Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters











Publication year range
1.
EMBO J ; 43(3): 339-361, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38238476

ABSTRACT

Hematopoietic stem cell (HSC) divisional fate and function are determined by cellular metabolism, yet the contribution of specific cellular organelles and metabolic pathways to blood maintenance and stress-induced responses in the bone marrow remains poorly understood. The outer mitochondrial membrane-localized E3 ubiquitin ligase MITOL/MARCHF5 (encoded by the Mitol gene) is known to regulate mitochondrial and endoplasmic reticulum (ER) interaction and to promote cell survival. Here, we investigated the functional involvement of MITOL in HSC maintenance by generating MX1-cre inducible Mitol knockout mice. MITOL deletion in the bone marrow resulted in HSC exhaustion and impairment of bone marrow reconstitution capability in vivo. Interestingly, MITOL loss did not induce major mitochondrial dysfunction in hematopoietic stem and progenitor cells. In contrast, MITOL deletion induced prolonged ER stress in HSCs, which triggered cellular apoptosis regulated by IRE1α. In line, dampening of ER stress signaling by IRE1α inihibitor KIRA6 partially rescued apoptosis of long-term-reconstituting HSC. In summary, our observations indicate that MITOL is a principal regulator of hematopoietic homeostasis and protects blood stem cells from cell death through its function in ER stress signaling.


Subject(s)
Endoribonucleases , Protein Serine-Threonine Kinases , Animals , Mice , Apoptosis , Hematopoietic Stem Cells/metabolism , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
2.
Genes (Basel) ; 14(10)2023 09 26.
Article in English | MEDLINE | ID: mdl-37895224

ABSTRACT

Mitochondrial dynamics, including fission and fusion processes, are essential for heart health. Mitochondria, the powerhouses of cells, maintain their integrity through continuous cycles of biogenesis, fission, fusion, and degradation. Mitochondria are relatively immobile in the adult heart, but their morphological changes due to mitochondrial morphology factors are critical for cellular functions such as energy production, organelle integrity, and stress response. Mitochondrial fusion proteins, particularly Mfn1/2 and Opa1, play multiple roles beyond their pro-fusion effects, such as endoplasmic reticulum tethering, mitophagy, cristae remodeling, and apoptosis regulation. On the other hand, the fission process, regulated by proteins such as Drp1, Fis1, Mff and MiD49/51, is essential to eliminate damaged mitochondria via mitophagy and to ensure proper cell division. In the cardiac system, dysregulation of mitochondrial dynamics has been shown to cause cardiac hypertrophy, heart failure, ischemia/reperfusion injury, and various cardiac diseases, including metabolic and inherited cardiomyopathies. In addition, mitochondrial dysfunction associated with oxidative stress has been implicated in atherosclerosis, hypertension and pulmonary hypertension. Therefore, understanding and regulating mitochondrial dynamics is a promising therapeutic tool in cardiac diseases. This review summarizes the role of mitochondrial morphology in heart diseases for each mitochondrial morphology regulatory gene, and their potential as therapeutic targets to heart diseases.


Subject(s)
Heart Diseases , Heart Failure , Humans , Mitochondrial Dynamics/genetics , Heart Diseases/genetics , Mitochondria/genetics , Mitochondria/metabolism , Apoptosis , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
3.
J Biochem ; 173(1): 1-11, 2022 Dec 27.
Article in English | MEDLINE | ID: mdl-36346121

ABSTRACT

Mitochondria are involved in various cellular processes, such as energy production, inflammatory responses and cell death. Mitochondrial dysfunction is associated with many age-related diseases, including neurological disorders and heart failure. Mitochondrial quality is strictly maintained by mitochondrial dynamics linked to an adequate supply of phospholipids and other substances from the endoplasmic reticulum (ER). The outer mitochondrial membrane-localized E3 ubiquitin ligase MITOL/MARCHF5 is responsible for mitochondrial quality control through the regulation of mitochondrial dynamics, formation of mitochondria-ER contacts and mitophagy. MITOL deficiency has been shown to impair mitochondrial function, cause an excessive inflammatory response and increase vulnerability to stress, resulting in the exacerbation of the disease. In this study, we overview the ubiquitin-mediated regulation of mitochondrial function by MITOL and the relationship between MITOL and diseases.


Subject(s)
Ubiquitin-Protein Ligases , Ubiquitin , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Endoplasmic Reticulum/metabolism , Cell Death , Mitochondria/metabolism , Mitochondrial Proteins/metabolism
4.
iScience ; 25(7): 104582, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35789860

ABSTRACT

Abnormal mitochondrial fragmentation by dynamin-related protein1 (Drp1) is associated with the progression of aging-associated heart diseases, including heart failure and myocardial infarction (MI). Here, we report a protective role of outer mitochondrial membrane (OMM)-localized E3 ubiquitin ligase MITOL/MARCH5 against cardiac senescence and MI, partly through Drp1 clearance by OMM-associated degradation (OMMAD). Persistent Drp1 accumulation in cardiomyocyte-specific MITOL conditional-knockout mice induced mitochondrial fragmentation and dysfunction, including reduced ATP production and increased ROS generation, ultimately leading to myocardial senescence and chronic heart failure. Furthermore, ischemic stress-induced acute downregulation of MITOL, which permitted mitochondrial accumulation of Drp1, resulted in mitochondrial fragmentation. Adeno-associated virus-mediated delivery of the MITOL gene to cardiomyocytes ameliorated cardiac dysfunction induced by MI. Our findings suggest that OMMAD activation by MITOL can be a therapeutic target for aging-associated heart diseases, including heart failure and MI.

5.
Cancer Discov ; 12(1): 250-269, 2022 01.
Article in English | MEDLINE | ID: mdl-34462274

ABSTRACT

Ineffective hematopoiesis is a fundamental process leading to the pathogenesis of myelodysplastic syndromes (MDS). However, the pathobiological mediators of ineffective hematopoiesis in MDS remain unclear. Here, we demonstrated that overwhelming mitochondrial fragmentation in mutant hematopoietic stem cells and progenitors (HSC/P) triggers ineffective hematopoiesis in MDS. Mouse modeling of CBL exon deletion with RUNX1 mutants, previously unreported comutations in patients with MDS, recapitulated not only clinically relevant MDS phenotypes but also a distinct MDS-related gene signature. Mechanistically, dynamin-related protein 1 (DRP1)-dependent excessive mitochondrial fragmentation in HSC/Ps led to excessive reactive oxygen species production, induced inflammatory signaling activation, and promoted subsequent dysplasia formation and impairment of granulopoiesis. Mitochondrial fragmentation was generally observed in patients with MDS. Pharmacologic inhibition of DRP1 attenuated mitochondrial fragmentation and rescued ineffective hematopoiesis phenotypes in mice with MDS. These findings provide mechanistic insights into ineffective hematopoiesis and indicate that dysregulated mitochondrial dynamics could be a therapeutic target for bone marrow failure in MDS. SIGNIFICANCE: We demonstrated that excessive mitochondrial fragmentation is a fundamental pathobiological phenomenon that could trigger dysplasia formation and ineffective hematopoiesis in MDS. Our findings provide mechanistic insights into ineffective hematopoiesis and suggest dysregulated mitochondrial dynamics as a therapeutic target for treating MDS.This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Hematopoietic Stem Cells/pathology , Myelodysplastic Syndromes/pathology , Animals , Disease Models, Animal , Female , Hematopoiesis , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
6.
J Biochem ; 171(5): 529-541, 2022 May 11.
Article in English | MEDLINE | ID: mdl-34964862

ABSTRACT

The transfer of phospholipids from the endoplasmic reticulum (ER) to mitochondria via the mitochondria-ER contact site (MERCS) is essential for maintaining mitochondrial function and integrity. Here, we identified RMDN3/PTPIP51, possessing phosphatidic acid (PA)-transfer activity, as a neighbouring protein of the mitochondrial E3 ubiquitin ligase MITOL/MARCH5 by proximity-dependent biotin labelling using APEX2. We found that MITOL interacts with and ubiquitinates RMDN3. Mutational analysis identified lysine residue 89 in RMDN3 as a site of ubiquitination by MITOL. Loss of MITOL or the substitution of lysine 89 to arginine in RMDN3 significantly reduced the PA-binding activity of RMDN3, suggesting that MITOL regulates the transport of PA to mitochondria by activating RMDN3. Our findings imply that ubiquitin signalling regulates phospholipid transport at the MERCS.


Subject(s)
Lysine , Mitochondrial Proteins , Endoplasmic Reticulum/metabolism , Lysine/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Phosphatidic Acids/metabolism , Ubiquitin-Protein Ligases/metabolism
7.
Mol Ther Oncolytics ; 22: 265-276, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34553018

ABSTRACT

Most oncolytic virotherapy has thus far employed viruses deficient in genes essential for replication in normal cells but not in cancer cells. Intra-tumoral injection of such viruses has resulted in clinically significant anti-tumor effects on the lesions in the vicinity of the injection sites but not on distant visceral metastases. To overcome this limitation, we have developed a receptor-retargeted oncolytic herpes simplex virus employing a single-chain antibody for targeting tumor-associated antigens (RR-oHSV) and its modified version with additional mutations conferring syncytium formation (RRsyn-oHSV). We previously showed that RRsyn-oHSV exhibits preserved antigen specificity and an ∼20-fold higher tumoricidal potency in vitro relative to RR-oHSV. Here, we investigated the in vivo anti-tumor effects of RRsyn-oHSV using human cancer xenografts in immunodeficient mice. With only a single intra-tumoral injection of RRsyn-oHSV at very low doses, all treated tumors regressed completely. Furthermore, intra-venous administration of RRsyn-oHSV resulted in robust anti-tumor effects even against large tumors. We found that these potent anti-tumor effects of RRsyn-oHSV may be associated with the formation of long-lasting tumor cell syncytia not containing non-cancerous cells that appear to trigger death of the syncytia. These results strongly suggest that cancer patients with distant metastases could be effectively treated with our RRsyn-oHSV.

8.
J Mol Cell Cardiol ; 161: 116-129, 2021 12.
Article in English | MEDLINE | ID: mdl-34390730

ABSTRACT

MITOL/MARCH5 is an E3 ubiquitin ligase that plays a crucial role in the control of mitochondrial quality and function. However, the significance of MITOL in cardiomyocytes under physiological and pathological conditions remains unclear. First, to determine the significance of MITOL in unstressed hearts, we assessed the cellular changes with the reduction of MITOL expression by siRNA in neonatal rat primary ventricular cardiomyocytes (NRVMs). MITOL knockdown in NRVMs induced cell death via ferroptosis, a newly defined non-apoptotic programmed cell death, even under no stress conditions. This phenomenon was observed only in NRVMs, not in other cell types. MITOL knockdown markedly reduced mitochondria-localized GPX4, a key enzyme associated with ferroptosis, promoting accumulation of lipid peroxides in mitochondria. In contrast, the activation of GPX4 in MITOL knockdown cells suppressed lipid peroxidation and cell death. MITOL knockdown reduced the glutathione/oxidized glutathione (GSH/GSSG) ratio that regulated GPX4 expression. Indeed, the administration of GSH or N-acetylcysteine improved the expression of GPX4 and viability in MITOL-knockdown NRVMs. MITOL-knockdown increased the expression of the glutathione-degrading enzyme, ChaC glutathione-specific γ-glutamylcyclotransferase 1 (Chac1). The knockdown of Chac1 restored the GSH/GSSG ratio, GPX4 expression, and viability in MITOL-knockdown NRVMs. Further, in cultured cardiomyocytes stressed with DOX, both MITOL and GPX4 were reduced, whereas forced-expression of MITOL suppressed DOX-induced ferroptosis by maintaining GPX4 content. Additionally, MITOL knockdown worsened vulnerability to DOX, which was almost completely rescued by treatment with ferrostatin-1, a ferroptosis inhibitor. In vivo, cardiac-specific depletion of MITOL did not produce obvious abnormality, but enhanced susceptibility to DOX toxicity. Finally, administration of ferrostatin-1 suppressed exacerbation of DOX-induced myocardial damage in MITOL-knockout hearts. The present study demonstrates that MITOL determines the cell fate of cardiomyocytes via the ferroptosis process and plays a key role in regulating vulnerability to DOX treatment. (288/300).


Subject(s)
Cardiomyopathies/chemically induced , Doxorubicin/pharmacology , Glutathione/metabolism , Mitochondrial Proteins/metabolism , Myocytes, Cardiac/drug effects , Ubiquitin-Protein Ligases/metabolism , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Death/drug effects , Cells, Cultured , Doxorubicin/adverse effects , Ferroptosis/drug effects , HEK293 Cells , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , Myocytes, Cardiac/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Rats , Ubiquitin-Protein Ligases/genetics , gamma-Glutamylcyclotransferase/genetics , gamma-Glutamylcyclotransferase/metabolism
9.
J Biol Chem ; 297(2): 100986, 2021 08.
Article in English | MEDLINE | ID: mdl-34298015

ABSTRACT

Radial migration during cortical development is required for formation of the six-layered structure of the mammalian cortex. Defective migration of neurons is linked to several developmental disorders such as autism and schizophrenia. A unique swollen structure called the dilation is formed in migrating neurons and is required for movement of the centrosome and nucleus. However, the detailed molecular mechanism by which this dilation forms is unclear. We report that CAMDI, a gene whose deletion is associated with psychiatric behavior, is degraded by cell division cycle protein 20 (Cdc20)-anaphase-promoting complex/cyclosome (APC/C) cell-cycle machinery after centrosome migration into the dilation in mouse brain development. We also show that CAMDI is restabilized in the dilation until the centrosome enters the dilation, at which point it is once again immediately destabilized. CAMDI degradation is carried out by binding to Cdc20-APC/C via the destruction box degron of CAMDI. CAMDI destruction box mutant overexpression inhibits dilation formation and neuronal cell migration via maintaining the stabilized state of CAMDI. These results indicate that CAMDI is a substrate of the Cdc20-APC/C system and that the oscillatory regulation of CAMDI protein correlates with dilation formation for proper cortical migration.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Brain/growth & development , Cdc20 Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Movement , Mental Disorders/pathology , Nerve Tissue Proteins/metabolism , Neurons/pathology , Animals , Brain/metabolism , Brain/pathology , Cells, Cultured , Centrosome/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Female , Gene Knockdown Techniques/methods , Humans , Mental Disorders/genetics , Mental Disorders/metabolism , Mice , Models, Animal , Neurons/metabolism
10.
Genes Cells ; 26(7): 513-529, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33971063

ABSTRACT

The lysine methyltransferase SETDB1, an enzyme responsible for methylation of histone H3 at lysine 9, plays a key role in H3K9 tri-methylation-dependent silencing of endogenous retroviruses and developmental genes. Recent studies have shown that ubiquitination of human SETDB1 complements its catalytic activity and the silencing of endogenous retroviruses in human embryonic stem cells. However, it is not known whether SETDB1 ubiquitination is essential for its other major role in epigenetic silencing of developmental gene programs. We previously showed that SETDB1 contributes to the formation of H3K4/H3K9me3 bivalent chromatin domains that keep adipogenic Cebpa and Pparg genes in a poised state for activation and restricts the differentiation potential of pre-adipocytes. Here, we show that ubiquitin-resistant K885A mutant of SETDB1 represses adipogenic genes and inhibits pre-adipocyte differentiation similar to wild-type SETDB1. We show this was due to a compensation mechanism for H3K9me3 chromatin modifications on the Cebpa locus by other H3K9 methyltransferases Suv39H1 and Suv39H2. In contrast, the K885A mutant did not repress other SETDB1 target genes such as Tril and Gas6 suggesting SETDB1 represses its target genes by two mechanisms; one that requires its ubiquitination and another that still requires SETDB1 but not its enzyme activity.


Subject(s)
Adipogenesis , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Ubiquitination , 3T3-L1 Cells , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , HEK293 Cells , Histone Code , Histone-Lysine N-Methyltransferase/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mutation, Missense
11.
J Biol Chem ; 296: 100620, 2021.
Article in English | MEDLINE | ID: mdl-33811862

ABSTRACT

Mouse models of various neuropsychiatric disorders, such as schizophrenia, often display an immature dentate gyrus, characterized by increased numbers of immature neurons and neuronal progenitors and a dearth of mature neurons. We previously demonstrated that the CRMP5-associated GTPase (CRAG), a short splice variant of Centaurin-γ3/AGAP3, is highly expressed in the dentate gyrus. CRAG promotes cell survival and antioxidant defense by inducing the activation of serum response factors at promyelocytic leukemia protein bodies, which are nuclear stress-responsive domains, during neuronal development. However, the physiological role of CRAG in neuronal development remains unknown. Here, we analyzed the role of CRAG using dorsal forebrain-specific CRAG/Centaurin-γ3 knockout mice. The mice revealed maturational abnormality of the hippocampal granule cells, including increased doublecortin-positive immature neurons and decreased calbindin-positive mature neurons, a typical phenotype of immature dentate gyri. Furthermore, the mice displayed hyperactivity in the open-field test, a common measure of exploratory behavior, suggesting that these mice may serve as a novel model for neuropsychiatric disorder associated with hyperactivity. Thus, we conclude that CRAG is required for the maturation of neurons in the dentate gyrus, raising the possibility that its deficiency might promote the development of psychiatric disorders in humans.


Subject(s)
Dentate Gyrus/pathology , GTP Phosphohydrolases/physiology , Neural Stem Cells/pathology , Neurogenesis , Neurons/pathology , Prosencephalon/pathology , Psychomotor Agitation/pathology , Animals , Dentate Gyrus/metabolism , Exploratory Behavior , Female , Male , Mice , Mice, Knockout , Neural Stem Cells/metabolism , Neurons/metabolism , Prosencephalon/metabolism , Psychomotor Agitation/etiology , Psychomotor Agitation/metabolism
12.
Biochem Biophys Res Commun ; 549: 67-74, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33667711

ABSTRACT

Amyloid-ß (Aß) plaques are strongly associated with the development of Alzheimer's disease (AD). However, it remains unclear how morphological differences in Aß plaques determine the pathogenesis of Aß. Here, we categorized Aß plaques into four types based on the macroscopic features of the dense core, and found that the Aß-plaque subtype containing a larger dense core showed the strongest association with neuritic dystrophy. Astrocytes dominantly accumulated toward these expanded/dense-core-containing Aß plaques. Previously, we indicated that deletion of the mitochondrial ubiquitin ligase MITOL/MARCH5 triggers mitochondrial impairments and exacerbates cognitive decline in a mouse model with AD-related Aß pathology. In this study, MITOL deficiency accelerated the formation of expanded/dense-core-containing Aß plaques, which showed reduced contacts with astrocytes, but not microglia. Our findings suggest that expanded/dense-core-containing Aß-plaque formation enhanced by the alteration of mitochondrial function robustly contributes to the exacerbation of Aß neuropathology, at least in part, through the reduced contacts between Aß plaques and astrocytes.


Subject(s)
Amyloid beta-Peptides/toxicity , Astrocytes/pathology , Neurotoxins/toxicity , Plaque, Amyloid/pathology , Animals , Astrocytes/drug effects , Gene Deletion , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Neurites/drug effects , Neurites/metabolism , Neurites/pathology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
13.
Commun Biol ; 4(1): 192, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33580194

ABSTRACT

Mitochondrial pathophysiology is implicated in the development of Alzheimer's disease (AD). An integrative database of gene dysregulation suggests that the mitochondrial ubiquitin ligase MITOL/MARCH5, a fine-tuner of mitochondrial dynamics and functions, is downregulated in patients with AD. Here, we report that the perturbation of mitochondrial dynamics by MITOL deletion triggers mitochondrial impairments and exacerbates cognitive decline in a mouse model with AD-related Aß pathology. Notably, MITOL deletion in the brain enhanced the seeding effect of Aß fibrils, but not the spontaneous formation of Aß fibrils and plaques, leading to excessive secondary generation of toxic and dispersible Aß oligomers. Consistent with this, MITOL-deficient mice with Aß etiology exhibited worsening cognitive decline depending on Aß oligomers rather than Aß plaques themselves. Our findings suggest that alteration in mitochondrial morphology might be a key factor in AD due to directing the production of Aß form, oligomers or plaques, responsible for disease development.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Brain/enzymology , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Behavior, Animal , Blood Proteins/genetics , Blood Proteins/metabolism , Brain/pathology , Cell Line, Tumor , Cognition , Disease Models, Animal , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Plaque, Amyloid , Poly(A)-Binding Proteins/genetics , Poly(A)-Binding Proteins/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Protein Aggregates , Protein Aggregation, Pathological , Ubiquitin-Protein Ligases/genetics
14.
EMBO Rep ; 22(3): e49097, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33565245

ABSTRACT

Parkin promotes cell survival by removing damaged mitochondria via mitophagy. However, although some studies have suggested that Parkin induces cell death, the regulatory mechanism underlying the dual role of Parkin remains unknown. Herein, we report that mitochondrial ubiquitin ligase (MITOL/MARCH5) regulates Parkin-mediated cell death through the FKBP38-dependent dynamic translocation from the mitochondria to the ER during mitophagy. Mechanistically, MITOL mediates ubiquitination of Parkin at lysine 220 residue, which promotes its proteasomal degradation, and thereby fine-tunes mitophagy by controlling the quantity of Parkin. Deletion of MITOL leads to accumulation of the phosphorylated active form of Parkin in the ER, resulting in FKBP38 degradation and enhanced cell death. Thus, we have shown that MITOL blocks Parkin-induced cell death, at least partially, by protecting FKBP38 from Parkin. Our findings unveil the regulation of the dual function of Parkin and provide a novel perspective on the pathogenesis of PD.


Subject(s)
Mitophagy , Ubiquitin-Protein Ligases , Cell Survival , HeLa Cells , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
15.
Int J Mol Sci ; 21(11)2020 May 27.
Article in English | MEDLINE | ID: mdl-32471110

ABSTRACT

The molecular pathology of diseases seen from the mitochondrial axis has become more complex with the progression of research. A variety of factors, including the failure of mitochondrial dynamics and quality control, have made it extremely difficult to narrow down drug discovery targets. We have identified MITOL (mitochondrial ubiquitin ligase: also known as MARCH5) localized on the mitochondrial outer membrane and previously reported that it is an important regulator of mitochondrial dynamics and mitochondrial quality control. In this review, we describe the pathological aspects of MITOL revealed through functional analysis and its potential as a drug discovery target.


Subject(s)
Mitochondria/metabolism , Mitochondrial Diseases/enzymology , Mitochondrial Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Endoplasmic Reticulum/metabolism , Humans , Mitochondrial Dynamics
16.
J Biochem ; 168(3): 305-312, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32302394

ABSTRACT

In mitochondrial disorders, short stature and growth failure are common symptoms, but their underlying mechanism remains unknown. In this study, we examined the cause of growth failure of mice induced by nestin promoter-driven knockout of the mitochondrial ubiquitin ligase MITOL (MARCH5), a key regulator of mitochondrial function. MITOL-knockout mice have congenital hypoplasia of the anterior pituitary caused by decreased expression of pituitary transcript factor 1 (Pit1). Consistently, both mRNA levels of growth hormone (GH) and prolactin levels were markedly decreased in the anterior pituitary of mutant mice. Growth failure of mutant mice was partly rescued by hypodermic injection of recombinant GH. To clarify whether this abnormality was induced by the primary effect of MITOL knockdown in the anterior pituitary or a secondary effect of other lesions, we performed lentiviral-mediated knockdown of MITOL on cultured rat pituitary GH3 cells, which secrete GH. GH production was severely compromised in MITOL-knockdown GH3 cells. In conclusion, MITOL plays a critical role in the development of the anterior pituitary; therefore, mice with MITOL dysfunction exhibited pituitary dwarfism caused by anterior pituitary hypoplasia. Our findings suggest that mitochondrial dysfunction is commonly involved in the unknown pathogenesis of pituitary dwarfism.


Subject(s)
Dwarfism/genetics , Dwarfism/metabolism , Mitochondrial Proteins/genetics , Pituitary Gland, Anterior/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Cell Line, Tumor , Dwarfism/drug therapy , Gene Knockdown Techniques , Growth Hormone/administration & dosage , Growth Hormone/genetics , Growth Hormone/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Prolactin/genetics , Prolactin/metabolism , RNA, Messenger/genetics , Rats , Signal Transduction/genetics , Transfection
17.
Biomolecules ; 10(3)2020 03 13.
Article in English | MEDLINE | ID: mdl-32183225

ABSTRACT

Mitochondria are highly dynamic organelles that constantly fuse, divide, and move, and their function is regulated and maintained by their morphologic changes. Mitochondrial disease (MD) comprises a group of disorders involving mitochondrial dysfunction. However, it is not clear whether changes in mitochondrial morphology are related to MD. In this study, we examined mitochondrial morphology in fibroblasts from patients with MD (mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) and Leigh syndrome). We observed that MD fibroblasts exhibited significant mitochondrial fragmentation by upregulation of Drp1, which is responsible for mitochondrial fission. Interestingly, the inhibition of mitochondrial fragmentation by Drp1 knockdown enhanced cellular toxicity and led to cell death in MD fibroblasts. These results suggest that mitochondrial fission plays a critical role in the attenuation of mitochondrial damage in MD fibroblasts.


Subject(s)
Dynamins/metabolism , Fibroblasts/metabolism , Leigh Disease/metabolism , MELAS Syndrome/metabolism , Mitochondria/metabolism , Skin/metabolism , Cell Death , Cells, Cultured , Fibroblasts/pathology , Humans , Leigh Disease/pathology , MELAS Syndrome/pathology , Mitochondria/pathology , Skin/pathology
18.
Sci Rep ; 9(1): 20107, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882856

ABSTRACT

CRMP-5-associated GTPase (CRAG), a short splicing variant of centaurin-γ3/AGAP3, is predominantly expressed in the developing brain. We previously demonstrated that CRAG, but not centaurin-γ3, translocates to the nucleus and activates the serum response factor (SRF)-c-Fos pathway in cultured neuronal cells. However, the physiological relevance of CRAG in vivo is unknown. Here, we found that CRAG/centaurin-γ3-knockout mice showed intensively suppressed kainic acid-induced c-fos expression in the hippocampus. Analyses of molecular mechanisms underlying CRAG-mediated SRF activation revealed that CRAG has an essential role in GTPase activity, interacts with ELK1 (a co-activator of SRF), and activates SRF in an ELK1-dependent manner. Furthermore, CRAG and ELK1 interact with promyelocytic leukaemia bodies through SUMO-interacting motifs, which is required for SRF activation. These results suggest that CRAG plays a critical role in ELK1-dependent SRF-c-fos activation at promyelocytic leukaemia bodies in the developing brain.


Subject(s)
Alternative Splicing , GTP-Binding Proteins/genetics , GTPase-Activating Proteins/genetics , Serum Response Factor/metabolism , ets-Domain Protein Elk-1/genetics , Animals , Hippocampus/metabolism , Kainic Acid/pharmacology , Mice , Mice, Knockout , Neurons/metabolism , Promyelocytic Leukemia Protein/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Sumoylation
19.
Mol Cell Oncol ; 6(6): e1659078, 2019.
Article in English | MEDLINE | ID: mdl-31692879

ABSTRACT

Unfolded protein response (UPRs) directs adaption or apoptosis depending on the severity of endoplasmic-reticulum (ER) stress. We found that apoptotic signaling by inositol requiring enzyme 1α (IRE1α), a transducer of UPRs, is suppressed by mitochondrial ubiquitin ligase MITOL/MARCH5 on ER-mitochondria contacts, suggesting that mitochondria regulate cell fate under ER stress.

20.
PLoS One ; 14(11): e0224967, 2019.
Article in English | MEDLINE | ID: mdl-31730661

ABSTRACT

Little is known about the molecular mechanisms of cognitive deficits in psychiatric disorders. CAMDI is a psychiatric disorder-related factor, the deficiency of which in mice results in delayed neuronal migration and psychiatrically abnormal behaviors. Here, we found that CAMDI-deficient mice exhibited impaired recognition memory and spatial reference memory. Knockdown of CAMDI in hippocampal neurons increased the amount of internalized alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor (AMPAR) and attenuated the chemical long-term potentiation (LTP)-dependent cell surface expression of AMPAR. KIBRA was identified as a novel CAMDI-binding protein that retains AMPAR in the cytosol after internalization. KIBRA inhibited CAMDI-dependent Rab11 activation, thereby attenuating AMPAR cell surface expression. These results suggest that CAMDI regulates AMPAR cell surface expression during LTP. CAMDI dysfunction may partly explain the mechanism underlying cognitive deficits in psychiatric diseases.


Subject(s)
Cell Membrane/metabolism , Cognition , Intracellular Signaling Peptides and Proteins/metabolism , Memory , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism , Receptors, AMPA/metabolism , Animals , Cell Line, Tumor , Endocytosis , Humans , Long-Term Potentiation , Mice, Knockout , Protein Binding , Spatial Memory , rab GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL