptose/efeitos dos fármacos , Organoides/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Via de Sinalização Wnt/efeitos dos fármacos
3.
Sci Total Environ ; 904: 166667, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37652374

RESUMO

Lead (Pb) is widely used in industrial and daily-use consumer products. Early-life exposure may increase the risk of lead-related heart problems in childhood. However, the effects of early-life lead exposure on fetal heart development and long-term cardiac outcomes are unknown. In this study, pregnant ICR mice were exposed to lead acetate trihydrate (50 mg/kg/d) via oral gavage from gestation day 1.5 until offspring weaning. Thereafter, the second hit model was established, two groups of offspring (4 weeks old) were either administered sterile saline or Angiotensin II (Ang II) for 4 weeks until euthanasia. We investigated lead-induced offspring heart damage from embryonic period to adulthood by echocardiographic analysis, pathological H&E staining, and ultrastructural examination, as well as mitochondrial function detection. The results showed early-life lead exposure predisposed offspring mice to decreased ejection fraction, increased left ventricular volume, accompanied by hypertrophy and dilation, cardiomyocyte sarcomere dysplasia, abnormal mitochondrial structure, mitochondrial dysfunction, and decreased expression of key sarcomeric and mitochondrial genes, rendering them more susceptible to cardiac hypertrophy, vascular wall thickening, cardiac fibrosis, apoptosis, and heart failure induced by Ang II infusion. This study elucidates early-life low dose lead exposure compromises cardiac development and exacerbates second hit-induced cardiac pathological responses in adulthood, which furnishes crucial scientific evidence pertaining to the cardiac toxicity and risk evaluation associated with early-life exposure to lead.


Assuntos
Cardiomegalia , Chumbo , Humanos , Gravidez , Feminino , Camundongos , Animais , Chumbo/toxicidade , Chumbo/metabolismo , Camundongos Endogâmicos ICR , Cardiomegalia/induzido quimicamente , Cardiomegalia/genética , Cardiomegalia/metabolismo , Miócitos Cardíacos , Pressão Sanguínea , Angiotensina II/farmacologia , Angiotensina II/toxicidade
4.
Elife ; 112022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35913115

RESUMO

DBC1 has been characterized as a key regulator of physiological and pathophysiological activities, such as DNA damage, senescence, and tumorigenesis. However, the mechanism by which the functional stability of DBC1 is regulated has yet to be elucidated. Here, we report that the ubiquitination-mediated degradation of DBC1 is regulated by the E3 ubiquitin ligase SIAH2 and deubiquitinase OTUD5 under hypoxic stress. Mechanistically, hypoxia promoted DBC1 to interact with SIAH2 but not OTUD5, resulting in the ubiquitination and subsequent degradation of DBC1 through the ubiquitin-proteasome pathway. SIAH2 knockout inhibited tumor cell proliferation and migration, which could be rescued by double knockout of SIAH2/CCAR2. Human tissue microarray analysis further revealed that the SIAH2/DBC1 axis was responsible for tumor progression under hypoxic stress. These findings define a key role of the hypoxia-mediated SIAH2-DBC1 pathway in the progression of human breast cancer and provide novel insights into the metastatic mechanism of breast cancer.


Assuntos
Neoplasias da Mama , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Hipóxia/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
5.
JCI Insight ; 7(3)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35132957

RESUMO

Takotsubo syndrome (TTS) is an acute, stress-induced cardiomyopathy that occurs predominantly in women after extreme physical and/or emotional stress. To date, our understanding of the molecular basis for TTS remains unknown and, consequently, specific therapies are lacking. Myocardial infiltration of monocytes and macrophages in TTS has been documented in clinical studies. However, the functional importance of these findings remains poorly understood. Here, we show that a single high dose of isoproterenol (ISO) in mice induced a TTS-like cardiomyopathy phenotype characterized by female predominance, severe cardiac dysfunction, and robust myocardial infiltration of macrophages. Single-cell RNA-Seq studies of myocardial immune cells revealed that TTS-like cardiomyopathy is associated with complex activation of innate and adaptive immune cells in the heart, and macrophages were identified as the dominant immune cells. Global macrophage depletion (via clodronate liposome administration) or blockade of macrophage infiltration (via a CCR2 antagonist or in CCR2-KO mice) resulted in recovery of cardiac dysfunction in ISO-challenged mice. In addition, damping myeloid cell activation by HIF1α deficiency or exposure to the immunomodulatory agent bortezomib ameliorated ISO-induced cardiac dysfunction. Collectively, our findings identify macrophages as a critical regulator of TTS pathogenesis that can be targeted for therapeutic gain.


Assuntos
Cardiomiopatias/genética , Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Macrófagos/patologia , Miócitos Cardíacos/patologia , Cardiomiopatia de Takotsubo/genética , Animais , Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Modelos Animais de Doenças , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA/genética , RNA/metabolismo , Cardiomiopatia de Takotsubo/complicações , Cardiomiopatia de Takotsubo/patologia
6.
JCI Insight ; 7(3)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-34914637

RESUMO

Systemic hypoxia is characterized by peripheral vasodilation and pulmonary vasoconstriction. However, the system-wide mechanism for signaling hypoxia remains unknown. Accumulating evidence suggests that hemoglobin (Hb) in RBCs may serve as an O2 sensor and O2-responsive NO signal transducer to regulate systemic and pulmonary vascular tone, but this remains unexamined at the integrated system level. One residue invariant in mammalian Hbs, ß-globin cysteine93 (ßCys93), carries NO as vasorelaxant S-nitrosothiol (SNO) to autoregulate blood flow during O2 delivery. ßCys93Ala mutant mice thus exhibit systemic hypoxia despite transporting O2 normally. Here, we show that ßCys93Ala mutant mice had reduced S-nitrosohemoglobin (SNO-Hb) at baseline and upon targeted SNO repletion and that hypoxic vasodilation by RBCs was impaired in vitro and in vivo, recapitulating hypoxic pathophysiology. Notably, ßCys93Ala mutant mice showed marked impairment of hypoxic peripheral vasodilation and developed signs of pulmonary hypertension with age. Mutant mice also died prematurely with cor pulmonale (pulmonary hypertension with right ventricular dysfunction) when living under low O2. Altogether, we identify a major role for RBC SNO in clinically relevant vasodilatory responses attributed previously to endothelial NO. We conclude that SNO-Hb transduces the integrated, system-wide response to hypoxia in the mammalian respiratory cycle, expanding a core physiological principle.


Assuntos
Cistatina C/genética , DNA/genética , Hemoglobinas/metabolismo , Hipertensão Pulmonar/genética , Hipóxia/complicações , Mutação , Vasodilatação/fisiologia , Animais , Cistatina C/metabolismo , Análise Mutacional de DNA , Modelos Animais de Doenças , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/fisiopatologia , Hipóxia/genética , Hipóxia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes
7.
Antioxid Redox Signal ; 32(12): 906-927, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-31969001

RESUMO

Significance: Mitochondria are the cellular powerhouses for ATP synthesis through oxidative phosphorylation, and the centers for fatty acid ß-oxidation, metabolite synthesis, reactive oxygen species production, innate immunity, and apoptosis. To fulfill these critical functions, mitochondrial quality and homeostasis must be well maintained. Abnormal mitochondrial quality contributes to aging and age-related disorders, such as metabolic syndrome, cancers, and neurodegenerative diseases. Recent Advances: Mitophagy is a cellular process that selectively removes damaged or superfluous mitochondria by autolysosomal degradation and is regarded as one of the major mechanisms responsible for mitochondrial quality control. Critical Issues: To date, distinct mitophagy pathways have been discovered, including receptor-mediated mitophagy and ubiquitin-dependent mitophagy. Emerging knowledge of these pathways shows that they play important roles in sensing mitochondrial stress and signaling for metabolic adaptations. Future Directions: Here, we provide a review on the molecular mechanisms for mitophagy and its interplay with cellular metabolism, with a particular focus on its role in metabolic and age-related disorders.


Assuntos
Envelhecimento/metabolismo , Síndrome Metabólica/metabolismo , Mitocôndrias/metabolismo , Mitofagia , Neoplasias/metabolismo , Doenças Neurodegenerativas/metabolismo , Envelhecimento/patologia , Animais , Senescência Celular , Humanos , Síndrome Metabólica/patologia , Mitocôndrias/patologia , Neoplasias/patologia , Doenças Neurodegenerativas/patologia
8.
Blood Adv ; 3(15): 2342-2354, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31391167

RESUMO

Platelet activation requires fully functional mitochondria, which provide a vital energy source and control the life span of platelets. Previous reports have shown that both general autophagy and selective mitophagy are critical for platelet function. However, the underlying mechanisms remain incompletely understood. Here, we show that Nix, a previously characterized mitophagy receptor that plays a role in red blood cell maturation, also mediates mitophagy in platelets. Genetic ablation of Nix impairs mitochondrial quality, platelet activation, and FeCl3-induced carotid arterial thrombosis without affecting the expression of platelet glycoproteins (GPs) such as GPIb, GPVI, and αIIbß3 Metabolic analysis revealed decreased mitochondrial membrane potential, enhanced mitochondrial reactive oxygen species level, diminished oxygen consumption rate, and compromised adenosine triphosphate production in Nix -/- platelets. Transplantation of wild-type (WT) bone marrow cells or transfusion of WT platelets into Nix-deficient mice rescued defects in platelet function and thrombosis, suggesting a platelet-autonomous role (acting on platelets, but not other cells) of Nix in platelet activation. Interestingly, loss of Nix increases the life span of platelets in vivo, likely through preventing autophagic degradation of the mitochondrial protein Bcl-xL. Collectively, our findings reveal a novel mechanistic link between Nix-mediated mitophagy, platelet life span, and platelet physiopathology. Our work suggests that targeting platelet mitophagy Nix might provide new antithrombotic strategies.


Assuntos
Plaquetas/metabolismo , Proteínas de Membrana/metabolismo , Mitofagia , Ativação Plaquetária , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Biomarcadores , Tempo de Sangramento , Plaquetas/ultraestrutura , Trombose das Artérias Carótidas/etiologia , Trombose das Artérias Carótidas/metabolismo , Trombose das Artérias Carótidas/patologia , Sobrevivência Celular/genética , Humanos , Imunofenotipagem , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Fenótipo , Ativação Plaquetária/genética , Testes de Função Plaquetária , Proteínas Proto-Oncogênicas/genética , Proteínas Supressoras de Tumor/genética
9.
Proc Natl Acad Sci U S A ; 115(20): E4661-E4669, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29712858

RESUMO

Nonischemic cardiomyopathy (NICM) resulting from long-standing hypertension, valvular disease, and genetic mutations is a major cause of heart failure worldwide. Recent observations suggest that myeloid cells can impact cardiac function, but the role of tissue-intrinsic vs. tissue-extrinsic myeloid cells in NICM remains poorly understood. Here, we show that cardiac resident macrophage proliferation occurs within the first week following pressure overload hypertrophy (POH; a model of heart failure) and is requisite for the heart's adaptive response. Mechanistically, we identify Kruppel-like factor 4 (KLF4) as a key transcription factor that regulates cardiac resident macrophage proliferation and angiogenic activities. Finally, we show that blood-borne macrophages recruited in late-phase POH are detrimental, and that blockade of their infiltration improves myocardial angiogenesis and preserves cardiac function. These observations demonstrate previously unappreciated temporal and spatial roles for resident and nonresident macrophages in the development of heart failure.


Assuntos
Cardiomegalia/patologia , Cardiomiopatias/patologia , Insuficiência Cardíaca/patologia , Fatores de Transcrição Kruppel-Like/metabolismo , Macrófagos/patologia , Miocárdio/patologia , Animais , Cardiomegalia/imunologia , Cardiomegalia/metabolismo , Cardiomiopatias/imunologia , Cardiomiopatias/metabolismo , Células Cultivadas , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/metabolismo , Fator 4 Semelhante a Kruppel , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Miocárdio/imunologia , Miocárdio/metabolismo , Pressão
10.
Cell Rep ; 13(11): 2368-2375, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26686628

RESUMO

Circadian rhythms offer temporal control of anticipatory physiologic adaptations in animals. In the mammalian cardiovascular system, the importance of these rhythms is underscored by increased cardiovascular disease in shift workers, findings recapitulated in experimental animal models. However, a nodal regulator that allows integration of central and peripheral information and coordinates cardiac rhythmic output has been elusive. Here, we show that kruppel-like factor 15 (KLF15) governs a biphasic transcriptomic oscillation in the heart with a maximum ATP production phase and a remodeling and repair phase corresponding to the active and resting phase of a rodent. Depletion of KLF15 in cardiomyocytes leads to a disorganized oscillatory behavior without phasic partition despite an intact core clock. Thus, KLF15 is a nodal connection between the clock and meaningful rhythmicity in the heart.


Assuntos
Proteínas de Ligação a DNA/genética , Miocárdio/metabolismo , Fatores de Transcrição/genética , Trifosfato de Adenosina/metabolismo , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/metabolismo , Aldeído-Desidrogenase Mitocondrial , Animais , Linhagem Celular , Ritmo Circadiano/fisiologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição Kruppel-Like , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Receptores dos Hormônios Tireóideos/genética , Receptores dos Hormônios Tireóideos/metabolismo , Análise de Sequência de RNA , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Transcriptoma
11.
J Clin Invest ; 125(9): 3461-76, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26241060

RESUMO

Mitochondrial homeostasis is critical for tissue health, and mitochondrial dysfunction contributes to numerous diseases, including heart failure. Here, we have shown that the transcription factor Kruppel-like factor 4 (KLF4) governs mitochondrial biogenesis, metabolic function, dynamics, and autophagic clearance. Adult mice with cardiac-specific Klf4 deficiency developed cardiac dysfunction with aging or in response to pressure overload that was characterized by reduced myocardial ATP levels, elevated ROS, and marked alterations in mitochondrial shape, size, ultrastructure, and alignment. Evaluation of mitochondria isolated from KLF4-deficient hearts revealed a reduced respiration rate that is likely due to defects in electron transport chain complex I. Further, cardiac-specific, embryonic Klf4 deletion resulted in postnatal premature mortality, impaired mitochondrial biogenesis, and altered mitochondrial maturation. We determined that KLF4 binds to, cooperates with, and is requisite for optimal function of the estrogen-related receptor/PPARγ coactivator 1 (ERR/PGC-1) transcriptional regulatory module on metabolic and mitochondrial targets. Finally, we found that KLF4 regulates autophagy flux through transcriptional regulation of a broad array of autophagy genes in cardiomyocytes. Collectively, these findings identify KLF4 as a nodal transcriptional regulator of mitochondrial homeostasis.


Assuntos
Fatores de Transcrição Kruppel-Like/metabolismo , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/metabolismo , Transcrição Gênica , Animais , Autofagia/genética , Células HEK293 , Cardiopatias/genética , Cardiopatias/metabolismo , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas/genética , Consumo de Oxigênio/genética , PPAR gama/genética , PPAR gama/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ratos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
12.
Int J Clin Exp Pathol ; 8(2): 2221-34, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25973133

RESUMO

OBJECTIVE: The combination of intracoronary transplantation and ultrasound-mediated microbubble destruction may promote effective and accurate delivery of bone marrow stem cells (BMSCs) into the infarct zone. To test this hypothesis in this study we examined the effectiveness of ultrasound-mediated microbubble destruction in combination with intracoronary transplantation of BMSCs for the treatment of myocardial infarction in canine model of acute myocardial infarction. METHOD: The dogs were randomly assigned to four groups: PBS, ultrasound-mediated microbubble destruction, BMSCs, BMSCs together with ultrasound-mediated microbubble destruction. At 28 days post-surgery, cardiac function and the percentage of perfusion defect area to total left ventricular perfusion area (DA%) were determined by myocardial contrast echocardiography. Nitro blue tetrazolium staining was performed to determine myocardial infarct size, hematoxylin and eosin staining for assessing microvascular injury, Masson's staining for analyzing myocardial tissue collagen, immunohistochemical analysis of α-actin to measure cardiac contractile function and of BrdU-labeled myocardial cells to measure the number of the BMSCs homing to the infarcted region. RESULTS: The transplantation of BMSCs significantly improved heart function and DA% (P < 0.05). The group that received ultrasound-mediated microbubble destruction with BMSCs transplantation showed the most improvement in heart function and DA% (P < 0.05). This group also showed a denser deposition of BMSCs in the coronary artery and more BrdU positive cells in the infarcted region, had the maximum number of α-actin positive cells, showed the smallest myocardial infarct area compared to other groups (P< 0.05). CONCLUSION: Ultrasound-mediated microbubble destruction increases the homing of BMSCs in the target area following intracoronary transplantation, which allows more BMSCs to differentiate into functional cardiomyocytes, thereby reducing myocardial infarct size and improving cardiac function.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais , Infarto do Miocárdio/terapia , Animais , Cães , Ecocardiografia , Microbolhas , Ultrassonografia de Intervenção
13.
J Biol Chem ; 289(17): 12016-12028, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24599951

RESUMO

Regulation of endothelial cell biology by the Notch signaling pathway (Notch) is essential to vascular development, homeostasis, and sprouting angiogenesis. Although Notch determines cell fate and differentiation in a wide variety of cells, the molecular basis of upstream regulation of Notch remains poorly understood. Our group and others have implicated the Krüppel-like factor family of transcription factors as critical regulators of endothelial function. Here, we show that Krüppel-like factor 4 (KLF4) is a central regulator of sprouting angiogenesis via regulating Notch. Using a murine model in which KLF4 is overexpressed exclusively in the endothelium, we found that sustained expression of KLF4 promotes ineffective angiogenesis leading to diminished tumor growth independent of endothelial cell proliferation or cell cycling effects. These tumors feature increased vessel density yet are hypoperfused, leading to tumor hypoxia. Mechanistically, we show that KLF4 differentially regulates expression of Notch receptors, ligands, and target genes. We also demonstrate that KLF4 limits cleavage-mediated activation of Notch1. Finally, we rescue Notch target gene expression and the KLF4 sprouting angiogenesis phenotype by supplementation of DLL4 recombinant protein. Identification of this hitherto undiscovered role of KLF4 implicates this transcription factor as a critical regulator of Notch, tumor angiogenesis, and sprouting angiogenesis.


Assuntos
Endotélio Vascular/metabolismo , Fatores de Transcrição Kruppel-Like/fisiologia , Neovascularização Patológica , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Sequência de Bases , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Primers do DNA , Técnicas de Silenciamento de Genes , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Transgênicos
14.
Arterioscler Thromb Vasc Biol ; 33(6): 1135-44, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23640482

RESUMO

Macrophages are key regulators of many organ systems, including innate and adaptive immunity, systemic metabolism, hematopoiesis, vasculogenesis, malignancy, and reproduction. The pleiotropic roles of macrophages are mirrored by similarly diverse cellular phenotypes. A simplified schema classifies macrophages as M1, classically activated macrophages, or M2, alternatively activated macrophages. These cells are characterized by their expression of cell surface markers, secreted cytokines and chemokines, and transcription and epigenetic pathways. Transcriptional regulation is central to the differential speciation of macrophages, and several major pathways have been described as essential for subset differentiation. In this review, we discuss the transcriptional regulation of macrophages.


Assuntos
Imunidade Adaptativa/genética , Imunidade Inata/genética , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Ativação Transcricional/genética , Imunidade Adaptativa/imunologia , Animais , Antígenos de Superfície/genética , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Humanos , Imunidade Inata/imunologia , Ativação de Macrófagos/genética , Macrófagos/citologia , Camundongos , Sensibilidade e Especificidade , Transdução de Sinais
15.
Arterioscler Thromb Vasc Biol ; 32(12): 2836-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23065827

RESUMO

OBJECTIVE: To investigate the role of Krüppel-like factor 4 (KLF4), an essential transcriptional regulator of macrophage polarization (M1/M2), in the pathogenesis of atherosclerosis. METHODS AND RESULTS: Despite the acknowledged importance of macrophages in atherosclerosis, the role of M1 (classically activated or proinflammatory) versus M2 (alternatively activated or anti-inflammatory) macrophages in this process remains incompletely understood. We recently identified KLF4 as a regulator of macrophage subset specification; that is, KLF4 promotes M2 and inhibits M1 phenotype. Here, we provide evidence that KLF4-deficient macrophages exhibit enhanced proinflammatory activation and foam cell formation in response to oxidized lipids. In vivo, myeloid KLF4-deficient mice (ApoE(-/-) background) develop significantly more vascular inflammation and atherosclerotic lesion formation. CONCLUSIONS: Our findings identify myeloid KLF4 as an essential regulator of vascular inflammation and experimental atherogenesis.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/fisiopatologia , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/fisiologia , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/fisiologia , Aterosclerose/patologia , Modelos Animais de Doenças , Células Espumosas/patologia , Células Espumosas/fisiologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Macrófagos/patologia , Macrófagos/fisiologia , Camundongos , Camundongos Knockout , Fenótipo
16.
J Clin Invest ; 121(7): 2736-49, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21670502

RESUMO

Current paradigms suggest that two macrophage subsets, termed M1 and M2, are involved in inflammation and host defense. While the distinct functions of M1 and M2 macrophages have been intensively studied - the former are considered proinflammatory and the latter antiinflammatory - the determinants of their speciation are incompletely understood. Here we report our studies that identify Krüppel-like factor 4 (KLF4) as a critical regulator of macrophage polarization. Macrophage KLF4 expression was robustly induced in M2 macrophages and strongly reduced in M1 macrophages, observations that were recapitulated in human inflammatory paradigms in vivo. Mechanistically, KLF4 was found to cooperate with Stat6 to induce an M2 genetic program and inhibit M1 targets via sequestration of coactivators required for NF-κB activation. KLF4-deficient macrophages demonstrated increased proinflammatory gene expression, enhanced bactericidal activity, and altered metabolism. Furthermore, mice bearing myeloid-specific deletion of KLF4 exhibited delayed wound healing and were predisposed to developing diet-induced obesity, glucose intolerance, and insulin resistance. Collectively, these data identify KLF4 as what we believe to be a novel regulator of macrophage polarization.


Assuntos
Polaridade Celular , Fatores de Transcrição Kruppel-Like/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Tecido Adiposo/citologia , Tecido Adiposo/fisiologia , Animais , Linhagem Celular , Escherichia coli/imunologia , Expressão Gênica , Humanos , Resistência à Insulina/fisiologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Obesidade/fisiopatologia , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo , Cicatrização/fisiologia
17.
J Biol Chem ; 282(38): 27728-35, 2007 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-17660514

RESUMO

The cardiac serine protease corin is the pro-atrial natriuretic peptide convertase. Corin is made as a zymogen, which is activated by proteolytic cleavage. Previous studies showed that recombinant human corin expressed in HEK 293 cells was biologically active, but activated corin fragments were not detectable, making it difficult to study corin activation. In this study, we showed that recombinant rat corin was activated in HEK 293 cells, murine HL-1 cardiomyocytes, and rat neonatal cardiomyocytes. In these cells, activated corin represented a small fraction of the total corin molecules. The activation of recombinant rat corin was inhibited by small molecule trypsin inhibitors but not inhibitors for matrix metalloproteinases or cysteine proteases, suggesting that a trypsin-like protease activated corin in these cells. Glycosidase digestion showed that rat and human corin proteins contained substantial N-glycans but little O-glycans. Treatment of HEK 293 cells expressing rat corin with tunicamycin prevented corin activation and inhibited its pro-atrial natriuretic peptide processing activity. Similar effects of tunicamycin on endogenous corin activity were found in HL-1 cells. Mutations altering the two N-glycosylation sites in the protease domain of rat corin prevented its activation in HEK 293 and HL-1 cells. Our results indicate that N-linked oligosaccharides play an important role in corin activation.


Assuntos
Precursores Enzimáticos/metabolismo , Serina Endopeptidases/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Glicosilação , Humanos , Camundongos , Modelos Biológicos , Miocárdio/metabolismo , Polissacarídeos/metabolismo , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/química
18.
Genes Dev ; 21(11): 1396-408, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17510282

RESUMO

gp130-linked cytokines such as interleukin-6 (IL-6) stimulate the formation of tyrosine-phosphorylated signal transducer and activator of transcription 3 (P-STAT3), which activates many genes, including the STAT3 gene itself. The resulting increase in the concentration of unphosphorylated STAT3 (U-STAT3) drives a second wave of expression of genes such as RANTES, IL6, IL8, MET, and MRAS that do not respond directly to P-STAT3. Thus, U-STAT3 sustains cytokine-dependent signaling at late times through a mechanism completely distinct from that used by P-STAT3. Many U-STAT3-responsive genes have kappaB elements that are activated by a novel transcription factor complex formed when U-STAT3 binds to unphosphorylated NFkappaB (U-NFkappaB), in competition with IkappaB. The U-STAT3/U-NFkappaB complex accumulates in the nucleus with help from the nuclear localization signal of STAT3, activating a subset of kappaB-dependent genes. Additional genes respond to U-STAT3 through an NFkappaB-independent mechanism. The role of signal-dependent increases in U-STAT3 expression in regulating gene expression is likely to be important in physiological responses to gp130-linked cytokines and growth factors that activate STAT3, and in cancers that have constitutively active P-STAT3.


Assuntos
Regulação da Expressão Gênica , Interleucina-6/farmacologia , NF-kappa B/metabolismo , Fator de Transcrição STAT3/metabolismo , Transcrição Gênica , Sítios de Ligação , Biomarcadores/metabolismo , Mama/citologia , Mama/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas/metabolismo , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Imunoprecipitação da Cromatina , Células Epiteliais/metabolismo , Perfilação da Expressão Gênica , Humanos , Imunoprecipitação , NF-kappa B/genética , Sinais de Localização Nuclear , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Regiões Promotoras Genéticas , Elementos Reguladores de Transcrição , Transdução de Sinais
19.
FASEB J ; 20(12): 2147-9, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16935937

RESUMO

Cells without Bak and Bax are largely resistant to apoptosis, despite the presence of other key components of the apoptotic machinery. We screened 7,800 natural compounds and found several that could specifically induce caspase activation and the release of cytochrome c (cyto c) in the bak(-/-)/bax(-/-) cells. One of these was gossypol, a polyphenolic compound naturally found in cottonseed that has been used in antifertility trials. We found that gossypol, but not other Bcl-2-interacting molecules, induced cyto c release and loss of mitochondrial membrane potential (delta psi m) independently of mPTP and Bak/Bax activation. Furthermore, we found that gossypol induced an allosteric change in Bcl-2 in both bak(-/-)/bax(-/-) cells and Bcl-2 overexpressing cells. This change in Bcl-2 conformation led to the release of cyto c in the presence of Bcl-2 and Bcl-xL in reconstituted proteoliposomes. We also observed that gossypol substantially reduced the growth of tumor xenografts from Bcl-2 overexpressing cells in nude mice. We conclude that gossypol converts the antiapoptotic molecule Bcl-2 into a proapoptotic molecule that can mediate the release of cyto c and induce apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Citocromos c/metabolismo , Gossipol/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Animais , Antineoplásicos/uso terapêutico , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , Conformação Proteica , Proteínas Proto-Oncogênicas c-bcl-2/química , Carga Tumoral/efeitos dos fármacos , Proteína X Associada a bcl-2 , Proteína bcl-X/metabolismo
20.
Exp Cell Res ; 306(1): 85-93, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15878335

RESUMO

Our previous studies have shown that SMAD5, an important intracellular mediator of transforming growth factor beta (TGF-beta) family, is required for normal development of the cardiovascular system in vivo. In the current study, we reported that the lack of the Smad5 gene resulted in apoptosis of cardiac myocytes in vivo. To further investigate the mechanism of the Smad5 gene in cardiomyocyte apoptosis, the embryonic stem (ES) cell differentiation system was employed. We found that the myotubes that differentiated from the homozygous Smad5ex6/ex6 mutant ES cells underwent collapse and degeneration during the late stages of in vitro differentiation, mimicking the in vivo observation. By electron microscopy, abnormal swollen mitochondria were observed in cardiomyocytes both from Smad5-deficient embryos and from ES-differentiated cells. There was also a significant reduction in mitochondrial membrane potential (Deltapsi m) and a leakage of cytochrome c from mitochondria into the cytosol of myocytes differentiated from Smad5 mutant ES cells. The expression of p53 and p21 was found to be elevated in the differentiated Smad5 mutant myocytes, and this was accompanied by an up-regulation in caspase 3 expression. These results suggest that the Smad5-mediated TGF-beta signals may protect cardiomyocytes from apoptosis by maintaining the integrity of the mitochondria, probably through suppression of p53 mediated pathways.


Assuntos
Apoptose/fisiologia , Proteínas de Ligação a DNA/fisiologia , Mitocôndrias/fisiologia , Miócitos Cardíacos/fisiologia , Fosfoproteínas/fisiologia , Transativadores/fisiologia , Animais , Apoptose/genética , Caspase 3 , Caspases/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miocárdio/ultraestrutura , Miócitos Cardíacos/citologia , Miócitos Cardíacos/ultraestrutura , Fosfoproteínas/genética , Proteína Smad5 , Transativadores/genética , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA