Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(16)2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39201277

RESUMO

The Chromodomain helicase DNA-binding protein 1-like (CHD1L) is a nucleosome remodeling enzyme, which plays a key role in chromatin relaxation during the DNA damage response. Genome editing has shown that deletion of CHD1L sensitizes cells to PARPi, but the effect of its pharmacological inhibition has not been defined. Triple-negative breast cancer SUM149PT, HCC1937, and MDA-MB-231 cells were used to assess the mechanism of action of the CHD1Li OTI-611. Cytotoxicity as a single agent or in combination with standard-of-care treatments was assessed in tumor organoids. Immunofluorescence was used to assess the translocation of PAR and AIF to the cytoplasm or the nucleus and to study markers of DNA damage or apoptosis. Trapping of PARP1/2 or CHD1L onto chromatin was also assessed by in situ subcellular fractionation and immunofluorescence and validated by Western blot. We show that the inhibition of CHD1L's ATPase activity by OTI-611 is cytotoxic to triple-negative breast cancer tumor organoids and synergizes with PARPi and chemotherapy independently of the BRCA mutation status. The inhibition of the remodeling function blocks the phosphorylation of H2AX, traps CHD1L on chromatin, and leaves PAR chains on PARP1/2 open for hydrolysis. PAR hydrolysis traps PARP1/2 at DNA damage sites and mediates PAR translocation to the cytoplasm, release of AIF from the mitochondria, and induction of PARthanatos. The targeted inhibition of CHD1L's oncogenic function by OTI-611 signifies an innovative therapeutic strategy for breast cancer and other cancers. This approach capitalizes on CHD1L-mediated DNA repair and cell survival vulnerabilities, thereby creating synergy with standard-of-care therapies.


Assuntos
Sobrevivência Celular , Dano ao DNA , DNA Helicases , Proteínas de Ligação a DNA , Parthanatos , Neoplasias de Mama Triplo Negativas , Humanos , Dano ao DNA/efeitos dos fármacos , Feminino , Linhagem Celular Tumoral , DNA Helicases/metabolismo , DNA Helicases/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Sobrevivência Celular/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Parthanatos/efeitos dos fármacos , Parthanatos/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/tratamento farmacológico , Apoptose/efeitos dos fármacos , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia
2.
Eur J Pharmacol ; 978: 176765, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-38906236

RESUMO

Subarachnoid hemorrhage (SAH) is a neurological condition with high mortality and poor prognosis, and there are currently no effective therapeutic drugs available. Poly (ADP-ribose) polymerase 1 (PARP-1) dependent cell death pathway-parthanatos is closely associated with stroke. We investigated improvements in neurological function, oxidative stress, blood-brain barrier and parthanatos-related protein expression in rats with SAH after intraperitoneal administration of PARP-1 inhibitor (AG14361). Our study found that the expression of parthanatos-related proteins was significantly increased after SAH. Immunofluorescence staining showed increased expression of apoptosis-inducing factor (AIF) in the nucleus after SAH. Administration of PARP-1 inhibitor significantly reduced malondialdehyde (MDA) level and the expression of parthanatos-related proteins. Immunofluorescence staining showed that PARP-1 inhibitor reduced the expression of 8-hydroxy-2' -deoxyguanosine (8-OHdG) and thus reduced oxidative stress. Moreover, PARP-1 inhibitor could inhibit inflammation-associated proteins level and neuronal apoptosis, protect the blood-brain barrier and significantly improve neurological function after SAH. These results suggest that PARP-1 inhibitor can significantly improve SAH, and the underlying mechanism may be through inhibiting parthanatos pathway.


Assuntos
Barreira Hematoencefálica , Lesões Encefálicas , Morte Celular , Parthanatos , Poli(ADP-Ribose) Polimerase-1 , Hemorragia Subaracnóidea , Animais , Masculino , Ratos , Fator de Indução de Apoptose/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Lesões Encefálicas/metabolismo , Lesões Encefálicas/etiologia , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/patologia , Morte Celular/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Parthanatos/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/tratamento farmacológico , Hemorragia Subaracnóidea/patologia
3.
Eur J Pharmacol ; 972: 176557, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38574839

RESUMO

Cerebral ischemia-reperfusion injury (CIRI) can induce massive death of ischemic penumbra neurons via oxygen burst, exacerbating brain damage. Parthanatos is a form of caspase-independent cell death involving excessive activation of PARP-1, closely associated with intense oxidative stress following CIRI. 4'-O-methylbavachalcone (MeBavaC), an isoprenylated chalcone component in Fructus Psoraleae, has potential neuroprotective effects. This study primarily investigates whether MeBavaC can act on SIRT3 to alleviate parthanatos of ischemic penumbra neurons induced by CIRI. MeBavaC was oral gavaged to the middle cerebral artery occlusion-reperfusion (MCAO/R) rats after occlusion. The effects of MeBavaC on cerebral injury were detected by the neurological deficit score and cerebral infarct volume. In vitro, PC-12 cells were subjected to oxygen and glucose deprivation/reoxygenation (OGD/R), and assessed cell viability and cell injury. Also, the levels of ROS, mitochondrial membrane potential (MMP), and intracellular Ca2+ levels were detected to reflect mitochondrial function. We conducted western blotting analyses of proteins involved in parthanatos and related signaling pathways. Finally, the exact mechanism between the neuroprotection of MeBavaC and parthanatos was explored. Our results indicate that MeBavaC reduces the cerebral infarct volume and neurological deficit scores in MCAO/R rats, and inhibits the decreased viability of PC-12 cells induced by OGD/R. MeBavaC also downregulates the expression of parthanatos-related death proteins PARP-1, PAR, and AIF. However, this inhibitory effect is weakened after the use of a SIRT3 inhibitor. In conclusion, the protective effect of MeBavaC against CIRI may be achieved by inhibiting parthanatos of ischemic penumbra neurons through the SIRT3-PARP-1 axis.


Assuntos
Chalconas , Fármacos Neuroprotetores , Parthanatos , Ratos Sprague-Dawley , Traumatismo por Reperfusão , Sirtuínas , Animais , Ratos , Masculino , Chalconas/farmacologia , Chalconas/uso terapêutico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/metabolismo , Parthanatos/efeitos dos fármacos , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/patologia , AVC Isquêmico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células PC12 , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neurônios/metabolismo , Cálcio/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/complicações , Sobrevivência Celular/efeitos dos fármacos , Sirtuína 3/metabolismo , Sirtuína 3/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo
4.
Br J Cancer ; 130(9): 1529-1541, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38461169

RESUMO

BACKGROUND: Several studies have described a potential anti-tumour effect of cannabinoids (CNB). CNB receptor 2 (CB2) is mostly present in hematopoietic stem cells (HSC). The present study evaluates the anti-leukaemic effect of CNB. METHODS: Cell lines and primary cells from acute myeloid leukaemia (AML) patients were used and the effect of the CNB derivative WIN-55 was evaluated in vitro, ex vivo and in vivo. RESULTS: We demonstrate a potent antileukemic effect of WIN-55 which is abolished with CB antagonists. WIN-treated mice, xenografted with AML cells, had better survival as compared to vehicle or cytarabine. DNA damage-related genes were affected upon exposure to WIN. Co-incubation with the PARP inhibitor Olaparib prevented WIN-induced cell death, suggesting PARP-mediated apoptosis which was further confirmed with the translocation of AIF to the nucleus observed in WIN-treated cells. Nicotinamide prevented WIN-related apoptosis, indicating NAD+ depletion. Finally, WIN altered glycolytic enzymes levels as well as the activity of G6PDH. These effects are reversed through PARP1 inhibition. CONCLUSIONS: WIN-55 exerts an antileukemic effect through Parthanatos, leading to translocation of AIF to the nucleus and depletion of NAD+, which are reversed through PARP1 inhibition. It also induces metabolic disruptions. These effects are not observed in normal HSC.


Assuntos
Leucemia Mieloide Aguda , Parthanatos , Humanos , Animais , Camundongos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Parthanatos/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Apoptose/efeitos dos fármacos , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Piperazinas/farmacologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Canabinoides/farmacologia , Ftalazinas/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Dano ao DNA/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Antineoplásicos/farmacologia
5.
Biochem Pharmacol ; 228: 116174, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38552851

RESUMO

Parthanatos is a cell death signaling pathway that has emerged as a compelling target for pharmaceutical intervention. It plays a pivotal role in the neuron loss and neuroinflammation that occurs in Parkinson's Disease (PD), Alzheimer's Disease (AD), Huntington's Disease (HD), Amyotrophic Lateral Sclerosis (ALS), and stroke. There are currently no treatments available to humans to prevent cell death in any of these diseases. This review provides an in-depth examination of the current understanding of the Parthanatos mechanism, with a particular focus on its implications in neuroinflammation and various diseases discussed herein. Furthermore, we thoroughly review potential intervention targets within the Parthanatos pathway. We dissect recent progress in inhibitory strategies, complimented by a detailed structural analysis of key Parthanatos executioners, PARP-1, AIF, and MIF, along with an assessment of their established inhibitors. We hope to introduce a new perspective on the feasibility of targeting components within the Parthanatos pathway, emphasizing its potential to bring about transformative outcomes in therapeutic interventions. By delineating therapeutic opportunities and known targets, we seek to emphasize the imperative of blocking Parthanatos as a precursor to developing disease-modifying treatments. This comprehensive exploration aims to catalyze a paradigm shift in our understanding of potential neurodegenerative disease therapeutics, advocating for the pursuit of effective interventions centered around Parthanatos inhibition.


Assuntos
Doenças Neurodegenerativas , Parthanatos , Acidente Vascular Cerebral , Humanos , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Animais , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/terapia , Parthanatos/efeitos dos fármacos , Parthanatos/fisiologia , Fármacos Neuroprotetores/uso terapêutico
6.
Mov Disord ; 39(4): 644-650, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38396375

RESUMO

Parkinson's disease (PD) is a progressive neurodegenerative condition that pathognomonically involves the death of dopaminergic neurons in the substantia nigra pars compacta, resulting in a myriad of motor and non-motor symptoms. Given the insurmountable burden of this disease on the population and healthcare system, significant efforts have been put forth toward generating disease modifying therapies. This class of treatments characteristically alters disease course, as opposed to current strategies that focus on managing symptoms. Previous literature has implicated the cell death pathway known as parthanatos in PD progression. Inhibition of this pathway by targeting poly (ADP)-ribose polymerase 1 (PARP1) prevents neurodegeneration in a model of idiopathic PD. However, PARP1 has a vast repertoire of functions within the body, increasing the probability of side effects with the long-term treatment likely necessary for clinically significant neuroprotection. Recent work culminated in the development of a novel agent targeting the macrophage migration inhibitory factor (MIF) nuclease domain, also named parthanatos-associated apoptosis-inducing factor nuclease (PAAN). This nuclease activity specifically executes the terminal step in parthanatos. Parthanatos-associated apoptosis-inducing factor nuclease inhibitor-1 was neuroprotective in multiple preclinical mouse models of PD. This piece will focus on contextualizing this discovery, emphasizing its significance, and discussing its potential implications for parthanatos-directed treatment. © 2024 International Parkinson and Movement Disorder Society.


Assuntos
Neurônios Dopaminérgicos , Fatores Inibidores da Migração de Macrófagos , Doença de Parkinson , Humanos , Neurônios Dopaminérgicos/metabolismo , Doença de Parkinson/metabolismo , Animais , Fatores Inibidores da Migração de Macrófagos/metabolismo , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/metabolismo , Parthanatos/efeitos dos fármacos
7.
Mol Cancer Ther ; 22(3): 306-316, 2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-36622760

RESUMO

Fusion of the E-26 transformation-specific (ETS)-related gene (ERG) with transmembrane serine protease 2 (TMPRSS2) is a crucial step in the occurrence and progression of approximately 50% of prostate cancers. Despite significant progress in drug discovery, ERG inhibitors have yet to be approved for the clinical treatment of prostate cancer. In this study, we used computer-aided drug design (CADD)-based virtual screening to screen for potential inhibitors of ERG. In vivo and in vitro methods revealed that nifuroxazide (NFZ) inhibited the proliferation of a TMPRSS2:ERG fusion-positive prostate cancer cell line (VCaP) with an IC50 lower than that of ERG-negative prostate cancer cell lines (LNCaP, DU145, and WPMY cells). Poly [ADP-ribose] polymerase 1, the critical mediator of parthanatos, is known to bind ERG and is required for ERG-mediated transcription. NFZ blocked this interaction and overly activated PARP1, leading to cell death that was reduced by olaparib, a PARP1 inhibitor. These results show that NFZ inhibits ERG, leading to parthanatic cell death.


Assuntos
Nitrofuranos , Proteínas de Fusão Oncogênica , Parthanatos , Neoplasias da Próstata , Humanos , Masculino , Linhagem Celular Tumoral , Proteínas de Fusão Oncogênica/genética , Parthanatos/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Transativadores/genética , Regulador Transcricional ERG/genética , Regulador Transcricional ERG/metabolismo , Nitrofuranos/farmacologia , Nitrofuranos/uso terapêutico
8.
PLoS Biol ; 19(11): e3001455, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34748530

RESUMO

Several studies have revealed a correlation between chronic inflammation and nicotinamide adenine dinucleotide (NAD+) metabolism, but the precise mechanism involved is unknown. Here, we report that the genetic and pharmacological inhibition of nicotinamide phosphoribosyltransferase (Nampt), the rate-limiting enzyme in the salvage pathway of NAD+ biosynthesis, reduced oxidative stress, inflammation, and keratinocyte DNA damage, hyperproliferation, and cell death in zebrafish models of chronic skin inflammation, while all these effects were reversed by NAD+ supplementation. Similarly, genetic and pharmacological inhibition of poly(ADP-ribose) (PAR) polymerase 1 (Parp1), overexpression of PAR glycohydrolase, inhibition of apoptosis-inducing factor 1, inhibition of NADPH oxidases, and reactive oxygen species (ROS) scavenging all phenocopied the effects of Nampt inhibition. Pharmacological inhibition of NADPH oxidases/NAMPT/PARP/AIFM1 axis decreased the expression of pathology-associated genes in human organotypic 3D skin models of psoriasis. Consistently, an aberrant induction of NAMPT and PARP activity, together with AIFM1 nuclear translocation, was observed in lesional skin from psoriasis patients. In conclusion, hyperactivation of PARP1 in response to ROS-induced DNA damage, fueled by NAMPT-derived NAD+, mediates skin inflammation through parthanatos cell death.


Assuntos
Inflamação/patologia , NAD/metabolismo , Nicotinamida Fosforribosiltransferase/metabolismo , Parthanatos , Poli(ADP-Ribose) Polimerases/metabolismo , Pele/patologia , Animais , Fator de Indução de Apoptose/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/genética , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/patologia , Larva/metabolismo , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/metabolismo , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Parthanatos/efeitos dos fármacos , Parthanatos/genética , Poli Adenosina Difosfato Ribose/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Proteínas Secretadas Inibidoras de Proteinases/deficiência , Proteínas Secretadas Inibidoras de Proteinases/metabolismo , Psoríase/genética , Psoríase/patologia , Espécies Reativas de Oxigênio/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/metabolismo
9.
Int J Mol Sci ; 22(18)2021 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-34575823

RESUMO

To investigate the effect of 14,15-EET on the parthanatos in neurons induced by cerebral ischemia and reperfusion, middle cerebral artery occlusion and reperfusion (MCAO/R) and oxygen glucose deprivation/reoxygenation (OGD/R) were used to simulate cerebral ischemia reperfusion in vivo and in vitro, respectively. TTC staining and the Tunel method were used to detect cerebral infarct volume and neuronal apoptosis. Western blot and immunofluorescence were used to detect poly (ADP-ribose) polymerase-1 (PARP-1) activation and AIF nuclear translocation. The production of reactive oxygen species (ROS) and the expression of antioxidant genes were detected by Mito SOX, DCFH-DA and qPCR methods. MCAO/R increased cerebral infarct volume and neuronal apoptosis in mice, while 14,15-EET pretreatment increased cerebral infarct volume and neuronal apoptosis. OGD/R induced reactive oxygen species generation, PARP-1 cleavage, and AIF nuclear translocation in cortical neurons. 14,15-EET pretreatment could enhance the antioxidant gene expression of glutathione peroxidase (GSH-Px), heme oxygenase-1 (HO-1) and superoxide dismutase (SOD) in cortical neurons after ischemia and reperfusion. 14,15-EET inhibits the neuronal parthanatos induced by MCAO/R through upregulation of the expression of antioxidant genes and by reducing the generation of reactive oxygen species. This study advances the EET neuroprotection theory and provides a scientific basis for targeted clinical drugs that reduce neuronal parthanatos following cerebral ischemia and reperfusion.


Assuntos
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Lesões Encefálicas/etiologia , Lesões Encefálicas/metabolismo , Isquemia Encefálica/complicações , Neurônios/efeitos dos fármacos , Parthanatos/efeitos dos fármacos , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Ácido 8,11,14-Eicosatrienoico/farmacologia , Animais , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/patologia , Isquemia Encefálica/etiologia , Modelos Animais de Doenças , Glucose/metabolismo , Masculino , Camundongos , Modelos Biológicos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/patologia
10.
Food Chem Toxicol ; 154: 112326, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34111490

RESUMO

Fumonisin B1 (FB1) is a well-known mycotoxin produced by Fusarium spp. and has a wide range of dose-dependent toxic effects, including nephrotoxicity, hepatotoxicity, and neurotoxicity. This research illustrated that FB1 exerts its toxicity in the neuroblastoma cell line through a distinct cell-death pathway called parthanatos. FB1 can cause excessive DNA strand breaks, leading to poly (ADP-ribose) polymerase-1 (PARP-1) overactivation and cell death. In this study, we used 50 µM FB1-treated SH-SY5Y neuroblastoma cells to elucidate the signaling pathway of FB1-induced parthanatos. We observed that FB1-induced cell death is caspase-independent and accompanied by rapid activation of PARP-1, c-Jun N-terminal kinase activation, reactive oxygen species (ROS) generation, and intracellular calcium increase. FB1 treatment also increased endoplasmic reticulum stress due to the rapid increase of calcium ions and ROS levels. In addition, FB1 induced massive DNA damage and chromatin decondensation. We also observed that apoptosis-inducing factor nuclear translocation and PAR accumulation were associated with the necroptosis signal.


Assuntos
Fumonisinas/farmacologia , Neuroblastoma/metabolismo , Parthanatos/efeitos dos fármacos , Cálcio/metabolismo , Linhagem Celular Tumoral , Cromatina/metabolismo , DNA/metabolismo , Dano ao DNA/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Mitocôndrias/efeitos dos fármacos , Necroptose/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Espécies Reativas de Oxigênio/metabolismo
12.
Aging (Albany NY) ; 13(3): 4242-4257, 2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33495407

RESUMO

Oral squamous cell carcinoma (OSCC) is one of the most common malignant tumors worldwide, and its prognosis is still not optimistic. Oxaliplatin is a type of platinum chemotherapeutic agent, but its treatment effects on OSCC and molecular mechanisms have not been fully elucidated. Parthanatos, a unique form of cell death, plays an important role in a variety of physiological and pathological processes. This study aims to investigate whether oxaliplatin inhibits OSCC by inducing parthanatos. Our results showed that oxaliplatin inhibited the proliferation and migration of OSCC cells in vitro, and also inhibited the tumorigenesis in vivo. Further experiments proved that oxaliplatin induced parthanatos in OSCC cells, characterized by depolarization of the mitochondrial membrane potential, up-regulation of PARP1, AIF and MIF in the nucleus, as well as the nuclear translocation of AIF. Meanwhile, PARP1 inhibitor rucaparib and siRNA against PARP1 attenuated oxaliplatin-induced parthanatos in OSCC cells. In addition, we found that oxaliplatin caused oxidative stress in OSCC cells, and antioxidant NAC not only relieved oxaliplatin-induced overproduction of reactive oxygen species (ROS) but also reversed parthanatos caused by oxaliplatin. In conclusion, our results indicate that oxaliplatin inhibits OSCC by activating PARP1-mediated parthanatos through increasing the production of ROS.


Assuntos
Antineoplásicos/farmacologia , Mitocôndrias/efeitos dos fármacos , Neoplasias Bucais/metabolismo , Oxaliplatina/farmacologia , Parthanatos/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Acetilcisteína/farmacologia , Animais , Antioxidantes/farmacologia , Linhagem Celular Tumoral , Glutationa/efeitos dos fármacos , Glutationa/metabolismo , Humanos , Camundongos , Mitocôndrias/metabolismo , Neoplasias Bucais/patologia , Transplante de Neoplasias , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Superóxido Dismutase/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Naunyn Schmiedebergs Arch Pharmacol ; 394(4): 783-796, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32989562

RESUMO

This study aims to explore the regulatory mechanisms of dexmedetomidine in parthanatos. MTT assay was applied to reveal cell viability; JC-1 staining assay was utilized to reveal mitochondrial membrane potential. Reactive oxygen species (ROS) probe, DCFH-DA, was used to detect intracellular ROS production. Luciferase activity assay was applied to measure the binding between miR-7-5p and PARP1. We first identified that bupivacaine inhibited the viability and induced the parthanatos of human neuroblastoma SH-SY5Y cells. In addition, dexmedetomidine, a potent α2-adrenoceptor agonist, reversed the regulatory effect of bupivacaine on parthanatos of SH-SY5Y. More importantly, dexmedetomidine counteracted bupivacaine-induced changes of mitochondrial membrane potential and ROS production in SH-SY5Y cells. Hyper-activation of PARP1 plays a vital role in parthanatos. Further exploration of our study identified that bupivacaine triggered overexpression of PARP1 in SH-SY5Y cells. Bioinformatics analysis revealed that miR-7-5p targeted the 3' untranslated region (3' UTR) of PARP1 to inhibit PARP1 expression. In addition, dexmedetomidine recovered the suppressive effects of bupivacaine on miR-7-5p expression. Dexmedetomidine suppressed bupivacaine-induced parthanatos in SH-SY5Y cells via the miR-7-5p/PARP1 axis, which may shed a new insight into parthanatos-dependent neuronal injury.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Anestésicos Locais , Bupivacaína , Dexmedetomidina/farmacologia , MicroRNAs/genética , Parthanatos/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/genética , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Oxirredutases/genética , Oxirredutases/metabolismo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Espécies Reativas de Oxigênio/metabolismo
14.
Curr Eye Res ; 46(1): 115-121, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32478624

RESUMO

PURPOSE: Our previous study discoveredreactive oxygen species (ROS) and apoptosis inducing factor (AIF) increased after retinal detachment. Parthanatos is a cell death form involving ROS and AIF, which is induced by poly (ADP-ribose) polymerase-1 (PARP-1). Therefore, we investigated whether PJ34 (a PARP-1 inhibitor) could inhibit parthanatos and protect the photoreceptors from cell death after retinal detachment (RD). METHODS: Experimental retinal detachment modelswere created in Sprague-Dawley rats by subretinal injection of sodium hyaluronate.PJ34 orDMSO were introduced into subretinal space at RD induction, respectively. The structure of retinas and the morphology of photoreceptors were observed by hematoxylin eosin (H&E) staining and transmission electron microscope (TEM). Parthanatos related proteins (PARP-1, PAR,AIF) were detected by Western blot. The vision-dependent behavior of rat was tested by Morris water maze. RESULTS: H&E staining and TEM results indicated that the structure and outer nuclear layer (ONL) thickness of retinas were preserved, and the photoreceptors death decreasedwith PJ34 treatment. Western blot showed that the expression of PARP-1, PAR and AIF were decreased withPJ34 treatment. In addition, administration of PJ34 also improved the vision-dependent behavior of rat. CONCLUSIONS: These findings suggested that PJ34 is a potential therapeutic agent that attenuated photoreceptor parthanatos death in retinal detachment through inhibition of PARP-1/AIF pathway.


Assuntos
Modelos Animais de Doenças , Parthanatos/efeitos dos fármacos , Fenantrenos/farmacologia , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Descolamento Retiniano/prevenção & controle , Animais , Fator de Indução de Apoptose/metabolismo , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/fisiologia , Microscopia Eletrônica de Transmissão , Células Fotorreceptoras de Vertebrados/ultraestrutura , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Descolamento Retiniano/fisiopatologia
15.
Apoptosis ; 25(7-8): 548-557, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32495124

RESUMO

Shikonin induced necroptosis in Jurkat cells were identified flow cytometrically by the up-regulation of RIP3 in live cells and that a proportion of these cells underwent other forms of regulated cell death (RCD) which included parthanatos (< 10%), or cleaved PARP (< 10%) and DNA Damage (> 30%). Live necroptotic cells also possessed functioning mitochondria with hyper-polarized mitochondria membrane potential and generated a fivefold increase in cellular reactive oxygen species (ROS) which was resistant to inhibition by zVAD and necrostatin-1 (Nec-1). After loss of plasma membrane integrity these dead necroptotic cells then showed a higher incidence of parthanatos (> 40%), or cleaved PARP (> 15%) but less DNA Damage (< 15%). Inhibition of shikonin induced apoptosis and necroptosis by zVAD and Nec-1 respectively resulted in live necroptotic cells with an increased incidence of cleaved PARP and reduced levels of DNA Damage respectively. Dead necroptotic cells then showed a reduced incidence of parthanatos and DNA Damage after inhibition by zVAD and Nec-1 respectively. A high proportion of these dead necroptotic cells (30%) which lacked plasma membrane integrity also displayed functioning hyper-polarized mitochondria with high levels of cellular ROS and thus had the capacity to influence the outcome of RCD processes rather than just been the end product of cell death, the necrotic cell. Flow cytometry can thus measure multiple forms of RCD and the level of cellular ROS and MMP which highlights the inter-connection between cell death processes and that a single cell may simultaneously display multiple forms of RCD.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Naftoquinonas/farmacologia , Necroptose/efeitos dos fármacos , Parthanatos/efeitos dos fármacos , Apoptose/genética , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Histonas/genética , Histonas/metabolismo , Humanos , Imidazóis/farmacologia , Indóis/farmacologia , Células Jurkat , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Necroptose/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Oligopeptídeos/farmacologia , Parthanatos/genética , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
16.
Nat Chem Biol ; 16(7): 791-800, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32251407

RESUMO

Cancer treatment generally involves drugs used in combinations. Most previous work has focused on identifying and understanding synergistic drug-drug interactions; however, understanding antagonistic interactions remains an important and understudied issue. To enrich for antagonism and reveal common features of these combinations, we screened all pairwise combinations of drugs characterized as activators of regulated cell death. This network is strongly enriched for antagonism, particularly a form of antagonism that we call 'single-agent dominance'. Single-agent dominance refers to antagonisms in which a two-drug combination phenocopies one of the two agents. Dominance results from differences in cell death onset time, with dominant drugs acting earlier than their suppressed counterparts. We explored mechanisms by which parthanatotic agents dominate apoptotic agents, finding that dominance in this scenario is caused by mutually exclusive and conflicting use of Poly(ADP-ribose) polymerase 1 (PARP1). Taken together, our study reveals death kinetics as a predictive feature of antagonism, due to inhibitory crosstalk between cell death pathways.


Assuntos
Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Parthanatos/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/genética , Apoptose/genética , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Antagonismo de Drogas , Sinergismo Farmacológico , Humanos , Cinética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Parthanatos/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo
17.
Reprod Sci ; 27(1): 75-86, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-32046374

RESUMO

Diminished ovarian reserve (DOR) is characterized by the depletion of the ovarian pool, which leads to reductions in oocyte quality and quantity. Studies have suggested that ovarian reserve or ovarian aging is tightly related to apoptosis. However, the cell death mechanism is not comprehensively understood. Parthanatos, a type of poly [ADP-ribose] polymerase 1(PARP1)-dependent and apoptosis-inducing factor (AIF)-mediated cell death, plays a crucial role in various disorders. In the present study, we aimed to investigate whether parthanatos is involved in the pathogenesis of DOR. We recruited 40 patients (20 DOR patients and 20 normal ovarian reserve (NOR) patients) and examined PAR expression and AIF translocation in their isolated cumulus GCs (granulosa cells) by fluorescence microscopy. Our results demonstrated that PAR expression and AIF nuclear translocation were significantly higher in cumulus GCs of DOR patients, suggesting that PARP1-dependent cell death may be associated with DOR pathophysiology. Moreover, we tested the protective function of melatonin on hydrogen peroxide (H2O2)-induced parthanatos in human ovarian cancer (IGROV1) cells. Our results demonstrated that H2O2 treatment of IGROV1 cells led to excessive protein PARylation and AIF translocation into the nuclei. Melatonin effectively inhibits PARylation, blocks translocation of AIF into the nucleus, and consequently decreases the risk of parthanatos in cumulus GCs.


Assuntos
Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Adenosina Difosfato Ribose/metabolismo , Fator de Indução de Apoptose/metabolismo , Melatonina/farmacologia , Reserva Ovariana/fisiologia , Parthanatos/fisiologia , Substâncias Protetoras/farmacologia , Adulto , Linhagem Celular Tumoral , Feminino , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Humanos , Parthanatos/efeitos dos fármacos
18.
JCI Insight ; 4(4)2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30830864

RESUMO

Poly(ADP-ribosyl)ation refers to the covalent attachment of ADP-ribose to protein, generating branched, long chains of ADP-ribose moieties, known as poly(ADP-ribose) (PAR). Poly(ADP-ribose) polymerase 1 (PARP1) is the main polymerase and acceptor of PAR in response to DNA damage. Excessive intracellular PAR accumulation due to PARP1 activation leads cell death in a pathway known as parthanatos. PAR degradation is mainly controlled by poly(ADP-ribose) glycohydrolase (PARG) and ADP-ribose-acceptor hydrolase 3 (ARH3). Our previous results demonstrated that ARH3 confers protection against hydrogen peroxide (H2O2) exposure, by lowering cytosolic and nuclear PAR levels and preventing apoptosis-inducing factor (AIF) nuclear translocation. We identified a family with an ARH3 gene mutation that resulted in a truncated, inactive protein. The 8-year-old proband exhibited a progressive neurodegeneration phenotype. In addition, parthanatos was observed in neurons of the patient's deceased sibling, and an older sibling exhibited a mild behavioral phenotype. Consistent with the previous findings, the patient's fibroblasts and ARH3-deficient mice were more sensitive, respectively, to H2O2 stress and cerebral ischemia/reperfusion-induced PAR accumulation and cell death. Further, PARP1 inhibition alleviated cell death and injury resulting from oxidative stress and ischemia/reperfusion. PARP1 inhibitors may attenuate the progression of neurodegeneration in affected patients with ARH3 deficiency.


Assuntos
Glicosídeo Hidrolases/genética , Doenças Neurodegenerativas/genética , Parthanatos/genética , Poli Adenosina Difosfato Ribose/metabolismo , Adulto , Animais , Fator de Indução de Apoptose/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Encéfalo/patologia , Células Cultivadas , Criança , Pré-Escolar , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/ética , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Fibroblastos , Glicosídeo Hidrolases/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Masculino , Camundongos , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Parthanatos/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Cultura Primária de Células , Traumatismo por Reperfusão/complicações , Pele/citologia
19.
Cell Microbiol ; 21(6): e13015, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30702185

RESUMO

Epithelial cell shedding is a defence mechanism against infectious microbes that use these cells as an infection foothold and that eliminate microbes from infection foci by removing infected cells. Mycoplasma pneumoniae, a causative agent of respiratory infections, is known to adhere to and colonise the surface of ciliated airway epithelial cells; it produces a large amount of hydrogen peroxide, indicating its capability of regulating hydrogen peroxide-induced infected cell detachment. In this study, we found that M. pneumoniae reduces exogenous hydrogen peroxide-induced detachment of the infected cells from culture plates. This cell detachment occurred dependently of DNA damage-initiated, poly (ADP-ribose) polymerase 1 (PARP1)-mediated cell death, or parthanatos. In cells infected with M. pneumoniae, exogenous hydrogen peroxide failed to induce DNA damage-initiated poly (ADP-ribose) (PAR) synthesis and concomitant increased cytoplasmic membrane rupture, both of which are biochemical hallmarks of parthanatos. The impairment of PAR synthesis was attributed to a reduction in the amount of cytosolic nicotinamide adenine dinucleotide (NAD), a substrate of PARP1, caused by M. pneumoniae. On the other hand, nonadherent mutant strains of M. pneumoniae showed a lower ability to reduce cell detachment than wild-type strains, but the extent to which NAD was decreased in infected cells was comparable to that seen in the wild-type strain. We found that NAD depletion could induce PARP1-independent cell detachment pathways following stimulation with hydrogen peroxide and that M. pneumoniae could also regulate PARP1-independent cell detachment in a cytoadhesion-dependent manner. These results suggest that M. pneumoniae might regulate infected cell detachment induced by hydrogen peroxide that it produces itself, and such a mechanism may contribute to sustaining the bacterial infection.


Assuntos
Células Epiteliais/microbiologia , Peróxido de Hidrogênio/toxicidade , Mycoplasma pneumoniae/metabolismo , Parthanatos/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Mycoplasma pneumoniae/genética , Mycoplasma pneumoniae/patogenicidade , NAD/metabolismo , Poli(ADP-Ribose) Polimerase-1/genética , Espécies Reativas de Oxigênio/metabolismo
20.
Free Radic Biol Med ; 131: 184-196, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30502458

RESUMO

In inflamed tissues or during ischemia-reperfusion episodes, activated macrophages produce large amounts of reactive species and are, thus, exposed to the damaging effects of reactive species. Here, our goal was to investigate the mechanism whereby activated macrophages protect themselves from oxidant stress-induced cell death. Hydrogen peroxide-treated mouse bone marrow-derived macrophages (BMDM) and THP-1 human monocyte-derived cells were chosen as models. We found a gradual development of resistance: first in monocyte-to-macrophage differentiation, and subsequently after lipopolysaccharide (LPS) exposure. Investigating the mechanism of the latter, we found that exposure to intense hydrogen peroxide stress causes poly(ADP-ribose) polymerase-1 (PARP-1) dependent programmed necrotic cell death, also known as parthanatos, as indicated by the protected status of PARP-1 knockout BMDMs and the protective effect of the PARP inhibitor PJ-34. In hydrogen peroxide-treated macrophages, however, apoptosis inducing factor (AIF) proved dispensable for parthanatos; nuclear translocation of AIF was not observed. A key event in LPS-mediated protection against the hydrogen peroxide-induced AIF independent parthanatos was downregulation of PARP1 mRNA and protein. The importance of this event was confirmed by overexpression of PARP1 in THP1 cells using a viral promoter, which lead to stable PARP1 levels even after LPS treatment and unresponsiveness to LPS-induced cytoprotection. In BMDMs, LPS-induced PARP1 suppression lead to prevention of NAD+ depletion. Moreover, LPS also induced expression of antioxidant proteins (superoxide dismutase-2, thioredoxin reductase 1 and peroxiredoxin) and triggered a metabolic shift to aerobic glycolysis, also known as the Warburg effect. In summary, we provide evidence that in macrophages intense hydrogen peroxide stress causes AIF-independent parthanatos from which LPS provides protection. The mechanism of LPS-mediated cytoprotection involves downregulation of PARP1, spared NAD+ and ATP pools, upregulation of antioxidant proteins, and a metabolic shift from mitochondrial respiration to aerobic glycolysis.


Assuntos
Fator de Indução de Apoptose/genética , Peróxido de Hidrogênio/farmacologia , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/genética , Superóxido Dismutase/genética , Animais , Fator de Indução de Apoptose/metabolismo , Regulação da Expressão Gênica , Glicólise/efeitos dos fármacos , Glicólise/genética , Humanos , Peróxido de Hidrogênio/antagonistas & inibidores , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , NAD/metabolismo , Fosforilação Oxidativa/efeitos dos fármacos , Parthanatos/efeitos dos fármacos , Parthanatos/genética , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Fenantrenos/farmacologia , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/metabolismo , Cultura Primária de Células , Regiões Promotoras Genéticas , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Superóxido Dismutase/metabolismo , Células THP-1 , Tiorredoxina Redutase 1/genética , Tiorredoxina Redutase 1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA