Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 11: 1171440, 2023.
Article in English | MEDLINE | ID: mdl-37745304

ABSTRACT

Endoplasmic reticulum-mitochondrial contact sites (ERMCS) play an important role in mitochondrial dynamics, calcium signaling, and autophagy. Disruption of the ERMCS has been linked to several neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). However, the etiological role of ERMCS in these diseases remains unclear. We previously established tyrosine hydroxylase reporter (TH-GFP) iPSC lines from a PD patient with a PRKN mutation to perform correlative light-electron microscopy (CLEM) analysis and live cell imaging in GFP-expressing dopaminergic neurons. Here, we analyzed ERMCS in GFP-expressing PRKN-mutant dopaminergic neurons from patients using CLEM and a proximity ligation assay (PLA). The PLA showed that the ERMCS were significantly reduced in PRKN-mutant patient dopaminergic neurons compared to the control under normal conditions. The reduction of the ERMCS in PRKN-mutant patient dopaminergic neurons was further enhanced by treatment with a mitochondrial uncoupler. In addition, mitochondrial calcium imaging showed that mitochondrial Ca2+ flux was significantly reduced in PRKN-mutant patient dopaminergic neurons compared to the control. These results suggest a defect in calcium flux from ER to mitochondria is due to the decreased ERMCS in PRKN-mutant patient dopaminergic neurons. Our study of ERMCS using TH-GFP iPSC lines would contribute to further understanding of the mechanisms of dopaminergic neuron degeneration in patients with PRKN mutations.

2.
Biochem Biophys Res Commun ; 676: 132-140, 2023 10 08.
Article in English | MEDLINE | ID: mdl-37516030

ABSTRACT

Insulin is essential in controlling blood glucose levels, and its synthesis and secretion have been well investigated. In contrast, how insulin secretory granules (ISGs) are degraded in pancreatic beta cells remains largely unknown. To clarify the mechanism, we constructed a fluorescent reporter detecting ISG degradation, where EGFP and mCherry are tandemly conjugated to a cytoplasmic region of ZnT8, an ISG membrane-localized protein. Depletion of serum and amino acid stimulated lysosomal ISG degradation detected with the reporter. Next, with MIN6 cells expressing Cas9 and the reporter, we investigated the involvement of conventional Atg5/7-dependent autophagy to show that it is dispensable for the ISG degradation process. Finally, we performed genome-wide screening by enriching the cells lacking the ISG degradation and showed that pathways regulating autophagy are not identified. These results suggest that alternative degradation in lysosomes, instead of conventional autophagy, may be involved in ISG degradation.


Subject(s)
Insulin-Secreting Cells , Insulin , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin Secretion , Membrane Proteins/metabolism , Coloring Agents/metabolism , Secretory Vesicles/metabolism , Cytoplasmic Granules/metabolism
3.
Stem Cells ; 41(8): 775-791, 2023 08 16.
Article in English | MEDLINE | ID: mdl-37228023

ABSTRACT

Cytochrome P450 3A4 (CYP3A4) is involved in first-pass metabolism in the small intestine and is heavily implicated in oral drug bioavailability and pharmacokinetics. We previously reported that vitamin D3 (VD3), a known CYP enzyme inducer, induces functional maturation of iPSC-derived enterocyte-like cells (iPSC-ent). Here, we identified a Notch activator and CYP modulator valproic acid (VPA), as a promotor for the maturation of iPSC-ent. We performed bulk RNA sequencing to investigate the changes in gene expression during the differentiation and maturation periods of these cells. VPA potentiated gene expression of key enterocyte markers ALPI, FABP2, and transporters such as SULT1B1. RNA-sequencing analysis further elucidated several function-related pathways involved in fatty acid metabolism, significantly upregulated by VPA when combined with VD3. Particularly, VPA treatment in tandem with VD3 significantly upregulated key regulators of enterohepatic circulation, such as FGF19, apical bile acid transporter SLCO1A2 and basolateral bile acid transporters SLC51A and SLC51B. To sum up, we could ascertain the genetic profile of our iPSC-ent cells to be specialized toward fatty acid absorption and metabolism instead of transporting other nutrients, such as amino acids, with the addition of VD3 and VPA in tandem. Together, these results suggest the possible application of VPA-treated iPSC-ent for modelling enterohepatic circulation.


Subject(s)
Induced Pluripotent Stem Cells , Valproic Acid , Humans , Valproic Acid/pharmacology , Valproic Acid/metabolism , Cholecalciferol/pharmacology , Cholecalciferol/metabolism , Induced Pluripotent Stem Cells/metabolism , Enterocytes/metabolism , Cells, Cultured
4.
PLoS One ; 16(8): e0256622, 2021.
Article in English | MEDLINE | ID: mdl-34432824

ABSTRACT

Based on the assumption that some progenitor cells in an organ might reside in neighboring adipose tissue, we investigated whether melanocyte progenitor cells reside in human subcutaneous adipose tissue. First, we examined the expression of human melanoma black 45 (HMB45) and microphthalmia-associated transcription factor (MITF) in undifferentiated adipose-derived stem cells (ADSCs) by immunostaining, RT-PCR, and western blotting. These two markers were detected in undifferentiated ADSCs, and their expression levels were increased in differentiated ADSCs in melanocyte-specific culture medium. Other melanocytic markers (Melan A, MATP, Mel2, Mel EM, tyrosinase, KIT, and PAX3) were also detected at variable levels in undifferentiated ADSCs, and the expression of some markers was increased during differentiation into the melanocyte lineage. We further showed that ADSCs differentiated in melanocyte-specific culture medium localized in the basal layer and expressed tyrosinase and HMB45 in a 3D epidermal culture system. Melanin deposits were also induced by ultraviolet-light-B (UVB) irradiation. These results demonstrate that melanocyte progenitor cells reside in human subcutaneous adipose tissue and that these cells might have the potential to differentiate into mature melanocytes. Melanocyte and keratinocyte progenitors residing in human subcutaneous tissue can be used for the treatment of skin diseases and skin rejuvenation in the future.


Subject(s)
Melanocytes/cytology , Stem Cells/cytology , Subcutaneous Tissue/anatomy & histology , Biomarkers/metabolism , Cell Differentiation , Cell Line, Tumor , Dihydroxyphenylalanine/metabolism , Down-Regulation , Epidermis/metabolism , Gene Expression Regulation , Humans , Keratinocytes/metabolism , Melanins/metabolism , Melanocytes/ultrastructure , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/metabolism , Models, Biological , Pigmentation , RNA, Small Interfering/metabolism , Stem Cells/ultrastructure
5.
Mol Brain ; 14(1): 58, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33757554

ABSTRACT

Mitochondrial structural changes are associated with the regulation of mitochondrial function, apoptosis, and neurodegenerative diseases. PRKN is known to be involved with various mechanisms of mitochondrial quality control including mitochondrial structural changes. Parkinson's disease (PD) with PRKN mutations is characterized by the preferential degeneration of dopaminergic neurons in the substantia nigra pars compacta, which has been suggested to result from the accumulation of damaged mitochondria. However, ultrastructural changes of mitochondria specifically in dopaminergic neurons derived from iPSC have rarely been analyzed. The main reason for this would be that the dopaminergic neurons cannot be distinguished directly among a mixture of iPSC-derived differentiated cells under electron microscopy. To selectively label dopaminergic neurons and analyze mitochondrial morphology at the ultrastructural level, we generated control and PRKN-mutated patient tyrosine hydroxylase reporter (TH-GFP) induced pluripotent stem cell (iPSC) lines. Correlative light-electron microscopy analysis and live cell imaging of GFP-expressing dopaminergic neurons indicated that iPSC-derived dopaminergic neurons had smaller and less functional mitochondria than those in non-dopaminergic neurons. Furthermore, the formation of spheroid-shaped mitochondria, which was induced in control dopaminergic neurons by a mitochondrial uncoupler, was inhibited in the PRKN-mutated dopaminergic neurons. These results indicate that our established TH-GFP iPSC lines are useful for characterizing mitochondrial morphology, such as spheroid-shaped mitochondria, in dopaminergic neurons among a mixture of various cell types. Our in vitro model would provide insights into the vulnerability of dopaminergic neurons and the processes leading to the preferential loss of dopaminergic neurons in patients with PRKN mutations.


Subject(s)
Dopaminergic Neurons/ultrastructure , Induced Pluripotent Stem Cells/cytology , Mitochondria/ultrastructure , Ubiquitin-Protein Ligases/genetics , Base Sequence , CRISPR-Cas Systems , Cells, Cultured , Gene Editing , Gene Knock-In Techniques , Genes, Reporter , Humans , Induced Pluripotent Stem Cells/drug effects , Microscopy, Electron/methods , Microscopy, Fluorescence , Neurogenesis , RNA, Guide, Kinetoplastida/genetics , Spheroids, Cellular , Tyrosine 3-Monooxygenase/genetics
6.
Autophagy ; 17(10): 2962-2974, 2021 10.
Article in English | MEDLINE | ID: mdl-33218272

ABSTRACT

Mitochondrial quality control, which is crucial for maintaining cellular homeostasis, has been considered to be achieved exclusively through mitophagy. Here we report an alternative mitochondrial quality control pathway mediated by extracellular mitochondria release. By performing time-lapse confocal imaging on a stable cell line with fluorescent-labeled mitochondria, we observed release of mitochondria from cells into the extracellular space. Correlative light-electron microscopy revealed that majority of the extracellular mitochondria are in free form and, on rare occasions, some are enclosed in membrane-surrounded vesicles. Rotenone- and carbonyl cyanide m-chlorophenylhydrazone-induced mitochondrial quality impairment promotes the extracellular release of depolarized mitochondria. Overexpression of PRKN (parkin RBR E3 ubiquitin protein ligase), which has a pivotal role in mitophagy regulation, suppresses the extracellular mitochondria release under basal and stress condition, whereas its knockdown exacerbates it. Correspondingly, overexpression of PRKN-independent mitophagy regulators, BNIP3 (BCL2 interacting protein 3) and BNIP3L/NIX (BCL2 interacting protein 3 like), suppress extracellular mitochondria release. Autophagy-deficient cell lines show elevated extracellular mitochondria release. These results imply that perturbation of mitophagy pathway prompts mitochondria expulsion. Presence of mitochondrial protein can also be detected in mouse sera. Sera of PRKN-deficient mice contain higher level of mitochondrial protein compared to that of wild-type mice. More importantly, fibroblasts and cerebrospinal fluid samples from Parkinson disease patients carrying loss-of-function PRKN mutations show increased extracellular mitochondria compared to control subjects, providing evidence in a clinical context. Taken together, our findings suggest that extracellular mitochondria release is a comparable yet distinct quality control pathway from conventional mitophagy.Abbreviations: ACTB: actin beta; ANXA5: annexin A5; ATP5F1A/ATP5A: ATP synthase F1 subunit alpha; ATG: autophagy related; BNIP3: BCL2 interacting protein 3; BNIP3L/NIX: BCL2 interacting protein 3 like; CCCP: carbonyl cyanide m-chlorophenylhydrazone; CM: conditioned media; CSF: cerebrospinal fluid; DMSO: dimethyl sulfoxide; EM: electron microscopy; HSPD1/Hsp60: heat shock protein family D (Hsp60) member 1; KD: knockdown; KO: knockout; MAP1LC3A/LC3: microtubule associated protein 1 light chain 3 alpha; MT-CO1: mitochondrially encoded cytochrome c oxidase I; NDUFB8: NADH:ubiquinone oxidoreductase subunit B8; OE: overexpression; OPA1: OPA1 mitochondrial dynamin like GTPase; OXPHOS: oxidative phosphorylation; PBS: phosphate-buffered saline; PB: phosphate buffer; PD: Parkinson disease; PINK1: PTEN induced kinase 1; PRKN: parkin RBR E3 ubiquitin protein ligase; RB1CC1/FIP200: RB1 inducible coiled-coil 1; SDHB: succinate dehydrogenase complex iron sulfur subunit B; TOMM20: translocase of outer mitochondrial membrane 20; TOMM40: translocase of outer mitochondrial membrane 40; UQCRC2: ubiquinol-cytochrome c reductase core protein 2; WT: wild-type.


Subject(s)
Autophagy , Mitophagy , Animals , Autophagy/physiology , Humans , Mice , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Mitophagy/genetics , Ubiquitin-Protein Ligases/metabolism
7.
Nat Chem Biol ; 17(3): 335-343, 2021 03.
Article in English | MEDLINE | ID: mdl-33168978

ABSTRACT

Mitochondrial DNA (mtDNA) mutations are the major cause of mitochondrial diseases. Cells harboring disease-related mtDNA mutations exhibit various phenotypic abnormalities, such as reduced respiration and elevated lactic acid production. Induced pluripotent stem cell (iPSC) lines derived from patients with mitochondrial disease, with high proportions of mutated mtDNA, exhibit defects in maturation into neurons or cardiomyocytes. In this study, we have discovered a small-molecule compound, which we name tryptolinamide (TLAM), that activates mitochondrial respiration in cybrids generated from patient-derived mitochondria and fibroblasts from patient-derived iPSCs. We found that TLAM inhibits phosphofructokinase-1 (PFK1), which in turn activates AMPK-mediated fatty-acid oxidation to promote oxidative phosphorylation, and redirects carbon flow from glycolysis toward the pentose phosphate pathway to reinforce anti-oxidative potential. Finally, we found that TLAM rescued the defect in neuronal differentiation of iPSCs carrying a high ratio of mutant mtDNA, suggesting that PFK1 represents a potential therapeutic target for mitochondrial diseases.


Subject(s)
Amides/pharmacology , Carbolines/pharmacology , Fibroblasts/drug effects , Induced Pluripotent Stem Cells/drug effects , Mitochondria/drug effects , Neurons/drug effects , Phosphofructokinase-1/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Amides/chemistry , Carbolines/chemistry , Cell Differentiation/drug effects , Cell Respiration/drug effects , Cell Respiration/genetics , Chimera/genetics , Chimera/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Glycolysis/drug effects , Glycolysis/genetics , HEK293 Cells , HeLa Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mutation , Neurons/metabolism , Neurons/pathology , Oxidative Phosphorylation/drug effects , Pentose Phosphate Pathway/genetics , Phosphofructokinase-1/antagonists & inhibitors , Phosphofructokinase-1/metabolism
8.
Sci Rep ; 10(1): 12187, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32699230

ABSTRACT

Insulin gene mutations have been identified to cause monogenic diabetes, and most of which developed permanent neonatal diabetes at young ages before 6 months of age in humans. To establish an animal model of permanent diabetes, we performed genome editing using the CRISPR/Cas9 system. We generated a novel Kuma mutant mice with p.Q104del in the Insulin2 (Ins2) gene in a BRJ background that exhibits a severe immune deficiency. Kuma mutant mice are non-obese and developed hyperglycemia from 3 weeks after birth in both males and females, which are inherited in a dominant mode. Kuma mutant mice displayed reduced insulin protein levels from 3-weeks-old, which seem to be caused by the low stability of the mutant insulin protein. Kuma mutant showed a reduction in islet size and islet mass. Electron microscopic analysis revealed a marked decrease in the number and size of insulin granules in the beta-cells of the mutant mice. Hyperglycemia of the mutant can be rescued by insulin administration. Our results present a novel insulin mutation that causes permanent early-onset diabetes, which provides a model useful for islet transplantation studies.


Subject(s)
Hyperglycemia/pathology , Insulin/genetics , Animals , CRISPR-Cas Systems/genetics , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Female , Gene Editing , Glucose Tolerance Test , Hyperglycemia/drug therapy , Hyperglycemia/genetics , Insulin/therapeutic use , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/physiology , Islets of Langerhans/ultrastructure , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Microscopy, Electron , Oligodeoxyribonucleotides/metabolism , Organ Size
9.
J Cell Sci ; 132(11)2019 05 31.
Article in English | MEDLINE | ID: mdl-31076512

ABSTRACT

Peroxisomes cooperate with mitochondria in the performance of cellular metabolic functions, such as fatty acid oxidation and the maintenance of redox homeostasis. However, whether peroxisomes also regulate mitochondrial fission-fusion dynamics or mitochondrion-dependent apoptosis remained unclear. We now show that genetic ablation of the peroxins Pex3 or Pex5, which are essential for peroxisome biogenesis, results in mitochondrial fragmentation in mouse embryonic fibroblasts (MEFs) in a manner dependent on Drp1 (also known as DNM1L). Conversely, treatment with 4-PBA, which results in peroxisome proliferation, resulted in mitochondrial elongation in wild-type MEFs, but not in Pex3-knockout MEFs. We further found that peroxisome deficiency increased the levels of cytosolic cytochrome c and caspase activity under basal conditions without inducing apoptosis. It also greatly enhanced etoposide-induced caspase activation and apoptosis, which is indicative of an enhanced cellular sensitivity to death signals. Taken together, our data unveil a previously unrecognized role for peroxisomes in the regulation of mitochondrial dynamics and mitochondrion-dependent apoptosis. Effects of peroxin gene mutations on mitochondrion-dependent apoptosis may contribute to pathogenesis of peroxisome biogenesis disorders.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Peroxisomes/metabolism , Animals , Butylamines/pharmacology , Caspases/metabolism , Cell Line , Cytochromes c/metabolism , Dynamins/metabolism , Humans , Lipoproteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Peroxins/genetics , Peroxisomal Disorders/pathology , Peroxisome-Targeting Signal 1 Receptor/genetics , RNA Interference , RNA, Small Interfering/genetics
10.
Cell Death Dis ; 8(1): e2551, 2017 01 12.
Article in English | MEDLINE | ID: mdl-28079893

ABSTRACT

Mitochondrial diseases are genetically heterogeneous and present a broad clinical spectrum among patients; in most cases, genetic determinants of mitochondrial diseases are heteroplasmic mitochondrial DNA (mtDNA) mutations. However, it is uncertain whether and how heteroplasmic mtDNA mutations affect particular cellular fate-determination processes, which are closely associated with the cell-type-specific pathophysiology of mitochondrial diseases. In this study, we established two isogenic induced pluripotent stem cell (iPSC) lines each carrying different proportions of a heteroplasmic m.3243A>G mutation from the same patient; one exhibited apparently normal and the other showed most likely impaired mitochondrial respiratory function. Low proportions of m.3243A>G exhibited no apparent molecular pathogenic influence on directed differentiation into neurons and cardiomyocytes, whereas high proportions of m.3243A>G showed both induced neuronal cell death and inhibited cardiac lineage commitment. Such neuronal and cardiac maturation defects were also confirmed using another patient-derived iPSC line carrying quite high proportion of m.3243A>G. In conclusion, mitochondrial respiratory dysfunction strongly inhibits maturation and survival of iPSC-derived neurons and cardiomyocytes; our presenting data also suggest that appropriate mitochondrial maturation actually contributes to cellular fate-determination processes during development.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria/genetics , Mitochondrial Diseases/genetics , Myocytes, Cardiac/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mutation , Myocytes, Cardiac/pathology , Neurons/metabolism , Neurons/pathology
11.
Hum Mol Genet ; 24(16): 4698-709, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26025377

ABSTRACT

Mitochondrial dysfunction caused by pathogenic mutations in mitochondrial tRNA genes emerges only when mutant mitochondrial DNA (mtDNA) proportions exceed intrinsic pathogenic thresholds; however, little is known about the actual proportions of mutant mtDNA that can affect particular cellular lineage-determining processes. Here, we mainly focused on the effects of mitochondrial respiratory dysfunction caused by m.3243A>G heteroplasmy in MT-TL1 gene on cellular reprogramming. We found that generation of induced pluripotent stem cells (iPSCs) was drastically depressed only by high proportions of mutant mtDNA (≥ 90% m.3243A>G), and these proportions were strongly associated with the degree of induced mitochondrial respiratory dysfunction. Nevertheless, all established iPSCs, even those carrying ∼ 100% m.3243A>G, exhibited an embryonic stem cell-like pluripotent state. Therefore, our findings clearly demonstrate that loss of physiological integrity in mitochondria triggered by mutant mtDNA constitute a roadblock to cellular rejuvenation, but do not affect the maintenance of the pluripotent state.


Subject(s)
Cellular Reprogramming/genetics , DNA, Mitochondrial/genetics , Induced Pluripotent Stem Cells , Mitochondrial Diseases/genetics , Mutation , Female , Humans , Male , Mitochondrial Diseases/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism
12.
Proc Natl Acad Sci U S A ; 109(26): 10528-33, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22689997

ABSTRACT

It has been hypothesized that respiration defects caused by accumulation of pathogenic mitochondrial DNA (mtDNA) mutations and the resultant overproduction of reactive oxygen species (ROS) or lactates are responsible for aging and age-associated disorders, including diabetes and tumor development. However, there is no direct evidence to prove the involvement of mtDNA mutations in these processes, because it is difficult to exclude the possible involvement of nuclear DNA mutations. Our previous studies resolved this issue by using an mtDNA exchange technology and showed that a G13997A mtDNA mutation found in mouse tumor cells induces metastasis via ROS overproduction. Here, using transmitochondrial mice (mito-mice), which we had generated previously by introducing G13997A mtDNA from mouse tumor cells into mouse embryonic stem cells, we provide convincing evidence supporting part of the abovementioned hypothesis by showing that G13997A mtDNA regulates diabetes development, lymphoma formation, and metastasis--but not aging--in this model.


Subject(s)
DNA, Mitochondrial/genetics , Diabetes Mellitus, Experimental/genetics , Lymphoma/genetics , Mitochondrial Diseases/genetics , Mutation , 3T3 Cells , Animals , Base Sequence , Cell Line, Transformed , DNA Primers , Mice , Phenotype , Polymerase Chain Reaction , Reactive Oxygen Species/metabolism
13.
Exp Anim ; 60(4): 397-404, 2011.
Article in English | MEDLINE | ID: mdl-21791879

ABSTRACT

This study determined pathogenicity of an A11181G mtDNA mutation found in a senescence-accelerated mouse strain, SAMP8. The mutation was at a highly conserved site of the mt-Nd4 gene, which encodes one of the respiratory complex I subunits. The young SAMP8 expressed reduced complex I activity, which is controlled by both nuclear and mitochondrial DNA (mtDNA). To exclude the nuclear effects, we isolated transmitochondrial cybrids that share the same nuclear background, but possess mtDNA with or without the mutation. The cybrids showed normal respiratory function irrespective of whether their mtDNA possessed the mutation or not, suggesting that the A11181G mutation is not responsible for respiration defects found in SAMP8.


Subject(s)
Cell Nucleus/genetics , DNA, Mitochondrial/genetics , Electron Transport Complex I/genetics , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Aging , Amino Acid Substitution , Animals , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cell Respiration/genetics , DNA, Mitochondrial/chemistry , DNA, Mitochondrial/metabolism , Electron Transport Complex I/metabolism , Male , Mice , Mice, Inbred Strains , NADH Dehydrogenase/chemistry , Nuclear Transfer Techniques , Sequence Alignment
14.
FEBS Lett ; 584(18): 3943-8, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20674568

ABSTRACT

To investigate the effects of respiration defects on the disease phenotypes, we generated trans-mitochondrial mice (mito-mice) by introducing a mutated G13997A mtDNA, which specifically induces respiratory complex I defects and metastatic potentials in mouse tumor cells. First, we obtained ES cells and chimeric mice containing the G13997A mtDNA, and then we generated mito-mice carrying the G13997A mtDNA via its female germ line transmission. The three-month-old mito-mice showed complex I defects and lactate overproduction, but showed no other phenotypes related to mitochondrial diseases or tumor formation, suggesting that aging or additional nuclear abnormalities are required for expression of other phenotypes.


Subject(s)
Carcinoma/genetics , DNA, Mitochondrial/genetics , DNA, Neoplasm/genetics , Lung Neoplasms/genetics , Mitochondria/genetics , Mitochondrial Diseases/genetics , Age Factors , Aging/genetics , Animals , Carcinoma/pathology , Chimera , Embryonic Stem Cells , Female , Lung Neoplasms/pathology , Male , Mice , Mice, Transgenic , Mitochondrial Diseases/pathology , Neoplasm Metastasis , Optic Atrophy, Hereditary, Leber/genetics
15.
FEBS Lett ; 581(9): 1910-6, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17434485

ABSTRACT

Anemia is a symptom in patients with Pearson syndrome caused by the accumulation of mutated mitochondrial DNA (mtDNA). Such mutated mtDNAs have been detected in patients with anemia. This suggested that respiration defects due to mutated mtDNA are responsible for the anemia. However, there has been no convincing experimental evidence to confirm the pathophysiological relation between respiration defects in hematopoietic cells and expression of anemia. We address this issue by transplanting bone marrow cells carrying pathogenic mtDNA with a large-scale deletion (DeltamtDNA) into normal mice. The bone marrow-transplanted mice carried high proportion of DeltamtDNA only in hematopoietic cells, and resultant the mice suffered from macrocytic anemia. They show abnormalities of erythroid differentiation and weak erythropoietic response to a stressful condition. These observations suggest that hematopoietic cell-specific respiration defects caused by mtDNAs with pathogenic mutations are responsible for anemia by inducing abnormalities in erythropoiesis.


Subject(s)
Anemia/genetics , DNA, Mitochondrial/genetics , Erythroid Cells/cytology , Hematopoietic Stem Cells/cytology , Adenosine Triphosphate/metabolism , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Respiration/genetics , Electron Transport Chain Complex Proteins/metabolism , Erythroid Cells/metabolism , Female , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial Diseases/genetics , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...