Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Circ Res ; 132(11): 1468-1485, 2023 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-37042252

RESUMO

BACKGROUND: The ability of the right ventricle (RV) to adapt to an increased pressure afterload determines survival in patients with pulmonary arterial hypertension. At present, there are no specific treatments available to prevent RV failure, except for heart/lung transplantation. The wingless/int-1 (Wnt) signaling pathway plays an important role in the development of the RV and may also be implicated in adult cardiac remodeling. METHODS: Molecular, biochemical, and pharmacological approaches were used both in vitro and in vivo to investigate the role of Wnt signaling in RV remodeling. RESULTS: Wnt/ß-catenin signaling molecules are upregulated in RV of patients with pulmonary arterial hypertension and animal models of RV overload (pulmonary artery banding-induced and monocrotaline rat models). Activation of Wnt/ß-catenin signaling leads to RV remodeling via transcriptional activation of FOSL1 and FOSL2 (FOS proto-oncogene [FOS] like 1/2, AP-1 [activator protein 1] transcription factor subunit). Immunohistochemical analysis of pulmonary artery banding -exposed BAT-Gal (ß-catenin-activated transgene driving expression of nuclear ß-galactosidase) reporter mice RVs exhibited an increase in ß-catenin expression compared with their respective controls. Genetic inhibition of ß-catenin, FOSL1/2, or WNT3A stimulation of RV fibroblasts significantly reduced collagen synthesis and other remodeling genes. Importantly, pharmacological inhibition of Wnt signaling using inhibitor of PORCN (porcupine O-acyltransferase), LGKK-974 attenuated fibrosis and cardiac hypertrophy leading to improvement in RV function in both, pulmonary artery banding - and monocrotaline-induced RV overload. CONCLUSIONS: Wnt- ß-Catenin-FOSL signaling is centrally involved in the hypertrophic RV response to increased afterload, offering novel targets for therapeutic interference with RV failure in pulmonary hypertension.


Assuntos
Insuficiência Cardíaca , Hipertensão Arterial Pulmonar , Ratos , Camundongos , Animais , Remodelação Ventricular , beta Catenina , Cateninas , Monocrotalina/toxicidade , Transdução de Sinais , Modelos Animais de Doenças , Função Ventricular Direita
2.
Nat Commun ; 14(1): 1980, 2023 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-37031213

RESUMO

Activation of endothelial YAP/TAZ signaling is crucial for physiological and pathological angiogenesis. The mechanisms of endothelial YAP/TAZ regulation are, however, incompletely understood. Here we report that the protocadherin FAT1 acts as a critical upstream regulator of endothelial YAP/TAZ which limits the activity of these transcriptional cofactors during developmental and tumor angiogenesis by promoting their degradation. We show that loss of endothelial FAT1 results in increased endothelial cell proliferation in vitro and in various angiogenesis models in vivo. This effect is due to perturbed YAP/TAZ protein degradation, leading to increased YAP/TAZ protein levels and expression of canonical YAP/TAZ target genes. We identify the E3 ubiquitin ligase Mind Bomb-2 (MIB2) as a FAT1-interacting protein mediating FAT1-induced YAP/TAZ ubiquitination and degradation. Loss of MIB2 expression in endothelial cells in vitro and in vivo recapitulates the effects of FAT1 depletion and causes decreased YAP/TAZ degradation and increased YAP/TAZ signaling. Our data identify a pivotal mechanism of YAP/TAZ regulation involving FAT1 and its associated E3 ligase MIB2, which is essential for YAP/TAZ-dependent angiogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Transativadores , Humanos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteólise , Transativadores/metabolismo , Células Endoteliais/metabolismo , Proteínas de Sinalização YAP , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Neovascularização Patológica/metabolismo , Fosfoproteínas/metabolismo , Caderinas/metabolismo
3.
Immunity ; 56(5): 979-997.e11, 2023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-37100060

RESUMO

Immune cell trafficking constitutes a fundamental component of immunological response to tissue injury, but the contribution of intrinsic RNA nucleotide modifications to this response remains elusive. We report that RNA editor ADAR2 exerts a tissue- and stress-specific regulation of endothelial responses to interleukin-6 (IL-6), which tightly controls leukocyte trafficking in IL-6-inflamed and ischemic tissues. Genetic ablation of ADAR2 from vascular endothelial cells diminished myeloid cell rolling and adhesion on vascular walls and reduced immune cell infiltration within ischemic tissues. ADAR2 was required in the endothelium for the expression of the IL-6 receptor subunit, IL-6 signal transducer (IL6ST; gp130), and subsequently, for IL-6 trans-signaling responses. ADAR2-induced adenosine-to-inosine RNA editing suppressed the Drosha-dependent primary microRNA processing, thereby overwriting the default endothelial transcriptional program to safeguard gp130 expression. This work demonstrates a role for ADAR2 epitranscriptional activity as a checkpoint in IL-6 trans-signaling and immune cell trafficking to sites of tissue injury.


Assuntos
Interleucina-6 , RNA , Células Endoteliais/metabolismo , Receptor gp130 de Citocina , Endotélio/metabolismo , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo
4.
Circ Res ; 131(7): 580-597, 2022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-36000401

RESUMO

BACKGROUND: ADAR1 (adenosine deaminase acting on RNA-1)-mediated adenosine to inosine (A-to-I) RNA editing plays an essential role for distinguishing endogenous from exogenous RNAs, preventing autoinflammatory ADAR1 also regulates cellular processes by recoding specific mRNAs, thereby altering protein functions, but may also act in an editing-independent manner. The specific role of ADAR1 in cardiomyocytes and its mode of action in the heart is not fully understood. To determine the role of ADAR1 in the heart, we used different mutant mouse strains, which allows to distinguish immunogenic, editing-dependent, and editing-independent functions of ADAR1. METHODS: Different Adar1-mutant mouse strains were employed for gene deletion or specific inactivation of ADAR1 enzymatic activity in cardiomyocytes, either alone or in combination with Ifih1 (interferon induced with helicase C domain 1) or Irf7 (interferon regulatory factor 7) gene inactivation. Mutant mice were investigated by immunofluorescence, Western blot, RNAseq, proteomics, and functional MRI analysis. RESULTS: Inactivation of Adar1 in cardiomyocytes resulted in late-onset autoinflammatory myocarditis progressing into dilated cardiomyopathy and heart failure at 6 months of age. Adar1 depletion activated interferon signaling genes but not NFκB (nuclear factor kappa B) signaling or apoptosis and reduced cardiac hypertrophy during pressure overload via induction of Irf7. Additional inactivation of the cytosolic RNA sensor MDA5 (melanoma differentiation-associated gene 5; encoded by the Ifih1 gene) in Adar1 mutant mice prevented activation of interferon signaling gene and delayed heart failure but did not prevent lethality after 8.5 months. In contrast, compound mutants only expressing catalytically inactive ADAR1 in an Ifih1-mutant background were completely normal. Inactivation of Irf7 attenuated the phenotype of Adar1-deficient cardiomyocytes to a similar extent as Ifih1 depletion, identifying IRF7 as the main mediator of autoinflammatory responses caused by the absence of ADAR1 in cardiomyocytes. CONCLUSIONS: Enzymatically active ADAR1 prevents IRF7-mediated autoinflammatory reactions in the heart triggered by endogenous nonedited RNAs. In addition to RNA editing, ADAR1 also serves editing-independent roles in the heart required for long-term cardiac function and survival.


Assuntos
Adenosina Desaminase , Insuficiência Cardíaca , Adenosina/metabolismo , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Animais , Inosina/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/metabolismo , Interferons/metabolismo , Camundongos , Camundongos Mutantes , NF-kappa B/metabolismo , RNA
6.
Int J Mol Sci ; 23(1)2021 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-35008777

RESUMO

Oncostatin M (OSM) and leukemia inhibitory factor (LIF) signaling protects the heart after myocardial infarction (MI). In mice, oncostatin M receptor (OSMR) and leukemia inhibitory factor receptor (LIFR) are selectively activated by the respective cognate ligands while OSM activates both the OSMR and LIFR in humans, which prevents efficient translation of mouse data into potential clinical applications. We used an engineered human-like OSM (hlOSM) protein, capable to signal via both OSMR and LIFR, to evaluate beneficial effects on cardiomyocytes and hearts after MI in comparison to selective stimulation of either LIFR or OSMR. Cell viability assays, transcriptome and immunoblot analysis revealed increased survival of hypoxic cardiomyocytes by mLIF, mOSM and hlOSM stimulation, associated with increased activation of STAT3. Kinetic expression profiling of infarcted hearts further specified a transient increase of OSM and LIF during the early inflammatory phase of cardiac remodeling. A post-infarction delivery of hlOSM but not mOSM or mLIF within this time period combined with cardiac magnetic resonance imaging-based strain analysis uncovered a global cardioprotective effect on infarcted hearts. Our data conclusively suggest that a simultaneous and rapid activation of OSMR and LIFR after MI offers a therapeutic opportunity to preserve functional and structural integrity of the infarcted heart.


Assuntos
Cardiotônicos/metabolismo , Infarto do Miocárdio/prevenção & controle , Oncostatina M/metabolismo , Receptores de OSM-LIF/metabolismo , Animais , Hipóxia Celular/genética , Sobrevivência Celular , Células Cultivadas , Humanos , Cinética , Fator Inibidor de Leucemia/metabolismo , Camundongos , Contração Miocárdica , Infarto do Miocárdio/genética , Miócitos Cardíacos/metabolismo , Engenharia de Proteínas , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptores de Oncostatina M/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Especificidade da Espécie , Transcriptoma/genética
7.
Cancer Res ; 80(19): 4199-4211, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32816854

RESUMO

Although NF-κB is known to play a pivotal role in lung cancer, contributing to tumor growth, microenvironmental changes, and metastasis, the epigenetic regulation of NF-κB in tumor context is largely unknown. Here we report that the IKK2/NF-κB signaling pathway modulates metastasis-associated protein 2 (MTA2), a component of the nucleosome remodeling and deacetylase complex (NuRD). In triple transgenic mice, downregulation of IKK2 (Sftpc-cRaf-IKK2DN) in cRaf-induced tumors in alveolar epithelial type II cells restricted tumor formation, whereas activation of IKK2 (Sftpc-cRaf-IKK2CA) supported tumor growth; both effects were accompanied by altered expression of MTA2. Further studies employing genetic inhibition of MTA2 suggested that in primary tumor growth, independent of IKK2, MTA2/NuRD corepressor complex negatively regulates NF-κB signaling and tumor growth, whereas later dissociation of MTA2/NuRD complex from the promoter of NF-κB target genes and IKK2-dependent positive regulation of MTA2 leads to activation of NF-κB signaling, epithelial-mesenchymal transition, and lung tumor metastasis. These findings reveal a previously unrecognized biphasic role of MTA2 in IKK2/NF-κB-driven primary-to-metastatic lung tumor progression. Addressing the interaction between MTA2 and NF-κB would provide potential targets for intervention of tumor growth and metastasis. SIGNIFICANCE: These findings strongly suggest a prominent role of MTA2 in primary tumor growth, lung metastasis, and NF-κB signaling modulatory functions.


Assuntos
Histona Desacetilases/metabolismo , Neoplasias Pulmonares/patologia , NF-kappa B/metabolismo , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Animais , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Histona Desacetilases/genética , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Inflamação/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/genética , Proteínas Repressoras/genética , Transdução de Sinais , Transativadores/genética , Microambiente Tumoral
8.
Nat Commun ; 10(1): 2130, 2019 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-31086178

RESUMO

Hypoxia signaling plays a major role in non-malignant and malignant hyperproliferative diseases. Pulmonary hypertension (PH), a hypoxia-driven vascular disease, is characterized by a glycolytic switch similar to the Warburg effect in cancer. Ras association domain family 1A (RASSF1A) is a scaffold protein that acts as a tumour suppressor. Here we show that hypoxia promotes stabilization of RASSF1A through NOX-1- and protein kinase C- dependent phosphorylation. In parallel, hypoxia inducible factor-1 α (HIF-1α) activates RASSF1A transcription via HIF-binding sites in the RASSF1A promoter region. Vice versa, RASSF1A binds to HIF-1α, blocks its prolyl-hydroxylation and proteasomal degradation, and thus enhances the activation of the glycolytic switch. We find that this mechanism operates in experimental hypoxia-induced PH, which is blocked in RASSF1A knockout mice, in human primary PH vascular cells, and in a subset of human lung cancer cells. We conclude that RASSF1A-HIF-1α forms a feedforward loop driving hypoxia signaling in PH and cancer.


Assuntos
Hipóxia Celular , Hipertensão Pulmonar/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Pulmonares/patologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Modelos Animais de Doenças , Glicólise , Células HEK293 , Células HeLa , Humanos , Hipertensão Pulmonar/cirurgia , Pulmão/irrigação sanguínea , Pulmão/patologia , Pulmão/cirurgia , Masculino , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso , NADPH Oxidase 1/metabolismo , Cultura Primária de Células , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteína Quinase C/metabolismo , Proteólise , Artéria Pulmonar/citologia , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
9.
Biomed Res Int ; 2018: 3293584, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29511676

RESUMO

Pulmonary arterial hypertension (PAH) is a progressive disorder characterized by remodeling of the pulmonary vasculature and a rise in right ventricular (RV) afterload. The increased RV afterload leads to right ventricular failure (RVF) which is the reason for the high morbidity and mortality in PAH patients. The objective was to evaluate the therapeutic efficacy and antiremodeling potential of the phosphodiesterase type 5 (PDE5) inhibitor sildenafil and the soluble guanylate cyclase stimulator riociguat in a model of pressure overload RV hypertrophy induced by pulmonary artery banding (PAB). Mice subjected to PAB, one week after surgery, were treated with either sildenafil (100 mg/kg/d, n = 5), riociguat (30 mg/kg/d, n = 5), or vehicle (n = 5) for 14 days. RV function and remodeling were assessed by right heart catheterization, magnetic resonance imaging (MRI), and histomorphometry. Both sildenafil and riociguat prevented the deterioration of RV function, as determined by a decrease in RV dilation and restoration of the RV ejection fraction (EF). Although both compounds did not decrease right heart mass and cellular hypertrophy, riociguat prevented RV fibrosis induced by PAB. Both compounds diminished TGF-beta1 induced collagen synthesis of RV cardiac fibroblasts in vitro. Treatment with either riociguat or sildenafil prevented the progression of pressure overload-induced RVF, representing a novel therapeutic approach.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Hipertensão Pulmonar/tratamento farmacológico , Pirazóis/administração & dosagem , Pirimidinas/administração & dosagem , Citrato de Sildenafila/administração & dosagem , Disfunção Ventricular Direita/tratamento farmacológico , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/genética , Modelos Animais de Doenças , Coração/efeitos dos fármacos , Coração/fisiopatologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/fisiopatologia , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Masculino , Camundongos , Inibidores da Fosfodiesterase 5/administração & dosagem , Pressão , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiopatologia , Remodelação Vascular/efeitos dos fármacos , Disfunção Ventricular Direita/genética , Disfunção Ventricular Direita/patologia
10.
EMBO J ; 36(9): 1199-1214, 2017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28314781

RESUMO

Control of energy homeostasis and metabolism is achieved by integrating numerous pathways, and miRNAs are involved in this process by regulating expression of multiple target genes. However, relatively little is known about the posttranscriptional processing of miRNAs and a potential role for the precursors they derive from. Here, we demonstrate that mature miRNA-22 is more abundant in muscle from male mice relative to females and that this enables sex-specific regulation of muscular lipid metabolism and body weight by repressing estrogen receptor alpha (ERα) expression. We found that the ERα adjusts its own activity by preventing processing of miR-22 via direct binding to a conserved ERα-binding element within the primary miR-22 precursor. Mutation of the ERα binding site within the pri-miR-22 in vivo eliminates sex-specific differences in miR-22 expression. We reason that the resulting tissue selective negative feedback regulation is essential to establish sex-specific differences in muscle metabolism and body weight development.


Assuntos
Receptor alfa de Estrogênio/biossíntese , Regulação da Expressão Gênica , Metabolismo dos Lipídeos , MicroRNAs/metabolismo , Músculos/metabolismo , Animais , Masculino , Camundongos , Fatores Sexuais
11.
JCI Insight ; 1(7): e81175, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27699262

RESUMO

Obesity is an increasing health problem worldwide, and nonsurgical strategies to treat obesity have remained rather inefficient. We here show that acute loss of TGF-ß-activated kinase 1 (TAK1) in adipocytes results in an increased rate of apoptotic adipocyte death and increased numbers of M2 macrophages in white adipose tissue. Mice with adipocyte-specific TAK1 deficiency have reduced adipocyte numbers and are resistant to obesity induced by a high-fat diet or leptin deficiency. In addition, adipocyte-specific TAK1-deficient mice under a high-fat diet showed increased energy expenditure, which was accompanied by enhanced expression of the uncoupling protein UCP1. Interestingly, acute induction of adipocyte-specific TAK1 deficiency in mice already under a high-fat diet was able to stop further weight gain and improved glucose tolerance. Thus, loss of TAK1 in adipocytes reduces the total number of adipocytes, increases browning of white adipose tissue, and may be an attractive strategy to treat obesity, obesity-dependent diabetes, and other associated complications.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Apoptose , MAP Quinase Quinase Quinases/deficiência , Obesidade/genética , Adipócitos/citologia , Animais , Dieta Hiperlipídica , MAP Quinase Quinase Quinases/genética , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína Desacopladora 1/metabolismo
12.
Circ Res ; 118(12): 1906-17, 2016 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-27140435

RESUMO

RATIONALE: Activated cardiac fibroblasts (CF) are crucial players in the cardiac damage response; excess fibrosis, however, may result in myocardial stiffening and heart failure development. Inhibition of activated CF has been suggested as a therapeutic strategy in cardiac disease, but whether this truly improves cardiac function is unclear. OBJECTIVE: To study the effect of CF ablation on cardiac remodeling. METHODS AND RESULTS: We characterized subgroups of murine CF by single-cell expression analysis and identified periostin as the marker showing the highest correlation to an activated CF phenotype. We generated bacterial artificial chromosome-transgenic mice allowing tamoxifen-inducible Cre expression in periostin-positive cells as well as their diphtheria toxin-mediated ablation. In the healthy heart, periostin expression was restricted to valvular fibroblasts; ablation of this population did not affect cardiac function. After chronic angiotensin II exposure, ablation of activated CF resulted in significantly reduced cardiac fibrosis and improved cardiac function. After myocardial infarction, ablation of periostin-expressing CF resulted in reduced fibrosis without compromising scar stability, and cardiac function was significantly improved. Single-cell transcriptional analysis revealed reduced CF activation but increased expression of prohypertrophic factors in cardiac macrophages and cardiomyocytes, resulting in localized cardiomyocyte hypertrophy. CONCLUSIONS: Modulation of the activated CF population is a promising approach to prevent adverse cardiac remodeling in response to angiotensin II and after myocardial infarction.


Assuntos
Moléculas de Adesão Celular/metabolismo , Fibroblastos/metabolismo , Ventrículos do Coração/metabolismo , Infarto do Miocárdio/metabolismo , Remodelação Ventricular , Angiotensinas/toxicidade , Animais , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/genética , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibrose , Ventrículos do Coração/patologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/etiologia , Miócitos Cardíacos/metabolismo
13.
Nat Med ; 21(4): 353-62, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25751817

RESUMO

Cardiac healing after myocardial ischemia depends on the recruitment and local expansion of myeloid cells, particularly macrophages. Here we identify Reg3ß as an essential regulator of macrophage trafficking to the damaged heart. Using mass spectrometry-based secretome analysis, we found that dedifferentiating cardiomyocytes release Reg3ß in response to the cytokine OSM, which signals through Jak1 and Stat3. Loss of Reg3ß led to a large decrease in the number of macrophages in the ischemic heart, accompanied by increased ventricular dilatation and insufficient removal of neutrophils. This defect in neutrophil removal in turn caused enhanced matrix degradation, delayed collagen deposition and increased susceptibility to cardiac rupture. Our data indicate that OSM, acting through distinct intracellular pathways, regulates both cardiomyocyte dedifferentiation and cardiomyocyte-dependent regulation of macrophage trafficking. Release of OSM from infiltrating neutrophils and macrophages initiates a positive feedback loop in which OSM-induced production of Reg3ß in cardiomyocytes attracts additional OSM-secreting macrophages. The activity of the feedback loop controls the degree of macrophage accumulation in the heart, which is instrumental in myocardial healing.


Assuntos
Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Isquemia Miocárdica/patologia , Miocárdio/patologia , Oncostatina M/metabolismo , Proteínas/metabolismo , Animais , Antígenos de Neoplasias/genética , Biomarcadores Tumorais/genética , Colágeno/metabolismo , Eletroforese em Gel Bidimensional , Feminino , Coração/fisiologia , Ventrículos do Coração/metabolismo , Inflamação , Interleucina-6/metabolismo , Lectinas Tipo C/genética , Macrófagos/citologia , Masculino , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Proteínas Associadas a Pancreatite , Proteínas/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
14.
Am J Respir Crit Care Med ; 191(4): 437-47, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25536148

RESUMO

RATIONALE: Recent studies indicate that tumor-associated macrophages (MΦ) with an M2 phenotype can influence cancer progression and metastasis, but the regulatory pathways remain poorly characterized. OBJECTIVES: This study investigated the role of tumor-associated MΦ in lung cancer. METHODS: Coculturing of MΦ with mouse Lewis lung carcinoma (LLC1) and 10 different human lung cancer cell lines (adenocarcinoma, squamous cell carcinoma, and large cell carcinoma) caused up-regulation of CCR2/CCL2 and CX3CR1/CX3CL1 in both the cancer cells and the MΦ. MEASUREMENTS AND MAIN RESULTS: In the MΦ-tumor cell system, IL-10 drove CCR2 and CX3CR1 up-regulation, whereas CCL1, granulocyte colony-stimulating factor, and MIP1α were required for the up-regulation of CCL2 and CX3CL1. Downstream phenotypic effects included enhanced LLC1 proliferation and migration and MΦ M2 polarization. In vivo, MΦ depletion (clodronate, MΦ Fas-induced apoptosis mice) and genetic ablation of CCR2 and CX3CR1 all inhibited LLC1 tumor growth and metastasis, shifted tumor-associated MΦ toward M1 polarization, suppressed tumor vessel growth, and enhanced survival (metastasis model). Furthermore, mice treated with CCR2 antagonist mimicked genetic ablation of CCR2, showing reduced tumor growth and metastasis. In human lung cancer samples, tumor MΦ infiltration and CCR2 expression correlated with tumor stage and metastasis. CONCLUSIONS: Tumor-associated MΦ play a central role in lung cancer growth and metastasis, with bidirectional cross-talk between MΦ and cancer cells via CCR2 and CX3CR1 signaling as a central underlying mechanism. These findings suggest that the therapeutic strategy of blocking CCR2 and CX3CR1 may prove beneficial for halting lung cancer progression.


Assuntos
Adenocarcinoma/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Grandes/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Adenocarcinoma/patologia , Animais , Receptor 1 de Quimiocina CX3C , Carcinoma de Células Grandes/patologia , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Quimiocina CCL2/metabolismo , Quimiocina CX3CL1/metabolismo , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Metástase Neoplásica , Estadiamento de Neoplasias , Receptor Cross-Talk , Receptores CCR2/metabolismo , Receptores de Quimiocinas/metabolismo , Regulação para Cima
15.
PLoS One ; 9(6): e98456, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24893162

RESUMO

AIMS: Mouse models of myocardial infarction (MI) are commonly used to explore the pathophysiological role of the monocytic response in myocardial injury and to develop translational strategies. However, no study thus far has examined the potential impact of inter-individual variability and sham surgical procedures on monocyte subset kinetics after experimental MI in mice. Our goal was to investigate determinants of systemic myeloid cell subset shifts in C57BL/6 mice following MI by developing a protocol for sequential extensive flow cytometry (FCM). METHODS AND RESULTS: Following cross-sectional multiplex FCM analysis we provide for the first time a detailed description of absolute quantities, relative subset composition, and biological variability of circulating classical, intermediate, and non-classical monocyte subsets in C57BL/6 mice. By using intra-individual longitudinal measurements after MI induction, a time course of classical and non-classical monocytosis was recorded. This approach disclosed a significant reduction of monocyte subset dispersion across all investigated time points following MI. We found that in the current invasive model of chronic MI the global pattern of systemic monocyte kinetics is mainly determined by a nonspecific inflammatory response to sham surgery and not by the extent of myocardial injury. CONCLUSIONS: Application of sequential multiplexed FCM may help to reduce the impact of biological variability in C57BL/6 mice. Furthermore, the confounding influence of sham surgical procedures should always be considered when measuring monocyte subset kinetics in a murine model of MI.


Assuntos
Monócitos/metabolismo , Infarto do Miocárdio/cirurgia , Animais , Estudos Transversais , Citometria de Fluxo , Imuno-Histoquímica , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL
16.
Cytokine ; 64(1): 43-5, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23764551

RESUMO

Cardiac pressure overload-induced hypertrophy and pathological remodelling frequently leads to right ventricular dysfunction, which is the most frequent cause of death in patients with pulmonary arterial hypertension. Nowadays, accumulating reports support the concept that proinflammatory cytokines and growth factors play crucial roles in the failing heart. We recently identified Fn14 as an endogenous key regulator in cardiac fibrosis in the PAB (Pulmonary Artery Banding) pressure-overload model. Right ventricular overload after PAB is also characterized by hypertrophy. The aim of this study was to determine whether right ventricular (RV) cardiac hypertrophy induced by PAB is mediated by the TWEAK/Fn14 axis. After baseline MRI, Fn14(-/-) mice and wild-type (WT) littermates were randomly assigned to two groups: (1) SHAM-operated (n⩾4, per genotype) and (2) PAB (n⩾11, per genotype). The results of MRI and histological analysis demonstrated that Fn14(-/-) mice exhibit less PAB-induced cardiac hypertrophy compared to WT littermates. Moreover, Fn14 overexpression in cultured adult rat cardiomyocytes enhanced cardiomyocyte size. Collectively, our studies demonstrate that Fn14 ablation attenuates RV hypertrophy after PAB and that activation of TWEAK/Fn14 signaling promotes cardiomyocyte growth in vitro. These results nominate Fn14 as a potential novel target for the treatment of heart hypertrophy.


Assuntos
Hipertrofia Ventricular Direita/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Animais , Células Cultivadas , Hipertensão Pulmonar Primária Familiar , Coração/crescimento & desenvolvimento , Hipertensão Pulmonar/cirurgia , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Artéria Pulmonar/cirurgia , Ratos , Ratos Sprague-Dawley , Receptores do Fator de Necrose Tumoral/genética , Transdução de Sinais , Receptor de TWEAK
17.
Basic Res Cardiol ; 108(2): 325, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23325387

RESUMO

Pulmonary arterial hypertension (PAH) is a fatal disease for which no cure is yet available. The leading cause of death in PAH is right ventricular (RV) failure. Previously, the TNF receptor superfamily member fibroblast growth factor-inducible molecule 14 (Fn14) has been associated with different fibrotic diseases. However, so far there is no study demonstrating a causal role for endogenous Fn14 signaling in RV or LV heart disease. The purpose of this study was to determine whether global ablation of Fn14 prevents RV fibrosis and remodeling improving heart function. Here, we provide evidence for a causative role of Fn14 in pulmonary artery banding (PAB)-induced RV fibrosis and dysfunction in mice. Fn14 expression was increased in the RV after PAB. Mice lacking Fn14 (Fn14(-/-)) displayed substantially reduced RV fibrosis and dysfunction following PAB compared to wild-type littermates. Cell culture experiments demonstrated that activation of Fn14 induces collagen expression via RhoA-dependent nuclear translocation of myocardin-related transcription factor-A (MRTF-A)/MAL. Furthermore, activation of Fn14 in vitro caused fibroblast proliferation and myofibroblast differentiation, which corresponds to suppression of PAB-induced RV fibrosis in Fn14(-/-) mice. Moreover, our findings suggest that Fn14 expression is regulated by endothelin-1 (ET-1) in cardiac fibroblasts. We conclude that Fn14 is an endogenous key regulator in cardiac fibrosis and suggest this receptor as potential new target for therapeutic interventions in heart failure.


Assuntos
Hipertrofia Ventricular Direita/prevenção & controle , Miocárdio/patologia , Receptores do Fator de Necrose Tumoral/fisiologia , Disfunção Ventricular Direita/prevenção & controle , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Western Blotting , Diferenciação Celular , Proliferação de Células , Colágeno/metabolismo , Citocina TWEAK , Endotelina-1/fisiologia , Hipertensão Pulmonar Primária Familiar , Fibrose/prevenção & controle , Imunofluorescência , Hipertensão Pulmonar/complicações , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/fisiopatologia , Imuno-Histoquímica , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Miofibroblastos , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Receptor de TWEAK , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Necrose Tumoral/genética , Fatores de Necrose Tumoral/metabolismo , Regulação para Cima , Disfunção Ventricular Direita/metabolismo , Disfunção Ventricular Direita/fisiopatologia
18.
Cardiovasc Res ; 91(4): 587-97, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21628326

RESUMO

AIMS: The transcription factor FoxO3 contributes to anti-hypertrophic signalling in the heart presumably by regulating autophagic-lysosomal and ubiquitin-proteasomal pathways. We wanted to study FoxO3 function in the adult heart in vivo by expressing a constitutively active mutant of FoxO3 in transgenic mice. METHODS AND RESULTS: We generated transgenic mice in which a tetracycline-regulated constitutively active FoxO3 transgene (FoxO3-CA) is controlled by the heart-specific α-myosin heavy chain promoter. Cardiac-specific expression in adult mice resulted in a decrease in heart weight by 25% and a reduction in stroke volume and cardiac output. The decrease in heart size was due to a reduction in the size of individual cardiomyocytes, whereas there was no evidence for increased cell death. FoxO3 activation was accompanied by the initiation of a foetal gene programme with increased expression of ß-myosin heavy chain and natriuretic peptides, and by the activation of AKT and mammalian target of rapamycin signalling. As shown by electron microscopy, FoxO3-CA massively stimulated destruction of sarcomeres and autophagy, and induced expression of LC3-II and BNIP3. When FoxO3-CA expression was shut off in affected mice, cardiac atrophy and dysfunction as well as molecular markers were normalized within 1 month. FoxO3-CA expression did not counteract hypertrophy induced by transverse aortic constriction. CONCLUSION: Heart-specific expression of constitutively active FoxO3 leads to reversible heart atrophy. The reversibility of the phenotype suggests a remarkable ability of the adult myocardium to respond to different regulatory cues.


Assuntos
Autofagia , Fatores de Transcrição Forkhead/fisiologia , Miocárdio/patologia , Animais , Atrofia , Peso Corporal , Modelos Animais de Doenças , Proteína Forkhead Box O3 , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/patologia , Tamanho do Órgão , Fenótipo , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais , Serina-Treonina Quinases TOR/fisiologia , Remodelação Ventricular
19.
Circ Res ; 103(11): 1327-34, 2008 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-18927463

RESUMO

Circulating blood-derived vasculogenic cells improve neovascularization of ischemic tissue by a broad repertoire of potential therapeutic actions. Whereas initial studies documented that the cells incorporate and differentiate to cardiovascular cells, other studies suggested that short-time paracrine mechanisms mediate the beneficial effects. The question remains to what extent a physical incorporation is contributing to the beneficial effects of cell therapy. By using the inducible suicide gene thymidine kinase to deplete transplanted cells, we determined the contribution of physical incorporation in 3 animal models. After acute myocardial infarction, depletion of cells 14 days after infusion resulted in a reduction of capillary density and a substantial deterioration of heart function. Likewise, neovascularization of Matrigel plugs and ischemic limbs was significantly suppressed when infused cells were depleted 7 days after infusion. Induction of cell death in the previously transplanted cells reduced perfusion and led to vascular leakage as evidenced by Evans blue extravasation. These results indicate that physical incorporation and persistence of cells contribute to cell-mediated improvement of neovascularization and cardiac function. Long-term paracrine activities and/or cell intrinsic mechanisms may have contributed to the maintenance of functional improvement.


Assuntos
Coração/fisiopatologia , Isquemia Miocárdica/terapia , Neovascularização Fisiológica/fisiologia , Transplante de Células-Tronco/métodos , Animais , Capilares/fisiopatologia , Técnicas de Cultura de Células , Terapia Baseada em Transplante de Células e Tecidos/métodos , Vasos Coronários/patologia , Feminino , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Humanos , Lentivirus , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/fisiologia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Nus , Isquemia Miocárdica/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA