Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.385
Filter
1.
Commun Biol ; 7(1): 967, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122870

ABSTRACT

The mitochondrial permeability transition pore (mPTP) is a supramolecular channel that regulates exchange of solutes across cristae membranes, with executive roles in mitochondrial function and cell death. The contribution of the mPTP to normal physiology remains debated, although evidence implicates the mPTP in mitochondrial inner membrane remodeling in differentiating progenitor cells. Here, we demonstrate that strict control over mPTP conductance shapes metabolic machinery as cells transit toward hematopoietic identity. Cells undergoing the endothelial-to-hematopoietic transition (EHT) tightly control chief regulatory elements of the mPTP. During EHT, maturing arterial endothelium restricts mPTP activity just prior to hematopoietic commitment. After transition in cellular identity, mPTP conductance is restored. In utero treatment with NIM811, a molecule that blocks sensitization of the mPTP to opening by Cyclophilin D (CypD), amplifies oxidative phosphorylation (OXPHOS) in hematopoietic precursors and increases hematopoiesis in the embryo. Additionally, differentiating pluripotent stem cells (PSCs) acquire greater organization of mitochondrial cristae and hematopoietic activity following knockdown of the CypD gene, Ppif. Conversely, knockdown of Opa1, a GTPase critical for proper cristae architecture, induces cristae irregularity and impairs hematopoiesis. These data elucidate a mechanism that regulates mitochondrial maturation in hematopoietic precursors and underscore a role for the mPTP in the acquisition of hematopoietic fate.


Subject(s)
Hematopoietic Stem Cells , Mitochondria , Mitochondrial Permeability Transition Pore , Animals , Mitochondria/metabolism , Hematopoietic Stem Cells/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Hematopoiesis , Peptidyl-Prolyl Isomerase F/metabolism , Peptidyl-Prolyl Isomerase F/genetics , Cell Differentiation , Oxidative Phosphorylation , Female , Mice, Inbred C57BL
2.
Afr Health Sci ; 24(1): 295-306, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38962330

ABSTRACT

Background: The Mediterranean thistle Atractylis gummifera L. (Asteraceae; AG) has diterpenoid glucosides; atractyloside and carboxyatractyloside that interact with mitochondrial protein adenine nucleotide translocator (ANT) and resulted in ATP inhibition. Despite its well-known toxicity, acute poisonings still occur with this plant. Although most symptoms are attributed to ANT and diterpenoids interaction, in-depth investigation of the effects of AG extract on various cellular processes has not been performed. Objective/method: We tested in vitro induction of mitochondrial permeability transition pore (MPTP) opening in bovine liver mitochondria and evaluated its cytotoxicity and genotoxicity using Allium cepa test. Cell division, mitotic index (MI) and total chromosomal and mitotic aberrations (TAs), that all seem potentially affected by ATP shortage, were studied in root cells of Allium cepa exposed to Atractylis gummifera extract. Results: With the two different doses of two purified AG fractions, stronger induction of MPTP was observed compared to the induction with the standard pure atracyloside. Aqueous AG extract exerted inhibition root growth in A. cepa at 6 different doses. The TAs was increased in a dose-dependent manner too, while mitotic index was decreased at the same doses. Evaluation of mitotic phases revealed mitodepressive effect of AG on A. cepa roots. Conclusion: this work highlights cellular and mitochondrial adverse effects of Atractylis gummifera extracts. A purified fraction that likely corresponds to ATR derivatives induces MPTP opening leading to swelling of mitochondria and its dysfunction. Allium cepa test provides the evidence for A. gummifera genotoxicity and cytotoxicity.


Subject(s)
Atractyloside , Plant Extracts , Plant Extracts/pharmacology , Plant Extracts/toxicity , Animals , Cattle , Atractyloside/pharmacology , Atractyloside/toxicity , Onions/drug effects , Mitochondria, Liver/drug effects , Mitochondrial Permeability Transition Pore , Mitochondrial Membrane Transport Proteins/drug effects
4.
Biomed Pharmacother ; 177: 117111, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39013220

ABSTRACT

Mitochondrial dysfunction is critical in the pathogenesis of asthma. Mitochondrial permeability transition pore (mPTP) regulates the release of mitochondrial damage-associated molecular patterns (mtDAMPs) to maintain mitochondrial homeostasis. Bongkrekic acid (BKA) is a highly selective inhibitor of mPTP opening, participates the progression of various diseases. This research investigated the exact roles of BKA and mPTP in the pathogenesis of asthma and elucidated its underlying mechanisms. In the present study, cytochrome c, one of the mtDAMPs, levels were elevated in asthmatic patients, and associated to airway inflammation and airway obstruction. BKA, the inhibitor of mPTP markedly reversed TDI-induced airway hyperresponsiveness, airway inflammation, and mitochondrial dysfunction. Pretreatment with mitochondrial precipitation, to simulate the release of mtDAMPs, further increased TDI-induced airway inflammation and the expression of RAGE in mice. Administration of the inhibitor of RAGE, FPS-ZM1, alleviated the airway inflammation, the abnormal open of mPTP and mitochondrial dysfunction induced by mtDAMPs and TDI. Furthermore, stimulation with different mtDAMPs activated RAGE signaling in human bronchial epithelial cells. Accordingly, our study indicated that mPTP was important and BKA was efficient in alleviating inflammation in TDI-induced asthma. A positive feedback loop involving mPTP, mtDAMPs and RAGE was present in TDI-induced asthma, indicating that mPTP might serve as a potential therapeutic target for asthma.


Subject(s)
Asthma , Disease Models, Animal , Mitochondrial Permeability Transition Pore , Asthma/drug therapy , Asthma/metabolism , Animals , Humans , Mice , Mitochondrial Permeability Transition Pore/metabolism , Male , Feedback, Physiological/drug effects , Receptor for Advanced Glycation End Products/metabolism , Female , Mice, Inbred BALB C , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Signal Transduction/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mice, Inbred C57BL , Adult
5.
Sci Rep ; 14(1): 16444, 2024 07 16.
Article in English | MEDLINE | ID: mdl-39013979

ABSTRACT

Colistin is a polymyxin antibiotic currently experiencing renewed clinical interest due to its efficacy in the treatment of multidrug resistant (MDR) bacterial infections. The frequent onset of acute dose-dependent kidney injury, with the potential of leading to long-term renal damage, has limited its use and hampered adequate dosing regimens, increasing the risk of suboptimal plasma concentrations during treatment. The mechanism of colistin-induced renal toxicity has been postulated to stem from mitochondrial damage, yet there is no direct evidence of colistin acting as a mitochondrial toxin. The aim of this study was to evaluate whether colistin can directly induce mitochondrial toxicity and, if so, uncover the underlying molecular mechanism. We found that colistin leads to a rapid permeability transition of mitochondria isolated from mouse kidney that was fully prevented by co-incubation of the mitochondria with desensitizers of the mitochondrial transition pore cyclosporin A or L-carnitine. The protective effect of L-carnitine was confirmed in experiments in primary cultured mouse tubular cells. Consistently, the relative risk of colistin-induced kidney damage, calculated based on histological analysis as well as by the early marker of tubular kidney injury, Kim-1, was halved under co-administration with L-carnitine in vivo. Notably, L-carnitine neither affected the pharmacokinetics of colistin nor its antimicrobial activity against relevant bacterial strains. In conclusion, colistin targets the mitochondria and induces permeability transition thereof. L-carnitine prevents colistin-induced permeability transition in vitro. Moreover, L-carnitine co-administration confers partial nephroprotection in mice treated with colistin, without interfering with its pharmacokinetics and antibacterial activity.


Subject(s)
Acute Kidney Injury , Anti-Bacterial Agents , Carnitine , Colistin , Mitochondria , Animals , Colistin/adverse effects , Colistin/administration & dosage , Acute Kidney Injury/prevention & control , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Carnitine/pharmacology , Carnitine/administration & dosage , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Male , Mitochondrial Permeability Transition Pore/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , Cyclosporine
6.
Cell Commun Signal ; 22(1): 328, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38872145

ABSTRACT

BACKGROUND: Kawasaki disease (KD) is an immune vasculitis of unknown origin, characterized by transient inflammation. The activation of the cGAS-STING pathway, triggered by mitochondrial DNA (mtDNA) release, has been implicated in the onset of KD. However, its specific role in the progression of inflammation during KD's acute phase remains unclear. METHODS: We measured mtDNA and 2'3'-cGAMP expression in KD patient serum using RT-qPCR and ELISA. A murine model of KD was induced by injecting Lactobacillus casei cell wall extract (LCWE), after which cGAS-STING pathway activation and inflammatory markers were assessed via immunohistochemistry, western blot, and RT-qPCR. Human umbilical vein endothelial cells (HUVECs) were treated with KD serum and modulators of the cGAS-STING pathway for comparative analysis. Mitochondrial function was evaluated using Mitosox staining, mPTP opening was quantified by fluorescence microscopy, and mitochondrial membrane potential (MMP) was determined with JC-1 staining. RESULTS: KD patient serum exhibited increased mtDNA and 2'3'-cGAMP expression, with elevated levels of pathway-related proteins and inflammatory markers observed in both in vivo and in vitro models. TEM confirmed mitochondrial damage, and further studies demonstrated that inhibition of mPTP opening reduced mtDNA release, abrogated cGAS-STING pathway activation, and mitigated inflammation. CONCLUSION: These findings indicate that mtDNA released through the mPTP is a critical activator of the cGAS-STING pathway, contributing significantly to KD-associated inflammation. Targeting mtDNA release or the cGAS-STING pathway may offer novel therapeutic approaches for KD management.


