Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Biomedicines ; 11(9)2023 Sep 18.
Article in English | MEDLINE | ID: mdl-37760999

ABSTRACT

The pancreas is a vital organ with exocrine and endocrine functions. Pancreatitis is an inflammation of the pancreas caused by alcohol consumption and gallstones. This condition can heighten the risk of pancreatic cancer (PC), a challenging disease with a high mortality rate. Genetic and epigenetic factors contribute significantly to PC development, along with other risk factors. Early detection is crucial for improving PC outcomes. Diagnostic methods, including imagining modalities and tissue biopsy, aid in the detection and analysis of PC. In contrast, liquid biopsy (LB) shows promise in early tumor detection by assessing biomarkers in bodily fluids. Understanding the function of the pancreas, associated diseases, risk factors, and available diagnostic methods is essential for effective management and early PC detection. The current clinical examination of PC is challenging due to its asymptomatic early stages and limitations of highly precise diagnostics. Screening is recommended for high-risk populations and individuals with potential benign tumors. Among various PC screening methods, the N-NOSE plus pancreas test stands out with its high AUC of 0.865. Compared to other commercial products, the N-NOSE plus pancreas test offers a cost-effective solution for early detection. However, additional diagnostic tests are required for confirmation. Further research, validation, and the development of non-invasive screening methods and standardized scoring systems are crucial to enhance PC detection and improve patient outcomes. This review outlines the context of pancreatic cancer and the challenges for early detection.

2.
Cancers (Basel) ; 15(15)2023 Jul 29.
Article in English | MEDLINE | ID: mdl-37568686

ABSTRACT

BACKGROUND: The nematode Caenorhabditis elegans (C. elegans) possesses a sophisticated sense of smell and is used for a novel cancer screening test that utilizes the chemotaxis index. We designed a single-institution, prospective study to confirm the ability of Nematode Nose (N-NOSE) to determine preoperative chemotherapy's efficacy for esophageal cancer patients. PATIENTS AND METHODS: We investigated the predictability of N-NOSE screening for the clinical effects of preoperative chemotherapy for esophageal cancer patients receiving radical surgery. The index reduction score (IRS) was calculated via the chemotaxis of C. elegans at three points: before treatment, before surgery, and after surgery, and its clinical relevance was examined. RESULT: Thirty-nine patients with esophageal cancer were enrolled from August 2020 to December 2021, and 30 patients receiving radical surgery were examined. Complete response or partial response was achieved in 23 cases (76.7%). When the target of the treatment effect was complete response only, the prediction accuracies of the IRS calculated by area under the curve was 0.85 (95% Confidence interval: 0.62-1) in clinically achieving complete response group, and the sensitivity and specificity were 1 and 0.63, respectively. CONCLUSION: Index reduction score using N-NOSE screening may reflect the efficacy of chemotherapy for esophageal cancer patients. A large-scale prospective study at multiple centers is desired in the future.

3.
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
4.
Mitochondrion ; 34: 43-48, 2017 05.
Article in English | MEDLINE | ID: mdl-28093354

ABSTRACT

Mitochondrial disease is associated with a wide variety of clinical presentations, even among patients carrying heteroplasmic mitochondrial DNA (mtDNA) mutations, probably because of variations in mutant mtDNA proportions at the tissue and organ levels. Although several case reports and clinical trials have assessed the effectiveness of various types of drugs and supplements for the treatment of mitochondrial diseases, there are currently no cures for these conditions. In this study, we demonstrated for the first time that low dose resveratrol (RSV) ameliorated mitochondrial respiratory dysfunction in patient-derived fibroblasts carrying homoplasmic mtDNA mutations. Furthermore, low dose RSV also facilitated efficient cellular reprogramming of the patient-derived fibroblasts into induced pluripotent stem cells, partly due to improved cellular viability. Our results highlight the potential of RSV as a new therapeutic drug candidate for the treatment of mitochondrial diseases.


Subject(s)
Antioxidants/metabolism , Cell Respiration/drug effects , Cellular Reprogramming/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Stilbenes/metabolism , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , Pluripotent Stem Cells/physiology , Resveratrol
5.
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
6.
Am J Pathol ; 187(1): 110-121, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27855277

ABSTRACT

The relationships between the molecular abnormalities in mitochondrial respiratory chain complexes and their negative contributions to mitochondrial and cellular functions have been proved to be essential for better understandings in mitochondrial medicine. Herein, we established the method to identify disease phenotypic differences among patients with muscle histopathological cytochrome c oxidase (COX) deficiency, as one of the representative clinical features in mitochondrial diseases, by using patients' myoblasts that are derived from biopsied skeletal muscle tissues. We identified two obviously different severities in molecular diagnostic criteria of COX deficiency among patients: structurally stable, but functionally mild/moderate defect and severe functional defect with the disrupted COX holoenzyme structure. COX holoenzyme disorganization actually triggered several mitochondrial dysfunctions, including the decreased ATP level, the increased oxidative stress level, and the damaged membrane potential level, all of which lead to the deteriorated cellular growth, the accelerated cellular senescence, and the induced apoptotic cell death. Our cell-based in vitro diagnostic approaches would be widely applicable to understanding patient-specific pathomechanism in various types of mitochondrial diseases, including other respiratory chain complex deficiencies and other mitochondrial metabolic enzyme deficiencies.


Subject(s)
Cytochrome-c Oxidase Deficiency/enzymology , Cytochrome-c Oxidase Deficiency/pathology , Electron Transport Complex IV/metabolism , Mitochondria/metabolism , Cytochrome-c Oxidase Deficiency/diagnosis , Cytochrome-c Oxidase Deficiency/genetics , Holoenzymes/metabolism , Homeostasis , Humans , Models, Biological , Muscle Development , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mutation/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Mol Biol Cell ; 27(10): 1684-93, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27009201

ABSTRACT

Mitochondria that contain a mixture of mutant and wild-type mitochondrial (mt) DNA copies are heteroplasmic. In humans, homoplasmy is restored during early oogenesis and reprogramming of somatic cells, but the mechanism of mt-allele segregation remains unknown. In budding yeast, homoplasmy is restored by head-to-tail concatemer formation in mother cells by reactive oxygen species (ROS)-induced rolling-circle replication and selective transmission of concatemers to daughter cells, but this mechanism is not obvious in higher eukaryotes. Here, using heteroplasmic m.3243A > G primary fibroblast cells derived from MELAS patients treated with hydrogen peroxide (H2O2), we show that an optimal ROS level promotes mt-allele segregation toward wild-type and mutant mtDNA homoplasmy. Enhanced ROS level reduced the amount of intact mtDNA replication templates but increased linear tandem multimers linked by head-to-tail unit-sized mtDNA (mtDNA concatemers). ROS-triggered mt-allele segregation correlated with mtDNA-concatemer production and enabled transmission of multiple identical mt-genome copies as a single unit. Our results support a mechanism by which mt-allele segregation toward mt-homoplasmy is mediated by concatemers.


Subject(s)
Alleles , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Chromosome Segregation , DNA Replication/physiology , Fibroblasts/metabolism , Genes, Mitochondrial , Genome, Mitochondrial , Humans , MELAS Syndrome/genetics , MELAS Syndrome/metabolism , Mutation , Primary Cell Culture
8.
Stem Cells ; 34(4): 801-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26850516

ABSTRACT

Mitochondria contain multiple copies of their own genome (mitochondrial DNA; mtDNA). Once mitochondria are damaged by mutant mtDNA, mitochondrial dysfunction is strongly induced, followed by symptomatic appearance of mitochondrial diseases. Major genetic causes of mitochondrial diseases are defects in mtDNA, and the others are defects of mitochondria-associating genes that are encoded in nuclear DNA (nDNA). Numerous pathogenic mutations responsible for various types of mitochondrial diseases have been identified in mtDNA; however, it remains uncertain why mitochondrial diseases present a wide variety of clinical spectrum even among patients carrying the same mtDNA mutations (e.g., variations in age of onset, in affected tissues and organs, or in disease progression and phenotypic severity). Disease-relevant induced pluripotent stem cells (iPSCs) derived from mitochondrial disease patients have therefore opened new avenues for understanding the definitive genotype-phenotype relationship of affected tissues and organs in various types of mitochondrial diseases triggered by mtDNA mutations. In this concise review, we briefly summarize several recent approaches using patient-derived iPSCs and their derivatives carrying various mtDNA mutations for applications in human mitochondrial disease modeling, drug discovery, and future regenerative therapeutics.


Subject(s)
DNA, Mitochondrial/genetics , Induced Pluripotent Stem Cells , Mitochondria/genetics , Mitochondrial Diseases/genetics , Genetic Therapy , Genome, Mitochondrial/genetics , Humans , Mitochondria/pathology , Mitochondrial Diseases/pathology , Mitochondrial Diseases/therapy , Mutation , Patients
9.
Acta Neuropathol Commun ; 3: 52, 2015 Aug 22.
Article in English | MEDLINE | ID: mdl-26297375

ABSTRACT

INTRODUCTION: Numerous pathogenic mutations responsible for mitochondrial diseases have been identified in mitochondrial DNA (mtDNA)-encoded tRNA genes. In most cases, however, the detailed molecular pathomechanisms and cellular pathophysiology of these mtDNA mutations -how such genetic defects determine the variation and the severity of clinical symptoms in affected individuals- remain unclear. To investigate the molecular pathomechanisms and to realize in vitro recapitulation of mitochondrial diseases, intracellular mutant mtDNA proportions must always be considered. RESULTS: We found a disease-causative mutation, m.5541C>T heteroplasmy in MT-TW gene, in a patient exhibiting mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) with multiple organ involvement. We identified the intrinsic molecular pathomechanisms of m.5541C>T. This mutation firstly disturbed the translation machinery of mitochondrial tRNA(Trp) and induced mitochondrial respiratory dysfunction, followed by severely injured mitochondrial homeostasis. We also demonstrated cell-type-specific disease phenotypes using patient-derived induced pluripotent stem cells (iPSCs) carrying ~100 % mutant m.5541C>T. Significant loss of terminally differentiated iPSC-derived neurons, but not their stem/progenitor cells, was detected most likely due to serious mitochondrial dysfunction triggered by m.5541C>T; in contrast, m.5541C>T did not apparently affect skeletal muscle development. CONCLUSIONS: Our iPSC-based disease models would be widely available for understanding the "definite" genotype-phenotype relationship of affected tissues and organs in various mitochondrial diseases caused by heteroplasmic mtDNA mutations, as well as for further drug discovery applications.


Subject(s)
MELAS Syndrome/genetics , MELAS Syndrome/pathology , Mutation/genetics , RNA, Transfer, Trp/genetics , Adenosine Triphosphate/metabolism , Brain/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Child , Citrate (si)-Synthase/metabolism , DNA Mutational Analysis , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Male , Membrane Potentials/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myoblasts/metabolism , Neurons/physiology , RNA, Messenger/metabolism , Transcription Factors/metabolism
10.
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
11.
FEBS Open Bio ; 5: 219-25, 2015.
Article in English | MEDLINE | ID: mdl-25853038

ABSTRACT

Mitochondrial diseases are heterogeneous disorders, caused by mitochondrial dysfunction. Mitochondria are not regulated solely by nuclear genomic DNA but by mitochondrial DNA. It is difficult to develop effective therapies for mitochondrial disease because of the lack of mitochondrial disease models. Mitochondrial myopathy, encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) is one of the major mitochondrial diseases. The aim of this study was to generate MELAS-specific induced pluripotent stem cells (iPSCs) and to demonstrate that MELAS-iPSCs can be models for mitochondrial disease. We successfully established iPSCs from the primary MELAS-fibroblasts carrying 77.7% of m.3243A>G heteroplasmy. MELAS-iPSC lines ranged from 3.6% to 99.4% of m.3243A>G heteroplasmy levels. The enzymatic activities of mitochondrial respiratory complexes indicated that MELAS-iPSC-derived fibroblasts with high heteroplasmy levels showed a deficiency of complex I activity but MELAS-iPSC-derived fibroblasts with low heteroplasmy levels showed normal complex I activity. Our data indicate that MELAS-iPSCs can be models for MELAS but we should carefully select MELAS-iPSCs with appropriate heteroplasmy levels and respiratory functions for mitochondrial disease modeling.

12.
Hum Mutat ; 34(3): 446-52, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23281071

ABSTRACT

Mitochondrial complex III (CIII) deficiency is a relatively rare disease with high clinical and genetic heterogeneity. CIII comprises 11 subunits encoded by one mitochondrial and 10 nuclear genes. Abnormalities of the nuclear genes such as BCS1L and TTC19 encoding mitochondrial assembly factors are well known, but an explanation of the majority of CIII deficiency remains elusive. Here, we report three patients from a consanguineous Mexican family presenting with neonatal onset of hypoglycemia, lactic acidosis, ketosis, and hyperammonemia. We found a homozygous missense mutation in UQCRC2 that encodes mitochondrial ubiquinol-cytochrome c reductase core protein II by whole-exome sequencing combined with linkage analysis. On the basis of structural modeling, the mutation (p.Arg183Trp) was predicted to destabilize the hydrophobic core at the subunit interface of the core protein II homodimer. In vitro studies using fibroblasts from the index patient clearly indicated CIII deficiency, as well as impaired assembly of the supercomplex formed from complexes I, III, and IV. This is the first described human disease caused by a core protein abnormality in mitochondrial CIII.


Subject(s)
Electron Transport Complex III/genetics , Homozygote , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/genetics , Mutation, Missense , ATPases Associated with Diverse Cellular Activities , Acidosis, Lactic/genetics , Adult , Blotting, Western , Electron Transport Complex III/deficiency , Exome , Female , Genetic Linkage , Humans , Hyperammonemia/genetics , Hypoglycemia/genetics , Ketosis/genetics , Male , Membrane Proteins/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Pedigree , Protein Conformation , Sequence Analysis, DNA
13.
J Med Genet ; 49(12): 777-84, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23188110

ABSTRACT

BACKGROUND: Autosomal recessive hereditary spastic paraplegias (AR-HSP) constitute a heterogeneous group of neurodegenerative diseases involving pyramidal tracts dysfunction. The genes responsible for many types of AR-HSPs remain unknown. We attempted to identify the gene responsible for AR-HSP with optic atrophy and neuropathy. METHODS: The present study involved two patients in a consanguineous Japanese family. Neurologic examination and DNA analysis were performed for both patients, and a skin biopsy for one. We performed genome-wide linkage analysis involving single nucleotide polymorphism arrays, copy-number variation analysis, and exome sequencing. To clarify the mitochondrial functional alteration resulting from the identified mutation, we performed immunoblot analysis, mitochondrial protein synthesis assaying, blue native polyacrylamide gel electrophoresis (BN-PAGE) analysis, and respiratory enzyme activity assaying of cultured fibroblasts of the patient and a control. RESULTS: We identified a homozygous nonsense mutation (c.394C>T, p.R132X) in C12orf65 in the two patients in this family. This C12orf65 mutation was not found in 74 Japanese AR-HSP index patients without any mutations in previously known HSP genes. This mutation resulted in marked reduction of mitochondrial protein synthesis, followed by functional and structural defects in respiratory complexes I and IV. CONCLUSIONS: This novel nonsense mutation in C12orf65 could cause AR-HSP with optic atrophy and neuropathy, resulting in a premature stop codon. The truncated C12orf65 protein must lead to a defect in mitochondrial protein synthesis and a reduction in the respiratory complex enzyme activity. Thus, dysfunction of mitochondrial translation could be one of the pathogenic mechanisms underlying HSPs.


Subject(s)
Homozygote , Mutation , Optic Atrophy/genetics , Peptide Termination Factors/genetics , Peripheral Nervous System Diseases/genetics , Spastic Paraplegia, Hereditary/genetics , Adult , Base Sequence , DNA Copy Number Variations , Exome , Genetic Linkage , Humans , Male , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins , Optic Atrophy/metabolism , Pedigree , Peripheral Nervous System Diseases/metabolism , Spastic Paraplegia, Hereditary/metabolism
14.
Cell Metab ; 16(2): 274-83, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-22819524

ABSTRACT

Although substantial progress has been made in understanding the mechanisms underlying the expression of mtDNA-encoded polypeptides, the regulatory factors involved in mitoribosome-mediated synthesis and simultaneous insertion of mitochondrial oxidative phosphorylation (OXPHOS) polypeptides into the inner membrane of mitochondria are still unclear. In the present study, disruption of the mouse Crif1 gene, which encodes a mitochondrial protein, resulted in a profound deficiency in OXPHOS caused by the disappearance of OXPHOS subunits and complexes in vivo. CRIF1 was associated with large mitoribosomal subunits that were located close to the polypeptide exit tunnel, and the elimination of CRIF1 led to both aberrant synthesis and defective insertion of mtDNA-encoded nascent OXPHOS polypeptides into the inner membrane. CRIF1 interacted with nascent OXPHOS polypeptides and molecular chaperones, e.g., Tid1. Taken together, these results suggest that CRIF1 plays a critical role in the integration of OXPHOS polypeptides into the mitochondrial membrane in mammals.


Subject(s)
Cell Cycle Proteins/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Oxidative Phosphorylation , Peptides/metabolism , Animals , Blotting, Western , Cell Fractionation , Immunohistochemistry , Mice
15.
Brain Dev ; 34(3): 196-200, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21550738

ABSTRACT

Alternating hemiplegia of childhood (AHC) is a rare disorder characterized by repeated plegic attacks, movement disorders, autonomic phenomena, and developmental delay. To obtain insights into the pathophysiology of AHC, we determined the concentrations of matrix metalloproteinase-9 (MMP-9), tissue inhibitor of MMP-1 (TIMP-1), calcitonin gene-related peptide (CGRP), and substance P (SP) in the serum/plasma of AHC patients (n=6) and control subjects (n=11) by performing enzyme-linked immunosorbent assay (ELISA). Decreased levels of serum SP (382±161 pg/ml), increased levels of plasma MMP-9 (111.0±99.3 ng/mL) and increased MMP-9/TIMP-1 ratio (0.65±0.44) were revealed, compared to those in control subjects (SP: 620±223 pg/mL, p<0.05; MMP-9: 33.5±20.3 ng/mL, p<0.05; MMP-9/TIMP-1 ratio 0.21±0.09, p<0.005). Serum CGRP levels in AHC patients (32.6±14.4 pg/mL) were comparable to those in control subjects (37.0±17.0 pg/mL). Increased MMP-9 levels may be linked to the vascular insult and is common in migraineurs. However, because AHC patients showed different changes in SP and CGRP levels compared to those shown by migraineurs, these results suggest that AHC has a pathomechanism different from the hypothesis of trigeminovascular theory. Decreased SP may represent the autonomic dysfunction in AHC, for which an etiology with progressive neuronal damage can be hypothesized.


Subject(s)
Biomarkers/blood , Hemiplegia/blood , Hemiplegia/physiopathology , Matrix Metalloproteinase 9/blood , Substance P/blood , Adolescent , Adult , Calcitonin Gene-Related Peptide/blood , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Tissue Inhibitor of Metalloproteinase-1/blood
16.
Hum Mol Genet ; 20(19): 3841-51, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21750112

ABSTRACT

Choline kinase is the first step enzyme for phosphatidylcholine (PC) de novo biosynthesis. Loss of choline kinase activity in muscle causes rostrocaudal muscular dystrophy (rmd) in mouse and congenital muscular dystrophy in human, characterized by distinct mitochondrial morphological abnormalities. We performed biochemical and pathological analyses on skeletal muscle mitochondria from rmd mice. No mitochondria were found in the center of muscle fibers, while those located at the periphery of the fibers were significantly enlarged. Muscle mitochondria in rmd mice exhibited significantly decreased PC levels, impaired respiratory chain enzyme activities, decreased mitochondrial ATP synthesis, decreased coenzyme Q and increased superoxide production. Electron microscopy showed the selective autophagic elimination of mitochondria in rmd muscle. Molecular markers of mitophagy, including Parkin, PINK1, LC3, polyubiquitin and p62, were localized to mitochondria of rmd muscle. Quantitative analysis shows that the number of mitochondria in muscle fibers and mitochondrial DNA copy number were decreased. We demonstrated that the genetic defect in choline kinase in muscle results in mitochondrial dysfunction and subsequent mitochondrial loss through enhanced activation of mitophagy. These findings provide a first evidence for a pathomechanistic link between de novo PC biosynthesis and mitochondrial abnormality.


Subject(s)
Choline Kinase/metabolism , Mitochondria/enzymology , Muscle, Skeletal/enzymology , Muscular Dystrophies/enzymology , Adenosine Triphosphate/metabolism , Animals , Choline Kinase/genetics , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism
17.
Acta Neuropathol ; 121(6): 775-83, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21424749

ABSTRACT

Axial myopathy is a rare neuromuscular disease that is characterized by paraspinal muscle atrophy and abnormal posture, most notably camptocormia (also known as bent spine). The genetic cause of familial axial myopathy is unknown. Described here are the clinical features and cause of late-onset predominant axial myopathy and encephalopathy. A 73-year-old woman presented with a 10-year history of severe paraspinal muscle atrophy and cerebellar ataxia. Her 84-year-old sister also developed late-onset paraspinal muscle atrophy and generalized seizures with encephalopathy. Computed tomography showed severe atrophy and fatty degeneration of their paraspinal muscles. Their mother and maternal aunt also developed bent spines. The existence of many ragged-red fibers and cytochrome c oxidase-negative fibers in the biceps brachii muscle of the proband indicated a mitochondrial abnormality. No significant abnormalities were observed in the respiratory chain enzyme activities; however, the activities of complexes I and IV were relatively low compared with the activities of other complexes. Sequence analysis of the mitochondrial DNA from the muscle revealed a novel heteroplasmic mutation (m.602C>T) in the mitochondrial tRNA(Phe) gene. This familial case of late-onset predominant axial myopathy and encephalopathy may represent a new clinical phenotype of a mitochondrial disease.


Subject(s)
Mitochondrial Diseases/pathology , Muscle, Skeletal/pathology , Neuromuscular Diseases/pathology , Aged , Aged, 80 and over , DNA Mutational Analysis , DNA, Mitochondrial/genetics , Electron Transport Complex IV/metabolism , Female , Humans , Mitochondrial Diseases/complications , Mitochondrial Diseases/genetics , Muscle, Skeletal/enzymology , Muscle, Skeletal/ultrastructure , Neuromuscular Diseases/complications , Neuromuscular Diseases/genetics , Succinate Dehydrogenase/metabolism
18.
Ann Neurol ; 68(6): 845-54, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21194154

ABSTRACT

OBJECTIVE: To characterize the clinical features and clarify the pathogenicity of "benign cytochrome c oxidase deficiency myopathy." METHODS: The study included 8 patients with the phenotype of this disease. Six patients underwent muscle biopsies and all the 8 underwent mitochondrial DNA analyses. To confirm the pathogenicity of the detected mitochondrial DNA mutation, we performed northern blot analysis, using muscle specimens, and blue native polyacrylamide gel electrophoresis and respiratory chain enzyme activity assay of transmitochondrial cell lines (cybrids). RESULTS: Clinical symptoms were limited to skeletal muscle and improved spontaneously in all cases; however, 2 siblings had basal ganglia lesions. In all patients, we identified a homoplasmic m.14674T>C or m.14674T>G mitochondrial transfer RNA-glutamate mutation. Northern blot analysis revealed decreased levels of mitochondrial transfer RNA-glutamate molecules. Muscle specimens and cybrids derived from patients showed decreased activity of respiratory complexes IV, and/or I, III; however, this was normal in naive myoblasts. INTERPRETATION: Identification of a novel m.14674T>G mutation in addition to m.14674T>C indicated the importance of this site for disease causation. Analyses of cybrids revealed the pathogenicity of m.14674T>C mutation, which resulted in defects of cytochrome c oxidase and multiple respiratory chain enzymes. Furthermore, patients with basal ganglia lesions provided new insights into this disease, in which only skeletal muscle was thought to be affected. Normal respiratory chain enzyme activities in naive myoblasts suggested the compensatory influence of nuclear factors, which may be a clue to understanding the mechanisms of spontaneous recovery and low penetrance in families carrying the mutation.


Subject(s)
Glutamic Acid/genetics , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Muscle, Skeletal/pathology , RNA, Transfer/genetics , Adolescent , Brain/pathology , Child , Child, Preschool , DNA Mutational Analysis/methods , DNA, Mitochondrial/genetics , Electron Transport Complex IV/genetics , Female , Humans , Infant , Magnetic Resonance Imaging/methods , Male , Mitochondria, Muscle/enzymology , Mitochondria, Muscle/pathology , Mutation/genetics
19.
Mitochondrion ; 9(2): 115-22, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19460299

ABSTRACT

Two novel mitochondrial DNA base changes were identified at both sides of the 3243A>G mutation, the most common mutation associated with mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS). One was a 3244G>A transition in a girl with MELAS. The other was a 3242G>A transition in a girl with a mitochondrial disorder without a MELAS phenotype. Although the two base changes were adjacent to the 3243A>G mutation, they had different effects on the clinical phenotype, muscle pathology, and respiratory chain enzyme activity. Investigations of the different effects of the 3244G>A and 3242G>A base changes may provide a better understanding of tRNA dysfunction in mitochondrial disorders.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondrial Diseases/genetics , Point Mutation , Polymorphism, Genetic , Child , Child, Preschool , Female , Humans , MELAS Syndrome/genetics
20.
Biomaterials ; 28(29): 4294-302, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17602737

ABSTRACT

Current methods including the use of various biological and synthetic sealants are ineffective in the closure of intraoperative air leaks that often occur during cardiothoracic surgeries, resulting in a decreased quality of life for patients. We present the development of a novel lung air leak sealant using tissue engineered cell sheets. In contrast to previous materials such as fibrin glue, these bioengineered cell sheets immediately and permanently seal air leaks in a dynamic fashion that allows for the extensive tissue contraction and expansion involved in respiration, without any postoperative recurrences. Additionally, we demonstrate that mesothelial cells migrate to cover the transplanted cells sheets, thereby confirming excellent biocompatibility and integration with the host tissues. Finally, we present the use of skin fibroblasts as an effective and readily available autologous cell source that can be easily applied. This study shows for the first time, the development of an immediate and permanent lung air leak sealant, suitable for future clinical applications.


Subject(s)
Fibroblasts/transplantation , Guided Tissue Regeneration/methods , Pneumonectomy/adverse effects , Pneumonectomy/methods , Postoperative Complications/etiology , Postoperative Complications/surgery , Tissue Engineering/methods , Animals , Male , Rabbits , Rats , Rats, Nude , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL