Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Exp Physiol ; 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38478872

RESUMO

Ischaemic heart diseases (IHD) are among the major causes of mortality in the elderly population. Although timely reperfusion is a common treatment for IHD, it causes additional damage to the ischaemic myocardium known as ischaemia-reperfusion (IR) injury. Considering the importance of preventing reperfusion injuries, we aimed to examine the combination effect of mitochondrial transplantation (MT) and coenzyme Q10 (CoQ10 ) in myocardial IR injury of aged male rats. Seventy-two aged male Wistar rats were randomly divided into six groups: Sham, IR, CoQ10 , MT, combination therapy (MT + CoQ10 ) and vehicle. Myocardial IR injury was established by occlusion of the left anterior descending coronary artery followed by reopening. Young male Wistar rats were used as mitochondria donors. Isolated mitochondria were injected intraventricularly (500 µL of a respiration buffer containing 6 × 106 ± 5 × 105  mitochondria/mL) in MT-receiving groups at the onset of reperfusion. CoQ10  (10 mg/kg/day) was injected intraperitoneally for 2 weeks before IR induction. Twenty-four hours after reperfusion, haemodynamic parameters, myocardial infarct size (IS), lactate dehydrogenase (LDH) release and cardiac mitochondrial function (mitochondrial reactive oxygen species (ROS) generation and membrane potential) were measured. The combination of MT and CoQ10  improved haemodynamic index changes and reduced IS and LDH release (P < 0.05). It also decreased mitochondrial ROS generation and increased membrane potential (P < 0.05). CoQ10 also showed a significant cardioprotective effect. Combination therapy displayed greater cardioprotective effects than single treatments. This study revealed that MT and CoQ10 combination treatment can be considered as a promising cardioprotective strategy to reduce myocardial IR injury in ageing, in part by restoring mitochondrial function.

2.
Mol Biol Rep ; 50(5): 3973-3983, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36829080

RESUMO

BACKGROUND: Aging, as a recognized risk factor for ischemic heart disease, interferes with protective mechanisms and abolishes the optimal effectiveness of cardioprotective interventions, leading to the loss of cardioprotection following myocardial ischemia/reperfusion (I/R) injury. This study was designed to explore the possible interaction of aging with cardioprotection induced by combination therapy with coenzyme Q10 (CoQ10) and mitochondrial transplantation in myocardial I/R injury of aged rats. METHODS: Male Wistar rats (n = 72, 400-450 g, 22-24 months old) were randomized into groups with/without I/R and/or CoQ10 and mitochondrial transplantation, alone or in a combinational mode. An in vivo model of myocardial I/R injury was established by left anterior descending coronary artery occlusion and re-opening. Mitochondria were isolated from donor rats and injected intramyocardially (150 µl of the mitochondrial suspension containing 2 × 105±0.3 × 105 mitochondria) at the onset of reperfusion in recipient groups. CoQ10 (20 mg/kg/day) was injected intramuscularly for 7 days before I/R operation. Lastly, myocardial function, cTn-I level, expression of autophagy-associated proteins (Beclin1, p62, and LC3-II/LC3-I), and the levels of pro-inflammatory cytokines (TNF-α, IL-1ß, and IL-6) were assessed. RESULTS: Co-application of CoQ10 and mitotherapy concomitantly improved myocardial function and decreased cTn-I level in aged reperfused hearts (P < .001). This combination therapy also modulated autophagic activity and decreased pro-inflammatory cytokines (P < .01 to P < .001). This combinational approach induced noticeable cardioprotection in comparison with monotherapies-received groups. CONCLUSION: We found that combination of CoQ10 and mitochondrial transplantation attenuated myocardial I/R injury in aged rats, in part by modulating autophagy and inflammatory response, hence, appears to restore aging-related loss of cardioprotection in aged patients.


Assuntos
Traumatismo por Reperfusão Miocárdica , Traumatismo por Reperfusão , Ratos , Masculino , Animais , Traumatismo por Reperfusão Miocárdica/metabolismo , Ratos Wistar , Mitocôndrias/metabolismo , Autofagia , Citocinas
3.
Mol Biol Rep ; 50(3): 2147-2158, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36565415

RESUMO

BACKGROUND: Sepsis-induced myocardial dysfunction is associated with worse clinical outcomes and high mortality, but no effective therapeutic intervention has been explored, reinforcing the urgent need to develop innovative strategies. Mitochondrial dysfunction underlies the pathogenesis of sepsis-induced myocardial dysfunction. Herein, we assessed the effect of mitochondrial transplantation on sepsis-induced myocardial dysfunction in a rat model of cecal ligation and puncture (CLP)-induced sepsis. METHODS: Male Wistar rats (n = 80, 12 weeks old, 250-300 g) were divided into groups with/without CLP-induced sepsis receiving mitochondrial transplantation in single or two repetitive injections (1 h or 1 and 7 h post-CLP, respectively). Mitochondria were isolated from donor rats and injected intravenously (400 µl of mitochondrial suspension containing 7.5 × 106 mitochondria/ml of respiration buffer) in recipient groups. Twenty-four hours post-operation, LDH and cTn-I levels, mitochondrial functional endpoints, expression of mitochondrial biogenesis (SIRT-1 and PGC-1α) and fission/fusion (Drp1/Mfn1 and Mfn2) genes, and inflammatory cytokines (TNF-α, IL-1ß, and IL-6) levels were evaluated. Survival was tested over 72 h post-operation. RESULTS: Mitotherapy significantly improved 72-hours survival (P < .05) and decreased LDH and cTn-I levels (P < .01). It also restored mitochondrial function and expression of mitochondrial biogenesis and fusion genes, and decreased the expression of mitochondrial fission gene and the levels of inflammatory cytokines (P < .05 to P < .01). Mitotherapy with repetitive injections at 1 and 7 h post-CLP provided noticeable mitoprotection in comparison with the group receiving mitotherapy at single injection. CONCLUSION: Mitotherapy improved mitochondrial function, biogenesis, and dynamic associated with SIRT-1/PGC-1α network and suppressed inflammatory response in CLP-induced sepsis model, therefore, offers a promising strategy to overcome life-threatening sepsis challenge.


Assuntos
Cardiomiopatias , Sepse , Ratos , Masculino , Animais , Ratos Wistar , Biogênese de Organelas , Mitocôndrias/metabolismo , Cardiomiopatias/metabolismo , Citocinas/metabolismo , Sepse/complicações , Sepse/terapia
4.
Mol Biol Rep ; 50(7): 5655-5665, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37199864

RESUMO

BACKGROUND: The aged myocardium experiences various forms of stress that cause reduction of its tolerance to injury induced by ischemia/reperfusion (I/R). Developing effective cardioprotective modalities to prevent the amplification of I/R injury during aging is under focus of investigation. Mesenchymal stem cells (MSCs) have the ability to regenerate infarcted myocardium mostly by producing multiple secretory factors. This study aimed to explore the mechanisms of mitoprotection by MSCs-conditioned medium (CM) in myocardial I/R injury of aged rats. METHODS: Male Wistar rats (n = 72, 400-450 g, 22-24 months old) were randomized into groups with/without I/R and/or MSCs-CM treatment. To establish myocardial I/R injury, the method of LAD occlusion and re-opening was employed. MSCs-CM was administered intramyocardially (150 µl) at the onset of reperfusion in recipient group. After 24 h reperfusion, myocardial infarct size, LDH level, mitochondrial functional endpoints, expression of mitochondrial biogenesis-associated genes, and the levels of pro-inflammatory cytokines were evaluated. After 28 days reperfusion, echocardiographic assessment of cardiac function was performed. RESULTS: MSCs-CM treatment improved myocardial function and decreased infarct size and LDH level in aged I/R rats (P < .05 to P < .001). It also decreased mitochondrial ROS formation, enhanced mitochondrial membrane potential and ATP content, upregulated mitochondrial biogenesis-related genes including SIRT-1, PGC-1α, and NRF-2, and lessened TNF-α, IL-1ß, and IL-6 levels (P < .05 to P < .01). CONCLUSIONS: MSCs-CM treatment attenuated myocardial I/R injury in aged rats, in part by improving mitochondrial function and biogenesis and restraining inflammatory reaction. the upregulation of SIRT-1/PGC-1α/NRF-2 profiles is a possible target for the mitoprotective effects of MSCs-CM following I/R injury during aging.


Assuntos
Células-Tronco Mesenquimais , Infarto do Miocárdio , Traumatismo por Reperfusão Miocárdica , Traumatismo por Reperfusão , Ratos , Masculino , Animais , Traumatismo por Reperfusão Miocárdica/metabolismo , Meios de Cultivo Condicionados/farmacologia , Meios de Cultivo Condicionados/metabolismo , Ratos Wistar , Infarto do Miocárdio/terapia , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão/metabolismo , Células-Tronco Mesenquimais/metabolismo
5.
Mol Biol Rep ; 50(4): 3525-3537, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36787055

RESUMO

BACKGROUND: The metabolic and intracellular abnormalities in aging and diabetes cause loss of cardioprotection by routine interventions against myocardial ischemia/reperfusion (I/R) injury. We aimed to evaluate the possible interaction of aging and type-2 diabetes mellitus with cardioprotection and the potential protective effect of a mitochondrial cocktail (melatonin/nicotinamide mononucleotide (NMN)/ubiquinol) on myocardial I/R injury in aged diabetic rats. METHODS: Male Wistar rats (n = 108, 22-24 months old, 400-450 g) received high-fat diet/low dose of streptozotocin to induce type-2 diabetes, then were randomized into 9 groups of 12 rats each with/without I/R and/or melatonin, NMN, and ubiquinol, alone or in dual or triple combinations. Myocardial I/R was induced by LAD occlusion for 30 min followed by 24 h reperfusion. NMN (100 mg/kg/48 h, intraperitoneally) was administered for 28 days before I/R operation. Melatonin (10 mg/kg, intraperitoneally) and/or ubiquinol (30 mg/kg, intravenously) were administered at early reperfusion. Finally, hemodynamic index changes, infarct size, CK-MB levels, mitochondrial functional endpoints, and expression of mitochondrial biogenesis genes (SIRT-1/PGC-1α/NRF-2/TFAM) were assessed. RESULTS: The solo and dual applications of melatonin, NMN, and ubiquinol did not exert remarkable cardioprotective impacts. However, the triple combination improved myocardial function and decreased infarct size and CK-MB levels following myocardial I/R (P < .05 to P < .01). It also improved mitochondrial function and restored mitochondrial biogenesis genes (P < .01). CONCLUSIONS: Combination therapy with melatonin, NMN, and ubiquinol exerted significant cardioprotection and improved mitochondrial function and biogenesis via upregulation of SIRT-1/PGC-1α/NRF-2/TFAM profiles in aged diabetic rats and, thus, offers a promising strategy for providing noticeable cardioprotection against I/R injury also in aged diabetic patients.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Melatonina , Isquemia Miocárdica , Traumatismo por Reperfusão Miocárdica , Ratos , Masculino , Animais , Melatonina/farmacologia , Melatonina/uso terapêutico , Mononucleotídeo de Nicotinamida/farmacologia , Mononucleotídeo de Nicotinamida/uso terapêutico , Diabetes Mellitus Experimental/tratamento farmacológico , Ratos Wistar , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Infarto/tratamento farmacológico , Morbidade , Isquemia
6.
Can J Physiol Pharmacol ; 101(12): 682-691, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37523770

RESUMO

This work evaluated the combined effects of alpha-lipoic acid (ALA) and ischemic postconditioning (Post) on myocardial infarction and cell death in rats with chronic type-II diabetes following ischemia/reperfusion injury. The rats received a high-fat diet and were given one intraperitoneal injection of 35 mg/kg streptozotocin to induce chronic diabetes. They were then pretreated with ALA (100 mg/kg/day, orally) for 5 weeks before undergoing ischemia/reperfusion (I/R) insult. The hearts experienced 35 min regional ischemia through ligating the left anterior descending coronary artery, followed by 60 min reperfusion. The Post protocol involved 6 cycles of a 10/10 s algorithm, applied during the early stage of reperfusion. The use of Post alone did not significantly alter lactate dehydrogenase and infarct size levels, while ALA showed positive effects. Similar findings were observed for apoptotic changes with single treatments. However, the concurrent administration of ALA and Post significantly reduced the protein expressions of Bax, Bax/Bcl2, and cleaved caspase-3 while increasing Bcl2 expression. Additionally, the histopathological findings of the combined therapy were superior to those of single treatments. The concomitant use of ALA and Post effectively inhibited apoptosis, leading to cardiac recovery after I/R injury in diabetic conditions. This strategy could improve outcomes for preserving diabetic hearts following I/R insults.


Assuntos
Diabetes Mellitus Experimental , Pós-Condicionamento Isquêmico , Infarto do Miocárdio , Traumatismo por Reperfusão Miocárdica , Ácido Tióctico , Ratos , Animais , Miocárdio/metabolismo , Ácido Tióctico/farmacologia , Ácido Tióctico/uso terapêutico , Pós-Condicionamento Isquêmico/métodos , Diabetes Mellitus Experimental/metabolismo , Proteína X Associada a bcl-2/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Infarto do Miocárdio/metabolismo , Apoptose
7.
J Res Med Sci ; 28: 35, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37213461

RESUMO

Background: Application of doxorubicin (DOX) in cancer patients is limited due to its dose-dependent toxicity to nontarget tissues such as testis and subsequent infertility. Due to limitation of our knowledge about the mechanisms of DOX toxicity in the reproductive system, reduction of DOX-induced testicular toxicity remains an actual and primary clinical challenge. Considering the potentials of troxerutin (TXR) in generating a protective phenotype in many tissues, we aimed to examine the effect of TXR on DOX-induced testicular toxicity by evaluating the histological changes and the expression of mitochondrial biogenesis genes and microRNA-140 (miR-140). Materials and Methods: Twenty-four adult male Wistar rats (250-300 g) were divided in groups with/without DOX and/or TXR. DOX was injected intraperitoneally at 6 consecutive doses over 12 days (cumulative dose: 12 mg/kg). TXR (150 mg/kg/day; orally) was administered for 4 weeks before DOX challenge. One week after the last injection of DOX, testicular histopathological changes, spermatogenesis activity, and expression of mitochondrial biogenesis genes and miR-140 were determined. Results: DOX challenge significantly increased testicular histopathological changes, decreased testicular expression profiles of sirtuin 1 (SIRT-1) and nuclear respiratory factor-2 (NRF-2), and increased expression of miR-140 (P < 0.05 to P < 0.01). Pretreatment of DOX-received rats with TXR significantly reversed testicular histopathological changes, spermatogenesis activity index, and the expression levels of SIRT-1, peroxisome proliferator-activated receptor-γ coactivator 1-alpha (PGC-1α), NRF-2, and miR-140 (P < 0.05 to P < 0.01). Conclusion: Reduction of DOX-induced testicular toxicity following TXR pretreatment was associated with upregulation of SIRT-1/PGC-1α/NRF-2 profiles and better regulation of miR-140 expression. It seems that improving microRNA-mitochondrial biogenesis network can play a role in the beneficial effect of TXR on DOX-induced testicular toxicity.

8.
Neurochem Res ; 47(2): 358-371, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34626305

RESUMO

Pathophysiology of depression in elderlies is linked to aging-associated increase in indoleamine 2,3-dioxygenase (IDO) levels and activity and kynurenine (Kyn) metabolites. Moreover, these aging-induced changes may alter the brain's responses to stress. Growing evidence suggested that young plasma can positively affect brain dysfunctions in old age. The present study aimed to investigate whether the antidepressant effects of young plasma administration in aged rats subjected to chronic unpredictable mild stress (CUMS) and underlying mechanisms, focusing on the prefrontal cortex (PFC). Young (3 months old) and aged (22 months old) male rats were divided into five groups; young control, aged control, aged rats subjected to CUMS (A + CUMS), aged rats subjected to CUMS and treated with young plasma (A + CUMS + YP), and aged rats subjected to CUMS and treated with old plasma (A + CUMS + OP). Plasma was injected (1 ml, intravenously) three times per week for four weeks. Young plasma significantly improved CUMS-induced depressive-like behaviors, evidenced by the increased sucrose consumption ratio in the sucrose preference test and the reduced immobility time in the forced swimming test. Furthermore, young plasma markedly reduced the levels of interferon-gamma (IFN-γ), IDO, Kyn, and Kyn to tryptophan (Kyn/Trp) ratio in PFC tissue. Expression levels of the serotonin transporter and growth-associated protein (GAP)-43 were also significantly increased after chronic administration of young plasma. These findings provide evidence for the antidepressant effect of young plasma in old age; however, whether it improves depressive behaviors or faster recovery from stress-induced deficits is required to be elucidated.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase , Cinurenina , Animais , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Depressão/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Cinurenina/metabolismo , Masculino , Córtex Pré-Frontal/metabolismo , Ratos , Estresse Psicológico/metabolismo
9.
Mol Biol Rep ; 49(11): 11081-11099, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36107370

RESUMO

INTRODUCTION:  Autophagy is known as a conserved mechanism in order to preserve cell survival under various stress conditions via maintaining cellular homeostasis. Although autophagy is active in the heart at baseline and plays a critical role in cell survival, inappropriate activation of autophagy following ischemia/reperfusion (I/R) injury leads to cell death. MAIN TEXT: The distinct functions of autophagy in myocardial I/R injury condition have been examined in numerous studies, however, contradicting results have been achieved in this field. These studies have documented that autophagy acts as a double-edged sword in myocardial I/R injury. Clarifying the exact role of autophagy in determining the health or death of cardiomyocytes under I/R injury is very helpful to achieve better cardioprotection in prospective clinical studies. Thus, autophagy may be an interesting target for the treatment or prevention of myocardial I/R injury. But before considering this matter, it is necessary to address the gaps in our knowledge about the complex role of autophagy in myocardial I/R injury. CONCLUSION: In this review, by providing updated data about the role of autophagy in the heart during ischemia and reperfusion, we tried to provide more insights in this context and encourage scientists to pay special attention towards manipulating autophagy as an intriguing and powerful approach in cardioprotection.


Assuntos
Traumatismo por Reperfusão Miocárdica , Humanos , Traumatismo por Reperfusão Miocárdica/metabolismo , Estudos Prospectivos , Autofagia/genética , Miócitos Cardíacos/metabolismo , Sobrevivência Celular
10.
Mol Biol Rep ; 49(9): 8209-8218, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35842854

RESUMO

BACKGROUND: Clinical application of doxorubicin (DOX) is restricted due to its cardiotoxicity, reinforcing the significance of exploring new strategies to counteract DOX-induced cardiotoxicity. The present work aimed to investigate the ameliorative impact of combination therapy with nicotinamide mononucleotide (NMN) and troxerutin (TXR) on DOX-induced cardiotoxicity, with mitochondrial function/biogenesis and inflammatory response approach. METHODS: Male Wistar rats (n = 30, 250-300 g) were divided into groups with/without DOX and/or NMN and TXR, alone or in combination. Rats underwent 6 consecutive intraperitoneal injections of DOX (cumulative dose: 12 mg/kg). NMN (100 mg/kg/day; intraperitoneally) and/or TXR (150 mg/kg/day; orally) were administered for 28 days before DOX challenge. Seven days following the last injection of DOX, evaluation of cardiac histopathological changes, BNP and LDH levels, mitochondrial function (membrane potential, ROS generation, and ATP levels), expression of proteins involved in mitochondrial biogenesis (PGC-1α, NRF1, and TFAM), and inflammatory cytokines (TNF-α, IL-1ß, and IL-6) was performed. RESULTS: Combination of NMN and TXR significantly decreased the severity of histopathological damages, and BNP and LDH levels (P < 0.01 to P < 0.001). It also restored mitochondrial functional endpoints, and expression of proteins involved in mitochondrial biogenesis (P < 0.05 to P < 0.001), and decreased inflammatory cytokines (P < 0.01 to P < 0.001). The positive impacts of this combination therapy were more potent as compared to monotherapies. CONCLUSIONS: These findings shed new light on the understanding of additive properties of NMN/TXR combination therapy in protecting against DOX-induced cardiotoxicity. The cardioprotective effect of this combination therapy may be mediated in part through the restoration of mitochondrial function/biogenesis associated with the PGC-1α/NRF1/TFAM pathway, and suppression of inflammatory response.


Assuntos
Cardiotoxicidade , Mononucleotídeo de Nicotinamida , Animais , Antibióticos Antineoplásicos/farmacologia , Apoptose , Cardiotoxicidade/tratamento farmacológico , Cardiotoxicidade/prevenção & controle , Citocinas , Doxorrubicina/toxicidade , Hidroxietilrutosídeo/análogos & derivados , Masculino , Mononucleotídeo de Nicotinamida/farmacologia , Biogênese de Organelas , Estresse Oxidativo , Ratos , Ratos Wistar
11.
Mol Biol Rep ; 49(3): 1773-1782, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35098396

RESUMO

BACKGROUND: Investigating the interaction of diabetes with ischemic postconditioning (IPostC)-associated cardioprotection in myocardial ischemia/reperfusion (I/R) damage is of great clinical importance. The present work was designed to determine the possible synergistic effects of alpha-lipoic acid (LA) preconditioning and IPostC on myocardial I/R damage in type-II diabetic rats through modulating autophagy, and the involvement of mitochondrial function. METHODS: High-fat diet/low dose of streptozotocin-induced type-II diabetic model with duration of 12 weeks was used in this study. LA (100 mg/kg/day) was administered orally in diabetic rats for 5 weeks before I/R. Myocardial I/R was established on Langendorff apparatus through the ligation of left anterior descending coronary artery for 35 min, then reperfusion for 60 min. IPostC was carried out immediately at the beginning of the reperfusion. At the end of the experiment, myocardial infarct size (IS), autophagy markers at both gene and protein levels, and mitochondrial ROS production and membrane potential were assessed. RESULTS: Combined conditioning with LA and ischemia significantly decreased the IS of diabetic hearts (P < 0.05), however, single therapies had no significant effects. LA in combination with IPostC more significantly decreased LC3 and p62 mRNA levels (P < 0.01), and LC3II/LC3I and p62 protein levels (P < 0.01). Also, this combined therapy decreased mitochondrial ROS generation and membrane depolarization (P < 0.01). CONCLUSIONS: Pretreatment with LA in diabetic rats notably restored cardioprotection by IPostC via modulating autophagy and restoring mitochondrial function. This combined conditioning might be an effective strategy to diminish I/R damage in diabetic hearts.


Assuntos
Diabetes Mellitus Experimental , Pós-Condicionamento Isquêmico , Precondicionamento Isquêmico Miocárdico , Traumatismo por Reperfusão Miocárdica , Ácido Tióctico , Animais , Autofagia , Diabetes Mellitus Experimental/metabolismo , Mitocôndrias/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Ratos , Ácido Tióctico/farmacologia
12.
Clin Exp Pharmacol Physiol ; 49(4): 474-482, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34854121

RESUMO

The present study aims to evaluate the combined effect of ischaemic postconditioning (IPostC) and nicotinamide mononucleotide (NMN) on cardioprotection and mitochondrial function in aged rats subjected to myocardial ischaemia-reperfusion (IR) injury. Sixty aged Wistar rats were randomly divided into five groups (n = 12), including sham, control, NMN, IPostC, and NMN + IPostC. Regional ischaemia was induced by 30-min occlusion of the left anterior descending coronary artery (LAD) followed by 60-min reperfusion. IPostC was applied at the onset of reperfusion, by six cycles of 10-s reperfusion/ischaemia. NMN (100 mg/kg) was intraperitoneally injected every other day for 28 days before IR. Myocardial haemodynamics and infarct size (IS) were measured, and the left ventricles samples were harvested to assess cardiac mitochondrial function. The results showed that all treatments reduced lactate dehydrogenase release compared to those of the control group. IPostC alone failed to reduce IS and myocardial function. However, NMN and combined therapy could significantly improve myocardial function and decrease the IS compared to the control animals. Moreover, the effects of combined therapy on the decrease of IS, mitochondrial reactive oxygen species (ROS), and improvement of mitochondrial membrane potential (MMP) were greater than those of stand-alone treatments. These results demonstrated that cardioprotection by combined therapy with NMN + IPostC was superior to individual treatments, and pretreatment of aged rats with NMN was able to correct the failure of IPostC in protecting the hearts of aged rats against IR injury.


Assuntos
Pós-Condicionamento Isquêmico , Traumatismo por Reperfusão Miocárdica , Traumatismo por Reperfusão , Animais , Pós-Condicionamento Isquêmico/métodos , Mitocôndrias , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Mononucleotídeo de Nicotinamida/farmacologia , Mononucleotídeo de Nicotinamida/uso terapêutico , Ratos , Ratos Wistar
13.
Can J Infect Dis Med Microbiol ; 2022: 3277274, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35706715

RESUMO

Sepsis is defined as a life-threatening organ failure due to dysregulated host response to infection. Despite current advances in our knowledge about sepsis, it is still considered as a major global health challenge. Myocardial dysfunction is a well-defined manifestation of sepsis which is related to worse outcomes in septic patients. Given that the heart is a mitochondria-rich organ and the normal function of mitochondria is essential for successful modulation of septic response, the contribution of mitochondrial damage in sepsis-related myocardial dysfunction has attracted the attention of many scientists. It is widely accepted that mitochondrial damage is involved in sepsis-related myocardial dysfunction; however, effective and potential treatment modalities in clinical setting are still lacking. Mitochondrial-based therapies are potential approaches in sepsis treatment. Although various therapeutic strategies have been used for mitochondrial function improvement, their effects are limited when mitochondria undergo irreversible alterations under septic challenge. Therefore, application of more effective approaches such as mitochondrial transplantation has been suggested. This review highlights the crucial role of mitochondrial damage in sepsis-related myocardial dysfunction, then provides an overview on mitochondrial-based therapies and current approaches to mitochondrial transplantation as a novel strategy, and proposes future directions for more researches in this field.

14.
Exp Physiol ; 106(7): 1621-1630, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34018261

RESUMO

NEW FINDINGS: What is the central question of this study? Young plasma contains several rejuvenating factors that exert beneficial effects in ageing and neurodegenerative diseases: can repeated transfusion of young plasma improve depressive behaviour in aged rats? What is the main finding and its importance? Following chronic transfusion of young plasma, depressive behaviour was improved in the depression model of aged rats, which was associated with reduced apoptosis process in the prefrontal cortex. ABSTRACT: Brain ageing alters brain responses to stress, playing an essential role in the pathophysiology of late-life depression. Moreover, apoptotic activity is up-regulated in the prefrontal cortex in ageing and stress-related mood disorders. Considerable evidence suggests that factors in young blood could reverse age-related dysfunctions in organs, especially in the brain. Therefore, this study investigated the effect of young plasma administration on depressive behaviours in aged rats exposed to chronic unpredictable mild stress (CUMS), with a focus on the apoptosis process. Young (3 months old) and aged (22 months old) male rats were randomly assigned into four groups: young control (YC), aged control (AC), aged rats subjected to CUMS (A+CUMS) and aged rats subjected to CUMS and treated with young plasma (A+CUMS+YP). In the A+CUMS and A+CUMS+YP groups, CUMS was used to generate the depression rat model. Moreover, the A+CUMS+YP group received pooled plasma (1 ml, intravenously), collected from young rats, three times per week for 4 weeks. Young plasma administration significantly improved CUMS-induced depression-like behaviours, including decreased sucrose consumption ratio, reduced locomotor activity and prolonged immobility time. Importantly, young plasma reduced neuronal apoptosis in the prefrontal cortex that was associated with reduced TUNEL-positive cells and cleaved caspase-3 protein levels in the A+CUMS+YP compared with the A+CUMS group. Young plasma can partially improve the neuropathology of late-life depression through the apoptotic signalling pathways.


Assuntos
Antidepressivos , Depressão , Animais , Antidepressivos/farmacologia , Apoptose , Depressão/metabolismo , Depressão/terapia , Modelos Animais de Doenças , Hipocampo , Masculino , Córtex Pré-Frontal/metabolismo , Ratos , Estresse Psicológico/metabolismo
15.
Mol Biol Rep ; 48(4): 3337-3348, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33895973

RESUMO

Human amniotic membrane mesenchymal stem cells-derived conditioned medium (hAM-MSCs-CM) has positive effects against myocardial ischemia/reperfusion (MI/R) injury. However, it needs further investigations how hAM-MSCs-CM leads to the cell survival under MI/R via modulation of autophagy. The purpose of this study is investigating the effects of hAM-MSCs-CM in a rat model of MI/R injury by focusing on the role of autophagy as one of its possible mechanisms. Male Wistar rats (44 rats, 175-200 g) were randomly divided into four groups: Sham, MI/R, culture media-receiving and conditioned medium-receiving. MI/R was induced by 30 min of left anterior descending coronary artery ligation. After 15 min reperfusion, culture media or hAM-MSCs-CM (150 µl) were injected intramyocardially. At the end of the experiment, CK-MB, autophagy markers, phosphorylated and total forms of mTOR and ULK1, cardiac function and fibrosis were measured. hAM-MSCs-CM significantly decreased CK-MB levels (P < 0.0001), and also the mRNA levels of Beclin1 (P < 0.0001), LC3 (P = 0.012) and p62 (P = 0.003). In addition, hAM-MSCs-CM significantly reduced Beclin1, LC3II/LC3I and p62 protein levels (P < 0.0001), and increased p-mTOR/mTOR (P = 0.022) and p-ULK1/ULK1 (P < 0.0001) expressions. Moreover, hAM-MSCs-CM improved cardiac function and decreased fibrosis (P < 0.0001). This study showed cardioprotective effects of hAM-MSCs-CM against MI/R injury through modulation of autophagy via mTOR/ULK1 pathway. Based on these findings, it can be concluded that hAM-MSCs-CM can be offered as an attractive candidate for attenuation of MI/R injury in future, but needs further investigations.


Assuntos
Autofagia , Meios de Cultivo Condicionados/farmacologia , Coração/efeitos dos fármacos , Células-Tronco Mesenquimais , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Animais , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Modelos Animais de Doenças , Fibrose , Coração/fisiopatologia , Humanos , Masculino , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Ratos , Ratos Wistar , Serina-Treonina Quinases TOR
16.
Mol Biol Rep ; 48(4): 3089-3096, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33866495

RESUMO

It has been documented that aging increases the risk of cardiovascular disease including myocardial ischemia/reperfusion (IR) injury and acute myocardial infarction. In this study, we aimed to investigate the individual or combined effects of nicotinamide mononucleotide (NMN) and melatonin (Mel) treatment on apoptotic markers, expression of SIRT3, and FOXO1, and infarct size of the aged myocardium subjected to IR injury. Sixty aged Wistar rats (22-24 months) were assigned to five groups including sham, IR, NMN+IR, Mel+IR, and NMN+Mel+IR (combination therapy). Isolated hearts were exposed to 30-min regional ischemia followed by 60-min reperfusion. NMN (100 mg/kg/day/i.p.) was injected every second day starting on day 28 before IR injury. Melatonin was added to the perfusion solution five minutes prior to and until 15 min after the start of reperfusion. The infarct size was assessed by computerized planimetry. The mRNA levels of SIRT3, FOXO1, and apoptotic genes Bax, Bcl-2, and Caspase-3 were estimated by real-time PCR. All treatments reduced infarct size as compared with the IR group. Melatonin and NMN upregulated the gene expression of Bcl-2, SIRT3, and FOXO1 and downregulated the gene expression of Bax, and Caspase-3, in comparison to the IR group. Also, the protein levels of SIRT3, quantified by Western blotting, were upregulated by the interventions. The effects of combination therapy were significantly greater than those of melatonin or NMN alone. These findings indicate that the combined administration of NMN and melatonin can protect the aged heart against IR injury by decreasing apoptosis and activating the SIRT3/FOXO1 pathway.


Assuntos
Apoptose/efeitos dos fármacos , Melatonina/farmacologia , Traumatismo por Reperfusão Miocárdica , Mononucleotídeo de Nicotinamida/farmacologia , Envelhecimento/efeitos dos fármacos , Animais , Combinação de Medicamentos , Proteína Forkhead Box O1/efeitos dos fármacos , Proteína Forkhead Box O1/metabolismo , Coração/fisiopatologia , Masculino , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Proteínas do Tecido Nervoso/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Sirtuína 3/efeitos dos fármacos , Sirtuína 3/metabolismo
17.
Biogerontology ; 20(4): 381-395, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30838484

RESUMO

Nicotinamide adenine dinucleotide (NAD+) has been described as central coenzyme of redox reactions and is a key regulator of stress resistance and longevity. Aging is a multifactorial and irreversible process that is characterized by a gradual diminution in physiological functions in an organism over time, leading to development of age-associated pathologies and eventually increasing the probability of death. Ischemia is the lack of nutritive blood flow that causes damage and mortality that mostly occurs in various organs during aging. During the process of aging and related ischemic conditions, NAD+ levels decline and lead to nuclear and mitochondrial dysfunctions, resulting in age-related pathologies. The majority of studies have shown that restoring of NAD+ using supplementation with intermediates such as nicotinamide mononucleotide and nicotinamide riboside can be a valuable strategy for recovery of ischemic injury and age-associated defects. This review summarizes the molecular mechanisms responsible for the reduction in NAD+ levels during ischemic disorders and aging, as well as a particular focus is given to the recent progress in the understanding of NAD+ precursor's effects on aging and ischemia.


Assuntos
Envelhecimento/fisiologia , Isquemia , NAD/metabolismo , Descoberta de Drogas , Humanos , Isquemia/metabolismo , Isquemia/fisiopatologia , Isquemia/terapia , Mitocôndrias/metabolismo , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Mononucleotídeo de Nicotinamida/farmacologia , Compostos de Piridínio
18.
J Electrocardiol ; 55: 9-15, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31048226

RESUMO

BACKGROUND: Because clinical use of doxorubicin (DOX) in chemotherapy is limited due to cardiotoxicity, finding new strategies to alleviate DOX burden and improving patients' health are necessary. Due to positive cardiovascular impacts of high-intensity interval training (HIIT), here we have investigated the effect of HIIT on DOX-induced cardiotoxicity by evaluating the myocardial apoptosis mechanism as well as microRNA-499a-5p expression. METHODS: Male Wistar rats (250-270 g) were randomly allocated into four groups: control, HIIT, DOX, and HIIT+DOX. HIIT was performed as 7 sets of alternative intervals of high and low trainings for 1 h a day, 5 days a week for 6 weeks using a rodent treadmill. After the last session of HIIT, the trained and time-matched control rats received intraperitoneal injection of DOX (20 mg/kg). Three days later, the left ventricular samples were obtained to determine the expression of microRNA and genes and proteins regulating apoptosis via real-time PCR. Myocardial apoptosis was also evaluated using TUNEL staining method. RESULTS: DOX administration significantly increased the expression levels of Bax and caspase-6 mRNAs, Bax protein and Bax/Bcl2 ratio, while reduced the expression levels of Bcl2 mRNA and protein in comparison to control group (P < 0.01). Pre-treatment of DOX-received rats with HIIT significantly up-regulated the Bcl2 and reduced the Bax, Bax/Bcl2, and caspase-6 expression profiles toward control values (P < 0.05), not affecting GSK-3ß expression. In addition, DOX toxicity significantly overexpressed microRNA-499, comparing to control rats (P < 0.01). HIIT significantly reversed this overexpression and also reduced TUNEL-positive apoptotic cells in DOX-received rats (P < 0.05). CONCLUSIONS: The data suggested that prior training of rats with HIIT had protective effect on DOX cardiotoxicity through reversing the expression profiles of pro- and anti-apoptotic factors and microRNA-499 and reducing myocardial apoptosis.


Assuntos
Treinamento Intervalado de Alta Intensidade , MicroRNAs , Animais , Cardiotoxicidade , Doxorrubicina , Eletrocardiografia , Glicogênio Sintase Quinase 3 beta , Humanos , Masculino , MicroRNAs/genética , Ratos , Ratos Wistar
19.
Eur J Clin Microbiol Infect Dis ; 37(12): 2225-2233, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30187247

RESUMO

Several risk factors have been described for the pathogenesis of atherosclerosis. Infectious diseases are suggested to be a causative factor, and some viruses have been studied for their relation with atherosclerotic diseases. Studies report two hypotheses, direct and indirect effects, for the role of viral infections in atherogenesis. Viruses are able to initiate atherosclerosis by two different pathways. They can exert their direct effects on atherogenesis by infecting vascular cells and then inducing inflammation in the endothelium and smooth muscle cells. Alternatively, they can also apply indirect effects by infecting non-vascular cells and inducing systemic inflammation. In this review, we consider the available data about the effects and correlations of DNA and RNA viruses on atherosclerosis.


Assuntos
Aterosclerose/fisiopatologia , Aterosclerose/virologia , Infecções por Vírus de DNA/complicações , Inflamação/virologia , Infecções por Vírus de RNA/complicações , Animais , Aterosclerose/etiologia , Vírus de DNA , Humanos , Inflamação/etiologia , Camundongos , Vírus de RNA , Fatores de Risco
20.
J Cell Mol Med ; 21(10): 2257-2267, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28402080

RESUMO

Ischaemic postconditioning (IPostC) was introduced for the first time by Zhao et al. as a feasible method for reduction of myocardial ischaemia-reperfusion (IR) injury. The cardioprotection by this protocol has been extensively evaluated in various species. Then, further research revealed that IPostC is a safe and convenient approach in limiting IR injury of non-myocardial tissues such as lung, liver, kidney, intestine, skeletal muscle, brain and spinal cord. IPostC has been conducted with different algorithms, resulting in diverse effects. The possible important factors leading to these differences are the difference in activation levels of signalling pathways and protective mediators by any algorithm, presence or absence of IPostC effectors in each tissue, or intrinsic characteristics of the tissues as well as the methodological biases. Also, the conflicting results have been shown with the application of the same algorithm of IPostC in certain tissues or animal species. The effectiveness of IPostC may depend upon various parameters including the species and the tissues characteristics. For example, different heart rates and metabolic rates of the species and unequal amounts of perfusion and blood flow of the tissues should be considered as the important determinants of IPostC effectiveness and should be thought about in designing IPostC algorithms for future studies. Due to these discrepancies, there is still no optimal single IPostC algorithm applicable to any tissue or any species. This issue is the main topic of the present article.


Assuntos
Algoritmos , Pós-Condicionamento Isquêmico/métodos , Especificidade de Órgãos , Traumatismo por Reperfusão/prevenção & controle , Animais , Humanos , Rim/irrigação sanguínea , Rim/fisiopatologia , Pulmão/irrigação sanguínea , Pulmão/fisiopatologia , Traumatismo por Reperfusão/fisiopatologia , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa