Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Arch Biochem Biophys ; 754: 109954, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38432564

RESUMO

Iron overload has detrimental effects on bone marrow mesenchymal stem cells (BMMSCs), cells crucial for bone marrow homeostasis and hematopoiesis support. Excessive iron accumulation leads to the production of reactive oxygen species (ROS), resulting in cell death, cell cycle arrest, and disruption of vital cellular pathways. Although apoptosis has been extensively studied, other programmed cell death mechanisms including autophagy, necroptosis, and ferroptosis also play significant roles in iron overload-induced bone marrow cell death. Studies have highlighted the involvement of ROS production, DNA damage, MAPK pathways, and mitochondrial dysfunction in apoptosis. In addition, autophagy and ferroptosis are activated, as shown by the degradation of cellular components and lipid peroxidation, respectively. However, several compounds and antioxidants show promise in mitigating iron overload-induced cell death by modulating ROS levels, MAPK pathways, and mitochondrial integrity. Despite early indications, more comprehensive research and clinical studies are needed to better understand the interplay between these programmed cell death mechanisms and enable development of effective therapeutic strategies. This review article emphasizes the importance of studying multiple cell death pathways simultaneously and investigating potential rescuers to combat iron overload-induced bone marrow cell death.


Assuntos
Sobrecarga de Ferro , Ferro , Humanos , Ferro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Medula Óssea/metabolismo , Sobrecarga de Ferro/metabolismo , Apoptose , Células da Medula Óssea/metabolismo
2.
Toxicol Appl Pharmacol ; 479: 116727, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37863361

RESUMO

Iron overload cardiomyopathy (IOC) is the leading cause of death in cases of iron overload in patients. Previous studies demonstrated that iron overload led to cardiomyocyte dysfunction and death through multiple pathways including apoptosis, necroptosis and ferroptosis. However, the dominant cell death pathway in the iron-overloaded heart needs clarification. We tested the hypothesis that ferroptosis, an iron-dependent cell death, plays a dominant role in IOC, and ferroptosis inhibitor exerts greater efficacy than inhibitors of apoptosis and necroptosis on improving cardiac function in iron-overloaded rats. Iron dextran was injected intraperitoneally into male Wistar rats for four weeks to induce iron overload. Then, the rats were divided into 5 groups: treated with vehicle, apoptosis inhibitor (z-VAD-FMK), necroptosis inhibitor (Necrostatin-1), ferroptosis inhibitor (Ferrostatin-1) or iron chelator (deferoxamine) for 2 weeks. Cardiac function, mitochondrial function, apoptosis, necroptosis and ferroptosis were determined. The increased expression of apoptosis-, necroptosis- and ferroptosis-related proteins, were associated with impaired cardiac and mitochondrial function in iron-overloaded rats. All cell death inhibitors attenuated cardiac apoptosis, necroptosis and ferroptosis in iron-overloaded rats. Ferrostatin-1 was more effective than the other drugs in diminishing mitochondrial dysfunction and Bax/Bcl-2 ratio. Moreover, both Ferrostatin-1 and deferoxamine reversed iron overload-induced cardiac dysfunction as indicated by restored left ventricular ejection fraction and E/A ratio, whereas z-VAD-FMK and Necrostatin-1 only partially improved this parameter. These results indicated that ferroptosis could be the predominant form of cardiomyocyte death in IOC, and that inhibiting ferroptosis might be a potential novel treatment for IOC.


Assuntos
Cardiomiopatias , Ferroptose , Sobrecarga de Ferro , Ratos , Humanos , Masculino , Animais , Desferroxamina/metabolismo , Desferroxamina/farmacologia , Desferroxamina/uso terapêutico , Necroptose , Volume Sistólico , Ratos Wistar , Função Ventricular Esquerda , Apoptose , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/metabolismo , Ferro/metabolismo , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/prevenção & controle , Cardiomiopatias/induzido quimicamente , Mitocôndrias , Miócitos Cardíacos/metabolismo
3.
Life Sci ; 313: 121269, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36493877

RESUMO

AIMS: The present study was aimed to investigate the effects of cell death inhibitors including ferroptosis inhibitor, ferrostatin-1 (FER-1) and a pan-caspase inhibitor, z-VAD-FMK on brain parameters and cognitive function in iron-overloaded rats. MAIN METHODS: Male Wistar rats (n = 30) were divided into 2 groups to receive an intraperitoneal injection with either 10 % dextrose in normal saline solution (NSS) (control group, n = 6) or 100 mg/kg iron dextran (Fe group, n = 24) for 6 weeks. After 4 weeks of injection, Fe-injected rats were subdivided into 4 subgroups (n = 6/subgroup) to subcutaneously receive with 1) vehicle (10 % DMSO in NSS), 2) deferoxamine (25 mg/kg), 3) FER-1 (2 mg/kg), or 4) z-VAD-FMK (1 mg/kg). Control group was received vehicle. All subgroups were received each treatment for 2 weeks. Behavioral tests including the Morris water maze test and novel object recognition test, were performed at the end of treatment. Then, circulating iron levels and brain parameters including blood-brain barrier proteins, iron level, synaptic proteins, and ferroptosis/apoptosis were determined. KEY FINDINGS: All treatment attenuated iron-overloaded condition, brain pathologies, and the cognitive impairment. FER-1 and z-VAD-FMK provided superior effects than deferoxamine by attenuating loss of synaptic proteins and restoring cognitive function in both hippocampal-dependent and hippocampal-independent manners. SIGNIFICANCE: These findings suggest that cell death inhibitors act as the novel therapeutic targets for neuroprotection in iron-overloaded condition.


Assuntos
Desferroxamina , Ferro , Ratos , Masculino , Animais , Desferroxamina/farmacologia , Ratos Wistar , Apoptose , Cognição
4.
Life Sci ; 307: 120871, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-35952729

RESUMO

AIMS: This study aimed to investigate the changes in gut microbiota in iron-overload thalassemia and the roles of an iron chelator on gut dysbiosis/inflammation, and metabolites, including short-chain fatty acids (SCFAs) and trimethylamine N-oxide (TMAO). MAIN METHODS: Adult male C57BL/6 mice both wild-type (WT: n = 15) and heterozygous ß-thalassemia (BKO: n = 15) were fed on either a normal (ND: n = 5/group) or a high­iron diet for four months (HFe: n = 10/group). HFe-treated WT and HFe-treated BKO groups were further subdivided into two subgroups and each subgroup given either vehicle (n = 5/subgroup) or deferiprone (n = 5/subgroup) during the last month. Gut microbiota profiles, gut barrier characteristics, levels of proinflammatory cytokines, and plasma SCFAs and TMAO were determined at the end of the study. KEY FINDINGS: HFe-fed WT mice showed distinct gut microbiota profiles from those of ND-fed WT mice, whereas HFe-fed BKO mice showed slightly different gut microbiota profiles from ND-fed BKO. Gut inflammation and barrier disruption were found only in HFe-fed BKO mice, however, an increase in plasma TMAO levels and decreased levels of SCFAs were observed in both WT and BKO mice with HFe-feeding. Treatment with deferiprone, gut dysbiosis and disturbance of metabolites were attenuated in HFe-fed WT mice, but not in HFe-fed BKO mice. Increased Verrucomicrobia and Ruminococcaceae were associated with the beneficial effects of deferiprone. SIGNIFICANCE: Iron-overload leads to gut dysbiosis/inflammation and disturbance of metabolites, and deferiprone alleviates those conditions more effectively in WT than in those that are thalassemic.


Assuntos
Microbioma Gastrointestinal , Sobrecarga de Ferro , Talassemia , Animais , Citocinas/uso terapêutico , Deferiprona/farmacologia , Dieta , Disbiose/tratamento farmacológico , Inflamação/tratamento farmacológico , Ferro/metabolismo , Quelantes de Ferro/farmacologia , Sobrecarga de Ferro/complicações , Masculino , Metilaminas , Camundongos , Camundongos Endogâmicos C57BL
5.
Arch Biochem Biophys ; 721: 109189, 2022 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-35307366

RESUMO

Protein tyrosine phosphatase non-receptor type 6 (PTPN6) is a key regulatory protein in cellular signal transduction in the control of inflammation and cell death. Impairment of PTPN6 is known to be associated with human inflammatory diseases including neutrophilic dermatosis; however, comprehensive studies of PTPN6-associated neutrophilic dermatosis have not clearly identified the relationships involved. Reports from in vitro and in vivo studies revealed that inflammatory cytokines have increased in the white blood cells from PTPN6-knocked out mice, and systemic inflammation was also increased in these mice, resulting in skin inflammation in this model. Reports of PTPN6 regulatory functions through five pathophysiological mechanisms are summarized and discussed here including inhibition of myeloid differentiation primary response 88, enhancement of the regulatory function of receptor-interacting protein kinase, inhibition of receptor-interacting serine/threonine-protein kinase 3/mixed lineage kinase domain-like protein-dependent necroptosis, inhibition of caspase-8-dependent apoptosis, and inhibition of p38/mitogen-activated protein kinase. Treatments by blocking the pathways involved in signal transduction and inflammatory cytokine release are also summarized. Understanding this underlying mechanism could improve therapeutic strategies for neutrophilic dermatosis.


Assuntos
Inflamação , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Dermatopatias , Animais , Humanos , Inflamação/metabolismo , Camundongos , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Transdução de Sinais
6.
Sci Rep ; 12(1): 4635, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-35302058

RESUMO

The association between Lipocalin-2 (LCN2) and cognition in patients with metabolic syndrome (MetS) has not been thoroughly investigated. We aimed to evaluate whether serum LCN2 levels are associated with the alteration of cognitive function in patients with MetS. The total of 191 non-demented participants with MetS were enrolled onto the study in 2015, and a cohort study was conducted in a subpopulation in 2020. After adjustment for sex, age, waist circumference, creatinine levels, and HbA1C, an association between the higher serum LCN2 levels and the lower Montreal cognitive assessment (MoCA) scores was observed (B = - 0.045; 95%CI - 0.087, - 0.004; p 0.030). A total of 30 participants were followed-up in 2020. Serum LCN2 levels were decreased in correlation with age (23.31 ± 12.32 ng/ml in 2015 and 15.98 ± 11.28 ng/ml in 2020, p 0.024), while other metabolic parameters were unchanged. Magnetic resonance imaging studies were conducted on a subsample of patients in 2020 (n = 15). Associations between high serum LCN2 levels from 2015 and 2020 and changes in brain volume of hippocampus and prefrontal cortex from 2020 have been observed. These findings suggest a relationship between changes of the level of circulating LCN2, cognitive impairment, and changes in brain volume in patients with MetS. However, further investigation is still needed to explore the direct effect of circulating LCN2 on the cognition of MetS patients.


Assuntos
Disfunção Cognitiva , Lipocalina-2 , Síndrome Metabólica , Encéfalo , Disfunção Cognitiva/sangue , Estudos de Coortes , Humanos , Lipocalina-2/sangue , Síndrome Metabólica/sangue , Síndrome Metabólica/psicologia , Tamanho do Órgão
7.
Exp Biol Med (Maywood) ; 247(7): 574-583, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35130741

RESUMO

Iron overload can be the result of either dysregulated iron metabolism in the case of hereditary hemochromatosis or repeated blood transfusions in the case of secondary hemochromatosis (e.g. in ß-thalassemia and sickle cell anemia patients). Under iron overload conditions, transferrin (Tf) saturation leads to an increase in non-Tf bound iron which can result in the generation of reactive oxygen species (ROS). These excess ROS can damage cellular components, resulting in the dysfunction of vital organs including iron overload cardiomyopathy (IOC). Multiple studies have demonstrated that L-type and T-type calcium channels are the main routes for iron uptake in the heart, and that calcium channel blockers, given either individually or in combination with standard iron chelators, confer cardioprotective effects under iron overload conditions. Treatment with antioxidants may also provide therapeutic benefits. Interestingly, recent studies have suggested that mitochondrial dynamics and regulated cell death (RCD) pathways are potential targets for pharmacological interventions against iron-induced cardiomyocyte injury. In this review, the potential therapeutic roles of iron chelators, antioxidants, iron uptake/metabolism modulators, mitochondrial dynamics modulators, and inhibitors of RCD pathways in IOC are summarized and discussed.


Assuntos
Cardiomiopatias , Hemocromatose , Sobrecarga de Ferro , Antioxidantes/uso terapêutico , Cardiomiopatias/tratamento farmacológico , Hemocromatose/complicações , Hemocromatose/tratamento farmacológico , Hemocromatose/metabolismo , Humanos , Ferro/metabolismo , Quelantes de Ferro/metabolismo , Quelantes de Ferro/farmacologia , Quelantes de Ferro/uso terapêutico , Sobrecarga de Ferro/complicações , Sobrecarga de Ferro/tratamento farmacológico , Miócitos Cardíacos/metabolismo , Espécies Reativas de Oxigênio/metabolismo
8.
Front Med (Lausanne) ; 9: 1015306, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36660002

RESUMO

Objective: To compare the levels of Non-transferrin bound iron (NTBI) in fetuses with anemia, using Hb Bart's disease as a study model, and those in unaffected fetuses and to determine the association between fetal cardiac function and the levels of NTBI. Patients and methods: A prospective study was conducted on pregnancies at risk of fetal Hb Bart's disease. All fetuses underwent standard ultrasound examination at 18-22 weeks of gestation for fetal biometry, anomaly screening and fetal cardiac function. After that, 2 ml of fetal blood was taken by cordocentesis to measure NTBI by Labile Plasma Iron (LPI), serum iron, hemoglobin and hematocrit. The NTBI levels of both groups were compared and the correlation between NTBI and fetal cardiac function was determined. Results: A total of 50 fetuses, including 20 fetuses with Hb Bart's disease and 30 unaffected fetuses were recruited. There was a significant increase in the level of serum iron in the affected group (median: 22.7 vs. 9.7; p-value: 0.013) and also a significant increase in NTBI when compared with those of the unaffected fetuses (median 0.11 vs. 0.07; p-value: 0.046). In comparisons of fetal cardiac function, myocardial performance (Tei) index of both sides was significantly increased in the affected group (left Tei: p = 0.001, Right Tei: p = 0.008). Also, isovolumetric contraction time (ICT) was also significantly prolonged (left ICT: p = 0.00, right ICT: p = 0.000). Fetal LPI levels were significantly correlated inversely with fetal hemoglobin levels (p = 0.030) but not significantly correlated with the fetal serum iron levels (p = 0.138). Fetal LPI levels were also significantly correlated positively with myocardial performance index (Tei) of both sides (right Tei: R = 0.000, left Tei: R = 0.000) and right ICT (R = 0.013), but not significantly correlated with left ICT (R = 0.554). Conclusion: Anemia caused by fetal Hb Bart's disease in pre-hydropic stage is significantly associated with fetal cardiac dysfunction and increased fetal serum NTBI levels which are significantly correlated with worsening cardiac dysfunction. Nevertheless, based on the limitations of the present study, further studies including long-term data are required to support a role of fetal anemia as well as increased fetal serum NTBI levels in development of subsequent heart failure or cardiac compromise among the survivors, possibly predisposing to cardiovascular disease in adult life.

9.
J Alzheimers Dis ; 82(3): 1315-1332, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34151811

RESUMO

BACKGROUND: Palmitic acid (PA) promotes brain pathologies including Alzheimer's disease (AD)-related proteins, neuroinflammation, and microglial activation. The activation of neurons and microglia via their Fc gamma receptors (FcγRs) results in producing inflammatory cytokines. OBJECTIVE: To investigate the expression of FcγRs, FcγR signaling proteins, AD-related proteins, proinflammatory cytokines, and cell viability of neurons and microglia in association with PA exposure as well as the effects of FcγR blockade on these parameters in response to PA. METHODS: 200 and 400µM PA-conjugated BSA were applied to SH-SY5Y and HMC3 cells for 24 h. For FcγR blockage experiment, both cells were exposed to FcγR blocker before receiving of 200 and 400µM of PA-conjugated BSA for 24 h. RESULTS: PA significantly increased AD-related proteins, including Aß and BACE1, as well as increasing TNFα, IL-1ß, and IL-6 in SH-SY5Y and HMC3 cells. However, the p-Tau/Tau ratio was only increased in SH-SY5Y cells. These results were associated with an increase in FcγRs activation and a decrease in cell viability in both cell types. FcγRs blockage diminished the activation of FcγR in SH-SY5Y and HMC3 cells. Interestingly, blocking FcγRs before PA exposure reduced the increment of AD-related proteins, proinflammatory cytokines caused by PA. FcγRs blocking also inhibits cell death for 23%of SH-SY5Y cells and 64%of HMC3 cells, respectively. CONCLUSION: These findings suggest that PA is a risk factor for AD via the increased AD-related pathologies, inflammation, FcγRs activation, and brain cell death, while FcγR blockage can alleviate these effects.


Assuntos
Microglia/metabolismo , Neurônios/metabolismo , Ácido Palmítico/toxicidade , Receptores de IgG/antagonistas & inibidores , Receptores de IgG/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Inibidores Enzimáticos/toxicidade , Humanos , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos
10.
Biomolecules ; 11(5)2021 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33946939

RESUMO

Human health hazards caused by bisphenol A (BPA), a precursor for epoxy resins and polycarbonate-based plastics, are well documented and are closely associated with mitochondrial impairment and oxidative imbalance. This study aimed to assess the therapeutic efficacy of N-acetylcysteine (NAC) on renal deterioration caused by long-term BPA exposure and examine the signaling transduction pathway involved. Male Wistar rats were given vehicle or BPA orally for 12 weeks then the BPA-treated group was subdivided to receive vehicle or NAC concurrently with BPA for a further 4 weeks, while the vehicle-treated normal control group continued to receive vehicle through to the end of experiment. Proteinuria, azotemia, glomerular filtration reduction and histopathological abnormalities caused by chronic BPA exposure were significantly reduced following NAC therapy. NAC also diminished nitric oxide and lipid peroxidation but enhanced renal glutathione levels, and counteracted BPA-induced mitochondrial swelling, increased mitochondrial reactive oxygen species production, and the loss of mitochondrial membrane potential. The benefit of NAC was related to the modulation of signaling proteins in the AMPK-SIRT3-SOD2 axis. The present study shows the potential of NAC to restore mitochondrial integrity and oxidative balance after long-term BPA exposure, and suggests that NAC therapy is an effective approach to tackle renal deterioration in this condition.


Assuntos
Acetilcisteína/administração & dosagem , Azotemia/tratamento farmacológico , Compostos Benzidrílicos/efeitos adversos , Fenóis/efeitos adversos , Proteinúria/tratamento farmacológico , Acetilcisteína/farmacologia , Administração Oral , Animais , Azotemia/induzido quimicamente , Modelos Animais de Doenças , Taxa de Filtração Glomerular/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Proteinúria/induzido quimicamente , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
11.
Biochem Pharmacol ; 188: 114587, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33932471

RESUMO

The standard chemotherapy regimens of ovarian cancer are platinum-based chemotherapy (carboplatin and paclitaxel) and bevacizumab (BEV). However, the effects of BEV alone or combined with carboplatin and paclitaxel on mitochondrial dynamics, mitochondrial function, mitophagy, apoptosis, inflammation and vascular endothelial growth factor (VEGF) in human ovarian cancer mitochondria and cells have not yet been investigated. Therefore, we aimed to test the hypothesis that 1) platinum-based chemotherapy and BEV equally damage isolated mitochondria from human ovarian cancers, and ovarian cancer cells through inducing mitochondrial dynamics dysregulation, mitochondrial dysfunction, increased mitophagy and apoptosis, as well as altered inflammation and VEGF; and 2) combined therapies exert greater damage than monotherapy. Each isolated human ovarian cancer mitochondria (n = 16) or CaOV3 cells (n = 6) were treated with either platinum-based chemotherapy (carboplatin 10 µM and paclitaxel 5 µM), BEV (2 mg/mL) or combined platinum-based chemotherapy and BEV for 60 min or 24 h, respectively. Following the treatment, mitochondrial dynamics, mitochondrial function, mitophagy, apoptosis, cytotoxicity, inflammation and VEGF were determined. Platinum-based chemotherapy caused ovarian cancer mitochondria and cell damage through mitochondrial dysfunction, increased cell death with impairment of membrane integrity, and enhanced VEGF reduction, while BEV did not. BEV caused deterioration of ovarian cancer mitochondria and cells through mitochondrial-dependent apoptosis, but it had no effect on cell viability. Interestingly, combined platinum-based chemotherapy and BEV treatments had no addictive effects on all parameters except mitochondrial maximal respiration, when compared to monotherapy. Collectively, these findings suggest that platinum-based chemotherapy and BEV caused human ovarian cancer mitochondrial and cell damage through different mechanisms.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Bevacizumab/administração & dosagem , Carboplatina/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Paclitaxel/administração & dosagem , Feminino , Humanos , Pessoa de Meia-Idade , Platina/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas
12.
J Cell Physiol ; 236(7): 5108-5120, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33319934

RESUMO

This study aimed to investigate the mechanistic roles of LCN-2 and LCN-2 receptors (LCN-2R) as iron transporters in cardiomyocytes under iron overload condition. H9c2 cardiomyocytes were treated with either LCN-2 small interfering RNA (siRNA) or LCN-2R siRNA or L-type or T-type calcium channel (LTCC or TTCC) blockers, or iron chelator deferiprone (DFP). After the treatments, the cells were exposed to Fe3+ or Fe2+ , after that biological parameters were determined. Silencing of lipocalin-2 or its receptor improved cardiomyocyte viability via decreasing iron uptake, mitochondrial fission, mitophagy and cleaved caspase-3 only in the Fe3+ overload condition. In contrast, treatments with LTCC blocker and TTCC blocker showed beneficial effects on those parameters only in conditions of Fe2+ overload. Treatment with DFP has been shown beneficial effects both in Fe2+ and Fe3+ overload condition. All of these findings suggested that LTCC and TTCC play crucial roles in the Fe2+ uptake, whereas LCN-2 and LCN-2R were essential for Fe3+ uptake into the cardiomyocytes under iron overload conditions.


Assuntos
Apoptose/fisiologia , Sobrecarga de Ferro/patologia , Lipocalina-2/genética , Mitocôndrias/patologia , Miócitos Cardíacos/metabolismo , Animais , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo T/metabolismo , Linhagem Celular , Sobrevivência Celular/genética , Deferiprona/farmacologia , Ferro/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos
13.
J Perinat Med ; 48(5): 471-476, 2020 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-32286249

RESUMO

Background The primary objective of this study was to compare the fetal cardiac performance index (Tei index) between the fetuses of gestational diabetes mellitus (GDM) mothers and non-GDM mothers; and the secondary objective was to compare various other parameters of fetal cardiac function as well as maternal oxidative stress levels between the groups of GDM and non-GDM mothers. Methods A cross-sectional study was conducted on pregnant women at 24-28 weeks of gestation. All of the participants underwent 100 g, 3-h oral glucose tolerance test (OGTT) as a diagnostic test for GDM and were categorized as non-GDM and GDM group. All participants had fetal echocardiography performed for cardiac function, and then maternal blood samples were collected for biomarker measurements. Results A total of 80 pregnant women, including 43 in the GDM group and 37 in the non-GDM group, were included in the study. The maternal serum 8-isoprostane (8IsoP), tumor necrosis factor-α (TNF-α) and interleukin (IL)-10 levels were significantly higher in the GDM group than those in the non-GDM group (P: 0.028, P: 0.019 and P: 0.031, respectively). The fetal cardiac function parameters were not significantly different between the two groups. Regardless of the GDM status, the fetuses with high levels of oxidative stress (8Isop ≥1000 pg/mg protein) had a significantly higher rate of impaired shortening fraction (SF) of the left ventricle (P: 0.001). Conclusion GDM is significantly associated with an increase in the oxidative stress process, and a high level of oxidative stress was significantly associated with left ventricular (LV) function impairment. Though a correlation between GDM and fetal cardiac function impairment was not clearly demonstrated in this study, this study suggests that GDM patients with a high level of oxidative stress should be evaluated for fetal cardiac function.


Assuntos
Glicemia/metabolismo , Diabetes Gestacional , Ecocardiografia/métodos , Coração Fetal , Estresse Oxidativo , Adulto , Diabetes Gestacional/sangue , Diabetes Gestacional/diagnóstico , Diabetes Gestacional/fisiopatologia , Dinoprosta/análogos & derivados , Dinoprosta/sangue , Feminino , Coração Fetal/diagnóstico por imagem , Coração Fetal/metabolismo , Coração Fetal/fisiopatologia , Teste de Tolerância a Glucose/métodos , Humanos , Interleucina-10/sangue , Gravidez , Segundo Trimestre da Gravidez/fisiologia , Cuidado Pré-Natal/métodos , Fator de Necrose Tumoral alfa/sangue , Ultrassonografia Pré-Natal/métodos
14.
Arch Biochem Biophys ; 680: 108241, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31891670

RESUMO

Excessive iron accumulation in the heart can lead to iron overload cardiomyopathy (IOC), the leading cause of death in hemochromatosis patients. Current understanding regarding the mechanism by which iron overload causes a deterioration in cardiac performance, mitochondrial dysfunction, and impaired mitochondrial dynamics remains limited. Ferroptosis, a newly identified form of regulated cell death, has recently been revealed influencing the pathophysiological process of IOC. Nevertheless, the direct effect of cardiac iron overload on ferroptotic cell death is incompletely characterized. This review article comprehensively summarizes and discusses the effects of iron overload on cardiac mitochondrial function, cardiac mitochondrial dynamics, ferroptosis of cardiomyocytes, and left ventricular function in in vitro and in vivo reports. This review also provides relevant consistent and controversial information which can facilitate further mechanistic investigation into iron-induced cardiac dysfunction in the clinical setting in the near future.


Assuntos
Ferroptose , Sobrecarga de Ferro/metabolismo , Dinâmica Mitocondrial , Miócitos Cardíacos/metabolismo , Animais , Morte Celular , Humanos , Ferro/metabolismo , Sobrecarga de Ferro/patologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Miócitos Cardíacos/patologia , Estresse Oxidativo
15.
Toxicology ; 427: 152289, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31542421

RESUMO

The morbidity and mortality in thalassemia patients are predominantly caused by iron overload cardiomyopathy (IOC). Iron-induced cardiac intracellular Ca2+ ([Ca2+]i) dysregulation is among the core pathophysiological processes in IOC-related heart failure. Although cardioprotective roles of deferiprone (DFP) and N-acetylcysteine (NAC) have been reported, their effect on cardiac [Ca2+]i transients and Ca2+-regulatory protein expression in thalassemic mice is unknown. In the present study, iron overload condition was induced in wild-type (WT) and heterozygous ß-thalassemic (HT) mice by a high-iron diet. The iron-overloaded mice subsequently received a vehicle, DFP, NAC, or DFP plus NAC co-therapy. In both WT and HT iron-overloaded mice, DFP and NAC had similar efficacy in decreasing plasma non-transferrin-bound iron, decreasing cardiac iron concentration (CIC) and relieving systolic dysfunction. DFP plus NAC co-therapy, however, was better than the monotherapy in reducing CIC and restoring cardiac [Ca2+]i transient amplitude and rising rate. All regimens produced no change in cardiac Ca2+-regulatory protein expression. We provided the first evidence regarding the synergistic effect of combined iron chelator-antioxidant therapy on cardiac [Ca2+]i homeostasis in iron-overloaded thalassemic mice, with consistent improvement of cardiac contractility.


Assuntos
Acetilcisteína/farmacologia , Cálcio/metabolismo , Deferiprona/farmacologia , Quelantes de Ferro/farmacologia , Sobrecarga de Ferro/metabolismo , Miocárdio/metabolismo , Talassemia/metabolismo , Animais , Coração/efeitos dos fármacos , Homeostase , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Função Ventricular Esquerda/efeitos dos fármacos
16.
EBioMedicine ; 34: 256-266, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30049609

RESUMO

Ovarian cancer is the major cause of death out of all the gynecologic cancers. The prognosis of this cancer is quite poor since patients only seek treatment when it is at an advanced stage. Any early biomarkers for this cancer are still unknown. Dysregulation of mitochondrial dynamics with associated resistance to apoptosis plays a crucial role in several types of human carcinogenesis, including ovarian cancers. Previous studies showed that increased mitochondrial fission occurred in ovarian cancer cells. However, several pharmacological interventions and therapeutic strategies, which modify the mitochondrial dynamics through the promotion of mitochondrial fission and apoptosis of cancer cells, have been shown to potentially provide beneficial effects in ovarian cancer treatment. Therefore the aim of the present review is to summarize and discuss the current findings from in vitro, in vivo and clinical studies associated with the alteration of mitochondrial dynamics and ovarian cancers with and without interventions.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Dinâmica Mitocondrial/efeitos dos fármacos , Neoplasias Ovarianas/fisiopatologia , Animais , Feminino , Humanos , Neoplasias Ovarianas/tratamento farmacológico
17.
Cell Calcium ; 72: 18-25, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29748130

RESUMO

Although disturbance of cardiac Ca2+ regulation is involved in the pathophysiology of iron-overload cardiomyopathy, the obvious mechanisms involved in the dysregulation of iron-induced cardiac Ca2+ are unclear. Moreover, the roles of the iron chelator deferiprone and the T-type calcium channel blocker efonidipine on cardiac intracellular Ca2+ transients and Ca2+ regulatory proteins in thalassemic mice are still unknown. We tested the hypothesis that treatment with either deferiprone or efonidipine attenuated cardiac Ca2+ dysregulation and led to improved left ventricular (LV) function in iron-overloaded thalassemic mice. Wild-type (WT) mice and ß-thalassemic (HT) mice were fed with either a normal diet (ND) or a high iron-diet (FE) for 90 days. Then, the FE-fed mice were treated with either deferiprone (75 mg/kg/day) or efonidipine (4 mg/kg/day) for 30 days. ND-fed HT mice had an increase in T-type calcium channels (TTCC) and an increased level of sarcoplasmic reticulum Ca2+-ATPase (SERCA), compared with ND-fed WT mice. Chronic iron feeding led to an increase in TTCC and expression of SERCA proteins in FE-fed WT mice. Moreover, chronic iron overload led to increased plasma non-transferrin bound iron (NTBI) and cardiac iron deposition, impaired cardiac intracellular Ca2+ transients including decreased intracellular Ca2+ transient amplitude, rising rate and decay rate, as well as impaired LV function as indicated by a decreased %LV ejection fraction (%LVEF) in both WT and HT mice. Our findings showed that treatment with either deferiprone (DFP) or efonidipine (EFO) showed similar benefits in reducing plasma NTBI and cardiac iron deposition, and improving %LVEF from 84.3 (WT) to 89.3 (DFP) and 89.2 (EFO) treatment; and from 84.2 (HT) to 88.8 (DFP) and 89.5 (EFO) treatment, however there was no improvement in the regulation of cardiac Ca2+ in iron-overloaded thalassemic mice. These findings provide the understanding of the effects of these drugs on the iron-overloaded heart in thalassemic mice and suggest that an alternative intervention that could improve calcium regulation under this condition is needed to improve the therapeutic outcome. Moreover, whether the benefits of the TTCC blocker is via its inhibition of the TTCC alone or together with its ability to chelate iron are still unclear and need further investigation.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Cálcio/metabolismo , Quelantes/farmacologia , Deferiprona/farmacologia , Di-Hidropiridinas/farmacologia , Coração/fisiopatologia , Sobrecarga de Ferro/fisiopatologia , Nitrofenóis/farmacologia , Talassemia/fisiopatologia , Animais , Canais de Cálcio Tipo T/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Separação Celular , Coração/efeitos dos fármacos , Ferro/metabolismo , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Compostos Organofosforados/farmacologia , Transferrina/metabolismo , Função Ventricular/efeitos dos fármacos
18.
Exp Gerontol ; 103: 107-114, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29331535

RESUMO

The incidence of cardiovascular disease and metabolic syndrome increases after the onset of menopause, suggesting estrogen has a vital role in their prevention. Mitochondrial dynamics are known to play an important role in the maintenance of cardiac physiological function. However, the effects of estrogen deprivation on cardiometabolic status and cardiac mitochondrial dynamics under conditions of obese-insulin resistance have never been investigated. We hypothesized that estrogen deprivation aggravates cardiac dysfunction through increased cardiac mitochondrial fission in obese-insulin resistant rats. Female rats were fed on either a high fat (HFD, 57.60% fat) or normal (ND, 19.77% fat) diet for 13 weeks. The rats were then divided into 4 groups. Two sham groups (HFS and NDS) and 2 operated or ovariectomized (HFO and NDO) groups (n = 8/group). Six weeks after surgery, metabolic status, heart rate variability (HRV), left ventricular (LV) function, cardiac mitochondrial function and dynamics, and metabolic parameters were determined. Insulin resistance developed in NDO, HFS and HFO rats as indicated by increased plasma insulin and HOMA index. Although rats in both NDO and HFS groups had markedly impaired LV function indicated by reduced %LVFS and impaired cardiac mitochondrial function, rats in the HFO group had the most severe impairments. Moreover, the estrogen deprived rats (NDO and HFO) had increased cardiac mitochondrial fission through activation of phosphorylation of Drp-1 at serine 616. Our findings indicated that estrogen deprivation caused the worsening of LV dysfunction through increased cardiac mitochondrial fission in obese-insulin resistant rats.


Assuntos
Estrogênios/fisiologia , Resistência à Insulina , Dinâmica Mitocondrial , Obesidade/fisiopatologia , Animais , Doenças Cardiovasculares/fisiopatologia , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Frequência Cardíaca , Mitocôndrias Cardíacas/fisiologia , Ovariectomia , Estresse Oxidativo , Ratos , Ratos Wistar , Função Ventricular Esquerda
19.
Eur J Pharmacol ; 822: 43-50, 2018 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-29366843

RESUMO

Although both iron chelators and T-type calcium channel (TTCC) blockers have been shown to exert cardioprotection by decreasing cardiac iron deposition and reducing left ventricular (LV) dysfunction via different channels in iron-overloaded rodent models, the cardioprotective effects of combined iron chelator and TTCC blocker treatment in thalassemic mice has not been investigated. We hypothesized that a combined iron chelator and TTCC blocker exerts better cardioprotection than monotherapy by decreasing cardiac iron accumulation, apoptosis and oxidative stress. An iron-overload condition was induced in heterozygous ßKO thalassemic (HT) mice and wild-type (WT) mice by high iron diet consumption (FE) for 3 months. Then, the iron chelator deferiprone (DFP), the TTCC blocker efonidipine (Efo), and combined DFP plus Efo were fed via oral gavage for 1 month whilst the high iron diet was continued. LV function, heart rate variability (HRV), apoptosis and cardiac iron accumulation were determined. Chronic iron-overload in mice led to increased cardiac iron deposition, oxidative stress, apoptosis, and impaired LV function and HRV. Although DFP and Efo showed similar cardioprotective efficacy, the combined DFP plus Efo therapy exerted greater efficacy in reducing cardiac iron deposition and cellular apoptosis than either of the monotherapies in iron-overload thalassemic mice.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/metabolismo , Cardiotônicos/farmacologia , Quelantes/farmacologia , Sobrecarga de Ferro/complicações , Ferro/metabolismo , Talassemia/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/uso terapêutico , Cardiotônicos/uso terapêutico , Quelantes/uso terapêutico , Interações Medicamentosas , Frequência Cardíaca/efeitos dos fármacos , Masculino , Malondialdeído/sangue , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Talassemia/complicações , Talassemia/metabolismo , Talassemia/patologia , Disfunção Ventricular Esquerda/tratamento farmacológico
20.
Neurorehabil Neural Repair ; 32(2): 142-149, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29353543

RESUMO

BACKGROUND: Effects of Tai Chi (TC) on specific cognitive function and mechanisms by which TC may improve cognition in older adults with amnestic mild cognitive impairment (a-MCI) remain unknown. OBJECTIVE: To examine the effects of TC on cognitive functions and plasma biomarkers (brain-derived neurotrophic factor [BDNF], tumor necrosis factor-α [TNF-α], and interleukin-10 [IL-10]) in a-MCI. METHODS: A total of 66 older adults with a-MCI (mean age = 67.9 years) were randomized to either a TC (n = 33) or a control group (n = 33). Participants in the TC group learned TC with a certified instructor and then practiced at home for 50 min/session, 3 times/wk for 6 months. The control group received educational material that covered information related to cognition. The primary outcome was cognitive performance, including Logical Memory (LM) delayed recall, Block Design, Digit Span, and Trail Making Test B minus A (TMT B-A). The secondary outcomes were plasma biomarkers, including BDNF, TNF-α, and IL-10. RESULTS: At the end of the trial, performance on the LM and TMT B-A was significantly better in the TC group compared with the control group after adjusting for age, gender, and education ( P < .05). Plasma BDNF level was significantly increased for the TC group, whereas the other outcome measures were similar between the 2 groups after adjusting for age and gender ( P < .05). CONCLUSIONS: TC training significantly improved memory and the mental switching component of executive function in older adults with a-MCI, possibly via an upregulation of BDNF.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/sangue , Cognição/fisiologia , Disfunção Cognitiva/reabilitação , Função Executiva/fisiologia , Tai Chi Chuan , Idoso , Biomarcadores/sangue , Disfunção Cognitiva/sangue , Disfunção Cognitiva/psicologia , Feminino , Humanos , Interleucina-10/sangue , Masculino , Rememoração Mental/fisiologia , Pessoa de Meia-Idade , Testes Neuropsicológicos , Resultado do Tratamento , Fator de Necrose Tumoral alfa/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA