Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Metab ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38703762

ABSTRACT

The mitochondrial genome transcribes 13 mRNAs coding for well-known proteins essential for oxidative phosphorylation. We demonstrate here that cytochrome b (CYTB), the only mitochondrial-DNA-encoded transcript among complex III, also encodes an unrecognized 187-amino-acid-long protein, CYTB-187AA, using the standard genetic code of cytosolic ribosomes rather than the mitochondrial genetic code. After validating the existence of this mtDNA-encoded protein arising from cytosolic translation (mPACT) using mass spectrometry and antibodies, we show that CYTB-187AA is mainly localized in the mitochondrial matrix and promotes the pluripotent state in primed-to-naive transition by interacting with solute carrier family 25 member 3 (SLC25A3) to modulate ATP production. We further generated a transgenic knockin mouse model of CYTB-187AA silencing and found that reduction of CYTB-187AA impairs females' fertility by decreasing the number of ovarian follicles. For the first time, we uncovered the novel mPACT pattern of a mitochondrial mRNA and demonstrated the physiological function of this 14th protein encoded by mtDNA.

2.
Biochem Soc Trans ; 50(6): 1773-1783, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36484629

ABSTRACT

Quality control of mitochondria is essential for their homeostasis and function. Light chain 3 (LC3) associated autophagosomes-mediated mitophagy represents a canonical mitochondrial quality control pathway. Alternative quality control processes, such as mitochondrial-derived vesicles (MDVs), have been discovered, but the intact mitochondrial quality control remains unknown. We recently discovered a novel mitolysosome exocytosis mechanism for mitochondrial quality control in flunarizine (FNZ)-induced mitochondria clearance, where autophagosomes are not required, but rather mitochondria are engulfed directly by lysosomes, mediating mitochondrial secretion. As FNZ results in parkinsonism, we propose that excessive mitolysosome exocytosis is the cause.


Subject(s)
Mitophagy , Parkinsonian Disorders , Humans , Mitochondria/metabolism , Autophagosomes/metabolism , Lysosomes/metabolism , Exocytosis , Parkinsonian Disorders/metabolism , Autophagy
3.
Sci Adv ; 8(15): eabk2376, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35417232

ABSTRACT

Mitochondrial quality control plays an important role in maintaining mitochondrial homeostasis and function. Disruption of mitochondrial quality control degrades brain function. We found that flunarizine (FNZ), a drug whose chronic use causes parkinsonism, led to a parkinsonism-like motor dysfunction in mice. FNZ induced mitochondrial dysfunction and decreased mitochondrial mass specifically in the brain. FNZ decreased mitochondrial content in both neurons and astrocytes, without affecting the number of nigral dopaminergic neurons. In human neural progenitor cells, FNZ also induced mitochondrial depletion. Mechanistically, independent of ATG5- or RAB9-mediated mitophagy, mitochondria were engulfed by lysosomes, followed by a vesicle-associated membrane protein 2- and syntaxin-4-dependent extracellular secretion. A genome-wide CRISPR knockout screen identified genes required for FNZ-induced mitochondrial elimination. These results reveal not only a previously unidentified lysosome-associated exocytosis process of mitochondrial quality control that may participate in the FNZ-induced parkinsonism but also a drug-based method for generating mitochondria-depleted mammal cells.

4.
Adv Sci (Weinh) ; 8(10): 2004680, 2021 05.
Article in English | MEDLINE | ID: mdl-34026460

ABSTRACT

Mitochondrial DNA depletion syndrome (MDS) is a group of severe inherited disorders caused by mutations in genes, such as deoxyribonucleoside kinase (DGUOK). A great majority of DGUOK mutant MDS patients develop iron overload progressing to severe liver failure. However, the pathological mechanisms connecting iron overload and hepatic damage remains uncovered. Here, two patients' skin fibroblasts are reprogrammed to induced pluripotent stem cells (iPSCs) and then corrected by CRISPR/Cas9. Patient-specific iPSCs and corrected iPSCs-derived high purity hepatocyte organoids (iHep-Orgs) and hepatocyte-like cells (iHep) are generated as cellular models for studying hepatic pathology. DGUOK mutant iHep and iHep-Orgs, but not control and corrected one, are more sensitive to iron overload-induced ferroptosis, which can be rescued by N-Acetylcysteine (NAC). Mechanically, this ferroptosis is a process mediated by nuclear receptor co-activator 4 (NCOA4)-dependent degradation of ferritin in lysosome and cellular labile iron release. This study reveals the underlying pathological mechanisms and the viable therapeutic strategies of this syndrome, and is the first pure iHep-Orgs model in hereditary liver diseases.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Liver Failure/pathology , Mitochondrial Diseases/pathology , Mutation , Organoids/pathology , Respiration Disorders/pathology , DNA, Mitochondrial/genetics , Ferritins/metabolism , Ferroptosis , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Iron Overload/physiopathology , Liver/metabolism , Liver/pathology , Liver Failure/genetics , Liver Failure/metabolism , Lysosomes/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Nuclear Receptor Coactivators/genetics , Nuclear Receptor Coactivators/metabolism , Organoids/metabolism , Respiration Disorders/etiology , Respiration Disorders/metabolism
5.
Autophagy ; 16(3): 562-574, 2020 03.
Article in English | MEDLINE | ID: mdl-31234709

ABSTRACT

Selective elimination of mitochondria by autophagy is a critical strategy for a variety of physiological processes, including development, cell-fate determination and stress response. Although several mechanisms have been identified as responsible for selective degradation of mitochondria, such as the PINK1-PRKN/PARKIN- and receptor-dependent pathways, aspects of the mechanisms and particularly the principles underlying the selection process of mitochondria remain obscure. Here, we addressed a new selection strategy in which the selective elimination of mitochondria is dependent on organellar topology. We found that populations of mitochondria undergo different topological transformations under serum starvation, either swelling or forming donut shapes. Swollen mitochondria are associated with mitochondrial membrane potential dissipation and PRKN recruitment, which promote their selective elimination, while the donut topology maintains mitochondrial membrane potential and helps mitochondria resist autophagy. Mechanistic studies show that donuts resist autophagy even after depolarization through preventing recruitment of autophagosome receptors CALCOCO2/NDP52 and OPTN even after PRKN recruitment. Our results demonstrate topology-dependent, bifurcated mitochondrial recycling under starvation, that is swollen mitochondria undergo removal by autophagy, while donut mitochondria undergo fission and fusion cycles for reintegration. This study reveals a novel morphological selection for control of mitochondrial quality and quantity under starvation.


Subject(s)
Mitochondria/metabolism , Animals , Autophagy/drug effects , Autophagy-Related Protein 5/metabolism , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Culture Media, Serum-Free , Humans , Membrane Potential, Mitochondrial/drug effects , Membrane Transport Proteins/metabolism , Mice , Mitochondria/ultrastructure , Mitophagy/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
6.
Brain ; 142(8): 2215-2229, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31199454

ABSTRACT

Charcot-Marie-Tooth disease is a hereditary motor and sensory neuropathy exhibiting great clinical and genetic heterogeneity. Here, the identification of two heterozygous missense mutations in the C1orf194 gene at 1p21.2-p13.2 with Charcot-Marie-Tooth disease are reported. Specifically, the p.I122N mutation was the cause of an intermediate form of Charcot-Marie-Tooth disease, and the p.K28I missense mutation predominately led to the demyelinating form. Functional studies demonstrated that the p.K28I variant significantly reduced expression of the protein, but the p.I122N variant increased. In addition, the p.I122N mutant protein exhibited the aggregation in neuroblastoma cell lines and the patient's peroneal nerve. Either gain-of-function or partial loss-of-function mutations to C1ORF194 can specify different causal mechanisms responsible for Charcot-Marie-Tooth disease with a wide range of clinical severity. Moreover, a knock-in mouse model confirmed that the C1orf194 missense mutation p.I121N led to impairments in motor and neuromuscular functions, and aberrant myelination and axonal phenotypes. The loss of normal C1ORF194 protein altered intracellular Ca2+ homeostasis and upregulated Ca2+ handling regulatory proteins. These findings describe a novel protein with vital functions in peripheral nervous systems and broaden the causes of Charcot-Marie-Tooth disease, which open new avenues for the diagnosis and treatment of related neuropathies.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Animals , Calcium/metabolism , Gene Knock-In Techniques , Humans , Mice , Mice, Transgenic , Mutation, Missense , Pedigree
8.
Cell Death Dis ; 9(10): 966, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30237514

ABSTRACT

Hexadimethrine bromide (Polybrene) was once used clinically as a heparin neutralizer and has recently found use as a promoter in virus-mediated gene therapy trials and gene transfer in research. However, the potential for tissue-specific toxicity of polybrene at low doses has been ignored so far. Here, we found that after intracerebroventricular (ICV) polybrene injection, mice showed disability of movement accompanied neural death and gliosis in brain, and in human neurons, polybrene induces concentration-dependent neuritic beading and fragmentation. Mechanistically, polybrene induces a rapid voltage-dependent calcium channel (VDCC)-mediated influx of extracellular Ca2+. The elevated cytoplasmic Ca2+ activates DRP1, which leads to mitochondrial fragmentation and metabolic dysfunction. At the same time, Ca2+ influx induces endoplasmic reticulum (ER) fragmentation and tightened associations between ER and mitochondria, which makes mitochondria prone to Ca2+ overloading and ensuing permeability transition. These results reveal an unexpected neuronal toxicity of polybrene, wherein Ca2+ influx serves as a regulator for both mitochondrial dynamics and ER-mitochondrial remodeling.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Hexadimethrine Bromide/toxicity , Mitochondria/metabolism , Nerve Degeneration/chemically induced , Neurons/cytology , Neurons/drug effects , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred BALB C , Mitochondrial Dynamics , Reactive Oxygen Species/metabolism
9.
Cell Metab ; 28(6): 935-945.e5, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30174306

ABSTRACT

Reprogramming of somatic cells to induced pluripotent stem cells reconfigures chromatin modifications. Whether and how this process is regulated by signals originating in the mitochondria remain unknown. Here we show that the mitochondrial permeability transition pore (mPTP), a key regulator of mitochondrial homeostasis, undergoes short-term opening during the early phase of reprogramming and that this transient activation enhances reprogramming. In mouse embryonic fibroblasts, greater mPTP opening correlates with higher reprogramming efficiency. The reprogramming-promoting function of mPTP opening is mediated by plant homeodomain finger protein 8 (PHF8) demethylation of H3K9me2 and H3K27me3, leading to reduction in their occupancies at the promoter regions of pluripotency genes. mPTP opening increases PHF8 protein levels downstream of mitochondrial reactive oxygen species production and miR-101c and simultaneously elevates levels of PHF8's cofactor, α-ketoglutarate. Our findings represent a novel mitochondria-to-nucleus pathway in cell fate determination by mPTP-mediated epigenetic regulation.


Subject(s)
Cellular Reprogramming , Fibroblasts/metabolism , Histone Demethylases/metabolism , Histones/metabolism , Lysine/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Membranes/metabolism , Mouse Embryonic Stem Cells/metabolism , Transcription Factors/metabolism , Animals , HEK293 Cells , Humans , Ketoglutaric Acids/metabolism , Methylation , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Mitochondrial Permeability Transition Pore , Reactive Oxygen Species/metabolism
10.
Autophagy ; 13(9): 1543-1555, 2017 Sep 02.
Article in English | MEDLINE | ID: mdl-28722510

ABSTRACT

Induced pluripotent stem cells (iPSCs) have fewer and immature mitochondria than somatic cells and mainly rely on glycolysis for energy source. During somatic cell reprogramming, somatic mitochondria and other organelles get remodeled. However, events of organelle remodeling and interaction during somatic cell reprogramming have not been extensively explored. We show that both SKP/SKO (Sox2, Klf4, Pou5f1/Oct4) and SKPM/SKOM (SKP/SKO plus Myc/c-Myc) reprogramming lead to decreased mitochondrial mass but with different kinetics and by divergent pathways. Rapid, MYC/c-MYC-induced cell proliferation may function as the main driver of mitochondrial decrease in SKPM/SKOM reprogramming. In SKP/SKO reprogramming, however, mitochondrial mass initially increases and subsequently decreases via mitophagy. This mitophagy is dependent on the mitochondrial outer membrane receptor BNIP3L/NIX but not on mitochondrial membrane potential (ΔΨm) dissipation, and this SKP/SKO-induced mitophagy functions in an important role during the reprogramming process. Furthermore, endosome-related RAB5 is involved in mitophagosome formation in SKP/SKO reprogramming. These results reveal a novel role of mitophagy in reprogramming that entails the interaction between mitochondria, macroautophagy/autophagy and endosomes.


Subject(s)
Cellular Reprogramming , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitophagy , Animals , Embryo, Mammalian/cytology , Endosomes/metabolism , Endosomes/ultrastructure , Fibroblasts/metabolism , Kruppel-Like Factor 4 , Membrane Potential, Mitochondrial , Mice , Mitochondria/ultrastructure , Models, Biological , Transcription Factors/metabolism , rab5 GTP-Binding Proteins/metabolism
11.
CNS Neurosci Ther ; 22(8): 648-60, 2016 08.
Article in English | MEDLINE | ID: mdl-27080255

ABSTRACT

BACKGROUND: Neuritic degeneration is an important early pathological step in neurodegeneration. AIM: The purpose of this study was to explore the mechanisms connecting neuritic degeneration to the functional and morphological remodeling of endoplasmic reticulum (ER) and mitochondria. METHODS: Here, we set up neuritic degeneration models by neurite cutting-induced neural degeneration in human-induced pluripotent stem cell-derived neurons. RESULTS: We found that neuritic ER becomes fragmented and forms complexes with mitochondria, which induces IP3R-dependent mitochondrial Ca(2+) elevation and dysfunction during neuritic degeneration. Furthermore, mitochondrial membrane potential is required for ER fragmentation and mitochondrial Ca(2+) elevation during neuritic degeneration. Mechanically, tightening of the ER-mitochondria associations by expression of a short "synthetic linker" and ER Ca(2+) releasing together could promote mitochondrial Ca(2+) elevation, dysfunction, and reactive oxygen species generation. CONCLUSION: Our study reveals a dynamic remodeling of the ER-mitochondria interface underlying neuritic degeneration.


Subject(s)
Endoplasmic Reticulum/physiology , Membrane Potential, Mitochondrial/physiology , Nerve Degeneration/physiopathology , Neurites/ultrastructure , Neurons/ultrastructure , Apoptosis , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Fetus , HEK293 Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Neurites/physiology , Neurons/drug effects , Oligodeoxyribonucleotides/pharmacology , Pluripotent Stem Cells/drug effects , Proton Ionophores/pharmacology , Reactive Oxygen Species
12.
Cell Metab ; 23(1): 220-6, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26549484

ABSTRACT

The mechanisms of somatic cell reprogramming have been revealed at multiple levels. However, the lack of tools to monitor different reactive oxygen species (ROS) has left their distinct signals and roles in reprogramming unknown. We hypothesized that mitochondrial flashes (mitoflashes), recently identified spontaneous bursts of mitochondrial superoxide signaling, play a role in reprogramming. Here we show that the frequency of mitoflashes transiently increases, accompanied by flash amplitude reduction, during the early stages of reprogramming. This transient activation of mitoflashes at the early stage enhances reprogramming, whereas sustained activation impairs reprogramming. The reprogramming-promoting function of mitoflashes occurs via the upregulation of Nanog expression that is associated with decreases in the methylation status of the Nanog promoter through Tet2 occupancy. Together our findings provide a previously unknown role for superoxide signaling mediated epigenetic regulation in cell fate determination.


Subject(s)
Cellular Reprogramming , Homeodomain Proteins/metabolism , Mitochondria/physiology , Animals , Cells, Cultured , DNA Methylation , DNA-Binding Proteins/metabolism , Dioxygenases , Epigenesis, Genetic , Fibroblasts/physiology , Homeodomain Proteins/genetics , Humans , Mice , Nanog Homeobox Protein , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/metabolism , Signal Transduction , Superoxides/metabolism , Up-Regulation
13.
Cell Mol Life Sci ; 72(13): 2585-98, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25708700

ABSTRACT

Heteroplasmic cells, harboring both mutant and normal mitochondrial DNAs (mtDNAs), must accumulate mutations to a threshold level before respiratory activity is affected. This phenomenon has led to the hypothesis of mtDNA complementation by inter-mitochondrial content mixing. The precise mechanisms of heteroplasmic complementation are unknown, but it depends both on the mtDNA nucleoid dynamics among mitochondria as well as the mitochondrial dynamics as influenced by mtDNA. We tracked nucleoids among the mitochondria in real time to show that they are shared after complete fusion but not 'kiss-and-run'. Employing a cell hybrid model, we further show that mtDNA-less mitochondria, which have little ATP production and extensive Opa1 proteolytic cleavage, exhibit weak fusion activity among themselves, yet remain competent in fusing with healthy mitochondria in a mitofusin- and OPA1-dependent manner, resulting in restoration of metabolic function. Depletion of mtDNA by overexpression of the matrix-targeted nuclease UL12.5 resulted in heterogeneous mitochondrial membrane potential (ΔΨm) at the organelle level in mitofusin-null cells but not in wild type. In this system, overexpression of mitofusins or application of the fusion-promoting drug M1 could partially rescue the metabolic damage caused by UL12.5. Interestingly, mtDNA transcription/translation is not required for normal mitochondria to restore metabolic function to mtDNA-less mitochondria by fusion. Thus, interplay between mtDNA and fusion capacity governs a novel 'initial metabolic complementation'.


Subject(s)
DNA, Mitochondrial/metabolism , Hybrid Cells/physiology , Mitochondrial Dynamics/physiology , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Cell Fusion/methods , DNA Primers/genetics , DNA, Mitochondrial/genetics , Fluorescent Antibody Technique , GTP Phosphohydrolases/metabolism , Genetic Vectors/genetics , HeLa Cells , Humans , Mice , Microscopy, Confocal , Mitochondrial Dynamics/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...