Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 595
Filtrar
1.
Int J Mol Sci ; 25(9)2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38731864

RESUMO

The human brain possesses three predominate phospholipids, phosphatidylcholine (PC), phosphatidylethanolamine (PE) and phosphatidylserine (PS), which account for approximately 35-40%, 35-40%, and 20% of the brain's phospholipids, respectively. Mitochondrial membranes are relatively diverse, containing the aforementioned PC, PE, and PS, as well as phosphatidylinositol (PI) and phosphatidic acid (PA); however, cardiolipin (CL) and phosphatidylglycerol (PG) are exclusively present in mitochondrial membranes. These phospholipid interactions play an essential role in mitochondrial fusion and fission dynamics, leading to the maintenance of mitochondrial structural and signaling pathways. The essential nature of these phospholipids is demonstrated through the inability of mitochondria to tolerate alteration in these specific phospholipids, with changes leading to mitochondrial damage resulting in neural degeneration. This review will emphasize how the structure of phospholipids relates to their physiologic function, how their metabolism facilitates signaling, and the role of organ- and mitochondria-specific phospholipid compositions. Finally, we will discuss the effects of global ischemia and reperfusion on organ- and mitochondria-specific phospholipids alongside the novel therapeutics that may protect against injury.


Assuntos
Encéfalo , Parada Cardíaca , Mitocôndrias , Fosfolipídeos , Humanos , Fosfolipídeos/metabolismo , Mitocôndrias/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Parada Cardíaca/metabolismo , Transdução de Sinais , Membranas Mitocondriais/metabolismo , Dinâmica Mitocondrial
2.
Eur J Pharmacol ; 974: 176633, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38703975

RESUMO

Cardiac arrest (CA) remains a leading cause of death, with suboptimal survival rates despite efforts involving cardiopulmonary resuscitation and advanced life-support technology. Post-resuscitation myocardial dysfunction (PRMD) is an important determinant of patient outcomes. Myocardial ischemia/reperfusion injury underlies this dysfunction. Previous reports have shown that ruthenium red (RR) has a protective effect against cardiac ischemia-reperfusion injury; however, its precise mechanism of action in PRMD remains unclear. This study investigated the effects of RR on PRMD and analyzed its underlying mechanisms. Ventricular fibrillation was induced in rats, which were then subjected to cardiopulmonary resuscitation to establish an experimental CA model. At the onset of return of spontaneous circulation, RR (2.5 mg/kg) was administered intraperitoneally. Our study showed that RR improved myocardial function and reduced the production of oxidative stress markers such as malondialdehyde (MDA), glutathione peroxidase (GSSG), and reactive oxygen species (ROS) production. RR also helped maintain mitochondrial structure and increased ATP and GTP levels. Additionally, RR effectively attenuated myocardial apoptosis. Furthermore, we observed downregulation of proteins closely related to mitophagy, including ubiquitin-specific protease 33 (USP33) and P62, whereas LC3B (microtubule-associated protein light chain 3B) was upregulated. The upregulation of mitophagy may play a critical role in reducing myocardial injury. These results demonstrate that RR may attenuate PRMD by promoting mitophagy through the inhibition of USP33. These effects are likely mediated through diverse mechanisms, including antioxidant activity, apoptosis suppression, and preservation of mitochondrial integrity and energy metabolism. Consequently, RR has emerged as a promising therapeutic approach for addressing post-resuscitation myocardial dysfunction.


Assuntos
Modelos Animais de Doenças , Parada Cardíaca , Mitofagia , Ratos Sprague-Dawley , Rutênio Vermelho , Animais , Mitofagia/efeitos dos fármacos , Parada Cardíaca/complicações , Parada Cardíaca/tratamento farmacológico , Parada Cardíaca/metabolismo , Parada Cardíaca/fisiopatologia , Ratos , Masculino , Rutênio Vermelho/farmacologia , Rutênio Vermelho/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Ubiquitina Tiolesterase/metabolismo , Reanimação Cardiopulmonar , Regulação para Cima/efeitos dos fármacos , Miocárdio/patologia , Miocárdio/metabolismo , Apoptose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/fisiopatologia
3.
Cells ; 13(9)2024 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-38727320

RESUMO

Cardiac arrest survivors suffer the repercussions of anoxic brain injury, a critical factor influencing long-term prognosis. This injury is characterised by profound and enduring metabolic impairment. Ketone bodies, an alternative energetic resource in physiological states such as exercise, fasting, and extended starvation, are avidly taken up and used by the brain. Both the ketogenic diet and exogenous ketone supplementation have been associated with neuroprotective effects across a spectrum of conditions. These include refractory epilepsy, neurodegenerative disorders, cognitive impairment, focal cerebral ischemia, and traumatic brain injuries. Beyond this, ketone bodies possess a plethora of attributes that appear to be particularly favourable after cardiac arrest. These encompass anti-inflammatory effects, the attenuation of oxidative stress, the improvement of mitochondrial function, a glucose-sparing effect, and the enhancement of cardiac function. The aim of this manuscript is to appraise pertinent scientific literature on the topic through a narrative review. We aim to encapsulate the existing evidence and underscore the potential therapeutic value of ketone bodies in the context of cardiac arrest to provide a rationale for their use in forthcoming translational research efforts.


Assuntos
Parada Cardíaca , Corpos Cetônicos , Corpos Cetônicos/metabolismo , Humanos , Parada Cardíaca/metabolismo , Animais , Dieta Cetogênica
4.
J Neurosci ; 44(19)2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38565288

RESUMO

Excitotoxicity and the concurrent loss of inhibition are well-defined mechanisms driving acute elevation in excitatory/inhibitory (E/I) balance and neuronal cell death following an ischemic insult to the brain. Despite the high prevalence of long-term disability in survivors of global cerebral ischemia (GCI) as a consequence of cardiac arrest, it remains unclear whether E/I imbalance persists beyond the acute phase and negatively affects functional recovery. We previously demonstrated sustained impairment of long-term potentiation (LTP) in hippocampal CA1 neurons correlating with deficits in learning and memory tasks in a murine model of cardiac arrest/cardiopulmonary resuscitation (CA/CPR). Here, we use CA/CPR and an in vitro ischemia model to elucidate mechanisms by which E/I imbalance contributes to ongoing hippocampal dysfunction in male mice. We reveal increased postsynaptic GABAA receptor (GABAAR) clustering and function in the CA1 region of the hippocampus that reduces the E/I ratio. Importantly, reduced GABAAR clustering observed in the first 24 h rebounds to an elevation of GABAergic clustering by 3 d postischemia. This increase in GABAergic inhibition required activation of the Ca2+-permeable ion channel transient receptor potential melastatin-2 (TRPM2), previously implicated in persistent LTP and memory deficits following CA/CPR. Furthermore, we find Ca2+-signaling, likely downstream of TRPM2 activation, upregulates Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity, thereby driving the elevation of postsynaptic inhibitory function. Thus, we propose a novel mechanism by which inhibitory synaptic strength is upregulated in the context of ischemia and identify TRPM2 and CaMKII as potential pharmacological targets to restore perturbed synaptic plasticity and ameliorate cognitive function.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Camundongos Endogâmicos C57BL , Transdução de Sinais , Canais de Cátion TRPM , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Masculino , Camundongos , Canais de Cátion TRPM/metabolismo , Transdução de Sinais/fisiologia , Isquemia Encefálica/metabolismo , Região CA1 Hipocampal/metabolismo , Receptores de GABA-A/metabolismo , Hipocampo/metabolismo , Inibição Neural/fisiologia , Neurônios GABAérgicos/metabolismo , Parada Cardíaca/complicações , Parada Cardíaca/metabolismo
5.
Am J Physiol Regul Integr Comp Physiol ; 326(1): R43-R52, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37899753

RESUMO

Hydrogen peroxide (H2O2) and calcium ions (Ca2+) are functional regulators of skeletal muscle contraction and metabolism. Although H2O2 is one of the activators of the type-1 ryanodine receptor (RyR1) in the Ca2+ release channel, the interdependence between H2O2 and Ca2+ dynamics remains unclear. This study tested the following hypotheses using an in vivo model of mouse tibialis anterior (TA) skeletal muscle. 1) Under resting conditions, elevated cytosolic H2O2 concentration ([H2O2]cyto) leads to a concentration-dependent increase in cytosolic Ca2+ concentration ([Ca2+]cyto) through its effect on RyR1; and 2) in hypoxia (cardiac arrest) and muscle contractions (electrical stimulation), increased [H2O2]cyto induces Ca2+ accumulation. Cytosolic H2O2 (HyPer7) and Ca2+ (Fura-2) dynamics were resolved by TA bioimaging in young C57BL/6J male mice under four conditions: 1) elevated exogenous H2O2; 2) cardiac arrest; 3) twitch (1 Hz, 60 s) contractions; and 4) tetanic (30 s) contractions. Exogenous H2O2 (0.1-100 mM) induced a concentration-dependent increase in [H2O2]cyto (+55% at 0.1 mM; +280% at 100 mM) and an increase in [Ca2+]cyto (+3% at 1.0 mM; +8% at 10 mM). This increase in [Ca2+]cyto was inhibited by pharmacological inhibition of RyR1 by dantrolene. Cardiac arrest-induced hypoxia increased [H2O2]cyto (+33%) and [Ca2+]cyto (+20%) 50 min postcardiac arrest. Compared with the exogenous 1.0 mM H2O2 condition, [H2O2]cyto after tetanic muscle contractions rose less than one-tenth as much, whereas [Ca2+]cyto was 4.7-fold higher. In conclusion, substantial increases in [H2O2]cyto levels evoke only modest Ca2+ accumulation via their effect on the sarcoplasmic reticulum RyR1. On the other hand, contrary to hypoxia secondary to cardiac arrest, increases in [H2O2]cyto from muscle contractions are small, indicating that H2O2 generation is unlikely to be a primary factor driving the significant Ca2+ accumulation after, especially tetanic, muscle contractions.NEW & NOTEWORTHY We developed an in vivo mouse myocyte H2O2 imaging model during exogenous H2O2 loading, ischemic hypoxia induced by cardiac arrest, and muscle contractions. In this study, the interrelationship between cytosolic H2O2 levels and Ca2+ homeostasis during muscle contraction and hypoxic conditions was revealed. These results contribute to the elucidation of the mechanisms of muscle fatigue and exercise adaptation.


Assuntos
Parada Cardíaca , Peróxido de Hidrogênio , Masculino , Animais , Camundongos , Peróxido de Hidrogênio/farmacologia , Peróxido de Hidrogênio/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Contração Muscular/fisiologia , Retículo Sarcoplasmático/metabolismo , Homeostase , Hipóxia/metabolismo , Parada Cardíaca/metabolismo , Cálcio/metabolismo , Fibras Musculares Esqueléticas
6.
Biomed Pharmacother ; 165: 115201, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37480828

RESUMO

BACKGROUND: Cardiac arrest (CA), characterized by sudden onset and high mortality rates, is one of the leading causes of death globally, with a survival rate of approximately 6-24%. Studies suggest that the restoration of spontaneous circulation (ROSC) hardly improved the mortality rate and prognosis of patients diagnosed with CA, largely due to ischemia-reperfusion injury. MAIN BODY: Mesenchymal stem cells (MSCs) exhibit self-renewal and strong potential for multilineage differentiation. Their effects are largely mediated by extracellular vesicles (EVs). Exosomes are the most extensively studied subgroup of EVs. EVs mainly mediate intercellular communication by transferring vesicular proteins, lipids, nucleic acids, and other substances to regulate multiple processes, such as cytokine production, cell proliferation, apoptosis, and metabolism. Thus, exosomes exhibit significant potential for therapeutic application in wound repair, tissue reconstruction, inflammatory reaction, and ischemic diseases. CONCLUSION: Based on similar pathological mechanisms underlying post-cardiac arrest syndrome involving various tissues and organs in many diseases, the review summarizes the therapeutic effects of MSC-derived exosomes and explores the prospects for their application in the treatment of CA.


Assuntos
Exossomos , Vesículas Extracelulares , Parada Cardíaca , Células-Tronco Mesenquimais , Humanos , Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Inflamação/metabolismo , Parada Cardíaca/metabolismo , Células-Tronco Mesenquimais/metabolismo
7.
J Clin Invest ; 133(9)2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37115695

RESUMO

Out-of-hospital cardiac arrest is a leading cause of death in the US, with a mortality rate over 90%. Preclinical studies demonstrate that cooling during cardiopulmonary resuscitation (CPR) is highly beneficial, but can be challenging to implement clinically. No medications exist for improving long-term cardiac arrest survival. We have developed a 20-amino acid peptide, TAT-PHLPP9c, that mimics cooling protection by enhancing AKT activation via PH domain leucine-rich repeat phosphatase 1 (PHLPP1) inhibition. Complementary studies were conducted in mouse and swine. C57BL/6 mice were randomized into blinded saline control and peptide-treatment groups. Following a 12-minute asystolic arrest, TAT-PHLPP9c was administered intravenously during CPR and significantly improved the return of spontaneous circulation, mean arterial blood pressure and cerebral blood flow, cardiac and neurological function, and survival (4 hour and 5 day). It inhibited PHLPP-NHERF1 binding, enhanced AKT but not PKC phosphorylation, decreased pyruvate dehydrogenase phosphorylation and sorbitol production, and increased ATP generation in heart and brain. TAT-PHLPP9c treatment also reduced plasma taurine and glutamate concentrations after resuscitation. The protective benefit of TAT-PHLPP9c was validated in a swine cardiac arrest model of ventricular fibrillation. In conclusion, TAT-PHLPP9c may improve neurologically intact cardiac arrest survival without the need for physical cooling.


Assuntos
Reanimação Cardiopulmonar , Peptídeos Penetradores de Células , Parada Cardíaca , Camundongos , Animais , Suínos , Reanimação Cardiopulmonar/efeitos adversos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Camundongos Endogâmicos C57BL , Parada Cardíaca/terapia , Parada Cardíaca/etiologia , Parada Cardíaca/metabolismo , Modelos Animais de Doenças
8.
Shock ; 59(6): 892-901, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36930651

RESUMO

ABSTRACT: Background : Systemic inflammation acts as a contributor to neurologic deficits after cardiac arrest (CA) and cardiopulmonary resuscitation (CPR). Extracellular cold-inducible RNA-binding, protein (CIRP) has been demonstrated to be responsible in part for the inflammation through binding to toll-like receptor 4 (TLR4) after cerebral ischemia. The short peptide C23 derived from CIRP has a high affinity for TLR4, we hypothesize that C23 reduces systemic inflammation after CA/CPR by blocking the binding of CIRP to TLR4. Methods : Adult male SD rats in experimental groups were subjected to 5 min of CA followed by resuscitation. C23 peptide (8 mg/kg) or normal saline was injected intraperitoneally at the beginning of the return of spontaneous circulation (ROSC). Results : The expressions of CIRP, TNF-α, IL-6, and IL-1ß in serum and brain tissues were significantly increased at 24 h after ROSC ( P < 0.05). C23 treatment could markedly decrease the expressions of TNF-α, IL-6, and IL-1ß in serum ( P < 0.05). Besides, it can decrease the expressions of TLR4, TNF-α, IL-6, and IL-1ß in the cortex and hippocampus and inhibit the colocalization of CIRP and TLR4 ( P < 0.05). In addition, C23 treatment can reduce the apoptosis of hippocampus neurons ( P < 0.05). Finally, the rats in the C23 group have improved survival rate and neurological prognosis ( P < 0.05). Conclusions: These findings suggest that C23 can reduce systemic inflammation and it has the potential to be developed into a possible therapy for post-CA syndrome.


Assuntos
Reanimação Cardiopulmonar , Parada Cardíaca , Animais , Ratos , Masculino , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Receptor 4 Toll-Like , Ratos Sprague-Dawley , Reanimação Cardiopulmonar/métodos , Peptídeos/farmacologia , Parada Cardíaca/metabolismo , Inflamação/metabolismo
9.
Mol Biotechnol ; 65(12): 2061-2070, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36944895

RESUMO

Ischemia/reperfusion (I/R) injury is the leading cause of death following cardiac arrest (CA) and cardiopulmonary resuscitation (CPR). γδT cells are suggested to aggravate blood-brain barrier (BBB) injury in various pathological processes. We herein investigate the effects of γδT cells inhibitor (UC7-13D5) against I/R injury post-CA/CPR. C57BL/6 mice were subjected to CA through injection of KCL (70 µL of 0.5 mol/L) and cessation of mechanical ventilation followed by CPR. Flow cytometry was performed to measure the proportion of CD3-positive cells after intraperitoneal injection of 200 µg UC7-13D5 at 6 h, 24 h, and 48 h post-resuscitation into mice. Neurological scores and modified neurological severity scores were assessed to examine neurological functions. Brain edema was estimated via brain water content measurements. Immunohistochemistry of caspase-3 and immunofluorescence staining of claudin-1, ZO-1 and CD31 were performed to detect neuronal apoptosis, BBB integrity and angiogenesis. Microvascular morphology in the cortical area was assessed via H&E staining. Oxidative stress was determined by measuring malondialdehyde, myeloperoxidase, xanthine oxidase, superoxide dismutase, and glutathione peroxidase activities. Western blotting was performed to measure the protein levels of Nuclear factor-E2-related factor 2 (Nrf2) and Heme oxygenase-1 (HO-1). UC7-13D5 effectively depleted γδT cells. Inhibition of γδT cells improved neurological deficits and reduced brain edema post-CA/CPR. γδT cells depletion attenuated neuronal apoptosis, BBB disruption and oxidative stress and promoted angiogenesis following CA/CPR. Inhibition of γδT cells facilitated the activation of the Nrf2/HO-1 pathway in CA/CPR-induced mice. Inhibition of γδT cells alleviates neurological deficits and cerebral edema in mice with CA/CPR by inhibiting neuronal apoptosis, BBB disruption and oxidative stress, and promoting angiogenesis via activation of the Nrf2/HO-1 pathway.


Assuntos
Edema Encefálico , Reanimação Cardiopulmonar , Parada Cardíaca , Camundongos , Animais , Barreira Hematoencefálica/metabolismo , Edema Encefálico/metabolismo , Fator 2 Relacionado a NF-E2 , Camundongos Endogâmicos C57BL , Parada Cardíaca/complicações , Parada Cardíaca/terapia , Parada Cardíaca/metabolismo , Estresse Oxidativo , Linfócitos T
10.
ACS Nano ; 17(4): 3528-3548, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36758159

RESUMO

Treatment of cardiac arrest/cardiopulmonary resuscitation (CA/CPR)-induced brain injury remains a challenging issue without viable therapeutic options. Octanoic acid (OA), a lipid oil that is mainly metabolized in the astrocytes of the brain, is a promising treatment for this type of injury owing to its potential functions against oxidative stress, apoptosis, inflammation, and ability to stabilize mitochondria. However, the application of OA is strictly limited by its short half-life and low available concentration in the target organ. Herein, based on our previous research, an OA-based nanotherapy coated with a neutrophil membrane highly expressing RVG29, RVG29-H-NPOA, was successfully constructed by computer simulation-guided supramolecular assembly of polyethylenimine and OA. The in vitro and in vivo experiments showed that RVG29-H-NPOA could target and be distributed in the injured brain focus via the relay-targeted delivery mediated by RVG29-induced blood-brain barrier (BBB) penetration and neutrophil membrane protein-induced BBB binding and injury targeting. This results in enhancements of the antioxidant, antiapoptotic, mitochondrial stability-promoting and anti-inflammatory effects of OA and exhibited systematic alleviation of astrocyte injury, neuronal damage, and inflammatory response in the brain. Due to their systematic intervention in multiple pathological processes, RVG29-H-NPOA significantly increased the 24 h survival rate of CA/CPR model rats from 40% to 100% and significantly improved their neurological functions. Thus, RVG29-H-NPOA are expected to be a promising therapeutic for the treatment of CA/CPR-induced brain injury.


Assuntos
Lesões Encefálicas , Reanimação Cardiopulmonar , Parada Cardíaca , Ratos , Animais , Simulação por Computador , Neutrófilos , Parada Cardíaca/tratamento farmacológico , Parada Cardíaca/metabolismo , Encéfalo/metabolismo , Reanimação Cardiopulmonar/métodos , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Modelos Animais de Doenças
11.
Cell Mol Neurobiol ; 43(5): 2179-2202, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36266523

RESUMO

Substantial morbidity and mortality are associated with postcardiac arrest brain injury (PCABI). MicroRNAs(miRNAs) are essential regulators of neuronal metabolism processes and have been shown to contribute to alleviated neurological injury after cardiac arrest. In this study, we identified miRNAs related to the prognosis of patients with neurological dysfunction after cardiopulmonary resuscitation based on data obtained from the Gene Expression Omnibus (GEO) database. Then, we explored the effects of miR-483-5p on mitochondrial biogenesis, mitochondrial-dependent apoptosis, and oxidative stress levels after ischemia‒reperfusion injury in vitro and in vivo. MiR-483-5p was downregulated in PC12 cells and hippocampal samples compared with that in normal group cells and hippocampi. Overexpression of miR-483-5p increased the viability of PC12 cells after ischemia‒reperfusion injury and reduced the proportion of dead cells. A western blot analysis showed that miR-483-5p increased the protein expression of PCG-1, NRF1, and TFAM and reduced the protein expression of Bax and cleaved caspase 3, inhibiting the release of cytochrome c from mitochondria and alleviating oxidative stress injury by inhibiting the production of ROS and reducing MDA activity. We confirmed that miR-483-5p targeted TNFSF8 to regulate the AMPK/JNK pathway, thereby playing a neuroprotective role after cardiopulmonary resuscitation. Hence, this study provides further insights into strategies for inhibiting neurological impairment after cardiopulmonary resuscitation and suggests a potential therapeutic target for PCABI.


Assuntos
Parada Cardíaca , MicroRNAs , Fármacos Neuroprotetores , Traumatismo por Reperfusão , Ratos , Animais , Humanos , Sistema de Sinalização das MAP Quinases , Fármacos Neuroprotetores/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Apoptose/genética , Traumatismo por Reperfusão/metabolismo , Mitocôndrias/metabolismo , Parada Cardíaca/complicações , Parada Cardíaca/genética , Parada Cardíaca/metabolismo
12.
Cell Mol Neurobiol ; 43(5): 1957-1974, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36006573

RESUMO

After restoration of spontaneous circulation (ROSC) following cardiac arrest, complements can be activated and excessive autophagy can contribute to the brain ischemia-reperfusion (I/R) injury. Mild hypothermia (HT) protects against brain I/R injury after ROSC, but the mechanisms have not been fully elucidated. Here, we found that HT significantly inhibited the increases in serum NSE, S100ß, and C5a, as well as neurologic deficit scores, TUNEL-positive cells, and autophagic vacuoles in the pig brain cortex after ROSC. The C5a receptor 1 (C5aR1) mRNA and the C5a, C5aR1, Beclin 1, LC3-II, and cleaved caspase-3 proteins were significantly increased, but the P62 protein and the PI3K/Akt/mTOR pathway-related proteins were significantly reduced in pigs after ROSC or neuronal oxygen-glucose deprivation/reoxygenation. HT could significantly attenuate the above changes in NT-treated neurons. Furthermore, C5a treatment induced autophagy and apoptosis and reduced the PI3K/Akt/mTOR pathway-related proteins in cultured neurons, which could be reversed by C5aR1 antagonist PMX205. Our findings demonstrated that C5a could bind to C5aR1 to induce neuronal autophagy during the brain I/R injury, which was associated with the inhibited PI3K/Akt/mTOR pathway. HT could inhibit C5a-induced neuronal autophagy by regulating the C5a-C5aR1 interaction and the PI3K/Akt/mTOR pathway, which might be one of the neuroprotective mechanisms underlying I/R injury. The C5a receptor 1 (C5aR1) mRNA and the C5a, C5aR1, Beclin 1, LC3-II, and cleaved caspase-3 proteins were significantly increased, but the P62 protein and the PI3K/Akt/mTOR pathway-related proteins were significantly reduced in pigs after ROSC or neuronal oxygen-glucose deprivation/reoxygenation. Mild hypothermia (HT) could significantly attenuate the above changes in NT-treated neurons. Furthermore, C5a treatment induced autophagy and apoptosis and reduced the PI3K/Akt/mTOR pathway-related proteins in cultured neurons, which could be reversed by C5aR1 antagonist PMX205. Proposed mechanism by which HT protects against brain I/R injury by repressing C5a-C5aR1-induced excessive autophagy. Complement activation in response to brain I/R injury generates C5a that can interact with C5aR1 to inactivate mTOR, probably through the PI3K-AKT pathway, which can finally lead to autophagy activation. The excessively activated autophagy ultimately contributes to cell apoptosis and brain injury. HT may alleviate complement activation and then reduce C5a-induced autophagy to protect against brain I/R injury. HT, mild hypothermia; I/R, ischemia reperfusion.


Assuntos
Parada Cardíaca , Hipotermia , Fármacos Neuroprotetores , Traumatismo por Reperfusão , Animais , Suínos , Caspase 3/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Hipotermia/metabolismo , Proteína Beclina-1/metabolismo , Receptor da Anafilatoxina C5a/metabolismo , Fármacos Neuroprotetores/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Encéfalo/metabolismo , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/metabolismo , Oxigênio/metabolismo , Parada Cardíaca/metabolismo , Parada Cardíaca/terapia , Autofagia , RNA Mensageiro/metabolismo , Glucose/metabolismo
13.
Transl Stroke Res ; 14(5): 723-739, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-35921049

RESUMO

Cardiac arrest (CA) is common and devastating, and neuroprotective therapies for brain injury after CA remain limited. Neuroinflammation has been a target for two promising but underdeveloped post-CA therapies: neural stem cell (NSC) engrafting and glibenclamide (GBC). It is critical to understand whether one therapy has superior efficacy over the other and to further understand their immunomodulatory mechanisms. In this study, we aimed to evaluate and compare the therapeutic effects of NSC and GBC therapies post-CA. In in vitro studies, BV2 cells underwent oxygen-glucose deprivation (OGD) for three hours and were then treated with GBC or co-cultured with human NSCs (hNSCs). Microglial polarization phenotype and TLR4/NLRP3 inflammatory pathway proteins were detected by immunofluorescence staining. Twenty-four Wistar rats were randomly assigned to three groups (control, GBC, and hNSCs, N = 8/group). After 8 min of asphyxial CA, GBC was injected intraperitoneally or hNSCs were administered intranasally in the treatment groups. Neurological-deficit scores (NDSs) were assessed at 24, 48, and 72 h after return of spontaneous circulation (ROSC). Immunofluorescence was used to track hNSCs and quantitatively evaluate microglial activation subtype and polarization. The expression of TLR4/NLRP3 pathway-related proteins was quantified via Western blot. The in vitro studies showed the highest proportion of activated BV2 cells with an increased expression of TLR4/NLRP3 signaling proteins were found in the OGD group compared to OGD + GBC and OGD + hNSCs groups. NDS showed significant improvement after CA in hNSC and GBC groups compared to controls, and hNSC treatment was superior to GBC treatment. The hNSC group had more inactive morphology and anti-inflammatory phenotype of microglia. The quantified expression of TLR4/NLRP3 pathway-related proteins was significantly suppressed by both treatments, and the suppression was more significant in the hNSC group compared to the GBC group. hNSC and GBC therapy regulate microglial activation and the neuroinflammatory response in the brain after CA through TLR4/NLRP3 signaling and exert multiple neuroprotective effects, including improved neurological function and shortened time of severe neurological deficit. In addition, hNSCs displayed superior inflammatory regulation over GBC.


Assuntos
Lesões Encefálicas , Parada Cardíaca , Células-Tronco Neurais , Ratos , Animais , Humanos , Neuroproteção , Glibureto/farmacologia , Glibureto/uso terapêutico , Glibureto/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Doenças Neuroinflamatórias , Ratos Wistar , Células-Tronco Neurais/transplante , Parada Cardíaca/complicações , Parada Cardíaca/tratamento farmacológico , Parada Cardíaca/metabolismo , Lesões Encefálicas/metabolismo , Microglia , Glucose/metabolismo , Oxigênio/metabolismo
14.
Physiol Rep ; 10(3): e15173, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35150207

RESUMO

Cardiorenal syndrome type 1 (CRS-1) acute kidney injury (AKI) is a critical complication of acute cardiovascular disease but is poorly understood. AKI induces acute albuminuria. As chronic albuminuria is associated with worsening kidney disease and albumin has been implicated in tubular epithelial injury, we investigated whether albumin participates in CRS-1, and whether CRS-1 alters renal albumin handling. We report the role of albumin in in vivo and in vitro CRS-1 models. An established translational model, cardiac arrest and cardiopulmonary resuscitation (CA/CPR) induced severe acute albuminuria which correlated with tubular epithelial cell death. In vivo microscopy demonstrated CA/CPR-induced glomerular filtration of exogenous albumin, while administration of exogenous albumin after CA/CPR worsened AKI compared to iso-oncotic control. Increased albumin signal was observed in the proximal tubules of CA/CPR mice compared to sham. Comparison of albumin flux from tubular lumen to epithelial cells revealed saturated albumin transport within minutes of albumin injection after CA/CPR. In vitro, HK2 cells (human kidney tubular epithelial cells), exposed to oxygen-glucose deprivation were injured by albumin in a dose dependent fashion. This interference was unchanged by the tubular endocytic receptor megalin. In conclusion, CRS-1 alters albumin filtration and tubular uptake, leading to increased tubular exposure to albumin, which is injurious to tubular epithelial cells, worsening AKI. Our findings shed light on the pathophysiology of renal albumin and may guide interventions such as albumin resuscitation to improve CRS-1 outcomes. This investigation may have important translational relevance for patients that receive exogenous albumin as part of their CRS-1 treatment regimen.


Assuntos
Albuminas/metabolismo , Síndrome Cardiorrenal/metabolismo , Parada Cardíaca/metabolismo , Animais , Reanimação Cardiopulmonar/efeitos adversos , Linhagem Celular , Parada Cardíaca/etiologia , Humanos , Túbulos Renais/metabolismo , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
15.
Ann N Y Acad Sci ; 1507(1): 23-36, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33040363

RESUMO

Cardiac arrest (CA) is a sudden and devastating disease process resulting in more deaths in the United States than many cancers, metabolic diseases, and even car accidents. Despite such a heavy mortality burden, effective treatments have remained elusive. The past century has been productive in establishing the guidelines for resuscitation, known as cardiopulmonary resuscitation (CPR), as well as developing a scientific field whose aim is to elucidate the underlying mechanisms of CA and develop therapies to save lives. CPR has been successful in reinitiating the heart after arrest, enabling a survival rate of approximately 10% in out-of-hospital CA. Although current advanced resuscitation methods, including hypothermia and extracorporeal membrane oxygenation, have improved survival in some patients, they are unlikely to significantly improve the national survival rate any further without a paradigm shift. Such a change is possible with sustained efforts in the basic and clinical sciences of resuscitation and their implementation. This review seeks to discuss the current landscape in resuscitation medicine-how we got here and where we are going.


Assuntos
Reanimação Cardiopulmonar/tendências , Oxigenação por Membrana Extracorpórea/tendências , Parada Cardíaca/terapia , Animais , Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Reanimação Cardiopulmonar/métodos , Oxigenação por Membrana Extracorpórea/métodos , Parada Cardíaca/metabolismo , Parada Cardíaca/fisiopatologia , Humanos , Taxa de Sobrevida/tendências , Fatores de Tempo
16.
Ann N Y Acad Sci ; 1507(1): 60-69, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34554586

RESUMO

While much has been observed regarding hypothermia by way of environmental exposure, it is modern day medicine that deployed hypothermia as a therapeutic. From the early 1930s, when Temple Fay deployed "refrigeration" to treat pain, to the work of Wilfred Bigelow and Charles Drew, who utilized hypothermia in open heart surgery-the opportunities seemed endless. However, questions arose surrounding appropriate temperatures to achieve best outcomes and how to minimize adverse events, such as coagulopathy and infection. In the 1980s, hypothermia underwent a resurgence through Peter Safar's critical studies in large animals, which quickly translated into feasibility studies and the landmark trials of 2002 that paved the way for postcardiac arrest care as we currently know it. Through clinical and observational trials, modern-day targeted temperature management continues to adapt, striving to improve patient outcomes. While hypothermia has come a long way from the writings of Hippocrates, the ideal therapy has not yet been defined, and more work is needed. While the history is long, there is more to be written and advances to be achieved as we optimize the neuroprotective effects of hypothermia for comatose survivors of cardiac arrest.


Assuntos
Parada Cardíaca/terapia , Hipotermia Induzida/métodos , Hipotermia Induzida/tendências , Doenças do Sistema Nervoso/prevenção & controle , Neuroproteção/fisiologia , Animais , Ensaios Clínicos como Assunto/métodos , Parada Cardíaca/complicações , Parada Cardíaca/metabolismo , Humanos , Hipotermia Induzida/normas , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/metabolismo , Guias de Prática Clínica como Assunto/normas
17.
Crit Care Med ; 50(2): e199-e208, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34259447

RESUMO

OBJECTIVES: Cardiac arrest and subsequent resuscitation have been shown to deplete plasma phospholipids. This depletion of phospholipids in circulating plasma may contribute to organ damage postresuscitation. Our aim was to identify the diminishment of essential phospholipids in postresuscitation plasma and develop a novel therapeutic approach of supplementing these depleted phospholipids that are required to prevent organ dysfunction postcardiac arrest, which may lead to improved survival. DESIGN: Clinical case control study followed by translational laboratory study. SETTING: Research institution. PATIENTS/SUBJECTS: Adult cardiac arrest patients and male Sprague-Dawley rats. INTERVENTIONS: Resuscitated rats after 10-minute asphyxial cardiac arrest were randomized to be treated with lysophosphatidylcholine specie or vehicle. MEASUREMENTS AND MAIN RESULTS: We first performed a phospholipid survey on human cardiac arrest and control plasma. Using mass spectrometry analysis followed by multivariable regression analyses, we found that plasma lysophosphatidylcholine levels were an independent discriminator of cardiac arrest. We also found that decreased plasma lysophosphatidylcholine was associated with poor patient outcomes. A similar association was observed in our rat model, with significantly greater depletion of plasma lysophosphatidylcholine with increased cardiac arrest time, suggesting an association of lysophosphatidylcholine levels with injury severity. Using a 10-minute cardiac arrest rat model, we tested supplementation of depleted lysophosphatidylcholine species, lysophosphatidylcholine(18:1), and lysophosphatidylcholine(22:6), which resulted in significantly increased survival compared with control. Furthermore, the survived rats treated with these lysophosphatidylcholine species exhibited significantly improved brain function. However, supplementing lysophosphatidylcholine(18:0), which did not decrease in the plasma after 10-minute cardiac arrest, had no beneficial effect. CONCLUSIONS: Our data suggest that decreased plasma lysophosphatidylcholine is a major contributor to mortality and brain damage postcardiac arrest, and its supplementation may be a novel therapeutic approach.


Assuntos
Parada Cardíaca/metabolismo , Lisofosfatidilcolinas/análise , Programas de Rastreamento/normas , Fosfolipídeos/análise , Idoso , Idoso de 80 Anos ou mais , Animais , Feminino , Parada Cardíaca/sangue , Parada Cardíaca/complicações , Humanos , Lisofosfatidilcolinas/sangue , Masculino , Programas de Rastreamento/métodos , Programas de Rastreamento/estatística & dados numéricos , Fosfolipídeos/sangue , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença
18.
Ann N Y Acad Sci ; 1507(1): 49-59, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34060087

RESUMO

Cardiac arrest has many implications for morbidity and mortality. Few interventions have been shown to improve return of spontaneous circulation (ROSC) and long-term outcomes after cardiac arrest. Ischemic-reperfusion injury upon achieving ROSC creates an imbalance between oxygen supply and demand. Multiple events occur in the postcardiac arrest period, including excitotoxicity, mitochondrial dysfunction, and oxidative stress and inflammation, all of which contribute to ongoing brain injury and cellular death. Given that complex pathophysiology underlies global brain hypoxic ischemia, neuroprotective strategies targeting multiple stages of the neuropathologic cascade should be considered as a means of mitigating secondary neuronal injury and improving neurologic outcomes and survival in cardiac arrest victims. In this review article, we discuss a number of different pharmacologic agents that may have a potential role in targeting these injurious pathways following cardiac arrest. Pharmacologic therapies most relevant for discussion currently include memantine, perampanel, magnesium, propofol, thiamine, methylene blue, vitamin C, vitamin E, coenzyme Q10 , minocycline, steroids, and aspirin.


Assuntos
Lesões Encefálicas/prevenção & controle , Isquemia Encefálica/prevenção & controle , Parada Cardíaca/tratamento farmacológico , Neuroproteção/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Animais , Antioxidantes/administração & dosagem , Lesões Encefálicas/etiologia , Lesões Encefálicas/metabolismo , Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Parada Cardíaca/complicações , Parada Cardíaca/metabolismo , Humanos , Memantina/administração & dosagem , Neuroproteção/fisiologia , Nitrilas/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Piridonas/administração & dosagem , Tiamina/administração & dosagem
19.
Neuromolecular Med ; 24(2): 97-112, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34019239

RESUMO

Cardiopulmonary arrest (CA) can greatly impact a patient's life, causing long-term disability and death. Although multi-faceted treatment strategies against CA have improved survival rates, the prognosis of CA remains poor. We previously reported asphyxial cardiac arrest (ACA) can cause excessive activation of the sympathetic nervous system (SNS) in the brain, which contributes to cerebral blood flow (CBF) derangements such as hypoperfusion and, consequently, neurological deficits. Here, we report excessive activation of the SNS can cause enhanced neuropeptide Y levels. In fact, mRNA and protein levels of neuropeptide Y (NPY, a 36-amino acid neuropeptide) in the hippocampus were elevated after ACA-induced SNS activation, resulting in a reduced blood supply to the brain. Post-treatment with peptide YY3-36 (PYY3-36), a pre-synaptic NPY2 receptor agonist, after ACA inhibited NPY release and restored brain circulation. Moreover, PYY3-36 decreased neuroinflammatory cytokines, alleviated mitochondrial dysfunction, and improved neuronal survival and neurological outcomes. Overall, NPY is detrimental during/after ACA, but attenuation of NPY release via PYY3-36 affords neuroprotection. The consequences of PYY3-36 inhibit ACA-induced 1) hypoperfusion, 2) neuroinflammation, 3) mitochondrial dysfunction, 4) neuronal cell death, and 5) neurological deficits. The present study provides novel insights to further our understanding of NPY's role in ischemic brain injury.


Assuntos
Lesões Encefálicas , Isquemia Encefálica , Parada Cardíaca , Animais , Lesões Encefálicas/etiologia , Isquemia Encefálica/complicações , Parada Cardíaca/complicações , Parada Cardíaca/metabolismo , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeo Y/agonistas , Receptores de Neuropeptídeo Y/genética
20.
J Neuroinflammation ; 18(1): 257, 2021 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-34740380

RESUMO

BACKGROUND: Activation of the absent in melanoma 2 (AIM2) inflammasome and impaired autophagosome clearance in neurons contribute significantly to cardiac arrest and return of spontaneous circulation (CA-ROSC) injury, while the mechanism by which the AIM2 inflammasome is regulated and relationship between the processes remain poorly understood. Recently, charged multivesicular body protein 2A (CHMP2A), a subunit of endosomal sorting complex required for transport (ESCRT), was shown to regulate phagophore closure, and its depletion led to the accumulation of autophagosomes and induced cell death. Here, we investigated whether CHMP2A-mediated autophagy was an underlying mechanism of AIM2-associated inflammation after CA-ROSC and explored the potential link between the AIM2 inflammasome and autophagy under ischemic conditions. METHODS: AIM2 inflammasome activation and autophagic flux in the cortex were assessed in the CA-ROSC rat model. We injected LV-Vector or LV-CHMP2A virus into the motor cortex with stereotaxic coordinates and divided the rats into four groups: Sham, CA, CA+LV-Vector, and CA+LV-CHMP2A. Neurologic deficit scores (NDSs), balance beam tests, histopathological injury of the brain, and expression of the AIM2 inflammasome and proinflammatory cytokines were analyzed. RESULTS: AIM2 inflammasome activation and increased interleukin 1 beta (IL-1ß) and IL-18 release were concurrent with reduced levels of CHMP2A-induced autophagy in CA-ROSC rat neurons. In addition, silencing CHMP2A resulted in autophagosome accumulation and decreased autophagic degradation of the AIM2 inflammasome. In parallel, a reduction in AIM2 contributed to autophagy activation and mitigated oxygen-glucose deprivation and reperfusion (OGD-Rep)-induced inflammation. Notably, CHMP2A overexpression in the cortex hindered neuroinflammation, protected against ischemic brain damage, and improved neurologic outcomes after CA. CONCLUSIONS: Our results support a potential link between autophagy and AIM2 signaling, and targeting CHMP2A may provide new insights into neuroinflammation in the early phase during CA-ROSC.


Assuntos
Isquemia Encefálica/metabolismo , Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Parada Cardíaca/complicações , Doenças Neuroinflamatórias/metabolismo , Animais , Autofagia/fisiologia , Isquemia Encefálica/etiologia , Parada Cardíaca/metabolismo , Doenças Neuroinflamatórias/etiologia , Ratos , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA