Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Environ Geochem Health ; 46(6): 206, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38724672

RESUMO

After confirming that formaldehyde (FA) is carcinogenic, many studies were conducted in different countries to investigate this finding. Therefore, according to the dispersion of related studies, a bibliometric review of the current literature was performed with the aim of better understanding the exposure to FA and the resulting health risk, for the first time, using the Scopus database and the two open-source software packages, Bibliometrix R package. After screening the documents in Excel, the data was analyzed based on three aspects including performance analysis, conceptual structure, and intellectual structure, and the results were presented in tables and diagrams. A total of 468 documents were analyzed over period 1977-2023, in which 1956 authors from 56 countries participated. The number of scientific publications has grown significantly from 1977 (n = 1) to 2022 (n = 19). Zhang Y., from the Yale School of Public Health (USA), was identified as the most impactful author in this field. The Science of the Total Environment journal was identified as the main source of articles related to exposure to formaldehyde by publishing 25 studies. The United States and China were the most active countries with the most international collaboration. The main topics investigated during these 46 years included "formaldehyde" and "health risk assessment", which have taken new directions in recent years with the emergence of the keyword "asthma". The present study provides a comprehensive view of the growth and evolution of studies related to formaldehyde and the resulting health risks, which can provide a better understanding of existing research gaps and new and emerging issues.


Assuntos
Exposição Ambiental , Formaldeído , Formaldeído/toxicidade , Humanos , Medição de Risco , Bibliometria
2.
AAPS PharmSciTech ; 24(3): 79, 2023 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-36918482

RESUMO

All-trans retinoic acid and arsenic trioxide are the leading choices for the treatment of acute promyelocytic leukemia. Notwithstanding the impressive differentiative properties of all-trans retinoic acid and the apoptotic properties of arsenic trioxide, some problems still occur in acute promyelocytic leukemia treatment. These problems are due to patients' relapses, mainly related to changes in the ligand-binding domain of RARα (retinoic acid receptor α) and the cardiotoxic effects caused by arsenic trioxide. We previously developed a self-nanoemulsifying drug delivery system enriched with tocotrienols to deliver all-trans retinoic acid (SNEDDS-TRF-ATRA). Herein, we have evaluated if tocotrienols can help revert ATRA resistance in an APL cell line (NB4-R2 compared to sensitive NB4 cells) and mitigate the cardiotoxic effects of arsenic trioxide in a murine model. SNEDDS-TRF-ATRA enhanced all-trans retinoic acid cytotoxicity in NB4-R2 (resistant) cells but not in NB4 (sensitive) cells. Moreover, SNEDDS-TRF-ATRA did not significantly change the differentiative properties of all-trans retinoic acid in both NB4 and NB4-R2 cells. Combined administration of SNEDDS-TRF-ATRA and arsenic trioxide could revert QTc interval prolongation caused by ATO but evoked other electrocardiogram alterations in mice, such as T wave flattening. Therefore, SNEDDS-TRF-ATRA may enhance the antileukemic properties of all-trans retinoic acid but may influence ECG changes caused by arsenic trioxide administration. SNEDDS-TRF-ATRA presents cytotoxicity in resistant APL cells (NB4-R2). Combined administration of ATO and SNEDDS-TRF-ATRA in mice prevented the prolongation of the QTc interval caused by ATO but evoked ECG abnormalities such as T wave flattening.


Assuntos
Leucemia Promielocítica Aguda , Tocotrienóis , Animais , Camundongos , Trióxido de Arsênio/farmacologia , Trióxido de Arsênio/uso terapêutico , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Tocotrienóis/uso terapêutico , Tretinoína/farmacologia , Tretinoína/uso terapêutico , Eletrocardiografia , Óxidos/farmacologia , Óxidos/uso terapêutico
3.
Cells ; 12(4)2023 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-36831249

RESUMO

BACKGROUND: The use of mesenchymal stem cells (MSCs) appears to be a promising therapeutic approach for cardiac repair after myocardial infarction. However, clinical trials have revealed the need to improve their therapeutic efficacy. Recent evidence demonstrated that mitochondria undergo spontaneous transfer from damaged cells to MSCs, resulting in the activation of the cytoprotective and pro-angiogenic functions of recipient MSCs. Based on these observations, we investigated whether the preconditioning of MSCs with mitochondria could optimize their therapeutic potential for ischemic heart disease. METHODS: Human MSCs were exposed to mitochondria isolated from human fetal cardiomyocytes. After 24 h, the effects of mitochondria preconditioning on the MSCs' function were analyzed both in vitro and in vivo. RESULTS: We found that cardiac mitochondria-preconditioning improved the proliferation and repair properties of MSCs in vitro. Mechanistically, cardiac mitochondria mediate their stimulatory effects through the production of reactive oxygen species, which trigger their own degradation in recipient MSCs. These effects were further confirmed in vivo, as the mitochondria preconditioning of MSCs potentiated their therapeutic efficacy on cardiac function following their engraftment into infarcted mouse hearts. CONCLUSIONS: The preconditioning of MSCs with the artificial transfer of cardiac mitochondria appears to be promising strategy to improve the efficacy of MSC-based cell therapy in ischemic heart disease.


Assuntos
Células-Tronco Mesenquimais , Infarto do Miocárdio , Isquemia Miocárdica , Camundongos , Animais , Humanos , Isquemia Miocárdica/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Mitocôndrias Cardíacas/metabolismo , Células-Tronco Mesenquimais/metabolismo
4.
Int J Mol Sci ; 24(2)2023 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-36674841

RESUMO

Acute heart failure (AHF) due to acute myocardial infarction (AMI) is likely to involve cardiogenic shock (CS), with neuro-hormonal activation. A relationship between AHF, CS and vasopressin response is suspected. This study aimed to investigate the implication of vasopressin on hemodynamic parameters and tissue perfusion at the early phase of CS complicating AMI. Experiments were performed on male Wistar rats submitted or not to left coronary artery ligation (AMI and Sham). Six groups were studied Sham and AMI treated or not with either a vasopressin antagonist SR-49059 (Sham-SR, AMI-SR) or agonist terlipressin (Sham-TLP, AMI-TLP). Animals were sacrificed one day after surgery (D1) and after hemodynamic parameters determination. Vascular responses to vasopressin were evaluated, ex vivo, on aorta. AHF was defined by a left ventricular ejection fraction below 40%. CS was defined by AHF plus tissue hypoperfusion evidenced by elevated serum lactate level or low mesenteric oxygen saturation (SmO2) at D1. Mortality rates were 40% in AMI, 0% in AMI-SR and 33% in AMI-TLP. Immediately after surgery, a sharp decrease in SmO2 was observed in all groups. At D1, SmO2 recovered in Sham and in SR-treated animals while it remained low in AMI and further decreased in TLP-treated groups. The incidence of CS among AHF animals was 72% in AMI or AMI-TLP while it was reduced to 25% in AMI-SR. Plasma copeptin level was increased by AMI. Maximal contractile response to vasopressin was decreased in AMI (32%) as in TLP- and SR- treated groups regardless of ligation. Increased vasopressin secretion occurring in the early phase of AMI may be responsible of mesenteric hypoperfusion resulting in tissue hypoxia. Treatment with a vasopressin antagonist enhanced mesenteric perfusion and improve survival. This could be an interesting therapeutic strategy to prevent progression to cardiogenic shock.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Masculino , Ratos , Animais , Choque Cardiogênico/etiologia , Volume Sistólico , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Antagonistas dos Receptores de Hormônios Antidiuréticos/uso terapêutico , Função Ventricular Esquerda , Ratos Wistar , Infarto do Miocárdio/complicações , Infarto do Miocárdio/terapia , Insuficiência Cardíaca/etiologia , Vasopressinas/farmacologia
5.
Stoch Environ Res Risk Assess ; 37(1): 175-184, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35965492

RESUMO

The main objectives of this study were to (i) assess variation within fine particles (PM2.5) and tropospheric ozone (O3) time series in Khorramabad (Iran) between 2019 (before) and 2020 (during COVID-19 pandemic); (ii) assess relationship between PM2.5 and O3, the PM2.5/O3 ratio, and energy consumption; and (iii) estimate the health effects of exposure to ambient PM2.5 and O3. From hourly PM2.5 and O3 concentrations, we applied both linear-log and integrated exposure-response functions, city-specific relative risk, and baseline incidence values to estimate the health effects over time. A significant correlation was found between PM2.5 and O3 (r =-0.46 in 2019, r =-0.55 in 2020, p < 0.05). The number of premature deaths for all non-accidental causes (27.5 and 24.6), ischemic heart disease (7.3 and 6.3), chronic obstructive pulmonary disease (17 and 19.2), and lung cancer (9.2 and 6.25) attributed to ambient PM2.5 exposure and for respiratory diseases (4.7 and 5.4) for exposure to O3 above 10 µg m-3 for people older than 30-year-old were obtained in 2019 and 2020. The number of years of life lost declined by 11.6% in 2020 and exposure to PM2.5 reduced the life expectancy by 0.58 and 0.45 years, respectively in 2019 and 2020. Compared to 2019, the restrictive measures associated to COVID-19 pandemic led to reduction in PM2.5 (-25.5%) and an increase of O3 concentration (+ 8.0%) in Khorramabad.

6.
Development ; 149(19)2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-36239412

RESUMO

The binding of 17ß-oestradiol to oestrogen receptor alpha (ERα) plays a crucial role in the control of reproduction, acting through both nuclear and membrane-initiated signalling. To study the physiological role of membrane ERα in the reproductive system, we used the C451A-ERα mouse model with selective loss of function of membrane ERα. Despite C451A-ERα mice being described as sterile, daily weighing and ultrasound imaging revealed that homozygous females do become pregnant, allowing the investigation of the role of ERα during pregnancy for the first time. All neonatal deaths of the mutant offspring mice resulted from delayed parturition associated with failure in pre-term progesterone withdrawal. Moreover, pregnant C451A-ERα females exhibited partial intrauterine embryo arrest at about E9.5. The observed embryonic lethality resulted from altered expansion of Tpbpa-positive spiral artery-associated trophoblast giant cells into the utero-placental unit, which is associated with an imbalance in expression of angiogenic factors. Together, these processes control the trophoblast-mediated spiral arterial remodelling. Hence, loss of membrane ERα within maternal tissues clearly alters the activity of invasive trophoblast cells during placentogenesis. This previously unreported function of membrane ERα could open new avenues towards a better understanding of human pregnancy-associated pathologies.


Assuntos
Receptor alfa de Estrogênio , Trofoblastos , Animais , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Feminino , Fertilidade , Humanos , Camundongos , Placenta/metabolismo , Gravidez , Progesterona/metabolismo , Receptores de Estrogênio/metabolismo , Trofoblastos/metabolismo
8.
Environ Res ; 207: 112218, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-34655608

RESUMO

This study investigates potential influence of urban trees on black carbon (BC) removal by Norway spruce and silver birch along with the BC formation, mass concentration in air, and source apportionment. The main sources of BC in urban areas are transport, household and industry. BC concentrations monitored in urban background station in Vilnius (Lithuania) showed that biomass burning was a significant contributor to BC emissions even during warm period of the year. Therefore, BC emission levels were determined for the most common biomass fuels (mixed wood pellets, oak, ash, birch and spruce firewood) and two types of agro-biomass (triticale and rapeseed straw pellets) burned in modern and old heating systems. The highest emissions were obtained for biomass fuels especially birch firewood. BC aerosol particles produced by the condensation mechanism during the combustion processes were found in all samples taken from the leaf surface. The short-term effect of BC exposure on photosynthetic pigments (chlorophyll a and b; and carotenoids) in the foliage of one-year-old Norway spruce and silver birch seedlings was evaluated by the experiment carried out in the phytotron greenhouse. The seedlings showed different short-term responses to BC exposure. All treatments applied in the phytotron greenhouse resulted in lower chlorophyll content in spruce foliage compared to natural conditions but not differed for birch seedlings. However, the exposure of BC particles on the spruce and birch seedlings in the phytotron increased the content of photosynthetic pigments compared to the control seedlings in the phytotron. Overall, urban trees can help improve air quality by reducing BC levels through dry deposition on tree foliage, and needle-like trees are more efficient than broad-leaved trees in capturing BC. Nevertheless, a further study could assess the longer-term effects of BC particles on tree biochemical and chemical reactions.


Assuntos
Poluentes Atmosféricos , Picea , Poluentes Atmosféricos/análise , Betula , Carbono/análise , Clorofila A , Fuligem
9.
Biomed Pharmacother ; 144: 112373, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34794238

RESUMO

Side effects often limit the use of doxorubicin (DOX) in cancer treatment. We have recently developed a nanostructured lipid carrier (NLC) formulation for synergistic chemotherapy, encapsulating DOX and the anticancer adjuvants docosahexaenoic acid (DHA) and α-tocopherol succinate (TS). Hydrophobic ion-pairing with TS allowed a high DOX entrapment in the nanocarrier. In this work, we investigated the pharmacokinetics of this formulation after intravenous administration in mice. The first data obtained led us to propose synthesizing covalent DOX-TS conjugates to increase DOX retention in the NLC. We successfully conjugated DOX to TS via an amide or hydrazone bond. In vitro studies in 4T1 tumor cells indicated low cytotoxicity of the amide derivative, while the hydrazone conjugate was effective in killing cancer cells. We encapsulated the hydrazone derivative in a DHA-based nanocarrier (DOX-hyd-TS/NLC), which had reduced particle size and high drug encapsulation efficiency. The pH-sensitive hydrazone bond allowed controlled DOX release from the NLC, with increased drug release at acidic conditions. In vivo studies revealed that DOX-hyd-TS/NLC had a better pharmacokinetic profile than free DOX and attenuated the short-term cardiotoxic effects caused by DOX, such as QT prolongation and impaired left ventricular systolic function. Moreover, this formulation showed excellent therapeutic performance by reducing tumor growth in 4T1 tumor-bearing mice and decreasing DOX-induced toxicity to the heart and liver, demonstrated by hematologic, biochemical, and histologic analyses. These results indicate that DOX-hyd-TS/NLC may be a promising nanocarrier for breast cancer treatment.


Assuntos
Antibióticos Antineoplásicos/farmacocinética , Ácidos Docosa-Hexaenoicos/química , Doxorrubicina/farmacocinética , Pró-Fármacos , alfa-Tocoferol/química , Animais , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Doxorrubicina/efeitos adversos , Portadores de Fármacos , Composição de Medicamentos , Liberação Controlada de Fármacos , Humanos , Hidrazonas/química , Concentração de Íons de Hidrogênio , Lipídeos/química , Síndrome do QT Longo/induzido quimicamente , Camundongos , Camundongos Endogâmicos BALB C , Nanoestruturas , Transplante de Neoplasias , Função Ventricular Esquerda/efeitos dos fármacos
10.
Front Cardiovasc Med ; 8: 690533, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34277738

RESUMO

Cancer and cardiovascular diseases are the leading causes of death and morbidity worldwide. Strikingly, cardiovascular disorders are more common and more severe in cancer patients than in the general population, increasing incidence rates. In this context, it is vital to consider the anticancer efficacy of a treatment and the devastating heart complications it could potentially cause. Oncocardiology has emerged as a promising medical and scientific field addressing these aspects from different angles. Interestingly, nanomedicine appears to have great promise in reducing the cardiotoxicity of anticancer drugs, maintaining or even enhancing their efficacy. Several studies have shown the benefits of nanocarriers, although with some flaws when considering the concept of oncocardiology. Herein, we discuss how preclinical studies should be designed as closely as possible to clinical protocols, considering various parameters intrinsic to the animal models used and the experimental protocols. The sex and age of the animals, the size and location of the tumors, the doses of the nanoformulations administered, and the acute vs. the long-term effects of treatments are essential aspects. We also discuss the perspectives offered by non-invasive imaging techniques to simultaneously assess both the anticancer effects of treatment and its potential impact on the heart. The overall objective is to accelerate the development and validation of nanoformulations through high-quality preclinical studies reproducing the clinical conditions.

11.
Eur J Cardiothorac Surg ; 59(1): 147-154, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-32974659

RESUMO

OBJECTIVES: Pulmonary hypertension and heart disease contribute to the high morbidity rate following pneumonectomy (PN). The pathophysiology is still poorly understood. The objective was to investigate the consequences of PN on cardiopulmonary function in rats and to explore in vitro the involved mechanisms. METHODS: Sixty Sprague-Dawley male rats randomly underwent either a right PN (PN group) or sham surgery. Ten rats per group were sacrificed on postoperative days 3, 7 and 28. Cardiopulmonary alterations were investigated by echocardiographic, haemodynamic and histological analyses. In vitro, the shear stress was reproduced using a Flexcell Tension™ cyclic stretch on cultured human pulmonary endothelial cells (P-ECs) to investigate the impact on pulmonary artery smooth muscle cell (PA-SMC) growth. Data are expressed as mean ± SD. RESULTS: Mean pulmonary arterial pressure gradually increased in the PN group to reach 35 ± 7 mmHg on postoperative day 28 vs 18 ± 4 in sham (P = 0.001), likewise the proportion of muscularized distal pulmonary arteries, 83 ± 1% vs 5 ± 1%, respectively (P < 0.001), related to in situ PA-SMC proliferation. The right ventricle area and lateral wall thickness were doubled in the PN group on postoperative day 28. The left ventricle ejection fraction decreased on postoperative days 7 and 28 while the right ventricle function was maintained. In vitro, the human PA-SMC growth was significantly greater when seeded with stretched vs non-stretched P-EC media, highlighting the role of shear stress on the P-EC paracrine function. CONCLUSIONS: Right PN led to pulmonary hypertension and proportional right heart remodelling in rats. The shear stress related to high blood flow alters the pulmonary endothelial paracrine control of SMC growth.


Assuntos
Hipertensão Pulmonar , Animais , Células Endoteliais , Humanos , Hipertensão Pulmonar/etiologia , Masculino , Pneumonectomia/efeitos adversos , Artéria Pulmonar/diagnóstico por imagem , Ratos , Ratos Sprague-Dawley
12.
J Cell Physiol ; 234(10): 18283-18296, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30912139

RESUMO

Endogenous progenitor cells may participate in cardiac repair after a myocardial infarction (MI). The beta 2 adrenergic receptor (ß2-AR) pathway induces proliferation of c-kit+ cardiac progenitor cells (CPC) in vitro. We investigated if ß2-AR pharmacological stimulation could ameliorate endogenous CPC-mediated regeneration after a MI. C-kit+ CPC ß1-AR and ß2-AR expression was evaluated in vivo and in vitro. A significant increase in the percentage of CPCs expressing ß1-AR and ß2-AR was measured 7 days post-MI. Accordingly, 24 hrs of low serum and hypoxia in vitro significantly increased CPC ß2-AR expression. Cell viability and differentiation assays validated a functional role of CPC ß2-AR. The effect of pharmacological activation of ß2-AR was studied in C57 mice using fenoterol administered in the drinking water 1 week before MI or sham surgery or at the time of the surgery. MI induced a significant increase in the percentage of c-kit+ progenitor cells at 7 days, whereas pretreatment with fenoterol prolonged this response resulting in a significant elevated number of CPC up to 21 days post-MI. This increased number of CPC correlated with a decrease in infarct size. The immunofluorescence analysis of the heart tissue for proliferation, apoptosis, macrophage infiltration, cardiomyocytes surface area, and vessel density showed significant changes on the basis of surgery but no benefit due to fenoterol treatment. Cardiac function was not ameliorated by fenoterol administration when evaluated by echocardiography. Our results suggest that ß2-AR stimulation may improve the cardiac repair process by supporting an endogenous progenitor cell response but is not sufficient to improve the cardiac function.


Assuntos
Infarto do Miocárdio/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Células-Tronco/metabolismo , Animais , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Transdução de Sinais/fisiologia
13.
Cancer Lett ; 432: 227-236, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-29935372

RESUMO

Neuregulin 1 (NRG1), a ligand for HER3 and HER4 receptors, is secreted by both pancreatic tumor cells (PC) and cancer-associated fibroblasts (CAFs), the latter representing the most abundant compound of pancreatic stroma. This desmoplastic stroma contributes to Pancreatic Ductal Adenocarcinoma (PDAC) aggressiveness and therapeutic failure by promoting tumor progression, invasion and resistance to chemotherapies. In the present work, we aimed at disrupting the complex crosstalk between PC and CAF in order to prevent tumor cell proliferation. To do so, we demonstrated the promising tumor growth inhibitory effect of the 7E3, an original antibody directed to NRG1. This antibody promotes antibody dependent cellular cytotoxicity in NRG1-positive PC and CAFs and inhibits NRG1-associated signaling pathway induction, by blocking NRG1-mediated HER3 activation. Moreover, 7E3 inhibits migration and growth of pancreatic cancer cells co-cultured with CAFs, both in vitro and in vivo using orthotopic pancreatic tumor xenografts. Our preclinical results demonstrate that the anti-NRG1 antibody 7E3 could represent a promising approach to target pancreatic stroma and cancer cells, thereby providing novel therapeutic options for PDAC.


Assuntos
Anticorpos Monoclonais/farmacologia , Fibroblastos Associados a Câncer/efeitos dos fármacos , Carcinoma Ductal Pancreático/prevenção & controle , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neuregulina-1/antagonistas & inibidores , Neoplasias Pancreáticas/prevenção & controle , Receptor ErbB-3/antagonistas & inibidores , Animais , Apoptose , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Proliferação de Células , Técnicas de Cocultura , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neuregulina-1/imunologia , Neuregulina-1/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Receptor ErbB-3/imunologia , Receptor ErbB-3/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Mol Ther ; 26(3): 902-916, 2018 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-29249393

RESUMO

Despite considerable advances in cardiovascular disease treatment, heart failure remains a public health challenge. In this context, gene therapy appears as an attractive approach, but clinical trials using single therapeutic molecules result in moderate benefit. With the objective of improving ischemic heart failure therapy, we designed a combined treatment, aimed to simultaneously stimulate angiogenesis, prevent cardiac remodeling, and restore contractile function. We have previously validated IRES-based vectors as powerful tools to co-express genes of interest. Mono- and multicistronic lentivectors expressing fibroblast growth factor 2 (angiogenesis), apelin (cardioprotection), and/or SERCA2a (contractile function) were produced and administrated by intramyocardial injection into a mouse model of myocardial infarction. Data reveal that combined treatment simultaneously improves vessel number, heart function parameters, and fibrosis prevention, due to FGF2, SERCA2a, and apelin, respectively. Furthermore, addition of SERCA2a in the combination decreases cardiomyocyte hypertrophy. Large-scale transcriptome analysis reveals that the triple treatment is the most efficient in restoring angiogenic balance as well as expression of genes involved in cardiac function and remodeling. Our study validates the concept of combined treatment of ischemic heart disease with apelin, FGF2, and SERCA2a and shows that such therapeutic benefit is mediated by a more effective recovery of gene network regulation.


Assuntos
Apelina/genética , Fator 2 de Crescimento de Fibroblastos/genética , Expressão Gênica , Redes Reguladoras de Genes , Isquemia Miocárdica/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Animais , Cardiomegalia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Fibrose , Ordem dos Genes , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/genética , Lentivirus/genética , Camundongos , Isquemia Miocárdica/patologia , Isquemia Miocárdica/terapia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Transcriptoma , Transdução Genética
15.
Antioxid Redox Signal ; 25(1): 10-27, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-26959532

RESUMO

AIMS: In heart failure (HF), mitochondrial quality control and autophagy are progressively impaired, but the role of oxidative stress in this process and its underlying mechanism remain to be defined. By degrading norepinephrine and serotonin, the mitochondrial enzyme, monoamine oxidase-A (MAO-A), is a potent source of reactive oxygen species (ROS) in the heart and its activation leads to the persistence of mitochondrial damage. In this study, we analyzed the consequences of ROS generation by MAO-A on the autophagy-lysosome pathway in the heart. RESULTS: Cardiomyocyte-driven expression of MAO-A in mice led to mitochondrial fission and translocation of Drp1 and Parkin in the mitochondrial compartment. Ventricles from MAO-A transgenic mice displayed accumulation of LC3-positive autophagosomes, together with p62 and ubiquitylated proteins, indicating impairment of autophagy. In vitro adenoviral delivery of MAO-A in cardiomyocytes and the consequent generation of ROS blocked autophagic flux with accumulation of LC3II, p62, and ubiquitylated proteins, leading to mitochondrial fission and cell necrosis. In addition, MAO-A activation induced accumulation of lysosomal proteins, cathepsin D and Lamp1, reduced lysosomal acidification, and blocked the nuclear translocation of transcription factor-EB (TFEB), a master regulator of autophagy and lysosome biogenesis. Most interestingly, overexpression of TFEB attenuated autophagosome buildup, mitochondrial fission, cardiomyocyte death, and HF associated with MAO-A activation. INNOVATION AND CONCLUSION: This study unravels a new link between MAO-dependent H2O2 production and lysosomal dysfunction. Altogether, our findings demonstrate that the MAO-A/H2O2 axis has a negative impact on the elimination and recycling of mitochondria through the autophagy-lysosome pathway, which participates in cardiomyocyte death and HF. Antioxid. Redox Signal. 25, 10-27.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Insuficiência Cardíaca/metabolismo , Monoaminoxidase/metabolismo , Necrose/metabolismo , Estresse Oxidativo , Animais , Apoptose/genética , Autofagossomos/metabolismo , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração/metabolismo , Ventrículos do Coração/fisiopatologia , Humanos , Peróxido de Hidrogênio/metabolismo , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Miócitos Cardíacos/patologia , Necrose/fisiopatologia , Norepinefrina/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Serotonina/metabolismo , Ativação Transcricional/genética
16.
Cardiovasc Res ; 108(2): 254-67, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26370247

RESUMO

AIMS: Post-infarction remodelling is accompanied and influenced by perturbations in mitogen-activated protein kinase (MAPK) signalling. The growth arrest and DNA-damage-inducible 45 (Gadd45) proteins are small acidic proteins involved in DNA repair and modulation of MAPK activity. Little is known about the role of Gadd45 in the heart. Here, we explored the potential contribution of Gadd45 gamma (γ) isoform to the acute and late phase of heart failure (HF) after myocardial infarction (MI) and determined the mechanisms underlying Gadd45γ actions. METHODS AND RESULTS: The Gadd45γ isoform is up-regulated in murine cardiomyocytes subjected to simulated ischaemia and in the mouse heart during MI. To mimic the situation observed during MI, we enhanced Gadd45γ content in cardiomyocytes with a single injection of an adeno-associated viral (AAV9) vector encoding Gadd45γ under the cTNT promoter. Gadd45γ overexpression induces cardiomyocyte apoptosis, fibrosis, left ventricular dysfunction, and HF. On the other hand, genetic deletion of Gadd45γ in knockout mice confers resistance to ischaemic injury, at least in part by limiting cardiomyocyte apoptosis. Mechanistically, Gadd45γ activates receptor-interacting protein 1 (RIP1) and caspase-8 in a p38 MAPK-dependent manner to promote cardiomyocyte death. CONCLUSION: This work is the first to demonstrate that Gadd45γ accumulation during MI promotes the development and persistence of HF by inducing cardiomyocyte apoptosis in a p38 MAPK-dependent manner. We clearly identify Gadd45γ as a therapeutic target in the development of HF.


Assuntos
Proteínas de Transporte/metabolismo , Insuficiência Cardíaca/etiologia , Sistema de Sinalização das MAP Quinases , Infarto do Miocárdio/complicações , Remodelação Ventricular , Animais , Caspase 8/metabolismo , Células Cultivadas , Proteínas Ativadoras de GTPase/metabolismo , Insuficiência Cardíaca/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos Knockout , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo
17.
Basic Res Cardiol ; 110(3): 23, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25837837

RESUMO

Cardiac troponins are released and cleared slowly after myocardial injury, complicating the diagnosis of early, and recurrent, acute myocardial infarction. Cardiac myosin-binding protein C (cMyC) is a similarly cardiac-restricted protein that may have different release/clearance kinetics. Using novel antibodies raised against the cardiac-specific N-terminus of cMyC, we used confocal microscopy, immunoblotting and immunoassay to document its location and release. In rodents, we demonstrate rapid release of cMyC using in vitro and in vivo models of acute myocardial infarction. In patients, with ST elevation myocardial infarction (STEMI, n = 20), undergoing therapeutic ablation of septal hypertrophy (TASH, n = 20) or having coronary artery bypass surgery (CABG, n = 20), serum was collected prospectively and frequently. cMyC appears in the serum as full-length and fragmented protein. Compared to cTnT measured using a contemporary high-sensitivity commercial assay, cMyC peaks earlier (STEMI, 9.3 ± 3.1 vs 11.8 ± 3.4 h, P < 0.007; TASH, 9.7 ± 1.4 vs 21.6 ± 1.4 h, P < 0.0001), accumulates more rapidly (during first 4 h after TASH, 25.8 ± 1.9 vs 4.0 ± 0.4 ng/L/min, P < 0.0001) and disappears more rapidly (post-CABG, decay half-time 5.5 ± 0.8 vs 22 ± 5 h, P < 0.0001). Our results demonstrate that following defined myocardial injury, the rise and fall in the serum of cMyC is more rapid than that of cTnT. We speculate that these characteristics could enable earlier diagnosis of myocardial infarction and reinfarction in suspected non-STEMI, a population not included in this early translational study.


Assuntos
Biomarcadores/sangue , Proteínas de Transporte/sangue , Infarto do Miocárdio/sangue , Idoso , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Immunoblotting , Masculino , Pessoa de Meia-Idade , Ratos , Ratos Wistar , Ressonância de Plasmônio de Superfície
18.
FASEB J ; 29(6): 2653-66, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25757563

RESUMO

Preterm birth is an inflammatory process resulting from the massive infiltration of innate immune cells and the production of proinflammatory cytokines in the myometrium. However, proinflammatory cytokines, which induce labor in vivo, fail to induce labor-associated features in human myometrial cells (MCs). We thus aimed to investigate if reactive oxygen species (ROS) production could be the missing step between immune cell activation and MC response. Indeed, we found that ROS production is increased in the human preterm laboring myometrium (27% ROS producing cells, respectively, versus 2% in nonlaboring controls), with 90% ROS production in macrophages. Using LPS-stimulated myometrial samples and cell coculture experiments, we demonstrated that ROS production is required for labor onset. Furthermore, we showed that ROS are required first in the NADPH oxidase (NADPHox)-2/NF-κB-dependent macrophage response to inflammatory stimuli but, more importantly, to trigger macrophage-induced MCs transactivation. Remarkably, in a murine model of LPS-induced preterm labor (inducing delivery within 17 hours, with no pup survival), cotreatment with glutathione delayed labor onset up to 94 hours and prevented in utero fetal distress, allowing 46% pups to survive. These results suggest that targeting ROS production with the macrophage-permeable antioxidant glutathione could constitute a promising strategy to prevent preterm birth.


Assuntos
Morte Fetal/prevenção & controle , Glutationa/farmacologia , Macrófagos/metabolismo , Miométrio/efeitos dos fármacos , Trabalho de Parto Prematuro/prevenção & controle , Espécies Reativas de Oxigênio/metabolismo , Adulto , Animais , Animais Recém-Nascidos , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Expressão Gênica , Glutationa/administração & dosagem , Humanos , Recém-Nascido , Lipopolissacarídeos , Masculino , Camundongos Endogâmicos C57BL , Miométrio/citologia , Miométrio/metabolismo , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , NF-kappa B/metabolismo , Trabalho de Parto Prematuro/induzido quimicamente , Trabalho de Parto Prematuro/metabolismo , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Adulto Jovem
19.
Nat Struct Mol Biol ; 20(10): 1182-90, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24037507

RESUMO

p38α mitogen-activated protein kinase (p38α) is activated by a variety of mechanisms, including autophosphorylation initiated by TGFß-activated kinase 1 binding protein 1 (TAB1) during myocardial ischemia and other stresses. Chemical-genetic approaches and coexpression in mammalian, bacterial and cell-free systems revealed that mouse p38α autophosphorylation occurs in cis by direct interaction with TAB1(371-416). In isolated rat cardiac myocytes and perfused mouse hearts, TAT-TAB1(371-416) rapidly activates p38 and profoundly perturbs function. Crystal structures and characterization in solution revealed a bipartite docking site for TAB1 in the p38α C-terminal kinase lobe. TAB1 binding stabilizes active p38α and induces rearrangements within the activation segment by helical extension of the Thr-Gly-Tyr motif, allowing autophosphorylation in cis. Interference with p38α recognition by TAB1 abolishes its cardiac toxicity. Such intervention could potentially circumvent the drawbacks of clinical pharmacological inhibitors of p38 catalytic activity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Ativação Enzimática/fisiologia , Humanos , Camundongos , Proteína Quinase 14 Ativada por Mitógeno/química , Modelos Moleculares , Dados de Sequência Molecular , Isquemia Miocárdica/enzimologia , Ressonância Magnética Nuclear Biomolecular , Fosforilação , Ratos , Homologia de Sequência de Aminoácidos
20.
Antioxid Redox Signal ; 18(1): 5-18, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22738191

RESUMO

AIMS: Oxidative stress and mitochondrial dysfunction participate together in the development of heart failure (HF). mRNA levels of monoamine oxidase-A (MAO-A), a mitochondrial enzyme that produces hydrogen peroxide (H(2)O(2)), increase in several models of cardiomyopathies. Therefore, we hypothesized that an increase in cardiac MAO-A could cause oxidative stress and mitochondrial damage, leading to cardiac dysfunction. In the present study, we evaluated the consequences of cardiac MAO-A augmentation on chronic oxidative damage, cardiomyocyte survival, and heart function, and identified the intracellular pathways involved. RESULTS: We generated transgenic (Tg) mice with cardiac-specific MAO-A overexpression. Tg mice displayed cardiac MAO-A activity levels similar to those found in HF and aging. As expected, Tg mice showed a significant decrease in the cardiac amounts of the MAO-A substrates serotonin and norepinephrine. This was associated with enhanced H(2)O(2) generation in situ and mitochondrial DNA oxidation. As a consequence, MAO-A Tg mice demonstrated progressive loss of cardiomyocytes by necrosis and ventricular failure, which were prevented by chronic treatment with the MAO-A inhibitor clorgyline and the antioxidant N-acetyl-cystein. Interestingly, Tg hearts exhibited p53 accumulation and downregulation of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), a master regulator of mitochondrial function. This was concomitant with cardiac mitochondrial ultrastructural defects and ATP depletion. In vitro, MAO-A adenovirus transduction of neonatal cardiomyocytes mimicked the results in MAO-A Tg mice, triggering oxidative stress-dependent p53 activation, leading to PGC-1α downregulation, mitochondrial impairment, and cardiomyocyte necrosis. INNOVATION AND CONCLUSION: We provide the first evidence that MAO-A upregulation in the heart causes oxidative mitochondrial damage, p53-dependent repression of PGC-1α, cardiomyocyte necrosis, and chronic ventricular dysfunction.


Assuntos
Mitocôndrias Cardíacas/enzimologia , Monoaminoxidase/metabolismo , Miócitos Cardíacos/patologia , Necrose/enzimologia , Transativadores/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Disfunção Ventricular Esquerda/enzimologia , Animais , Cardiomiopatia Dilatada/enzimologia , Células Cultivadas , Doença Crônica , Indução Enzimática , Fibrose , Ventrículos do Coração/enzimologia , Ventrículos do Coração/patologia , Hipertrofia Ventricular Esquerda/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monoaminoxidase/genética , Miócitos Cardíacos/metabolismo , Oxirredução , Estresse Oxidativo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição , Regulação para Cima , Disfunção Ventricular Esquerda/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA