Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 30(7): 552-563, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33693705

ABSTRACT

Facioscapulohumeral muscular dystrophy (FSHD) is an inherited muscle disease caused by misexpression of the DUX4 gene in skeletal muscle. DUX4 is a transcription factor, which is normally expressed in the cleavage-stage embryo and regulates gene expression involved in early embryonic development. Recent studies revealed that DUX4 also activates the transcription of repetitive elements such as endogenous retroviruses (ERVs), mammalian apparent long terminal repeat (LTR)-retrotransposons and pericentromeric satellite repeats (Human Satellite II). DUX4-bound ERV sequences also create alternative promoters for genes or long non-coding RNAs, producing fusion transcripts. To further understand transcriptional regulation by DUX4, we performed nanopore long-read direct RNA sequencing (dRNA-seq) of human muscle cells induced by DUX4, because long reads show whole isoforms with greater confidence. We successfully detected differential expression of known DUX4-induced genes and discovered 61 differentially expressed repeat loci, which are near DUX4-ChIP peaks. We also identified 247 gene-ERV fusion transcripts, of which 216 were not reported previously. In addition, long-read dRNA-seq clearly shows that RNA splicing is a common event in DUX4-activated ERV transcripts. Long-read analysis showed non-LTR transposons including Alu elements are also transcribed from LTRs. Our findings revealed further complexity of DUX4-induced ERV transcripts. This catalogue of DUX4-activated repetitive elements may provide useful information to elucidate the pathology of FSHD. Also, our results indicate that nanopore dRNA-seq has complementary strengths to conventional short-read complementary DNA sequencing.


Subject(s)
Homeodomain Proteins/genetics , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Nanopores , Repetitive Sequences, Nucleic Acid/genetics , Sequence Analysis, RNA/methods , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation , Humans , Muscle Cells/metabolism , Muscular Dystrophy, Facioscapulohumeral/pathology , Protein Isoforms/genetics , RNA Isoforms/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA/statistics & numerical data
2.
Hum Mol Genet ; 28(2): 320-331, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30307508

ABSTRACT

Facioscapulohumeral dystrophy type 1 (FSHD-1) is the most common autosomal dominant form of muscular dystrophy with a prevalence of ∼1 in 8000 individuals. It is considered a late-onset form of muscular dystrophy and leads to asymmetric muscle weakness in the facial, scapular, trunk and lower extremities. The prevalent hypothesis on disease pathogenesis is explained by misexpression of a germ line, primate-specific transcription factor DUX4-fl (double homeobox 4, full-length isoform) linked to the chromosome 4q35. In vitro and in vivo studies have demonstrated that very low levels of DUX4-fl expression are sufficient to induce an apoptotic and/or lethal phenotype, and therefore modeling of the disease has proved challenging. In this study, we expand upon our previously established injection model of DUX4 misexpression in zebrafish and describe a DUX4-inducible transgenic zebrafish model that better recapitulates the expression pattern and late onset phenotype characteristic of FSHD patients. We show that an induced burst of DUX4 expression during early development results in the onset of FSHD-like phenotypes in adulthood, even when DUX4 is no longer detectable. We also utilize our injection model to study long-term consequences of DUX4 expression in those that fail to show a developmental phenotype. Herein, we introduce a hypothesis that DUX4 expression during developmental stages is sufficient to induce FSHD-like phenotypes in later adulthood. Our findings point to a developmental role of DUX4 misexpression in the pathogenesis of FSHD and should be factored into the design of future therapies.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Gene Expression , Gene Expression Regulation, Developmental , Muscle Contraction , Muscle, Skeletal/embryology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal , Muscular Dystrophy, Facioscapulohumeral/embryology , Muscular Dystrophy, Facioscapulohumeral/etiology , Muscular Dystrophy, Facioscapulohumeral/genetics , Zebrafish , Zebrafish Proteins/genetics
3.
Biochem Biophys Res Commun ; 512(1): 22-28, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30853177

ABSTRACT

The C-terminal Ig-domain of lamin A plays critical roles in cell function via interaction with proteins, DNA, and chromatin. Mutations in this domain are known to cause various diseases including Emery-Dreifuss muscular dystrophy (EDMD) and familial partial lipodystrophy (FPLD). Here we examined the biophysical and biochemical properties of mutant Ig-domains identified in patients with EDMD and FPLD. EDMD-related mutant Ig-domain showed decreased stability to heat and denaturant. This result was also confirmed by experiments using full-length mutant lamin A, although the decrease in melting temperature was much less than that of the mutant Ig-domain alone. The unstable EDMD Ig-domain disrupted the proper assembly of lamin A, resulting in abnormal paracrystal formation and decreased viscosity. In contrast, FPLD-related mutant Ig-domains were thermally stable, although they lost DNA binding function. Alanine substitution experiments revealed a functional domain of DNA binding in the Ig-domain. Thus, the overall biophysical property of Ig-domains is closely associated with clinical phenotype.


Subject(s)
Lamin Type A/chemistry , Muscular Dystrophy, Emery-Dreifuss/metabolism , Amino Acid Substitution , Biophysical Phenomena , DNA/chemistry , DNA/metabolism , Humans , In Vitro Techniques , Lamin Type A/genetics , Lamin Type A/metabolism , Lipodystrophy, Familial Partial/genetics , Lipodystrophy, Familial Partial/metabolism , Microscopy, Electron, Transmission , Models, Molecular , Muscular Dystrophy, Emery-Dreifuss/genetics , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Domains , Protein Stability , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
4.
J Minim Access Surg ; 14(2): 99-104, 2018.
Article in English | MEDLINE | ID: mdl-28695876

ABSTRACT

INTRODUCTION: Single-incision laparoscopic cholecystectomy (SILC) is widely used as a treatment option for gallbladder disease. However, obesity has been considered a relative contraindication to this approach due to more advanced technical difficulties. The aim of this report was to review our experience with SILC to evaluate the impact of body mass index (BMI) on the surgical outcome. PATIENTS AND METHODS: Between May 2009 and February 2013, 237 patients underwent SILC at our institute. Pre- and post-operative data of the 17 obese patients (O-group) (BMI ≥30 kg/m2) and 220 non-obese patients (NO-group) (BMI <29.9 kg/m2) were compared retrospectively. SILC was performed under general anaesthesia, using glove technique. Indications for surgery included benign gallbladder disease, except for emergent surgeries. RESULTS: Mean age of patients was significantly higher in the NO-group than O-group (58.9 ± 13.5 years vs. 50.8 ± 14.0 years, P = 0.025). SILC was successfully completed in 233 patients (98.3%). Four patients (1.7%) in the NO-group required an additional port, and one patient was converted to an open procedure. The median operative time was 70 ± 25 min in the NO-group and 75.2 ± 18.3 min in the O-group. All complications were minor, except for one case in the NO-group that suffered with leakage of the cystic duct stump, for which endoscopic nasobiliary drainage was need. CONCLUSION: Our findings show that obesity, intended as a BMI ≥30 kg/m2, does not have an adverse impact on the technical difficulty and post-operative outcomes of SILC. Obesity-related comorbidities did not increase the risks for SILC.

5.
Anal Bioanal Chem ; 409(6): 1531-1545, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27913831

ABSTRACT

In order to facilitate reliable and efficient determination of both the particle number concentration (PNC) and the size of nanoparticles (NPs) by single-particle ICP-MS (spICP-MS) without the need to correct for the particle transport efficiency (TE, a possible source of bias in the results), a total-consumption sample introduction system consisting of a large-bore, high-performance concentric nebulizer and a small-volume on-axis cylinder chamber was utilized. Such a system potentially permits a particle TE of 100 %, meaning that there is no need to include a particle TE correction when calculating the PNC and the NP size. When the particle TE through the sample introduction system was evaluated by comparing the frequency of sharp transient signals from the NPs in a measured NP standard of precisely known PNC to the particle frequency for a measured NP suspension, the TE for platinum NPs with a nominal diameter of 70 nm was found to be very high (i.e., 93 %), and showed satisfactory repeatability (relative standard deviation of 1.0 % for four consecutive measurements). These results indicated that employing this total consumption system allows the particle TE correction to be ignored when calculating the PNC. When the particle size was determined using a solution-standard-based calibration approach without an NP standard, the particle diameters of platinum and silver NPs with nominal diameters of 30-100 nm were found to agree well with the particle diameters determined by transmission electron microscopy, regardless of whether a correction was performed for the particle TE. Thus, applying the proposed system enables NP size to be accurately evaluated using a solution-standard-based calibration approach without the need to correct for the particle TE.

6.
Biochim Biophys Acta ; 1852(10 Pt A): 2042-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26170059

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a genetic disorder in which multiple genes are aberrantly spliced. Sarco/endoplasmic reticulum Ca(2+)-ATPase 1 (SERCA1) is one of these genes, and it encodes a P-type ATPase. SERCA1 transports Ca(2+) from the cytosol to the lumen, and is involved in muscular relaxation. It has two splice variants (SERCA1a and SERCA1b) that differ in the last eight amino acids, and the contribution of these variants to DM1 pathology is unclear. Here, we show that SERCA1b protein is highly expressed in DM1 muscle tissue, mainly localised at fast twitch fibres. Additionally, when SERCA1a and SERCA1b were overexpressed in cells, we found that the ATPase and Ca(2+) uptake activity of SERCA1a was almost double that of SERCA1b. Although the affinity for both ATP and Ca(2+) was similar between the two variants, SERCA1b was more sensitive to the inner microsomal environment. Thus, we hypothesise that aberrant expression of SERCA1b in DM1 patients is the cause of abnormal intracellular Ca(2+) homeostasis.

7.
Genes Cells ; 20(2): 121-34, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25403273

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an RNA-mediated disorder characterized by muscle weakness, cardiac defects and multiple symptoms and is caused by expanded CTG repeats within the 3' untranslated region of the DMPK gene. In this study, we found abnormal splicing of actin-binding LIM protein 1 (ABLIM1) in skeletal muscles of patients with DM1 and a DM1 mouse model (HSA(LR) ). An exon 11 inclusion isoform is expressed in skeletal muscle and heart of non-DM1 individuals, but not in skeletal muscle of patients with DM1 or other adult human tissues. Moreover, we determined that ABLIM1 splicing is regulated by several splice factors, including MBNL family proteins, CELF1, 2 and 6, and PTBP1, using a cellular splicing assay. MBNL proteins promoted the inclusion of ABLIM1 exon 11, but other proteins and expanded CUG repeats repressed exon 11 of ABLIM1. This result is consistent with the hypothesis that MBNL proteins are trapped by expanded CUG repeats and inactivated in DM1 and that CELF1 is activated in DM1. However, activation of PTBP1 has not been reported in DM1. Our results suggest that the exon 11 inclusion isoform of ABLIM1 may have a muscle-specific function, and its abnormal splicing could be related to muscle symptoms of DM1.


Subject(s)
CCAAT-Enhancer-Binding Protein-delta/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , LIM Domain Proteins/metabolism , Microfilament Proteins/metabolism , Muscle, Skeletal/metabolism , Myotonic Dystrophy/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , RNA Splicing , RNA-Binding Proteins/metabolism , Animals , Cell Line , Disease Models, Animal , Exons , Humans , Male , Mice
8.
Gan To Kagaku Ryoho ; 43(3): 327-30, 2016 Mar.
Article in Japanese | MEDLINE | ID: mdl-27067848

ABSTRACT

This study was aimed to assess the feasibility and short-term outcomes of adjuvant systemic chemotherapy with either S-1/oxaliplatin (SOX) or mFOLFOX6 (FOLFOX)after curative resection of distant metastases from colorectal cancer. We retrospectively examined 16 patients who underwent R0 resection of colorectal metastases, including the liver (n=6), lung (n=5), lymph node (n=3), and peritoneum (n=2), followed by chemotherapy with SOX (n=7) or FOLFOX (n=9) until disease progression. The mean recurrence-free survival was 13.2 months in the SOX group and 16.9 months in the FOLFOX group. The mean overall survival was 17.9 and 22.9 months, respectively. The number of given courses were 6.5 and 11.0, respectively. Although sensory neuropathy was observed in 38% of the patients, relative dose intensity was higher than 80%. Adjuvant chemotherapy with SOX or FOLFOX was feasible and effective. Further randomized prospective trials are warranted to confirm these results.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/drug therapy , Rectal Neoplasms/drug therapy , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemotherapy, Adjuvant , Colonic Neoplasms/pathology , Colonic Neoplasms/surgery , Drug Combinations , Female , Fluorouracil/therapeutic use , Humans , Leucovorin/therapeutic use , Male , Middle Aged , Neoplasm Metastasis , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Oxonic Acid/administration & dosage , Rectal Neoplasms/pathology , Rectal Neoplasms/surgery , Recurrence , Retrospective Studies , Tegafur/administration & dosage
9.
Hum Mol Genet ; 22(3): 568-77, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23108159

ABSTRACT

Facioscapulohumeral muscular dystrophy (FSHD) is a common form of muscular dystrophy characterized by an asymmetric progressive weakness and wasting of the facial, shoulder and upper arm muscles, frequently accompanied by hearing loss and retinal vasculopathy. FSHD is an autosomal dominant disease linked to chromosome 4q35, but the causative gene remains controversial. DUX4 is a leading candidate gene as causative of FSHD. However, DUX4 expression is extremely low in FSHD muscle, and there is no DUX4 animal model that mirrors the pathology in human FSHD. Here, we show that the misexpression of very low levels of human DUX4 in zebrafish development recapitulates the phenotypes seen in human FSHD patients. Microinjection of small amounts of human full-length DUX4 (DUX4-fl) mRNA into fertilized zebrafish eggs caused asymmetric abnormalities such as less pigmentation of the eyes, altered morphology of ears, developmental abnormality of fin muscle, disorganization of facial musculature and/or degeneration of trunk muscle later in development. Moreover, DUX4-fl expression caused aberrant localization of myogenic cells marked with α-actin promoter-driven enhanced green fluorescent protein outside somite boundary, especially in head region. These abnormalities were rescued by coinjection of the short form of DUX4 (DUX4-s). Our results suggest that the misexpression of DUX4-fl, even at extremely low level, can recapitulate the phenotype observed in FSHD patients in a vertebrate model. These results strongly support the current hypothesis for a role of DUX4 in FSHD pathogenesis. We also propose that DUX4 expression during development is important for the pathogenesis of FSHD.


Subject(s)
Homeodomain Proteins/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Zebrafish/genetics , Actins/genetics , Actins/metabolism , Animals , Disease Models, Animal , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , Microscopy, Electron, Transmission , Muscle, Skeletal/abnormalities , Muscular Dystrophy, Facioscapulohumeral/pathology , Ovum/growth & development , Phenotype , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Shoulder/abnormalities
10.
Biochem Biophys Res Commun ; 461(3): 481-6, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25888793

ABSTRACT

CHRNA1 encodes the α subunit of nicotinic acetylcholine receptors (nAChRs) and is expressed at the neuromuscular junction. Moreover, it is one of the causative genes of Congenital Myasthenic Syndromes (CMS). CHRNA1 undergoes alternative splicing to produce two splice variants: P3A(-), without exon P3A, and P3A(+), with the exon P3A. Only P3A(-) forms functional nAChR. Aberrant alternative splicing caused by intronic or exonic point mutations in patients leads to an extraordinary increase in P3A(+) and a concomitant decrease in P3A(-). Consequently this resulted in a shortage of functional receptors. Aiming to restore the imbalance between the two splice products, antisense oligonucleotides (AONs) were employed to induce exon P3A skipping. Three AON sequences were designed to sterically block the putative binding sequences for splicing factors necessary for exon recognition. Herein, we show that AON complementary to the 5' splice site of the exon was the most effective at exon skipping of the minigene with causative mutations, as well as endogenous wild-type CHRNA1. We conclude that single administration of the AON against the 5' splice site is a promising therapeutic approach for patients based on the dose-dependent effect of the AON and the additive effect of combined AONs. This conclusion is favorable to patients with inherited diseases of uncertain etiology that arise from aberrant splicing leading to a subsequent loss of functional translation products because our findings encourage the option of AON treatment as a therapeutic for these prospectively identified diseases.


Subject(s)
Exons , Myasthenic Syndromes, Congenital/therapy , Oligonucleotides, Antisense/metabolism , RNA Precursors/genetics , RNA, Messenger/genetics , Receptors, Nicotinic/genetics , HEK293 Cells , Humans , RNA Splicing
11.
Biosci Biotechnol Biochem ; 79(6): 912-8, 2015.
Article in English | MEDLINE | ID: mdl-25612552

ABSTRACT

According to the amyloid hypothesis, amyloid ß accumulates in brains with Alzheimer's disease (AD) and triggers cell death and memory deficit. Previously, we developed a rice Aß vaccine expressing Aß, which reduced brain Aß levels in the Tg2576 mouse model of familial AD. We used senescence-accelerated SAMP8 mice as a model of sporadic AD and investigated the relationship between Aß and oxidative stress. Insoluble Aß and 4-hydroxynonenal (4-HNE) levels tended to be reduced in SAMP8 mice-fed the rice Aß vaccine. We attempted to clarify the relationship between oxidative stress and Aß in vitro. Addition of Aß peptide to the culture medium resulted in an increase in 4-HNE levels in SH-SY5Y cells. Tg2576 mice, which express large amounts of Aß in their brain, also exhibited increased 4-HNE levels; this increase was inhibited by the Aß vaccine. These results indicate that Aß induces oxidative stress in cultured cells and in the mouse brain.


Subject(s)
Aging , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Oxidative Stress , Peptide Fragments/metabolism , Aldehydes/metabolism , Alzheimer Disease/physiopathology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Animals , Brain/metabolism , Buffers , Humans , Male , Maze Learning , Mice , Mice, Transgenic , Oryza/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Solubility , Vaccines/genetics
12.
Am J Hum Genet ; 88(6): 845-851, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21665002

ABSTRACT

Congenital muscular dystrophy is a heterogeneous group of inherited muscle diseases characterized clinically by muscle weakness and hypotonia in early infancy. A number of genes harboring causative mutations have been identified, but several cases of congenital muscular dystrophy remain molecularly unresolved. We examined 15 individuals with a congenital muscular dystrophy characterized by early-onset muscle wasting, mental retardation, and peculiar enlarged mitochondria that are prevalent toward the periphery of the fibers but are sparse in the center on muscle biopsy, and we have identified homozygous or compound heterozygous mutations in the gene encoding choline kinase beta (CHKB). This is the first enzymatic step in a biosynthetic pathway for phosphatidylcholine, the most abundant phospholipid in eukaryotes. In muscle of three affected individuals with nonsense mutations, choline kinase activities were undetectable, and phosphatidylcholine levels were decreased. We identified the human disease caused by disruption of a phospholipid de novo biosynthetic pathway, demonstrating the pivotal role of phosphatidylcholine in muscle and brain.


Subject(s)
Choline Kinase/genetics , Mitochondria, Muscle/pathology , Muscular Dystrophies/congenital , Muscular Dystrophies/pathology , Phosphatidylcholines/biosynthesis , Adolescent , Child , Child, Preschool , Female , Humans , Intellectual Disability/genetics , Male , Mitochondria, Muscle/genetics , Muscular Dystrophies/genetics , Mutation , Pedigree , Phosphatidylcholines/genetics , Polymorphism, Genetic , Young Adult
13.
Dev Biol ; 361(1): 79-89, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22020047

ABSTRACT

Filamin C is an actin-crosslinking protein that is specifically expressed in cardiac and skeletal muscles. Although mutations in the filamin C gene cause human myopathy with cardiac involvement, the function of filamin C in vivo is not yet fully understood. Here we report a medaka mutant, zacro (zac), that displayed an enlarged heart, caused by rupture of the myocardiac wall, and progressive skeletal muscle degeneration in late embryonic stages. We identified zac to be a homozygous nonsense mutation in the filamin C (flnc) gene. The medaka filamin C protein was found to be localized at myotendinous junctions, sarcolemma, and Z-disks in skeletal muscle, and at intercalated disks in the heart. zac embryos showed prominent myofibrillar degeneration at myotendinous junctions, detachment of myofibrils from sarcolemma and intercalated disks, and focal Z-disk destruction. Importantly, the expression of γ-actin, which we observed to have a strong subcellular localization at myotendinous junctions, was specifically reduced in zac mutant myotomes. Inhibition of muscle contraction by anesthesia alleviated muscle degeneration in the zac mutant. These results suggest that filamin C plays an indispensable role in the maintenance of the structural integrity of cardiac and skeletal muscles for support against mechanical stress.


Subject(s)
Cardiomegaly/genetics , Contractile Proteins/genetics , Contractile Proteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Disorders, Atrophic/genetics , Myocardium/metabolism , Oryzias/embryology , Actins/metabolism , Animals , Birefringence , Cloning, Molecular , Codon, Nonsense/genetics , DNA Primers/genetics , Filamins , Fluorescent Antibody Technique , In Situ Hybridization , Microscopy, Electron, Transmission , Oligonucleotides, Antisense/genetics , Oryzias/genetics , Polymorphism, Restriction Fragment Length/genetics , Sarcolemma/pathology
14.
Am J Pathol ; 180(4): 1570-80, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22349301

ABSTRACT

Myofibrillar myopathy (MFM) is a group of disorders that are pathologically defined by the disorganization of the myofibrillar alignment associated with the intracellular accumulation of Z-disk-associated proteins. MFM is caused by mutations in genes encoding Z-disk-associated proteins, including myotilin. Although a number of MFM mutations have been identified, it has been difficult to elucidate the precise roles of the mutant proteins. Here, we present a useful method for the characterization of mutant proteins associated with MFM. Expression of mutant myotilins in mouse tibialis anterior muscle by in vivo electroporation recapitulated both the pathological changes and the biochemical characteristics observed in patients with myotilinopathy. In mutant myotilin-expressing muscle fibers, myotilin aggregates and is costained with polyubiquitin, and Z-disk-associated proteins and myofibrillar disorganization were commonly seen. In addition, the expressed S60C mutant myotilin protein displayed marked detergent insolubility in electroporated mouse muscle, similar to that observed in human MFM muscle with the same mutation. Thus, in vivo electroporation can be a useful method for evaluating the pathogenicity of mutations identified in MFM.


Subject(s)
Cytoskeletal Proteins/genetics , Muscle Proteins/genetics , Muscular Diseases/genetics , Mutation , Animals , Cells, Cultured , Connectin , Cytoskeletal Proteins/metabolism , Electroporation/methods , Female , Humans , Mice , Microfilament Proteins , Microscopy, Electron , Middle Aged , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Muscular Diseases/metabolism , Muscular Diseases/pathology , Myoblasts/metabolism , Myofibrils/metabolism , Polyubiquitin/metabolism , Protein Unfolding
15.
Surg Innov ; 20(6): NP30-4, 2013 Dec.
Article in English | MEDLINE | ID: mdl-22441807

ABSTRACT

Schwannomas are benign tumors that arise from neural sheath Schwann cells. Solitary benign schwannoma is generally located in the head and neck and is a rare neoplasm among the tumors of the retroperitoneal space. Reports of laparoscopic excision of retroperitoneal schwannomas have recently been on the increase. However, few cases of single-port laparoscopic excision of these tumors have been reported. Moreover, there are no reports of single-port excision of schwannomas attached to the body of pancreas and around the splenic vessels. This is the first report of a schwannoma lying adjacent to the body of the pancreas between the splenic artery and vein that was excised by single-port laparoscopic surgery. The most notable aspect of our procedure is the use of bipolar forceps. Single-port laparoscopic excision using bipolar forceps is a feasible and safe procedure for retroperitoneal solitary tumors, even when they are close to the splenic artery and vein.


Subject(s)
Laparoscopy/methods , Neurilemmoma/surgery , Pancreatic Neoplasms/surgery , Retroperitoneal Neoplasms/surgery , Female , Humans , Middle Aged , Retroperitoneal Space/surgery
16.
J Cell Sci ; 123(Pt 22): 3893-900, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20980393

ABSTRACT

Mutations in LMNA, which encodes A-type nuclear lamins, cause various human diseases, including myopathy, cardiomyopathy, lipodystrophy and progeria syndrome. To date, little is known about how mutations in a single gene cause a wide variety of diseases. Here, by characterizing an antibody that specifically recognizes the phosphorylation of Ser458 of A-type lamins, we uncover findings that might contribute to our understanding of laminopathies. This antibody only reacts with nuclei in muscle biopsies from myopathy patients with mutations in the Ig-fold motif of A-type lamins. Ser458 phosphorylation is not seen in muscles from control patients or patients with any other neuromuscular diseases. In vitro analysis confirmed that only lamin A mutants associated with myopathy induce phosphorylation of Ser458, whereas lipodystrophy- or progeria-associated mutants do not. We also found that Akt1 directly phosphorylates Ser458 of lamin A with myopathy-related mutations in vitro. These results suggest that Ser458 phosphorylation of A-type lamins correlates with striated muscle laminopathies; this might be useful for the early diagnosis of LMNA-associated myopathies. We propose that disease-specific phosphorylation of A-type lamins by Akt1 contributes to myopathy caused by LMNA mutations.


Subject(s)
Lamin Type A/metabolism , Muscular Dystrophies/metabolism , Adult , Animals , COS Cells , Child , Child, Preschool , Chlorocebus aethiops , Female , Humans , Immunohistochemistry , Lamin Type A/genetics , Male , Mice , Middle Aged , Muscular Dystrophies/genetics , Phosphorylation , Transfection
17.
Ann Neurol ; 69(6): 1005-13, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21391237

ABSTRACT

OBJECTIVE: Emery-Dreifuss muscular dystrophy (EDMD) is a genetically heterogeneous muscular disease that presents with muscular dystrophy, joint contractures, and cardiomyopathy with conduction defects. Mutations in several nuclear envelope protein genes have been associated with EDMD in less than half of patients, implying the existence of other causative and modifier genes. We therefore analyzed TMEM43, which encodes LUMA, a newly identified nuclear membrane protein and also a binding partner of emerin and lamins, to investigate whether LUMA may contribute to the pathomechanism of EDMD-related myopathy. METHODS: Forty-one patients with EDMD-related myopathy were enrolled. In vitro and in vivo transfection analyses were performed to assay the binding partners and oligomerization of mutant LUMA. RESULTS: We identified heterozygous missense mutations, p.Glu85Lys and p.Ile91Val in TMEM43, in 2 EDMD-related myopathy patients. Reduced nuclear staining of LUMA was observed in the muscle from the patient with p.Glu85Lys mutation. By in vitro transfection analysis, p.Glu85Lys mutant LUMA resulted to failure in oligomerization, a process that may be important for protein complex formation on nuclear membrane. Furthermore, we demonstrated for the first time that LUMA can interact with another nuclear membrane protein, SUN2, in addition to emerin. Cells expressing mutant LUMA revealed reduced nuclear staining with or without aggregates of emerin and SUN2 together with a higher proportion of abnormally shaped nuclei. In vivo expression of mutant LUMA by electroporation in mouse tibialis anterior muscles likewise demonstrated the decreased staining of emerin and SUN2 on myonuclei. INTERPRETATION: Our results suggest that mutant LUMAs may be associated with EDMD-related myopathy.


Subject(s)
Membrane Proteins/genetics , Muscle, Skeletal/pathology , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/pathology , Mutation, Missense/genetics , Adult , Aged , Animals , Cell Line, Transformed , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Mutational Analysis/methods , Electroporation/methods , Female , Gene Expression Regulation/genetics , Humans , Immunoprecipitation/methods , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice , Muscle, Skeletal/metabolism , Mutagenesis, Site-Directed/methods , Transfection/methods
18.
Gan To Kagaku Ryoho ; 38(1): 121-4, 2011 Jan.
Article in Japanese | MEDLINE | ID: mdl-21368472

ABSTRACT

Cetuximab is an agent approved as epidermal growth factor receptor (EGFR)-positive for unresectable advanced or recurrent colorectal cancer. A 58-year-old man with liver metastasis had relapsed after resection of rectal cancer. We treated him with cetuximab monotherapy as third-line chemotherapy after treatment failures with infusional 5-FU, LV and oxaliplatin (FOLFOX4 regi- men); and infusional 5-FU, LV and irinotecan(FOLFIRI regimen). The patient was administered cetuximab (400 mg/m² initial dose and 250 mg/m²/ week thereafter). After sixteen weeks of treatment, a computed tomography scan revealed reduced sizes of the liver metastases. The tumor response has still been maintained after thirty courses of treatment, and the chemotherapeutic response was evaluated as a partial response according to the Response Evaluation Criteria In Solid Tumor guidelines. The main toxicity was a grade 2 rash, but was manageable by topical steroid and moisturizing agent. We have added some review of the literature, and the cetuximab therapy is reported.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Drug Resistance, Neoplasm/drug effects , Rectal Neoplasms/drug therapy , Salvage Therapy , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Cetuximab , Fluorouracil/therapeutic use , Humans , Leucovorin/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Male , Middle Aged , Organoplatinum Compounds/therapeutic use , Rectal Neoplasms/pathology , Recurrence
19.
Stem Cell Reports ; 16(6): 1527-1541, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34048688

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an adult-onset incurable motor neuron (MN) disease. The reasons for selective MN vulnerability in ALS are unknown. Axonal pathology is among the earliest signs of ALS. We searched for novel modulatory genes in human MN axon shortening affected by TARDBP mutations. In transcriptome analysis of RNA present in the axon compartment of human-derived induced pluripotent stem cell (iPSC)-derived MNs, PHOX2B (paired-like homeobox protein 2B) showed lower expression in TARDBP mutant axons, which was consistent with axon qPCR and in situ hybridization. PHOX2B mRNA stability was reduced in TARDBP mutant MNs. Furthermore, PHOX2B knockdown reduced neurite length in human MNs. Finally, phox2b knockdown in zebrafish induced short spinal axons and impaired escape response. PHOX2B is known to be highly express in other types of neurons maintained after ALS progression. Collectively, TARDBP mutations induced loss of axonal resilience, which is an important ALS-related phenotype mediated by PHOX2B downregulation.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Axons/metabolism , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Motor Neurons/metabolism , Transcription Factors/metabolism , Zebrafish/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , DNA-Binding Proteins/genetics , Gene Expression Regulation , Gene Knockdown Techniques/methods , Homeodomain Proteins/genetics , Humans , Mutation , Phenotype , Transcription Factors/genetics , Transcriptome
20.
Mob DNA ; 11: 29, 2020.
Article in English | MEDLINE | ID: mdl-32963593

ABSTRACT

BACKGROUND: Endogenous retroviruses (ERVs) are remnants of ancient retroviral infections of mammalian germline cells. A large proportion of ERVs lose their open reading frames (ORFs), while others retain them and become exapted by the host species. However, it remains unclear what proportion of ERVs possess ORFs (ERV-ORFs), become transcribed, and serve as candidates for co-opted genes. RESULTS: We investigated characteristics of 176,401 ERV-ORFs containing retroviral-like protein domains (gag, pro, pol, and env) in 19 mammalian genomes. The fractions of ERVs possessing ORFs were overall small (~ 0.15%) although they varied depending on domain types as well as species. The observed divergence of ERV-ORF from their consensus sequences showed bimodal distributions, suggesting that a large proportion of ERV-ORFs either recently, or anciently, inserted themselves into mammalian genomes. Alternatively, very few ERVs lacking ORFs were found to exhibit similar divergence patterns. To identify candidates for ERV-derived genes, we estimated the ratio of non-synonymous to synonymous substitution rates (dN/dS) for ERV-ORFs in human and non-human mammalian pairs, and found that approximately 42% of the ERV-ORFs showed dN/dS < 1. Further, using functional genomics data including transcriptome sequencing, we determined that approximately 9.7% of these selected ERV-ORFs exhibited transcriptional potential. CONCLUSIONS: These results suggest that purifying selection operates on a certain portion of ERV-ORFs, some of which may correspond to uncharacterized functional genes hidden within mammalian genomes. Together, our analyses suggest that more ERV-ORFs may be co-opted in a host-species specific manner than we currently know, which are likely to have contributed to mammalian evolution and diversification.

SELECTION OF CITATIONS
SEARCH DETAIL