Subject(s)
DNA, Mitochondrial , Inflammation , Membrane Proteins , Mitochondrial Permeability Transition Pore , Mucocutaneous Lymph Node Syndrome , Nucleotidyltransferases , Signal Transduction , Mucocutaneous Lymph Node Syndrome/metabolism , Mucocutaneous Lymph Node Syndrome/pathology , Mucocutaneous Lymph Node Syndrome/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Inflammation/pathology , Inflammation/metabolism , Inflammation/genetics , Animals , Mitochondrial Permeability Transition Pore/metabolism , Male , Mice , Human Umbilical Vein Endothelial Cells/metabolism , Female , Acute Disease , Mice, Inbred C57BL , Child, Preschool
7.
Int J Mol Sci ; 25(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38892381

ABSTRACT

Metabolic dysfunction-associated fatty liver disease (MAFLD) is one of the most common chronic liver diseases worldwide. Some patients with MAFLD develop metabolic dysfunction-associated steatohepatitis (MASH), which can lead to severe liver fibrosis. However, the molecular mechanisms underlying this progression remain unknown, and no effective treatment for MASH has been developed so far. In this study, we performed a longitudinal detailed analysis of mitochondria in the livers of choline-deficient, methionine-defined, high-fat-diet (CDAHFD)-fed mice, which exhibited a MASH-like pathology. We found that FoF1-ATPase activity began to decrease in the mitochondria of CDAHFD-fed mice prior to alterations in the activity of mitochondrial respiratory chain complex, almost at the time of onset of liver fibrosis. In addition, the decrease in FoF1-ATPase activity coincided with the accelerated opening of the mitochondrial permeability transition pore (PTP), for which FoF1-ATPase might be a major component or regulator. As fibrosis progressed, mitochondrial permeability transition (PT) induced in CDAHFD-fed mice became less sensitive to cyclosporine A, a specific PT inhibitor. These results suggest that episodes of fibrosis might be related to the disruption of mitochondrial function via PTP opening, which is triggered by functional changes in FoF1-ATPase. These novel findings could help elucidate the pathogenesis of MASH and lead to the development of new therapeutic strategies.


Subject(s)
Choline Deficiency , Diet, High-Fat , Disease Models, Animal , Fatty Liver , Animals , Diet, High-Fat/adverse effects , Mice , Choline Deficiency/metabolism , Choline Deficiency/complications , Male , Fatty Liver/metabolism , Fatty Liver/etiology , Fatty Liver/pathology , Mitochondrial Permeability Transition Pore/metabolism , Mitochondria, Liver/metabolism , Choline/metabolism , Mice, Inbred C57BL , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/etiology , Amino Acids/metabolism , Mitochondria/metabolism , Methionine/deficiency , Methionine/metabolism
8.
J Agric Food Chem ; 72(26): 14975-14983, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38898562

ABSTRACT

Enniatins (ENNs) A1 and B1, previously considered ionophores, are emerging mycotoxins with effects on Ca2+ homeostasis. However, their exact mechanism of action remains unclear. This study investigated how these toxins affect Ca2+ flux in SH-SY5Y cells. ENN A1 induced Ca2+ influx through store-operated channels (SOC). The mitochondrial uncoupler FCCP reduced this influx, suggesting that the mitochondrial status influences the toxin effect. Conversely, ENN B1 did not affect SOC but acted on another Ca2+ channel, as shown when nickel, which directly blocks the Ca2+ channel pore, is added. Mitochondrial function also influenced the effects of ENN B1, as treatment with FCCP reduced toxin-induced Ca2+ depletion and uptake. In addition, both ENNs altered mitochondrial function by producing the opening of the mitochondrial permeability transition pore. This study describes for the first time that ENN A1 and B1 are not Ca2+ ionophores and suggests a different mechanism of action for each toxin.


Subject(s)
Calcium , Depsipeptides , Mitochondria , Mitochondria/metabolism , Mitochondria/drug effects , Calcium/metabolism , Humans , Depsipeptides/pharmacology , Mycotoxins/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Cell Line, Tumor
9.
Life Sci ; 351: 122802, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38857656

ABSTRACT

Adenosine nucleotide translocases (ANTs) are a family of proteins abundant in the inner mitochondrial membrane, primarily responsible for shuttling ADP and ATP across the mitochondrial membrane. Additionally, ANTs are key players in balancing mitochondrial energy metabolism and regulating cell death. ANT2 isoform, highly expressed in undifferentiated and proliferating cells, is implicated in the development and drug resistance of various tumors. We conduct a detailed analysis of the potential mechanisms by which ANT2 may influence tumorigenesis and drug resistance. Notably, the significance of ANT2 extends beyond oncology, with roles in non-tumor cell processes including blood cell development, gastrointestinal motility, airway hydration, nonalcoholic fatty liver disease, obesity, chronic kidney disease, and myocardial development, making it a promising therapeutic target for multiple pathologies. To better understand the molecular mechanisms of ANT2, this review summarizes the structural properties, expression patterns, and basic functions of the ANT2 protein. In particular, we review and analyze the controversy surrounding ANT2, focusing on its role in transporting ADP/ATP across the inner mitochondrial membrane, its involvement in the composition of the mitochondrial permeability transition pore, and its participation in apoptosis.


Subject(s)
Adenine Nucleotide Translocator 2 , Humans , Animals , Adenine Nucleotide Translocator 2/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Apoptosis , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Adenosine Triphosphate/metabolism
10.
Cell Biol Int ; 48(9): 1266-1284, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38837523

ABSTRACT

Platelets are essential component of circulation that plays a major role in hemostasis and thrombosis. During activation and its demise, platelets release platelet-derived microvesicles, with lysophosphatidylcholine (LPC) being a prominent component in their lipid composition. LPC, an oxidized low-density lipoprotein, is involved in cellular metabolism, but its higher level is implicated in pathologies like atherosclerosis, diabetes, and inflammatory disorders. Despite this, its impact on platelet function remains relatively unexplored. To address this, we studied LPC's effects on washed human platelets. A multimode plate reader was employed to measure reactive oxygen species and intracellular calcium using H2DCF-DA and Fluo-4-AM, respectively. Flow cytometry was utilized to measure phosphatidylserine expression, mitochondrial membrane potential (ΔΨm), and mitochondrial permeability transition pore (mPTP) formation using FITC-Annexin V, JC-1, and CoCl2/calcein-AM, respectively. Additionally, platelet morphology and its ultrastructure were observed via phase contrast and electron microscopy. Sonoclot and light transmission aggregometry were employed to examine fibrin formation and platelet aggregation, respectively. The findings demonstrate that LPC induced oxidative stress and increased intracellular calcium in platelets, resulting in increased phosphatidylserine expression and reduced ΔΨm. LPC triggered caspase-independent platelet death and mPTP opening via cytosolic and mitochondrial calcium, along with microvesiculation and reduced platelet counts. LPC increased the platelet's size, adopting a balloon-shaped morphology, causing membrane fragmentation and releasing its cellular contents, while inducing a pro-coagulant phenotype with increased fibrin formation and reduced integrin αIIbß3 activation. Conclusively, this study reveals LPC-induced oxidative stress and calcium-mediated platelet death, necrotic in nature with pro-coagulant properties, potentially impacting inflammation and repair mechanisms during vascular injury.


Subject(s)
Blood Platelets , Calcium , Cell Death , Lysophosphatidylcholines , Oxidative Stress , Reactive Oxygen Species , Humans , Oxidative Stress/drug effects , Lysophosphatidylcholines/pharmacology , Lysophosphatidylcholines/metabolism , Calcium/metabolism , Blood Platelets/metabolism , Blood Platelets/drug effects , Reactive Oxygen Species/metabolism , Cell Death/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Platelet Aggregation/drug effects , Mitochondrial Permeability Transition Pore/metabolism
11.
Mol Med ; 30(1): 77, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840035

ABSTRACT

BACKGROUND: Ischemic stroke presents a significant threat to human health due to its high disability rate and mortality. Currently, the clinical treatment drug, rt-PA, has a narrow therapeutic window and carries a high risk of bleeding. There is an urgent need to find new effective therapeutic drugs for ischemic stroke. Icariin (ICA), a key ingredient in the traditional Chinese medicine Epimedium, undergoes metabolism in vivo to produce Icaritin (ICT). While ICA has been reported to inhibit neuronal apoptosis after cerebral ischemia-reperfusion (I/R), yet its underlying mechanism remains unclear. METHODS: PC-12 cells were treated with 200 µM H2O2 for 8 h to establish a vitro model of oxidative damage. After administration of ICT, cell viability was detected by Thiazolyl blue tetrazolium Bromide (MTT) assay, reactive oxygen species (ROS) and apoptosis level, mPTP status and mitochondrial membrane potential (MMP) were detected by flow cytometry and immunofluorescence. Apoptosis and mitochondrial permeability transition pore (mPTP) related proteins were assessed by Western blotting. Middle cerebral artery occlusion (MCAO) model was used to establish I/R injury in vivo. After the treatment of ICA, the neurological function was scored by ZeaLonga socres; the infarct volume was observed by 2,3,5-Triphenyltetrazolium chloride (TTC) staining; HE and Nissl staining were used to detect the pathological state of the ischemic cortex; the expression changes of mPTP and apoptosis related proteins were detected by Western blotting. RESULTS: In vitro: ICT effectively improved H2O2-induced oxidative injury through decreasing the ROS level, inhibiting mPTP opening and apoptosis. In addition, the protective effects of ICT were not enhanced when it was co-treated with mPTP inhibitor Cyclosporin A (CsA), but reversed when combined with mPTP activator Lonidamine (LND). In vivo: Rats after MCAO shown cortical infarct volume of 32-40%, severe neurological impairment, while mPTP opening and apoptosis were obviously increased. Those damage caused was improved by the administration of ICA and CsA. CONCLUSIONS: ICA improves cerebral ischemia-reperfusion injury by inhibiting mPTP opening, making it a potential candidate drug for the treatment of ischemic stroke.


Subject(s)
Apoptosis , Flavonoids , Ischemic Stroke , Membrane Potential, Mitochondrial , Mitochondrial Permeability Transition Pore , Oxidative Stress , Reactive Oxygen Species , Animals , Oxidative Stress/drug effects , Rats , Flavonoids/pharmacology , Flavonoids/therapeutic use , Mitochondrial Permeability Transition Pore/metabolism , Apoptosis/drug effects , Ischemic Stroke/drug therapy , Ischemic Stroke/metabolism , Ischemic Stroke/etiology , PC12 Cells , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial/drug effects , Male , Reperfusion Injury/metabolism , Reperfusion Injury/drug therapy , Disease Models, Animal , Hydrogen Peroxide/metabolism , Cell Survival/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats, Sprague-Dawley
12.
Basic Res Cardiol ; 119(4): 569-585, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38890208

ABSTRACT

Mitochondrial calcium (Ca2+) signals play a central role in cardiac homeostasis and disease. In the healthy heart, mitochondrial Ca2+ levels modulate the rate of oxidative metabolism to match the rate of adenosine triphosphate consumption in the cytosol. During ischemia/reperfusion (I/R) injury, pathologically high levels of Ca2+ in the mitochondrial matrix trigger the opening of the mitochondrial permeability transition pore, which releases solutes and small proteins from the matrix, causing mitochondrial swelling and ultimately leading to cell death. Pharmacological and genetic approaches to tune mitochondrial Ca2+ handling by regulating the activity of the main Ca2+ influx and efflux pathways, i.e., the mitochondrial Ca2+ uniporter and sodium/Ca2+ exchanger, represent promising therapeutic strategies to protect the heart from I/R injury.


Subject(s)
Calcium , Mitochondria, Heart , Myocardial Reperfusion Injury , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/pathology , Humans , Animals , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Calcium/metabolism , Calcium Signaling , Mitochondrial Permeability Transition Pore/metabolism , Calcium Channels/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Cardiotonic Agents/metabolism
13.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731874

ABSTRACT

The mitochondrial protein IF1 is upregulated in many tumors and acts as a pro-oncogenic protein through its interaction with the ATP synthase and the inhibition of apoptosis. We have recently characterized the molecular nature of the IF1-Oligomycin Sensitivity Conferring Protein (OSCP) subunit interaction; however, it remains to be determined whether this interaction could be targeted for novel anti-cancer therapeutic intervention. We generated mitochondria-targeting peptides to displace IF1 from the OSCP interaction. The use of one selective peptide led to displacement of the inhibitor IF1 from ATP synthase, as shown by immunoprecipitation. NMR spectroscopy analysis, aimed at clarifying whether these peptides were able to directly bind to the OSCP protein, identified a second peptide which showed affinity for the N-terminal region of this subunit overlapping the IF1 binding region. In situ treatment with the membrane-permeable derivatives of these peptides in HeLa cells, that are silenced for the IF1 inhibitor protein, showed significant inhibition in mitochondrial permeability transition and no effects on mitochondrial respiration. These peptides mimic the effects of the IF1 inhibitor protein in cancer HeLa cells and confirm that the IF1-OSCP interaction inhibits apoptosis. A third peptide was identified which counteracts the anti-apoptotic role of IF1, showing that OSCP is a promising target for anti-cancer therapies.


Subject(s)
Mitochondria , Mitochondrial Proton-Translocating ATPases , Peptides , Humans , Apoptosis/drug effects , ATPase Inhibitory Protein/drug effects , ATPase Inhibitory Protein/metabolism , HeLa Cells , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Peptides/pharmacology , Peptides/chemistry , Peptides/metabolism , Protein Binding
14.
Physiol Rep ; 12(10): e16056, 2024 May.
Article in English | MEDLINE | ID: mdl-38777811

ABSTRACT

Permeability transition pore (PTP) opening dissipates ion and electron gradients across the internal mitochondrial membrane (IMM), including excess Ca2+ in the mitochondrial matrix. After opening, immediate PTP closure must follow to prevent outer membrane disruption, loss of cytochrome c, and eventual apoptosis. Flickering, defined as the rapid alternative opening/closing of PTP, has been reported in heart, which undergoes frequent, large variations in Ca2+. In contrast, in tissues that undergo depolarization events less often, such as the liver, PTP would not need to be as dynamic and thus these tissues would not be as resistant to stress. To evaluate this idea, it was decided to follow the reversibility of the permeability transition (PT) in isolated murine mitochondria from two different tissues: the very dynamic heart, and the liver, which suffers depolarizations less frequently. It was observed that in heart mitochondria PT remained reversible for longer periods and at higher Ca2+ loads than in liver mitochondria. In all cases, Ca2+ uptake was inhibited by ruthenium red and PT was delayed by Cyclosporine A. Characterization of this phenomenon included measuring the rate of oxygen consumption, organelle swelling and Ca2+ uptake and retention. Results strongly suggest that there are tissue-specific differences in PTP physiology, as it resists many more Ca2+ additions before opening in a highly active organ such as the heart than in an organ that seldom suffers Ca2+ loading, such as the liver.


Subject(s)
Calcium , Mitochondria, Heart , Mitochondria, Liver , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Rats, Wistar , Animals , Mitochondrial Permeability Transition Pore/metabolism , Male , Calcium/metabolism , Mitochondria, Heart/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Rats , Oxygen Consumption , Liver/metabolism , Mitochondrial Swelling/drug effects , Cyclosporine/pharmacology
15.
Cells ; 13(9)2024 May 06.
Article in English | MEDLINE | ID: mdl-38727324

ABSTRACT

Norbormide (NRB) is a Rattus-selective toxicant, which was serendipitously discovered in 1964 and formerly marketed as an eco-friendly rodenticide that was deemed harmless to non-Rattus species. However, due to inconsistent efficacy and the emergence of second-generation anticoagulants, its usage declined, with registration lapsing in 2003. NRBs' lethal action in rats entails irreversible vasoconstriction of peripheral arteries, likely inducing cardiac damage: however, the precise chain of events leading to fatality and the target organs involved remain elusive. This unique contractile effect is exclusive to rat arteries and is induced solely by the endo isomers of NRB, hinting at a specific receptor involvement. Understanding NRB's mechanism of action is crucial for developing species-selective toxicants as alternatives to the broad-spectrum ones currently in use. Recent research efforts have focused on elucidating its cellular mechanisms and sites of action using novel NRB derivatives. The key findings are as follows: NRB selectively opens the rat mitochondrial permeability transition pore, which may be a factor that contributes to its lethal effect; it inhibits rat vascular KATP channels, which potentially controls its Rattus-selective vasoconstricting activity; and it possesses intracellular binding sites in both sensitive and insensitive cells, as revealed by fluorescent derivatives. These studies have led to the development of a prodrug with enhanced pharmacokinetic and toxicological profiles, which is currently undergoing registration as a novel efficacious eco-sustainable Rattus-selective toxicant. The NRB-fluorescent derivatives also show promise as non-toxic probes for intracellular organelle labelling. This review documents in more detail these developments and their implications.


Subject(s)
Rodenticides , Animals , Rats , Rodenticides/toxicity , Humans , Vasoconstriction/drug effects , Mitochondrial Permeability Transition Pore/metabolism
16.
Cell Mol Life Sci ; 81(1): 236, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38795203

ABSTRACT

Chemoresistance is the main obstacle in the clinical treatment of osteosarcoma (OS). In this study, we investigated the role of EF-hand domain-containing protein 1 (EFHD1) in OS chemotherapy resistance. We found that the expression of EFHD1 was highly correlated with the clinical outcome after chemotherapy. We overexpressed EFHD1 in 143B cells and found that it increased their resistance to cell death after drug treatment. Conversely, knockdown of EFHD1 in 143BR cells (a cisplatin-less-sensitive OS cell line derived from 143B cells) increased their sensitivity to treatment. Mechanistically, EFHD1 bound to adenine nucleotide translocase-3 (ANT3) and inhibited its conformational change, thereby inhibiting the opening of the mitochondrial membrane permeability transition pore (mPTP). This effect could maintain mitochondrial function, thereby favoring OS cell survival. The ANT3 conformational inhibitor carboxyatractyloside (CATR), which can promote mPTP opening, enhanced the chemosensitivity of EFHD1-overexpressing cells when combined with cisplatin. The ANT3 conformational inhibitor bongkrekic acid (BKA), which can inhibit mPTP opening, restored the resistance of EFHD1 knockdown cells. In conclusion, our results suggest that EFHD1-ANT3-mPTP might be a promising target for OS therapy in the future.


Subject(s)
Cell Proliferation , Cisplatin , Drug Resistance, Neoplasm , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Osteosarcoma , Humans , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Mitochondrial Permeability Transition Pore/metabolism , Drug Resistance, Neoplasm/drug effects , Cell Proliferation/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Adenine Nucleotide Translocator 3/metabolism , Adenine Nucleotide Translocator 3/genetics , Antineoplastic Agents/pharmacology , Mitochondria/metabolism , Mitochondria/drug effects , Animals , Mice , Protein Binding
17.
Free Radic Biol Med ; 220: 222-235, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38735540

ABSTRACT

Studies have highlighted oxidative damage in the inner ear as a critical pathological basis for sensorineural hearing loss, especially the presbycusis. Poly(ADP-ribose) polymerase-1 (PARP1) activation responds to oxidative stress-induced DNA damage with pro-repair and pro-death effects resembling two sides of the same coin. PARP1-related cell death, known as parthanatos, whose underlying mechanisms are attractive research hotspots but remain to be clarified. In this study, we observed that aged rats showed stria vascularis degeneration and oxidative damage, and PARP1-dependent cell death was prominent in age-related cochlear disorganization and dysfunction. Based on oxidative stress model of primary cultured stria marginal cells (MCs), we revealed that upregulated PARP1 and PAR (Poly(ADP-ribose)) polymers are responsible for MCs oxidative death with high mitochondrial permeability transition pore (mPTP) opening and mitochondrial membrane potential (MMP) collapse, while inhibition of PARP1 ameliorated the adverse outcomes. Importantly, the PARylation of apoptosis-inducing factor (AIF) is essential for its conformational change and translocation, which subsequently causes DNA break and cell death. Concretely, the interaction of PAR and truncated AIF (tAIF) is the mainstream in the parthanatos pathway. We also found that the effects of AIF cleavage and release were achieved through calpain activity and mPTP opening, both of which could be regulated by PARP1 via mediation of mitochondria Ca2+ concentration. In conclusion, the PAR-Ca2+-tAIF signaling pathway in parthanatos contributes to the oxidative stress damage observed in MCs. Targeting PAR-Ca2+-tAIF might be a potential therapeutic strategy for the early intervention of presbycusis and other oxidative stress-associated sensorineural deafness.


Subject(s)
Apoptosis Inducing Factor , Calcium , Oxidative Stress , Poly (ADP-Ribose) Polymerase-1 , Presbycusis , Animals , Apoptosis Inducing Factor/metabolism , Apoptosis Inducing Factor/genetics , Rats , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Calcium/metabolism , Presbycusis/metabolism , Presbycusis/pathology , Presbycusis/genetics , Parthanatos/genetics , Membrane Potential, Mitochondrial , Stria Vascularis/metabolism , Stria Vascularis/pathology , Apoptosis , Mitochondrial Permeability Transition Pore/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Rats, Sprague-Dawley , DNA Damage , Aging/metabolism , Aging/pathology , Cochlea/metabolism , Cochlea/pathology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Male , Humans , Cells, Cultured
18.
J Pharmacol Sci ; 155(2): 35-43, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677784

ABSTRACT

Imeglimin is a novel oral antidiabetic drug for treating type 2 diabetes. However, the effect of imeglimin on NLRP3 inflammasome activation has not been investigated yet. Here, we aimed to investigate whether imeglimin reduces LPS-induced NLRP3 inflammasome activation in THP-1 macrophages and examine the associated underlying mechanisms. We analyzed the mRNA and protein expression levels of NLRP3 inflammasome components and IL-1ß secretion. Additionally, reactive oxygen species (ROS) generation, mitochondrial membrane potential, and mitochondrial permeability transition pore (mPTP) opening were measured by flow cytometry. Imeglimin inhibited NLRP3 inflammasome-mediated IL-1ß production in LPS-stimulated THP-1-derived macrophages. In addition, imeglimin reduced LPS-induced mitochondrial ROS production and mitogen-activated protein kinase phosphorylation. Furthermore, imeglimin restored the mitochondrial function by modulating mitochondrial membrane depolarization and mPTP opening. We demonstrated for the first time that imeglimin reduces LPS-induced NLRP3 inflammasome activation by inhibiting mPTP opening in THP-1 macrophages. These results suggest that imeglimin could be a promising new anti-inflammatory agent for treating diabetic complications.


