Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.599
Filter
1.
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)
Mitochondrial Proton-Translocating ATPases , Peptides , Humans , HeLa Cells , Mitochondrial Proton-Translocating ATPases/metabolism , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Peptides/pharmacology , Peptides/chemistry , Peptides/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Apoptosis/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , ATPase Inhibitory Protein , Protein Binding , Mitochondrial Membrane Transport Proteins/metabolism
2.
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 , Interleukin-1beta , Lipopolysaccharides , Macrophages , Membrane Potential, Mitochondrial , Mitochondria , NLR Family, Pyrin Domain-Containing 3 Protein , Reactive Oxygen Species , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Macrophages/drug effects , Macrophages/metabolism , Interleukin-1beta/metabolism , Membrane Potential, Mitochondrial/drug effects , Anti-Inflammatory Agents/pharmacology , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Hypoglycemic Agents/pharmacology , Phosphorylation/drug effects , THP-1 Cells , Mitogen-Activated Protein Kinases/metabolism
3.
EMBO Rep ; 25(4): 2071-2096, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38565738

ABSTRACT

Most mitochondrial proteins are synthesized on cytosolic ribosomes and imported into mitochondria in a post-translational reaction. Mitochondrial precursor proteins which use the ER-SURF pathway employ the surface of the endoplasmic reticulum (ER) as an important sorting platform. How they reach the mitochondrial import machinery from the ER is not known. Here we show that mitochondrial contact sites play a crucial role in the ER-to-mitochondria transfer of precursor proteins. The ER mitochondria encounter structure (ERMES) and Tom70, together with Djp1 and Lam6, are part of two parallel and partially redundant ER-to-mitochondria delivery routes. When ER-to-mitochondria transfer is prevented by loss of these two contact sites, many precursors of mitochondrial inner membrane proteins are left stranded on the ER membrane, resulting in mitochondrial dysfunction. Our observations support an active role of the ER in mitochondrial protein biogenesis.


Subject(s)
Mitochondria , Saccharomyces cerevisiae Proteins , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Protein Transport , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Endoplasmic Reticulum/metabolism , Saccharomyces cerevisiae Proteins/metabolism
4.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(4): 461-466, 2024 Apr 10.
Article in Chinese | MEDLINE | ID: mdl-38565513

ABSTRACT

OBJECTIVE: To explore the prevalence, clinical features, genetic characteristics and prognosis of Citrin deficiency in Henan province of China. METHODS: A total of 986 565 neonates screened by tandem mass spectrometry at the Third Affiliated Hospital of Zhengzhou University from January 2013 to December 2021 were retrospectively analyzed. Analysis of SLC25A13 gene variants and parental verification were carried out for neonates suspected for Citrin deficiency by next-generation sequencing. The clinical, biochemical and genetic characteristics of Citrin deficiency patients were integrated to guide the diet treatment and follow up the growth and development. Paired-t test was used to compare the amino acid levels in the peripheral blood samples before and after the treatment. RESULTS: Nine cases of Citrin deficiency were diagnosed among the 986 565 neonates. Specific elevation of citrulline was observed in all of the 9 cases. Six variants were detected by genetic sequencing, among which c.852_855delTATG, c.615+5G>A, c.550C>T and IVS16ins3kb were known pathogenic variants, whilst c.1111_1112delAT and c.837T>A were unreported previously. The detection rate for c. 852_855delTATG was the highest (61.6%, 11/18), followed by IVS16ins3kb (16.7%, 3/18). The clinical symptoms of all patients were relieved after the treatment, and the blood amino acid profile and biochemical parameters were significantly improved by gradually falling within the normal range. By June 2022, all patients had shown a good prognosis. CONCLUSION: The prevalence of Citrin deficiency among neonates from Henan Province by tandem mass spectrometry is 1/109 618, and the carrier rate for the pathogenic variants of the SLC25A13 gene was 1/166. The c.852_855delTATG may be a hot spot variant among the patients. Discovery of the novel variants has enriched the mutational spectrum of the SLC25A13 gene. Above results have provided a basis for the early diagnosis, treatment, prognosis and genetic counseling for the affected families.


Subject(s)
Citrullinemia , Neonatal Screening , Infant, Newborn , Humans , Neonatal Screening/methods , Citrullinemia/diagnosis , Citrullinemia/genetics , Retrospective Studies , Mutation , Citrulline , Mitochondrial Membrane Transport Proteins/genetics
5.
Orphanet J Rare Dis ; 19(1): 159, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38610036

ABSTRACT

BACKGROUND: Clinicians traditionally aim to identify a singular explanation for the clinical presentation of a patient; however, in some cases, the diagnosis may remain elusive or fail to comprehensively explain the clinical findings. In recent years, advancements in next-generation sequencing, including whole-exome sequencing, have led to the incidental identification of dual diagnoses in patients. Herein we present the cases of five pediatric patients diagnosed with dual rare genetic diseases. Their natural history and diagnostic process were explored, and lessons learned from utilizing next-generation diagnostic technologies have been reported. RESULTS: Five pediatric cases (3 boys, 2 girls) with dual diagnoses were reported. The age at diagnosis was from 3 months to 10 years. The main clinical presentations were psychomotor retardation and increased muscular tension, some accompanied with liver dysfunction, abnormal appearance, precocious puberty, dorsiflexion restriction and varus of both feet, etc. After whole-exome sequencing, nine diseases were confirmed in these patients: Angelman syndrome and Krabbe disease in case 1, Citrin deficiency and Kabuki syndrome in case 2, Homocysteinemia type 2 and Copy number variant in case 3, Isolated methylmalonic acidemia and Niemann-Pick disease type B in case 4, Isolated methylmalonic acidemia and 21-hydroxylase deficiency in case 5. Fifteen gene mutations and 2 CNVs were identified. Four novel mutations were observed, including c.15292de1A in KMT2D, c.159_164inv and c.1427G > A in SLC25A13, and c.591 C > G in MTHFR. CONCLUSIONS: Our findings underscore the importance of clinicians being vigilant about the significance of historical and physical examination. Comprehensive clinical experience is crucial for identifying atypical clinical features, particularly in cases involving dual rare genetic diseases.


Subject(s)
Abnormalities, Multiple , Amino Acid Metabolism, Inborn Errors , Angelman Syndrome , Citrullinemia , Male , Female , Humans , Child , Mitochondrial Membrane Transport Proteins
6.
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
7.
Cancer Lett ; 590: 216847, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38583647

ABSTRACT

Tamoxifen (TAM) resistance presents a major clinical obstacle in the management of estrogen-sensitive breast cancer, highlighting the need to understand the underlying mechanisms and potential therapeutic approaches. We showed that dysregulated mitochondrial dynamics were involved in TAM resistance by protecting against mitochondrial apoptosis. The dysregulated mitochondrial dynamics were associated with increased mitochondrial fusion and decreased fission, thus preventing the release of mitochondrial cytochrome c to the cytoplasm following TAM treatment. Dynamin-related GTPase protein mitofusin 1 (MFN1), which promotes fusion, was upregulated in TAM-resistant cells, and high MFN1 expression indicated a poor prognosis in TAM-treated patients. Mitochondrial translocation of MFN1 and interaction between MFN1 and mitofusin 2 (MFN2) were enhanced to promote mitochondrial outer membrane fusion. The interaction of MFN1 and cristae-shaping protein optic atrophy 1 (OPA1) and OPA1 oligomerization were reduced due to augmented OPA1 proteolytic cleavage, and their apoptosis-promoting function was reduced due to cristae remodeling. Furthermore, the interaction of MFN1 and BAK were increased, which restrained BAK activation following TAM treatment. Knockdown or pharmacological inhibition of MFN1 blocked mitochondrial fusion, restored BAK oligomerization and cytochrome c release, and amplified activation of caspase-3/9, thus sensitizing resistant cells to apoptosis and facilitating the therapeutic effects of TAM both in vivo and in vitro. Conversely, overexpression of MFN1 alleviated TAM-induced mitochondrial apoptosis and promoted TAM resistance in sensitive cells. These results revealed that dysregulated mitochondrial dynamics contributes to the development of TAM resistance, suggesting that targeting MFN1-mediated mitochondrial fusion is a promising strategy to circumvent TAM resistance.


Subject(s)
Apoptosis , Breast Neoplasms , Drug Resistance, Neoplasm , GTP Phosphohydrolases , Mitochondrial Dynamics , Tamoxifen , Humans , Tamoxifen/pharmacology , Mitochondrial Dynamics/drug effects , Apoptosis/drug effects , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Drug Resistance, Neoplasm/drug effects , Female , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Animals , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Cell Line, Tumor , Antineoplastic Agents, Hormonal/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , MCF-7 Cells , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Xenograft Model Antitumor Assays
8.
J Nat Prod ; 87(4): 1187-1196, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38632902

ABSTRACT

Psammaplins are sulfur containing bromotyrosine alkaloids that have shown antitumor activity through the inhibition of class I histone deacetylases (HDACs). The cytotoxic properties of psammaplin A (1), the parent compound, are related to peroxisome proliferator-activated receptor γ (PPARγ) activation, but the mechanism of action of its analogs psammaplin K (2) and bisaprasin (3) has not been elucidated. In this study, the protective effects against oxidative stress of compounds 1-3, isolated from the sponge Aplysinella rhax, were evaluated in SH-SY5Y cells. The compounds improved cell survival, recovered glutathione (GSH) content, and reduced reactive oxygen species (ROS) release at nanomolar concentrations. Psammaplins restored mitochondrial membrane potential by blocking mitochondrial permeability transition pore opening and reducing cyclophilin D expression. This effect was mediated by the capacity of 1-3 to activate PPARγ, enhancing gene expression of the antioxidant enzymes catalase, nuclear factor E2-related factor 2 (Nrf2), and glutathione peroxidase. Finally, HDAC3 activity was reduced by 1-3 under oxidative stress conditions. This work is the first description of the neuroprotective activity of 1 at low concentrations and the mechanism of action of 2 and 3. Moreover, it links for the first time the previously described effects of 1 in HDAC3 and PPARγ signaling, opening a new research field for the therapeutic potential of this compound family.


Subject(s)
Disulfides , Oxidative Stress , PPAR gamma , Tyrosine/analogs & derivatives , PPAR gamma/metabolism , Oxidative Stress/drug effects , Humans , Animals , Molecular Structure , Reactive Oxygen Species/metabolism , Neurons/drug effects , Histone Deacetylases/metabolism , Histone Deacetylases/drug effects , NF-E2-Related Factor 2/metabolism , Porifera/chemistry , Membrane Potential, Mitochondrial/drug effects , Antioxidants/pharmacology , Antioxidants/chemistry , Glutathione/metabolism , Alkaloids/pharmacology , Alkaloids/chemistry , Catalase/metabolism , Glutathione Peroxidase/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Membrane Transport Proteins/metabolism
9.
Biochem Soc Trans ; 52(2): 911-922, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38629718

ABSTRACT

To date, there is no general physical model of the mechanism by which unfolded polypeptide chains with different properties are imported into the mitochondria. At the molecular level, it is still unclear how transit polypeptides approach, are captured by the protein translocation machinery in the outer mitochondrial membrane, and how they subsequently cross the entropic barrier of a protein translocation pore to enter the intermembrane space. This deficiency has been due to the lack of detailed structural and dynamic information about the membrane pores. In this review, we focus on the recently determined sub-nanometer cryo-EM structures and our current knowledge of the dynamics of the mitochondrial two-pore outer membrane protein translocation machinery (TOM core complex), which provide a starting point for addressing the above questions. Of particular interest are recent discoveries showing that the TOM core complex can act as a mechanosensor, where the pores close as a result of interaction with membrane-proximal structures. We highlight unusual and new correlations between the structural elements of the TOM complexes and their dynamic behavior in the membrane environment.


Subject(s)
Mitochondria , Mitochondrial Membranes , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Protein Transport , Cryoelectron Microscopy/methods , Humans , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/chemistry , Models, Molecular , Protein Conformation , Animals
10.
Chem Biodivers ; 21(5): e202301916, 2024 May.
Article in English | MEDLINE | ID: mdl-38511277

ABSTRACT

BACKGROUND: Emodin has been shown to exert anti-inflammatory and cytoprotective effects. Our study aimed to identify a novel anti-inflammatory mechanism of emodin. METHODS: An LPS-induced model of microvascular endothelial cell (HMEC-1) injury was constructed. Cell proliferation was examined using a CCK-8 assay. The effects of emodin on reactive oxygen species (ROS), cell migration, the mitochondrial membrane potential (MMP), and the opening of the mitochondrial permeability transition pore (mPTP) were evaluated. Actin-Tracker Green was used to examine the relationship between cell microfilament reconstruction and ATP5A1 expression. The effects of emodin on the expression of ATP5A1, NALP3, and TNF-α were determined. After treatment with emodin, ATP5A1 and inflammatory factors (TNF-α, IL-1, IL-6, IL-13 and IL-18) were examined by Western blotting. RESULTS: Emodin significantly increased HMEC-1 cell proliferation and migration, inhibited the production of ROS, increased the mitochondrial membrane potential, and blocked the opening of the mPTP. Moreover, emodin could increase ATP5A1 expression, ameliorate cell microfilament remodeling, and decrease the expression of inflammatory factors. In addition, when ATP5A1 was overexpressed, the regulatory effect of emodin on inflammatory factors was not significant. CONCLUSION: Our findings suggest that emodin can protect HMEC-1 cells against inflammatory injury. This process is modulated by the expression of ATP5A1.


Subject(s)
Cell Proliferation , Emodin , Lipopolysaccharides , Up-Regulation , Emodin/pharmacology , Emodin/chemistry , Lipopolysaccharides/pharmacology , Humans , Cell Proliferation/drug effects , Up-Regulation/drug effects , Membrane Potential, Mitochondrial/drug effects , Cell Movement/drug effects , Reactive Oxygen Species/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Cell Line , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry
11.
Cardiovasc Pathol ; 70: 107630, 2024.
Article in English | MEDLINE | ID: mdl-38490313

ABSTRACT

A female neonate born with normal Apgar scores at 38+2 weeks of gestational age unexpectedly passed away within less than 30 hours after birth. The situation mirrored her brother's earlier demise within 24 hours post-delivery, suggesting a possible genetic disorder. Gross examination revealed widespread cyanosis and distinct yellowish changes on the cardiac ventricles. Histopathological examination disclosed lipid accumulation in the liver, heart, and kidneys. Tandem mass spectrometry detected elevated levels of 10 amino acids and 14 carnitines in cardiac blood. Trio-whole genome sequencing (Trio-WGS) identified the SLC25A20 c.199-10T>G mutation associated with carnitine-acylcarnitine translocase disease (CACTD), a type of fatty acid oxidation disorders (FAODs) with a potential for sudden death. Further validation of gene expression confirmed the functional deficiency of SLC25A20, ultimately diagnosing CACTD as the underlying cause of the neonate's demise. This case highlights the importance of prenatal metabolic and genetic screening for prospective parents and emphasizes the need for forensic doctors to integrate metabolomic and genomic investigations into autopsies for suspected inherited metabolic diseases.


Subject(s)
Carnitine Acyltransferases , Lipid Metabolism, Inborn Errors , Mutation , Humans , Infant, Newborn , Female , Carnitine Acyltransferases/deficiency , Carnitine Acyltransferases/genetics , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Lipid Metabolism, Inborn Errors/complications , Lipid Metabolism, Inborn Errors/diagnosis , Phenotype , Fatal Outcome , Genetic Predisposition to Disease , Sudden Infant Death/genetics , Sudden Infant Death/pathology , Sudden Infant Death/etiology , Autopsy , Death, Sudden, Cardiac/etiology , Death, Sudden, Cardiac/pathology , Cause of Death , Carnitine/analogs & derivatives , Carnitine/deficiency , Mitochondrial Membrane Transport Proteins/genetics , Myocardium/pathology , Myocardium/metabolism , Membrane Transport Proteins
12.
Life Sci ; 345: 122567, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38492919

ABSTRACT

The aim was to understand the direct impact of aerobic short-term exercise on lipid metabolism, specifically in regulating the mitochondrial carrier homolog 2 (MTCH2) and how it interferes with lipid metabolism in mesenteric adipose tissue. Swiss mice were divided into three groups: control, sedentary obese, and exercised obese. The obese groups were induced into obesity for fourteen weeks of a high-fat diet, and the trained submitted to seven aerobic exercise sessions. The exercise proved the significant increase of the pPerilipin-1, a hormone-sensitive lipase gene, and modulates lipid metabolism by increasing the expression of Mtch2 and acetyl Co-A carboxylase, perhaps occurring as feedback to regulate lipid metabolism in adipose tissue. In conclusion, we demonstrate, for the first time, how aerobic physical exercise increases Mtch2 transcription in mesenteric adipose tissue. This increase was due to changes in energy demand caused by exercise, confirmed by observing the significant reduction in mesenteric adipose tissue mass in the exercised group. Also, we showed that physical exercise increased the phosphorylative capacity of PLIN1, a protein responsible for the degradation of fatty acids in the lipid droplet, providing acyl and glycerol for cellular metabolism. Although our findings demonstrate evidence of MTCH2 as a protein that regulates lipid homeostasis, scant knowledge exists concerning the signaling of the MTCH2 pathway in regulatingfatty acid metabolism. Therefore, unveiling the means of molecular signaling of MTCH2 demonstrates excellent potential for treating obesity.


Subject(s)
Adipose Tissue , Lipid Metabolism , Mitochondrial Membrane Transport Proteins , Obesity , Physical Conditioning, Animal , Animals , Mice , Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Lipids , Mice, Obese , Mitochondrial Membrane Transport Proteins/metabolism , Obesity/metabolism , Physical Conditioning, Animal/physiology , Lipid Metabolism/genetics , Lipid Metabolism/physiology
13.
Sci Rep ; 14(1): 6751, 2024 03 21.
Article in English | MEDLINE | ID: mdl-38514795

ABSTRACT

Mitochondrial Ca2+ overload can mediate mitochondria-dependent cell death, a major contributor to several human diseases. Indeed, Duchenne muscular dystrophy (MD) is driven by dysfunctional Ca2+ influx across the sarcolemma that causes mitochondrial Ca2+ overload, organelle rupture, and muscle necrosis. The mitochondrial Ca2+ uniporter (MCU) complex is the primary characterized mechanism for acute mitochondrial Ca2+ uptake. One strategy for preventing mitochondrial Ca2+ overload is deletion of the Mcu gene, the pore forming subunit of the MCU-complex. Conversely, enhanced MCU-complex Ca2+ uptake is achieved by deleting the inhibitory Mcub gene. Here we show that myofiber-specific Mcu deletion was not protective in a mouse model of Duchenne MD. Specifically, Mcu gene deletion did not reduce muscle histopathology, did not improve muscle function, and did not prevent mitochondrial Ca2+ overload. Moreover, myofiber specific Mcub gene deletion did not augment Duchenne MD muscle pathology. Interestingly, we observed MCU-independent Ca2+ uptake in dystrophic mitochondria that was sufficient to drive mitochondrial permeability transition pore (MPTP) activation and skeletal muscle necrosis, and this same type of activity was observed in heart, liver, and brain mitochondria. These results demonstrate that mitochondria possess an uncharacterized MCU-independent Ca2+ uptake mechanism that is sufficient to drive MPTP-dependent necrosis in MD in vivo.


Subject(s)
Muscular Dystrophy, Duchenne , Animals , Humans , Mice , Calcium/metabolism , Calcium Channels/metabolism , Cell Death , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Muscular Dystrophy, Duchenne/pathology , Necrosis/metabolism
14.
Eur Rev Med Pharmacol Sci ; 28(5): 1873-1880, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38497870

ABSTRACT

BACKGROUND: This study aimed to evaluate clinical, biochemical, and genotypic findings of patients diagnosed with urea cycle mitochondrial transporter disorders. CASE SERIES: In this study, patients followed up with the diagnosis of urea cycle mitochondrial transporter disorders in the pediatric metabolism outpatient clinic of Diyarbakir Children's Hospital were retrospectively examined. Height, weight, head circumference, gender, age at diagnosis, follow-up period, consanguinity history between parents, and treatments of the patients included in the study were evaluated. Eight patients suffering from urea cycle mitochondrial transporter disorders were enrolled in the study. Five patients were found to have biallelic variants of the SLC25A15 gene. Two patients were found to have biallelic variants of the SLC25A13 gene. Two of our patients presented with gait disturbances and were diagnosed with HHH syndrome. One patient presented with liver failure and was diagnosed with HHH syndrome. The other three patients were identified by family screening. Citrin deficiency was detected in two patients with cholestasis and hepatomegaly in the infantile period. Ornithine levels increased in three of our patients with HHH syndrome during the first month of treatment despite a protein-restricted diet and adequate caloric intake. CONCLUSIONS: Increasing patients' caloric intake with HHH syndrome improves their ornithine levels. Our patients with citrin deficiency recovered clinically and biochemically before seven months.


Subject(s)
Citrullinemia , Hyperammonemia , Ornithine/deficiency , Urea Cycle Disorders, Inborn , Child , Humans , Retrospective Studies , Mitochondrial Membrane Transport Proteins/genetics , Urea
15.
Nanotoxicology ; 18(2): 122-133, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38436290

ABSTRACT

Food-grade titanium dioxide (E171) and zinc oxide nanoparticles (ZnO NPs) are found in diverse products for human use. E171 is used as whitening agent in food and cosmetics, and ZnO NPs in food packaging. Their potential multi-organ toxicity has raised concerns on their safety. Since mitochondrial dysfunction is a key aspect of cardio-pathologies, here, we evaluate the effect of chronic exposure to E171 and ZnO NPs in rats on cardiac mitochondria. Changes in cardiac electrophysiology and body weight were measured. E171 reduced body weight more than 10% after 5 weeks. Both E171 and ZnO NPs increased systolic blood pressure (SBP) from 110-120 to 120-140 mmHg after 45 days of treatment. Both NPs altered the mitochondrial permeability transition pore (mPTP), reducing calcium requirement for permeability by 60% and 93% in E171- and ZnO NPs-exposed rats, respectively. Treatments also affected conformational state of adenine nucleotide translocase (ANT). E171 reduced the binding of EMA to Cys 159 in 30% and ZnO NPs in 57%. Mitochondrial aconitase activity was reduced by roughly 50% with both NPs, indicating oxidative stress. Transmission electron microscopy (TEM) revealed changes in mitochondrial morphology including sarcomere discontinuity, edema, and hypertrophy in rats exposed to both NPs. In conclusion, chronic oral exposure to NPs induces functional and morphological damage in cardiac mitochondria, with ZnO NPs being more toxic than E171, possibly due to their dissociation in free Zn2+ ion form. Therefore, chronic intake of these food additives could increase risk of cardiovascular disease.


Subject(s)
Mitochondria, Heart , Titanium , Zinc Oxide , Animals , Titanium/toxicity , Zinc Oxide/toxicity , Zinc Oxide/chemistry , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Male , Rats , Administration, Oral , Permeability/drug effects , Mitochondrial Permeability Transition Pore/metabolism , Metal Nanoparticles/toxicity , Nanoparticles/toxicity , Nanoparticles/chemistry , Rats, Sprague-Dawley , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Blood Pressure/drug effects
16.
Methods Mol Biol ; 2778: 201-220, 2024.
Article in English | MEDLINE | ID: mdl-38478280

ABSTRACT

Mitochondrial ß-barrel proteins fulfill crucial roles in the biogenesis and function of the cell organelle. They mediate the import and membrane insertion of proteins and transport of small metabolites and ions. All ß-barrel proteins are made as precursors on cytosolic ribosomes and are imported into mitochondria. The ß-barrel proteins fold and assemble with partner proteins in the outer membrane. The in vitro import of radiolabelled proteins into isolated mitochondria is a powerful tool to investigate the import of ß-barrel proteins, the folding of the ß-barrel proteins, and their assembly into protein complexes. Altogether, the in vitro import assay is a versatile and crucial assay to analyze the mechanisms of the biogenesis of mitochondrial ß-barrel proteins.


Subject(s)
Mitochondrial Proteins , Saccharomyces cerevisiae Proteins , Mitochondrial Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Mitochondria/metabolism , Protein Transport , Mitochondrial Membrane Transport Proteins/metabolism
17.
Methods Mol Biol ; 2778: 221-236, 2024.
Article in English | MEDLINE | ID: mdl-38478281

ABSTRACT

Total interference reflection fluorescence (TIRF) microscopy of lipid bilayers is an effective technique for studying the lateral movement and ion channel activity of single integral membrane proteins. Here we describe how to integrate the mitochondrial outer membrane preprotein translocase TOM-CC and its ß-barrel protein-conducting channel Tom40 into supported lipid bilayers to identify possible relationships between movement and channel activity. We propose that our approach can be readily applied to membrane protein channels where transient tethering to either membrane-proximal or intramembrane structures is accompanied by a change in channel permeation.


Subject(s)
Mitochondrial Proteins , Saccharomyces cerevisiae Proteins , Mitochondrial Proteins/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Lipid Bilayers/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ion Channels/metabolism
18.
JCI Insight ; 9(3)2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38329125

ABSTRACT

Although cold preservation remains the gold standard in organ transplantation, cold stress-induced cellular injury is a significant problem in clinical orthotopic liver transplantation (OLT). Because a recent study showed that cold stress activates ferroptosis, a form of regulated cell death, we investigated whether and how ferroptosis determines OLT outcomes in mice and humans. Treatment with ferroptosis inhibitor (ferrostatin-1) during cold preservation reduced lipid peroxidation (malondialdehyde; MDA), primarily in liver sinusoidal endothelial cells (LSECs), and alleviated ischemia/reperfusion injury in mouse OLT. Similarly, ferrostatin-1 reduced cell death in cold-stressed LSEC cultures. LSECs deficient in nuclear factor erythroid 2-related factor 2 (NRF2), a critical regulator of ferroptosis, were susceptible to cold stress-induced cell death, concomitant with enhanced endoplasmic reticulum (ER) stress and expression of mitochondrial Ca2+ uptake regulator (MICU1). Indeed, supplementing MICU1 inhibitor reduced ER stress, MDA expression, and cell death in NRF2-deficient but not WT LSECs, suggesting NRF2 is a critical regulator of MICU1-mediated ferroptosis. Consistent with murine data, enhanced liver NRF2 expression reduced MDA levels, hepatocellular damage, and incidence of early allograft dysfunction in human OLT recipients. This translational study provides a clinically applicable strategy in which inhibition of ferroptosis during liver cold preservation mitigates OLT injury by protecting LSECs from peritransplant stress via an NRF2-regulatory mechanism.


Subject(s)
Cyclohexylamines , Ferroptosis , Liver Transplantation , Phenylenediamines , Mice , Humans , Animals , Liver Transplantation/adverse effects , Endothelial Cells/metabolism , NF-E2-Related Factor 2/metabolism , Cold-Shock Response , Liver/metabolism , Calcium-Binding Proteins/metabolism , Mitochondrial Membrane Transport Proteins/metabolism
19.
Cell Rep ; 43(3): 113805, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38377000

ABSTRACT

The majority of mitochondrial precursor proteins are imported through the Tom40 ß-barrel channel of the translocase of the outer membrane (TOM). The sorting and assembly machinery (SAM) is essential for ß-barrel membrane protein insertion into the outer membrane and thus required for the assembly of the TOM complex. Here, we demonstrate that the α-helical outer membrane protein Mco6 co-assembles with the mitochondrial distribution and morphology protein Mdm10 as part of the SAM machinery. MCO6 and MDM10 display a negative genetic interaction, and a mco6-mdm10 yeast double mutant displays reduced levels of the TOM complex. Cells lacking Mco6 affect the levels of Mdm10 and show assembly defects of the TOM complex. Thus, this work uncovers a role of the SAMMco6 complex for the biogenesis of the mitochondrial outer membrane.


Subject(s)
Membrane Transport Proteins , Saccharomyces cerevisiae Proteins , Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Carrier Proteins/metabolism , Protein Transport
20.
Mol Cell ; 84(4): 616-618, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38364779

ABSTRACT

Two recent studies by Liu et al.1 in Science and Shi et al.2 in this issue of Molecular Cell identify a mitochondrial GSH-sensing mechanism that couples SLC25A39-mediated GSH import to iron metabolism, advancing our understanding of nutrient sensing within organelles.


Subject(s)
Iron , Mitochondria , Mitochondrial Membrane Transport Proteins , Glutathione/metabolism , Iron/metabolism , Mitochondria/genetics , Mitochondria/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...