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










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 550, 2023 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-36754961

RESUMO

Myofibroblasts cause tissue fibrosis by producing extracellular matrix proteins, such as collagens. Humoral factors like TGF-ß, and matrix stiffness are important for collagen production by myofibroblasts. However, the molecular mechanisms regulating their ability to produce collagen remain poorly characterised. Here, we show that vestigial-like family member 3 (VGLL3) is specifically expressed in myofibroblasts from mouse and human fibrotic hearts and promotes collagen production. Further, substrate stiffness triggers VGLL3 translocation into the nucleus through the integrin ß1-Rho-actin pathway. In the nucleus, VGLL3 undergoes liquid-liquid phase separation via its low-complexity domain and is incorporated into non-paraspeckle NONO condensates containing EWS RNA-binding protein 1 (EWSR1). VGLL3 binds EWSR1 and suppresses miR-29b, which targets collagen mRNA. Consistently, cardiac fibrosis after myocardial infarction is significantly attenuated in Vgll3-deficient mice, with increased miR-29b expression. Overall, our results reveal an unrecognised VGLL3-mediated pathway that controls myofibroblasts' collagen production, representing a novel therapeutic target for tissue fibrosis.


Assuntos
MicroRNAs , Miocárdio , Humanos , Camundongos , Animais , Miocárdio/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fibrose , Colágeno/metabolismo , Miofibroblastos/metabolismo , Fatores de Transcrição/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Fibroblastos/metabolismo , Células Cultivadas
2.
FEBS Open Bio ; 13(2): 380-391, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36633120

RESUMO

During myocardial infarction (MI), cardiac cells at the infarcted area undergo cell death. In response, cardiac myofibroblasts, which are mainly differentiated from resident fibroblasts upon inflammation, produce extracellular matrix proteins such as collagen to fill the damaged areas of the heart to prevent cardiac rupture. In this study, we identified a cardioprotective role of G-protein-coupled receptor kinase 5 (GRK5) in MI. GRK5 expression was found to increase in the mouse heart after MI and was highly expressed in cardiac fibroblasts/myofibroblasts. In fibroblasts/myofibroblasts, GRK5 promoted the expression of inflammation-related genes through nuclear factor-κB activation, leading to an increase in the expression levels of fibrosis-related genes. Bone marrow transfer experiments confirmed that GRK5 in fibroblasts/myofibroblasts, but not in infiltrated macrophages in the infarcted area, is mainly responsible for GRK5-mediated inflammation in infarcted hearts. In addition, inflammation and fibrosis at the infarcted area were significantly suppressed in GRK5 knockout mice, resulting in increased mortality compared with that in wild-type mice. These data indicate that GRK5 in cardiac fibroblasts/myofibroblasts promotes inflammation and fibrosis to ameliorate the damage after MI.


Assuntos
Infarto do Miocárdio , Miocárdio , Animais , Camundongos , Colágeno/metabolismo , Fibrose , Inflamação/metabolismo , Camundongos Knockout , Infarto do Miocárdio/genética , Miocárdio/metabolismo
3.
J Biol Chem ; 299(3): 102934, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36690273

RESUMO

Fibrosis is mainly triggered by inflammation in various tissues, such as heart and liver tissues, and eventually leads to their subsequent dysfunction. Fibrosis is characterized by the excessive accumulation of extracellular matrix proteins (e.g., collagens) produced by myofibroblasts. The well-developed actin cytoskeleton of myofibroblasts, one of the main features differentiating them from resident fibroblasts in tissues under inflammatory conditions, contributes to maintaining their ability to produce excessive extracellular matrix proteins. However, the molecular mechanisms via which the actin cytoskeleton promotes the production of fibrosis-related genes in myofibroblasts remain unclear. In this study, we found, via single-cell analysis, that developmentally regulated brain protein (drebrin), an actin-binding protein, was specifically expressed in cardiac myofibroblasts with a well-developed actin cytoskeleton in fibrotic hearts. Moreover, our immunocytochemistry analysis revealed that drebrin promoted actin cytoskeleton formation and myocardin-related transcription factor-serum response factor signaling. Comprehensive single-cell analysis and RNA-Seq revealed that the expression of collagen triple helix repeat containing 1 (Cthrc1), a fibrosis-promoting secreted protein, was regulated by drebrin in cardiac myofibroblasts via myocardin-related transcription factor-serum response factor signaling. Furthermore, we observed the profibrotic effects of drebrin exerted via actin cytoskeleton formation and the Cthrc1 expression regulation by drebrin in liver myofibroblasts (hepatic stellate cells). Importantly, RNA-Seq demonstrated that drebrin expression levels increased in human fibrotic heart and liver tissues. In summary, our results indicated that the well-developed actin cytoskeleton and Cthrc1 expression due to drebrin in myofibroblasts promoted cardiac and hepatic fibrosis, suggesting that drebrin is a therapeutic target molecule for fibrosis.


Assuntos
Citoesqueleto de Actina , Proteínas da Matriz Extracelular , Fibrose , Miofibroblastos , Neuropeptídeos , Humanos , Citoesqueleto de Actina/metabolismo , Miofibroblastos/patologia , Fibrose/fisiopatologia , Análise da Expressão Gênica de Célula Única , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Diferenciação Celular/fisiologia , Transdução de Sinais , Células Estreladas do Fígado/metabolismo , Cardiopatias/fisiopatologia , Cirrose Hepática/fisiopatologia
4.
Biochem Biophys Res Commun ; 561: 180-186, 2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-34023784

RESUMO

Fibrosis is a condition characterized by the overproduction of extracellular matrix (ECM) components (e.g., collagen) in the myofibroblasts, causing tissue hardening and eventual organ dysfunction. Currently, the molecular mechanisms that regulate ECM production in the myofibroblasts are still obscure. In this study, we investigated the function of GPRC5B in the cardiac and lung myofibroblasts using real-time RT-PCR and siRNA-mediated knockdown. We discovered a significantly high expression of Gprc5b in the tissues of the fibrosis mice models and confirmed that Gprc5b was consistently expressed in the myofibroblasts of fibrotic hearts and lungs. We also found that Gprc5b expression was associated and may be dependent on the actin-MRTF-SRF signaling pathway. Notably, we observed that Gprc5b knockdown reduced the expression of collagen genes in the cardiac and lung myofibroblasts. Therefore, our findings reveal that GPRC5B enhances collagen production in the myofibroblasts, which directly promotes fibrosis in the tissues.


Assuntos
Colágeno/metabolismo , Fibrose/patologia , Coração/fisiopatologia , Fígado/metabolismo , Pulmão/metabolismo , Miofibroblastos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Fibrose/metabolismo , Fígado/patologia , Pulmão/patologia , Masculino , Camundongos , Miofibroblastos/patologia , Transdução de Sinais
5.
Nat Commun ; 12(1): 2783, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33986294

RESUMO

Dysfunction of the circadian clock has been implicated in the pathogenesis of cardiovascular disease. The CLOCK protein is a core molecular component of the circadian oscillator, so that mice with a mutated Clock gene (Clk/Clk) exhibit abnormal rhythms in numerous physiological processes. However, here we report that chronic kidney disease (CKD)-induced cardiac inflammation and fibrosis are attenuated in Clk/Clk mice even though they have high blood pressure and increased serum angiotensin II levels. A search for the underlying cause of the attenuation of heart disorder in Clk/Clk mice with 5/6 nephrectomy (5/6Nx) led to identification of the monocytic expression of G protein-coupled receptor 68 (GPR68) as a risk factor of CKD-induced inflammation and fibrosis of heart. 5/6Nx induces the expression of GPR68 in circulating monocytes via altered CLOCK activation by increasing serum levels of retinol and its binding protein (RBP4). The high-GPR68-expressing monocytes have increased potential for producing inflammatory cytokines, and their cardiac infiltration under CKD conditions exacerbates inflammation and fibrosis of heart. Serum retinol and RBP4 levels in CKD patients are also sufficient to induce the expression of GPR68 in human monocytes. Our present study reveals an uncovered role of monocytic clock genes in CKD-induced heart failure.


Assuntos
Proteínas CLOCK/genética , Relógios Circadianos/genética , Ritmo Circadiano/fisiologia , Cardiopatias/patologia , Monócitos/metabolismo , Insuficiência Renal Crônica/patologia , Fatores de Transcrição ARNTL/metabolismo , Animais , Proteínas CLOCK/metabolismo , Células Cultivadas , Ritmo Circadiano/genética , Citocinas/biossíntese , Fibrose/patologia , Hipertensão/genética , Hipertensão/patologia , Inflamação/genética , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/metabolismo
6.
Medicine (Baltimore) ; 99(29): e20867, 2020 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-32702828

RESUMO

RATIONALE: Phagocytosis is an important physiological process for eliminating unnecessary substances or dead cells after tissue damage, such as inflammation or infarction. Phagocytosis was previously considered to be mainly performed by professional phagocytotic cells, such as macrophages. In contrast, we previously demonstrated that the phagocytosis of dead cells and unnecessary substances by myofibroblasts is as important as that by professional phagocytotic cells in myocardial infarction. Based on our discovery, we speculated that phagocytosis by myofibroblasts may be a more common pathological phenomenon also in other diseases than previously believed. PATIENT CONCERNS: A 44-year-old male patient with atopic dermatitis developed a cutaneous reddish nodule with an underlying induration on his thigh. INTERVENTIONS: The cutaneous lesion was surgically removed. DIAGNOSES: Histopathological examination demonstrated that the cutaneous lesion had solid infiltration by inflammatory cells, namely, plasma cells, histiocytes, and lymphocytes, in the dermis. The cutaneous lesion included mucinosis in the dermis. Inside the mucinosis, we detected cells with clear areas of mucinous substances. Some of the cells were α-smooth muscle actin-positive myofibroblasts. Electron microscopic images demonstrated that there were collagen bands in the cells with mucinous engulfment. Based on these pieces of evidence, we conclude that these mucinous phagocytotic cells were myofibroblasts, not professional phagocytotic cells, such as macrophages. OUTCOMES: There was no recurrence of the lesion. LESSONS: The clinical appearance of this case resembled that of previously reported solitary cutaneous focal mucinoses. However, our case had distinctive characteristics, such as the phagocytosis of mucinous substances by myofibroblasts, multiple mucinous lesions in a single eruption, and the presence of inflammatory cells, which have not been previously reported. For this distinct cutaneous lesion, a clear dermatological and pathological name has yet to be determined. We propose "myofibroblast phagocytic cutaneous mucinosis" as a candidate name. In addition, our discoveries suggest that phagocytosis by myofibroblasts is not rare but rather is a common pathological phenomenon that has been undetected or unrecognized.


Assuntos
Mucinoses/patologia , Miofibroblastos/fisiologia , Fagocitose , Dermatopatias/patologia , Adulto , Humanos , Masculino , Microscopia Eletrônica , Mucinoses/cirurgia , Dermatopatias/cirurgia
7.
Biochem Biophys Res Commun ; 529(2): 224-230, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32703415

RESUMO

Fibrosis is attributed to excess deposition of extracellular matrix (ECM) proteins including collagen and is associated with various organ dysfunction. This excessive ECM is produced by myofibroblasts, which are differentiated from various cells by a variety of stimuli, represented by TGF-ß. However, molecular mechanisms for the regulation of ECM production in myofibroblasts remain obscure. In this study, we demonstrate that the expression of drebrin, which binds to and increases the stability of actin filament in neurons, is increased in mouse hearts and lungs upon fibrosis. Drebrin is mainly expressed in myofibroblasts in the fibrotic hearts and lungs and promotes the expression of fibrosis-related genes, such as Acta2 and Col1a1. Taken together, our study identifies drebrin as a molecule that promotes the production of fibrosis-related genes in myofibroblasts.


Assuntos
Pulmão/patologia , Miocárdio/patologia , Miofibroblastos/patologia , Neuropeptídeos/genética , Animais , Diferenciação Celular , Células Cultivadas , Fibrose , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Miofibroblastos/metabolismo , Células NIH 3T3 , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia , Regulação para Cima
8.
FASEB J ; 34(6): 8749-8763, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32385915

RESUMO

Leukotriene B4 receptor 1 (BLT1), a high-affinity G-protein-coupled receptor for leukotriene B4 (LTB4 ), is expressed on various inflammatory cells and plays critical roles in several inflammatory diseases. In myocardial infarction (MI), various inflammatory cells are known to be recruited to the infarcted area, but the function of BLT1 in MI is poorly understood. Here, we investigated the role of BLT1 in MI and the therapeutic effect of a BLT1 antagonist, ONO-4057, on MI. Mice with infarcted hearts showed increased BLT1 expression and LTB4 levels. BLT1-knockout mice with infarcted hearts exhibited attenuated leukocyte infiltration, proinflammatory cytokine production, and cell death, which led to reduced mortality and improved cardiac function after MI. Bone-marrow transplantation studies showed that BLT1 expressed on bone marrow-derived cells was responsible for the exacerbation of inflammation in infarcted hearts. Furthermore, ONO-4057 administration attenuated the inflammatory responses in hearts surgically treated for MI, which resulted in reduced mortality and improved cardiac function after MI. Our study demonstrated that BLT1 contributes to excessive inflammation after MI and could represent a new therapeutic target for MI.


Assuntos
Inflamação/metabolismo , Infarto do Miocárdio/metabolismo , Receptores do Leucotrieno B4/metabolismo , Animais , Modelos Animais de Doenças , Leucotrieno B4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/fisiologia
9.
J Biochem ; 168(1): 1-6, 2020 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-32302392

RESUMO

Myocardial infarction is one of the major causes of death worldwide. Many heart cells die during myocardial infarction through various processes such as necrosis, apoptosis, necroptosis, autophagy-related cell death, pyroptosis and ferroptosis. These dead cells in infarcted hearts expose the so-called 'eat-me' signals, such as phosphatidylserine, on their surfaces, enhancing their removal by professional and non-professional phagocytes. Clearance of dead cells by phagocytes in the diseased hearts plays a crucial role in the pathology of myocardial infarction by inhibiting the inflammatory responses caused by the leakage of contents from dead cells. This review focuses on the rapidly growing understanding of the molecular mechanisms of dead cell phagocytosis, termed efferocytosis, during myocardial infarction, which contributes to the pathophysiology of myocardial infarction.


Assuntos
Apoptose , Inflamação/fisiopatologia , Macrófagos/fisiologia , Infarto do Miocárdio/fisiopatologia , Fagócitos/fisiologia , Fagocitose , Humanos , Transdução de Sinais
11.
Medicine (Baltimore) ; 98(24): e15888, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31192921

RESUMO

RATIONALE: Suppression and of cancer metastasis is one of the most important issues in cancer care. Considering the typical clinical course of metastases, cancer cells might prefer certain environments or conditions. However, favorable environments for cancer metastasis have not been clearly identified. We had previously described a case of dual, yet separate, pancreatic and colon cancer, in which the metastatic pancreatic cancer was localized at the invasive portion of the colon cancer. We hypothesized that metastatic pancreatic cancer took over the colon cancer microenvironment. PATIENT CONCERNS: We experienced an another case of double cancer in a 65-year-old man who had lung squamous cell carcinoma and an independent pancreatic adenocarcinoma that metastasized to the liver as well as to the lung cancer lesion and pulmonary fibrotic regions associated with pneumothorax and bronchiolization. INTERVENTIONS: The pneumothorax could not be controlled by conservative treatment. Thus, an emergency surgery with partial resection of the lower lobe of right lung was performed. DIAGNOSES: We found multiple pancreatic cancer metastases in the lung cancer and fibrotic lesions in the surgical specimen. However, we detected no metastasis in normal lung tissues except inside small arteries, although the lung cancer and fibrotic tissue areas were smaller than the normal lung tissue areas in the surgical specimen. OUTCOMES: The patient died 50 days after the surgery. LESSONS: This case may thus provide evidence to strengthen our hypothesis that pancreatic cancer prefers to metastasize to other independent cancer lesions, overtaking the cancer microenvironment constructed by other independent cancers. The lung cancer microenvironment, rich in myofibroblasts and/or cancer-associated fibroblasts, might be suitable for pancreatic carcinoma metastasis. In addition, we propose the hypothesis that compared with normal tissues, noncancerous fibrotic lesions are preferable destinations for cancer metastasis. Furthermore, metastasis of pancreatic carcinoma to lung cancer and fibrotic tissues might be more common, although such cases have not been previously reported.


Assuntos
Carcinoma de Células Escamosas/cirurgia , Neoplasias Pulmonares/cirurgia , Segunda Neoplasia Primária/cirurgia , Neoplasias Pancreáticas/cirurgia , Pneumotórax/cirurgia , Idoso , Carcinoma de Células Escamosas/diagnóstico , Evolução Fatal , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/secundário , Masculino , Segunda Neoplasia Primária/diagnóstico , Neoplasias Pancreáticas/diagnóstico , Pneumotórax/diagnóstico , Pneumotórax/etiologia , Fibrose Pulmonar/diagnóstico , Fibrose Pulmonar/cirurgia , Microambiente Tumoral , Neoplasias Pancreáticas
12.
Development ; 145(14)2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-29986870

RESUMO

Erk5 belongs to the mitogen-activated protein kinase (MAPK) family. Following its phosphorylation by Mek5, Erk5 modulates several signaling pathways in a number of cell types. In this study, we demonstrated that Erk5 inactivation in mesenchymal cells causes abnormalities in skeletal development by inducing Sox9, an important transcription factor of skeletogenesis. We further demonstrate that Erk5 directly phosphorylates and activates Smurf2 (a ubiquitin E3 ligase) at Thr249, which promotes the proteasomal degradation of Smad proteins and phosphorylates Smad1 at Ser206 in the linker region known to trigger its proteasomal degradation by Smurf1. Smads transcriptionally activated the expression of Sox9 in mesenchymal cells. Accordingly, removal of one Sox9 allele in mesenchymal cells from Erk5-deficient mice rescued some abnormalities of skeletogenesis. These findings highlight the importance of the Mek5-Erk5-Smurf-Smad-Sox9 axis in mammalian skeletogenesis.


Assuntos
Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Osteogênese , Fatores de Transcrição SOX9/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Diferenciação Celular , Condrogênese , Humanos , Mesoderma/citologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteólise , Crânio/anormalidades , Ubiquitina/metabolismo , Ubiquitinação
13.
Nat Neurosci ; 21(6): 820-833, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29713080

RESUMO

Chronic morphine exposure upregulates adenylate cyclase signaling and reduces analgesic efficacy, a condition known as opioid tolerance. Nonopioid neurotransmitters can enhance morphine tolerance, but the mechanism for this is poorly understood. We show that morphine tolerance was delayed in mice lacking vasopressin 1b receptors (V1bRs) or after administration of V1bR antagonist into the rostral ventromedial medulla, where transcripts for V1bRs and µ-opioid receptors are co-localized. Vasopressin increased morphine-binding affinity in cells expressing both V1bR and µ-opioid receptors. Complex formation among V1bR, ß-arrestin-2, and µ-opioid receptor resulted in vasopressin-mediated upregulation of ERK phosphorylation and adenylate cyclase sensitization. A leucine-rich segment in the V1bR C-terminus was necessary for the association with ß-arrestin-2. Deletion of this leucine-rich segment increased morphine analgesia and reduced vasopressin-mediated adenylate cyclase sensitization. These findings indicate that inhibition of µ-opioid-receptor-associated V1bR provides an approach for enhancing morphine analgesia without increasing analgesic tolerance.


Assuntos
Tolerância a Medicamentos/genética , Morfina/farmacologia , Entorpecentes/farmacologia , Receptores Opioides mu/metabolismo , Receptores de Vasopressinas/metabolismo , beta-Arrestina 2/metabolismo , Adenilil Ciclases/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Injeções , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Bulbo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/farmacocinética , Dependência de Morfina/psicologia , Entorpecentes/farmacocinética , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Fosforilação , Receptores Opioides mu/genética , Receptores de Vasopressinas/genética , beta-Arrestina 2/genética
14.
Cardiovasc Res ; 113(10): 1243-1255, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28898995

RESUMO

AIMS: Transient receptor potential cation channel subfamily melastatin member 4 (TRPM4), a Ca2+-activated nonselective cation channel abundantly expressed in the heart, has been implicated in conduction block and other arrhythmic propensities associated with cardiac remodelling and injury. The present study aimed to quantitatively evaluate the arrhythmogenic potential of TRPM4. METHODS AND RESULTS: Patch clamp and biochemical analyses were performed using expression system and an immortalized atrial cardiomyocyte cell line (HL-1), and numerical model simulation was employed. After rapid desensitization, robust reactivation of TRPM4 channels required high micromolar concentrations of Ca2+. However, upon evaluation with a newly devised, ionomycin-permeabilized cell-attached (Iono-C/A) recording technique, submicromolar concentrations of Ca2+ (apparent Kd = ∼500 nM) were enough to activate this channel. Similar submicromolar Ca2+ dependency was also observed with sharp electrode whole-cell recording and in experiments coexpressing TRPM4 and L-type voltage-dependent Ca2+ channels. Numerical simulations using a number of action potential (AP) models (HL-1, Nygren, Luo-Rudy) incorporating the Ca2+- and voltage-dependent gating parameters of TRPM4, as assessed by Iono-C/A recording, indicated that a few-fold increase in TRPM4 activity is sufficient to delay late AP repolarization and further increases (≥ six-fold) evoke early afterdepolarization. These model predictions are consistent with electrophysiological data from angiotensin II-treated HL-1 cells in which TRPM4 expression and activity were enhanced. CONCLUSIONS: These results collectively indicate that the TRPM4 channel is activated by a physiological range of Ca2+ concentrations and its excessive activity can cause arrhythmic changes. Moreover, these results demonstrate potential utility of the first AP models incorporating TRPM4 gating for in silico assessment of arrhythmogenicity in remodelling cardiac tissue.


Assuntos
Potenciais de Ação , Arritmias Cardíacas/metabolismo , Simulação por Computador , Átrios do Coração/metabolismo , Frequência Cardíaca , Modelos Cardiovasculares , Miócitos Cardíacos/metabolismo , Análise Numérica Assistida por Computador , Canais de Cátion TRPM/metabolismo , Potenciais de Ação/efeitos dos fármacos , Angiotensina II/farmacologia , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Sinalização do Cálcio , Células HEK293 , Átrios do Coração/efeitos dos fármacos , Átrios do Coração/fisiopatologia , Frequência Cardíaca/efeitos dos fármacos , Humanos , Cinética , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp , Período Refratário Eletrofisiológico , Canais de Cátion TRPM/genética
15.
Sci Rep ; 7(1): 7812, 2017 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-28798316

RESUMO

Myocardial infarction (MI) is an ischaemic heart condition caused by the occlusion of coronary arteries. Following MI, lactic acid from anaerobic glycolysis increases and infiltrating immune cells produce severe inflammation, which leads to acidosis in the ischaemic heart. However, the physiological implication of this pH reduction remains largely unknown. T-cell death-associated gene 8 (TDAG8) is a proton-sensing G protein-coupled receptor found on cardiac macrophages that recognise increases in extracellular protons. We demonstrated that TDAG8 negatively regulates the transcription of the chemokine Ccl20. The infarcted hearts of TDAG8 KO mice showed an increase in CCL20 expression and the number of infiltrating IL-17A-producing γδT cells that express CCR6, a receptor for CCL20. Accordingly, excessive IL-17A production, which is linked to the functional deterioration after MI, was observed in MI-operated TDAG8 KO mice. The survival rate and cardiac function significantly decreased in TDAG8 KO mice compared with those in wild-type mice after MI. Thus, our results suggest that TDAG8 is a key regulator of MI and a potential therapeutic target.


Assuntos
Quimiocina CCL20/genética , Infarto do Miocárdio/genética , Animais , Quimiocina CCL20/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Interleucina-17/metabolismo , Linfócitos Intraepiteliais/imunologia , Camundongos , Camundongos Knockout , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/metabolismo , Receptores CCR6/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Análise de Sobrevida , Transcrição Gênica
16.
Prog Biophys Mol Biol ; 130(Pt B): 315-322, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28668597

RESUMO

Mitochondria are an important source of reactive oxygen species (ROS). Although it has been reported that myocardial stretch increases cellular ROS production by activating nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2), referred to as X-ROS signalling, the involvement of mitochondria in X-ROS is not clear. Mitochondria are organelles that generate adenosine triphosphate (ATP) for cellular energy needs, which are mechanical-load-dependent. Therefore, it would not be surprising if these organelles had mechano-sensitive functions associated with stretch-induced ROS production. In the present study, we investigated the relation between X-ROS and mitochondrial stretch-sensitive responses in isolated mouse cardiac myocytes. The cells were subjected to 10% axial stretch using computer-controlled, piezo-manipulated carbon fibres attached to both cell ends. Cellular ROS production and mitochondrial membrane potential (Δψm) were assessed optically by confocal microscopy. The axial stretch increased ROS production and hyperpolarised Δψm. Treatment with a mitochondrial metabolic uncoupler, carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP), at 0.5 µM did not suppress stretch-induced ROS production, whereas treatment with a respiratory Complex III inhibitor, antimycin A (5 µM), blunted the response. Although NOX inhibition by apocynin abrogated the stretch-induced ROS production, it did not suppress stretch-induced hyperpolarisation of Δψm. These results suggest that stretch causes activation of the respiratory chain to hyperpolarise Δψm, followed by NOX activation, which increases ROS production.


Assuntos
Fenômenos Mecânicos , Mitocôndrias/metabolismo , Miócitos Cardíacos/citologia , Animais , Fenômenos Biomecânicos , Potencial da Membrana Mitocondrial , Camundongos , Espécies Reativas de Oxigênio/metabolismo
17.
J Biol Chem ; 292(25): 10586-10599, 2017 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-28487374

RESUMO

Dioxin and related chemicals alter the expression of a number of genes by activating the aryl hydrocarbon receptors (AHR) to produce a variety of disorders including hepatotoxicity. However, it remains largely unknown how these changes in gene expression are linked to toxicity. To address this issue, we initially examined the effect of 2,3,7,8-tetrachrolodibenzo-p-dioxin (TCDD), a most toxic dioxin, on the hepatic and serum metabolome in male pubertal rats and found that TCDD causes many changes in the level of fatty acids, bile acids, amino acids, and their metabolites. Among these findings was the discovery that TCDD increases the content of leukotriene B4 (LTB4), an inducer of inflammation due to the activation of leukocytes, in the liver of rats and mice. Further analyses suggested that an increase in LTB4 comes from a dual mechanism consisting of an induction of arachidonate lipoxygenase-5, a rate-limiting enzyme in LTB4 synthesis, and the down-regulation of LTC4 synthase, an enzyme that converts LTA4 to LTC4. The above changes required AHR activation, because the same was not observed in AHR knock-out rats. In agreement with LTB4 accumulation, TCDD caused the marked infiltration of neutrophils into the liver. However, deleting LTB4 receptors (BLT1) blocked this effect. A TCDD-produced increase in the mRNA expression of inflammatory markers, including tumor-necrosis factor and hepatic damage, was also suppressed in BLT1-null mice. The above observations focusing on metabolomic changes provide novel evidence that TCDD accumulates LTB4 in the liver by an AHR-dependent induction of LTB4 biosynthesis to cause hepatotoxicity through neutrophil activation.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Dioxinas/toxicidade , Leucotrieno B4/biossíntese , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/patologia , Leucotrieno B4/genética , Ativação de Neutrófilo/efeitos dos fármacos , Infiltração de Neutrófilos/genética , Neutrófilos/patologia , Ratos , Ratos Mutantes , Receptores de Hidrocarboneto Arílico/genética , Receptores do Leucotrieno B4/genética , Receptores do Leucotrieno B4/metabolismo
18.
Bio Protoc ; 7(18): e2552, 2017 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-34541198

RESUMO

In myocardial infarction (MI), a plenty of cardiomyocytes undergo necrosis and necroptosis due to the lack of oxygen and nutrients. The dead cardiomyocytes are promptly engulfed by phagocytes. When the dead cells are not engulfed, the noxious contents of the cells are released outside, and thus, induce inflammation, and obstruct the function of organs. Therefore, phagocytosis is crucial for maintaining homeostasis of organs. Herein, we describe a protocol of an in vitro phagocytosis assay of necroptotic cells.

19.
Bio Protoc ; 7(18): e2553, 2017 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-34541199

RESUMO

In myocardial infarction (MI), a number of cardiomyocytes undergo apoptosis. These apoptotic cardiomyocytes are promptly engulfed by phagocytes. If the dead cells are not engulfed, their noxious contents are released outside, resulting in induction of inflammation. Therefore, the removal of these dead cells is necessary. However, the contribution of each phagocyte type to the removal of apoptotic cells in infarcted hearts remains unresolved. Here, we describe an in vitro protocol for a phagocytosis assay to compare the engulfment ability of cardiac macrophages and cardiac myofibroblasts.

20.
J Clin Invest ; 127(1): 383-401, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-27918308

RESUMO

Myocardial infarction (MI) results in the generation of dead cells in the infarcted area. These cells are swiftly removed by phagocytes to minimize inflammation and limit expansion of the damaged area. However, the types of cells and molecules responsible for the engulfment of dead cells in the infarcted area remain largely unknown. In this study, we demonstrated that cardiac myofibroblasts, which execute tissue fibrosis by producing extracellular matrix proteins, efficiently engulf dead cells. Furthermore, we identified a population of cardiac myofibroblasts that appears in the heart after MI in humans and mice. We found that these cardiac myofibroblasts secrete milk fat globule-epidermal growth factor 8 (MFG-E8), which promotes apoptotic engulfment, and determined that serum response factor is important for MFG-E8 production in myofibroblasts. Following MFG-E8-mediated engulfment of apoptotic cells, myofibroblasts acquired antiinflammatory properties. MFG-E8 deficiency in mice led to the accumulation of unengulfed dead cells after MI, resulting in exacerbated inflammatory responses and a substantial decrease in survival. Moreover, MFG-E8 administration into infarcted hearts restored cardiac function and morphology. MFG-E8-producing myofibroblasts mainly originated from resident cardiac fibroblasts and cells that underwent endothelial-mesenchymal transition in the heart. Together, our results reveal previously unrecognized roles of myofibroblasts in regulating apoptotic engulfment and a fundamental importance of these cells in recovery from MI.


Assuntos
Antígenos de Superfície/metabolismo , Apoptose , Transição Epitelial-Mesenquimal , Proteínas do Leite/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Miofibroblastos/metabolismo , Animais , Antígenos de Superfície/genética , Sobrevivência Celular/genética , Masculino , Camundongos , Camundongos Knockout , Proteínas do Leite/genética , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miocárdio/patologia , Miofibroblastos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...