Subject(s)
Inflammasomes , Macrophages , Mitochondria , Triazines , Humans , Anti-Inflammatory Agents/pharmacology , Hypoglycemic Agents/pharmacology , Inflammasomes/metabolism , Inflammasomes/drug effects , Interleukin-1beta/metabolism , Lipopolysaccharides , Macrophages/drug effects , Macrophages/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Mitogen-Activated Protein Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , THP-1 Cells , Triazines/pharmacology
19.
Circ Res ; 134(10): 1292-1305, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38618716

ABSTRACT

BACKGROUND: During myocardial ischemia/reperfusion (I/R) injury, high levels of matrix Ca2+ and reactive oxygen species (ROS) induce the opening of the mitochondrial permeability transition pore (mPTP), which causes mitochondrial dysfunction and ultimately necrotic death. However, the mechanisms of how these triggers individually or cooperatively open the pore have yet to be determined. METHODS: Here, we use a combination of isolated mitochondrial assays and in vivo I/R surgery in mice. We challenged isolated liver and heart mitochondria with Ca2+, ROS, and Fe2+ to induce mitochondrial swelling. Using inhibitors of the mPTP (cyclosporine A or ADP) lipid peroxidation (ferrostatin-1, MitoQ), we determined how the triggers elicit mitochondrial damage. Additionally, we used the combination of inhibitors during I/R injury in mice to determine if dual inhibition of these pathways is additivity protective. RESULTS: In the absence of Ca2+, we determined that ROS fails to trigger mPTP opening. Instead, high levels of ROS induce mitochondrial dysfunction and rupture independently of the mPTP through lipid peroxidation. As expected, Ca2+ in the absence of ROS induces mPTP-dependent mitochondrial swelling. Subtoxic levels of ROS and Ca2+ synergize to induce mPTP opening. Furthermore, this synergistic form of Ca2+- and ROS-induced mPTP opening persists in the absence of CypD (cyclophilin D), suggesting the existence of a CypD-independent mechanism for ROS sensitization of the mPTP. These ex vivo findings suggest that mitochondrial dysfunction may be achieved by multiple means during I/R injury. We determined that dual inhibition of the mPTP and lipid peroxidation is significantly more protective against I/R injury than individually targeting either pathway alone. CONCLUSIONS: In the present study, we have investigated the relationship between Ca2+ and ROS, and how they individually or synergistically induce mitochondrial swelling. Our findings suggest that Ca2+ mediates mitochondrial damage through the opening of the mPTP, although ROS mediates its damaging effects through lipid peroxidation. However, subtoxic levels both Ca2+ and ROS can induce mPTP-mediated mitochondrial damage. Targeting both of these triggers to preserve mitochondria viability unveils a highly effective therapeutic approach for mitigating I/R injury.


Subject(s)
Lipid Peroxidation , Mice, Inbred C57BL , Mitochondria, Heart , Mitochondria, Liver , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury , Reactive Oxygen Species , Animals , Lipid Peroxidation/drug effects , Mitochondrial Permeability Transition Pore/metabolism , Reactive Oxygen Species/metabolism , Mice , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/pathology , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Mitochondria, Liver/drug effects , Calcium/metabolism , Mitochondrial Swelling/drug effects
20.
Plant Sci ; 345: 112107, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38685455

ABSTRACT

Programmed cell death (PCD) is an important factor to reduces the viability of plant germplasm after cryopreservation. However, the pathways by which PCD occurs is not fully understood. To investigate whether there is a mitochondrial pathway for pollen PCD after cryopreservation, the pollen of Paeonia lactiflora two cultivars with different PCD levels after cryopreservation was used as test material and the changes of mitochondrial calcium ions (Ca2+), structure, function and their relationship with PCD were compared. The results showed that compared with fresh pollen, the PCD of 'Feng Huang Nie Pan' was significantly reduced after cryopreservation. Their mitochondrial Ca2+ content decreased by 74.27%, mitochondrial permeability transition pore (MPTP) opening reduced by 25.41%, mitochondrial membrane potential slightly decreased by 5.02%, cardiolipin oxidation decreased by 65.31%, and oxygen consumption remained stable, with a slightly ATP production increase. On the contrary, compared with fresh pollen, 'Zi Feng Chao Yang' showed severe PCD after cryopreservation. The decline in mitochondrial Ca2+-ATPase activity led to an accumulation of excessive Ca2+ within mitochondria, triggering widespread opening of MPTP, significantly affecting mitochondrial respiration and energy synthesis. These results suggest the mitochondrial pathway of PCD exists in pollen cryopreservation.


Subject(s)
Apoptosis , Calcium , Cryopreservation , Mitochondria , Paeonia , Pollen , Mitochondria/metabolism , Paeonia/physiology , Paeonia/metabolism , Pollen/physiology , Pollen/metabolism , Cryopreservation/methods , Calcium/metabolism , Membrane Potential, Mitochondrial/physiology , Mitochondrial Permeability Transition Pore/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL