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










Publication year range
1.
Sci Adv ; 9(48): eadj4605, 2023 12.
Article in English | MEDLINE | ID: mdl-38019907

ABSTRACT

Protein degradation in eukaryotic cells is mainly carried out by the 26S proteasome, a macromolecular complex not only present in the cytosol and nucleus but also associated with various membranes. How proteasomes are anchored to the membrane and the biological meaning thereof have been largely unknown in higher organisms. Here, we show that N-myristoylation of the Rpt2 subunit is a general mechanism for proteasome-membrane interaction. Loss of this modification in the Rpt2-G2A mutant cells leads to profound changes in the membrane-associated proteome, perturbs the endomembrane system, and undermines critical cellular processes such as cell adhesion, endoplasmic reticulum-associated degradation and membrane protein trafficking. Rpt2G2A/G2A homozygous mutation is embryonic lethal in mice and is sufficient to abolish tumor growth in a nude mice xenograft model. These findings have defined an evolutionarily conserved mechanism for maintaining membrane protein homeostasis and underscored the significance of compartmentalized protein degradation by myristoyl-anchored proteasomes in health and disease.


Subject(s)
Membrane Proteins , Proteasome Endopeptidase Complex , Humans , Animals , Mice , Proteasome Endopeptidase Complex/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proteostasis , Endoplasmic Reticulum-Associated Degradation , Mice, Nude , Lipids
2.
bioRxiv ; 2023 May 12.
Article in English | MEDLINE | ID: mdl-37214852

ABSTRACT

Protein degradation in eukaryotic cells is mainly carried out by the 26S proteasome, a macromolecular complex not only present in the cytosol and nucleus but also associated with various membranes. How proteasomes are anchored to the membrane and the biological meaning thereof have been largely unknown in higher organisms. Here we show that N-myristoylation of the Rpt2 subunit is a general mechanism for proteasome-membrane interaction. Loss of this modification in the Rpt2-G2A mutant cells leads to profound changes in the membrane-associated proteome, perturbs the endomembrane system and undermines critical cellular processes such as cell adhesion, endoplasmic reticulum-associated degradation (ERAD) and membrane protein trafficking. Rpt2 G2A/G2A homozygous mutation is embryonic lethal in mice and is sufficient to abolish tumor growth in a nude mice xenograft model. These findings have defined an evolutionarily conserved mechanism for maintaining membrane protein homeostasis and underscored the significance of compartmentalized protein degradation by m yristoyl- a nchored p roteasomes (MAPs) in health and disease.

3.
Anal Methods ; 14(46): 4842-4850, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36398599

ABSTRACT

Copper acetate/silver nitrate/polyvinylpyrrolidone was first prepared into nano-hybrid silver-doped copper oxide by electrospinning, and then nano-honeycomb particles were produced through heat-treatment. For the first time, honeycomb Ag@CuO nanoparticles were prepared by electrospinning, and a H2O2 sensor was constructed by modifying the carbon paste electrode (CPE) with the honeycomb Ag@CuO nanoparticles. This work performed the structural, morphological, and phase analysis of the Ag@CuO nanoparticles by scanning electron microscopy (SEM) and X-ray diffraction (XRD). The results indicated the synthesis of Ag@CuO hybrid nanoparticles with high purity, and cyclic voltammetry and amperometry show that the Ag@CuO modified electrode has high electrocatalytic performances with fast voltammetric responses and a notably decreased overpotential compared to that of even the CuO modified CPE. In addition, the Ag/CuO-CPE based H2O2 sensor has the highest sensitivity of 1982.14 µA (mmol L-1)-1 cm-2, the lowest detection limit of 0.01 µmol L-1 ((S/N) = 3), and the measured linear response for H2O2 oxidation ranged from 0.05 µmol L-1 to 100 µmol L-1 and 100 µmol L-1 to 1.5 mmol L-1. The proposed method was applied to the determination of H2O2 in coconut fruit samples from canned coconut, and the satisfactory results confirmed the applicability of this sensor in practical analysis.


Subject(s)
Nanoparticles , Porifera , Animals , Hydrogen Peroxide , Copper , Carbon , Fruit
4.
Ann Palliat Med ; 11(4): 1391-1400, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35523747

ABSTRACT

BACKGROUND: Trigeminal neuralgia (TN) is a type of transient and paroxysmal recurrent severe pain confined to the trigeminal nerve region. This study systematically evaluated the efficacy and safety of microvascular decompression (MVD) and percutaneous balloon compression (PBC) in the treatment of TN. METHODS: PubMed, Embase, The Cochrane Library, China National Knowledge Infrastructure, Wanfang, and Weipu databases were searched for articles published on the use of MVD and PBC in the treatment of TN from the dates of inception of the databases to October 2019. Articles on MVD and PBC in the treatment of TN were selected, and a meta-analysis was performed using RevMan 5.2 software. RESULTS: Eighteen studies (comprising 1,932 patients) were included in the study. MVD and PBC had similar overall effective rates in treating TN [odds ratio (OR) =0.79, 95% confidence interval (CI): 0.55-1.13, P=0.19]. Patients treated with PBC had a higher recurrence rate of TN than those treated MVD (OR =3.50, 95% CI: 2.25-5.44; P<0.00001), and patients treated with PBC experienced more adverse reactions than those treated with MVD (OR =17.79, 95% CI: 10.17-31.11; P<0.00001). DISCUSSION: The overall effective rates of PBC and MVD in the treatment of TN ewer similar, but MVD was associated with better recurrence and a lower rate of adverse reactions. Thus, both MVD and PBC can be used to effectively treat TN patients.


Subject(s)
Balloon Occlusion , Microvascular Decompression Surgery , Trigeminal Neuralgia , Balloon Occlusion/adverse effects , Humans , Pain/etiology , Retrospective Studies , Treatment Outcome , Trigeminal Neuralgia/etiology , Trigeminal Neuralgia/surgery
5.
Materials (Basel) ; 12(24)2019 Dec 09.
Article in English | MEDLINE | ID: mdl-31835348

ABSTRACT

This article selects studies on the preparation of fluorinated polyurethane-nano-alumina composite coating materials, and analyzes the anti-wear, water resistantance, and surface microstructure. Attenuated total reflection-Fourier transform infrared spectroscopy (ATR-FTIR) shows that the polyurethane synthesized in this study does not contain hydrophilic -CH2OH groups. The cavitation wear test depicts that the actual cavitation amount C of the Al2O3-FPU (4) (fluorinated polyurethane) coating is 0.9035 × 10-3 kg, and the anti-wear ability increases by 61.9% compared with FPU-0.5. The water-resistant test shows that the contact angle of water droplets on the surface of the coating increase from 95.3° of FPU-0.5 to 123.1° of Al2O3-FPU (4), and the water absorption decreases from 2.52% to 1.04%. Scanning electron microscopy (SEM) observation confirms that alumina particles can protrude on the coating surface and resist strong wear, while the C-F chain with high bond energy at the near-surface exhibits high strength and water resistance, which prevents wear from spreading deep into the coating. Differential scanning calorimetry (DSC) results show that the Tg(HS) value of the hard segment phase decreases with higher external force. Notably, when the coating is subjected to erosion, which enhances the crystallinity of the hard segment phase, the tensile strength of the hard segment phase of the coating surface is improved, which supports the wear resistance. Herein, we show that the addition of nano-alumina to fluorinated polyurethanes can control high water and abrasion resistance.

6.
J Cell Mol Med ; 23(4): 2536-2548, 2019 04.
Article in English | MEDLINE | ID: mdl-30672133

ABSTRACT

Evidence indicated that GATA5 may suppress hepatocellular carcinoma (HCC) cell malignant transformation, but the mechanism of how GATA5 affects cancer cell reprogramming to inhibit HCC malignant behaviour is still unclear. In this study, we report that the expression of ß-catenin and reprogramming genes p-Oct4, Nanog, Klf4, c-myc and EpCAM was significantly higher in HCC tissues compared to normal liver tissues. In contrast, the expression of GATA5 was significantly lower in HCC tissues compared to normal liver tissues. Transfection of CDH-GATA5 vectors into HCC cells (HLE, Bel 7402 and PLC/PRF/5 cells) increased the GATA5 expression and decreased the expression of ß-catenin and reprogramming genes p-Oct4, Nanog, Klf4, c-myc and EpCAM. Increased GATA5 expression by transfection with its expression vectors was also able to inhibit the cell growth, colony formation and capability of migration, invasion, while promoting apoptosis in HCC cells. Results revealed that GATA5 co-localization with ß-catenin in the cytoplasm, preventing ß-catenin from entering the nucleus. Treatment with the specific Wnt/ß-catenin pathway inhibitor salinomycin was able to reduce the expression of ß-catenin and reprogramming genes. Salinomycin exerted a similar influence as GATA5, and siRNA-GATA5 restored ß-catenin and reprogramming gene expression. This study demonstrates that an increase in the expression of GATA5 inhibits the expression of ß-catenin and reprogramming genes and suppresses tumour growth, colony formation, metastasis and invasion, while promoting apoptosis in HCC cells. The mechanism of GATA5 inhibiting the malignant behaviours of HCC cells may involve in the disruption of the Wnt/ß-catenin pathway and the reduction of reprogramming gene expression.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Transformation, Neoplastic/genetics , GATA5 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , beta Catenin/genetics , Adult , Aged , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Case-Control Studies , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cell Adhesion Molecule/antagonists & inhibitors , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Female , GATA5 Transcription Factor/antagonists & inhibitors , GATA5 Transcription Factor/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Male , Middle Aged , Nanog Homeobox Protein/antagonists & inhibitors , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/antagonists & inhibitors , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Pyrans/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Wnt Signaling Pathway , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
7.
Cell Tissue Res ; 364(3): 637-646, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26711913

ABSTRACT

Fibulin-4 is an extracellular matrix glycoprotein essential for elastic fiber formation. Mice deficient in fibulin-4 die perinatally because of severe pulmonary and vascular defects associated with the lack of intact elastic fibers. Patients with fibulin-4 mutations demonstrate similar defects, and a significant number die shortly after birth or in early childhood from cardiopulmonary failure. The patients also demonstrate skeletal and other systemic connective tissue abnormalities, including joint laxity and flexion contractures of the wrist. A fibulin-4 null mouse strain was generated and used to analyze the roles of fibulin-4 in tendon fibrillogenesis. This mouse model displayed bilateral forelimb contractures, in addition to pulmonary and cardiovascular defects. The forelimb and hindlimb tendons exhibited disruption in collagen fibrillogenesis in the absence of fibulin-4 as analyzed by transmission electron microscopy. Fewer fibrils were assembled, and fibrils were disorganized compared with wild-type controls. The organization of developing tenocytes and compartmentalization of the extracellular space was also disrupted. Fibulin-4 was co-localized with fibrillin-1 and fibrillin-2 in limb tendons by using immunofluorescence microscopy. Thus, fibulin-4 seems to play a role in regulating tendon collagen fibrillogenesis, in addition to its essential function in elastogenesis.


Subject(s)
Collagen/metabolism , Contracture/metabolism , Contracture/pathology , Extracellular Matrix Proteins/deficiency , Forelimb/pathology , Tendons/abnormalities , Animals , Contracture/complications , Extracellular Matrix Proteins/metabolism , Fibrillins/metabolism , Hernia/complications , Hernia/pathology , Phenotype , Protein Transport , Tendons/metabolism , Tendons/ultrastructure
8.
J Biol Chem ; 289(15): 10293-10307, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24563484

ABSTRACT

Dominant and recessive mutations in collagen VI genes, COL6A1, COL6A2, and COL6A3, cause a continuous spectrum of disorders characterized by muscle weakness and connective tissue abnormalities ranging from the severe Ullrich congenital muscular dystrophy to the mild Bethlem myopathy. Herein, we report the development of a mouse model for dominant collagen VI disorders by deleting exon 16 in the Col6a3 gene. The resulting heterozygous mouse, Col6a3(+/d16), produced comparable amounts of normal Col6a3 mRNA and a mutant transcript with an in-frame deletion of 54 bp of triple-helical coding sequences, thus mimicking the most common molecular defect found in dominant Ullrich congenital muscular dystrophy patients. Biosynthetic studies of mutant fibroblasts indicated that the mutant α3(VI) collagen protein was produced and exerted a dominant-negative effect on collagen VI microfibrillar assembly. The distribution of the α3(VI)-like chains of collagen VI was not altered in mutant mice during development. The Col6a3(+/d16) mice developed histopathologic signs of myopathy and showed ultrastructural alterations of mitochondria and sarcoplasmic reticulum in muscle and abnormal collagen fibrils in tendons. The Col6a3(+/d16) mice displayed compromised muscle contractile functions and thereby provide an essential preclinical platform for developing treatment strategies for dominant collagen VI disorders.


Subject(s)
Collagen Type VI/chemistry , Collagen Type VI/genetics , Disease Models, Animal , Muscular Diseases/physiopathology , Alleles , Animals , Exons , Extracellular Matrix/metabolism , Female , Fibroblasts/metabolism , Genes, Dominant , Heterozygote , Mice , Mice, Transgenic , Mitochondria/pathology , Mitochondria/ultrastructure , Muscle Contraction , Muscles/physiopathology , Muscular Diseases/genetics , Muscular Dystrophies/genetics , Phenotype , Sarcoplasmic Reticulum/pathology , Sequence Deletion , Tendons/pathology
9.
J Biol Chem ; 288(20): 14320-14331, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23564457

ABSTRACT

Collagen VI is a ubiquitously expressed extracellular microfibrillar protein. Its most common molecular form is composed of the α1(VI), α2(VI), and α3(VI) collagen α chains encoded by the COL6A1, COL6A2, and COL6A3 genes, respectively. Mutations in any of the three collagen VI genes cause congenital muscular dystrophy types Bethlem and Ullrich as well as intermediate phenotypes characterized by muscle weakness and connective tissue abnormalities. The α3(VI) collagen α chain has much larger N- and C-globular domains than the other two chains. Its most C-terminal domain can be cleaved off after assembly into microfibrils, and the cleavage product has been implicated in tumor angiogenesis and progression. Here we characterize a Col6a3 mutant mouse that expresses a very low level of a non-functional α3(VI) collagen chain. The mutant mice are deficient in extracellular collagen VI microfibrils and exhibit myopathic features, including decreased muscle mass and contractile force. Ultrastructurally abnormal collagen fibrils were observed in tendon, but not cornea, of the mutant mice, indicating a distinct tissue-specific effect of collagen VI on collagen I fibrillogenesis. Overall, the mice lacking normal α3(VI) collagen chains displayed mild musculoskeletal phenotypes similar to mice deficient in the α1(VI) collagen α chain, suggesting that the cleavage product of the α3(VI) collagen does not elicit essential functions in normal growth and development. The Col6a3 mouse mutant lacking functional α3(VI) collagen chains thus serves as an animal model for COL6A3-related muscular dystrophy.


Subject(s)
Collagen Type VI/deficiency , Collagen Type VI/genetics , Muscle, Skeletal/metabolism , Tendons/metabolism , Animals , Collagen Type VI/physiology , Disease Models, Animal , Extracellular Matrix/metabolism , Genotype , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microfibrils/metabolism , Muscle, Skeletal/physiopathology , Mutation , Phenotype , Tendons/physiopathology
10.
Surgery ; 150(2): 306-15, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21719059

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDA) is a highly lethal disease; a prominent desmoplastic reaction is a defining characteristic. Fibrillar collagens, such as collagen I and to a lesser extent, collagens III and V, comprise the majority of this stromal fibrosis. Type VI collagen (COL6) forms a microfibrillar network associated with type I collagen fibrils. The expression of COL6 has been linked with inflammation and survival. Importantly, tumor-specific alternative splicing in COL6A3 has been identified in several cancers by genome exon arrays. We evaluated the expression and localization of COL6A3 in PDA and premalignant lesions and explored the presence of alternative splicing events. METHODS: We analyzed paired PDA-normal (n = 18), intraductal papillary mucinous neoplasms (IPMN; n = 5), pancreatic cystadenoma (n = 5), and 8 PDA cell lines with reverse transcriptase polymerase chain reaction, using unique primers that identify total COL6A3 gene and alternative splicing sites in several of its exons. Western blot analysis and immunohistochemistry were used to analyze the expression levels and localization of COL6A3 protein in the different lesions, and in 2 animal models of PDA. RESULTS: COL6A3 protein levels were significantly upregulated in 77% of the paired PDA-adjacent tissue examined. COL6A3 was mainly present in the desmoplastic stroma of PDA, with high deposition around the malignant ducts and in between the sites of stromal fatty infiltration. Analysis of the COL6A3 splice variants showed tumor-specific consistent inclusion of exons 3 and 6 in 17 of the 18 (94%) paired PDA-adjacent tissues. Inclusion of exon 4 was exclusively tumor specific, with barely detectable expression in the adjacent tissues. IPMN and pancreatic cystadenomas showed no expression of any of the examined exons. Total COL6A3 mRNA and exon 6 were identified in 6 PDA cell lines, but only 2 cell lines (MIA PACA-2 and ASPC-1) expressed exons 3 and 4. In both the xenograft and transgenic models of PDA, COL6A3 immunoreactivity was present in the stroma and some PDA cells. CONCLUSION: We have described, for the first time, a dynamic process of tumor-specific alternative splicing in several exons of stromal COL6A3. Alternatively spliced proteins may contribute to the etiology or progression of cancer and may serve as markers for cancer diagnosis. Identification of COL6A3 isoforms as PDA-specific provides the basis for future studies to explore the oncogenic and diagnostic potential of these alternative splicing events.


Subject(s)
Alternative Splicing , Carcinoma, Pancreatic Ductal/genetics , Collagen Type VI/genetics , Pancreatic Neoplasms/genetics , Aged , Animals , Female , Humans , Male , Mice , Mice, Nude , Middle Aged , Precancerous Conditions/genetics
11.
J Biol Chem ; 285(13): 10005-10015, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20106987

ABSTRACT

Ullrich congenital muscular dystrophy (UCMD) is a disabling and life-threatening disorder resulting from either recessive or dominant mutations in genes encoding collagen VI. Although the majority of the recessive UCMD cases have frameshift or nonsense mutations in COL6A1, COL6A2, or COL6A3, recessive structural mutations in the COL6A2 C-globular region are emerging also. However, the underlying molecular mechanisms have remained elusive. Here we identified a homozygous COL6A2 E624K mutation (C1 subdomain) and a homozygous COL6A2 R876S mutation (C2 subdomain) in two UCMD patients. The consequences of the mutations were investigated using fibroblasts from patients and cells stably transfected with the mutant constructs. In contrast to expectations based on the clinical severity of these two patients, secretion and assembly of collagen VI were moderately affected by the E624K mutation but severely impaired by the R876S substitution. The E624K substitution altered the electrostatic potential of the region surrounding the metal ion-dependent adhesion site, resulting in a collagen VI network containing thick fibrils and spots with densely packed microfibrils. The R876S mutation prevented the chain from assembling into triple-helical collagen VI molecules. The minute amount of collagen VI secreted by the R876S fibroblasts was solely composed of a faster migrating chain corresponding to the C2a splice variant with an alternative C2 subdomain. In transfected cells, the C2a splice variant was able to assemble into short microfibrils. Together, the results suggest that the C2a splice variant may functionally compensate for the loss of the normal COL6A2 chain when mutations occur in the C2 subdomain.


Subject(s)
Alternative Splicing , Collagen Type VI/genetics , Genes, Recessive , Muscular Dystrophies/congenital , Muscular Dystrophies/genetics , Mutation, Missense , Adult , Amino Acid Sequence , Biopsy , Child , Collagen/chemistry , Female , Fibroblasts/metabolism , Homozygote , Humans , Ions , Male , Molecular Sequence Data , Sequence Homology, Amino Acid
12.
J Neuropathol Exp Neurol ; 67(2): 144-54, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18219255

ABSTRACT

Mutations in the extracellular matrix molecule collagen VI underlie the congenital muscular dystrophy types Ullrich and Bethlem. Establishing the origin of collagen VI in muscle is important for understanding the pathophysiology of these diseases and for developing future treatment approaches involving cell-specific delivery. Because the cells that produce collagen VI cannot be identified by histologic analysis, we examined the production of collagen VI in pure cultures of primary myogenic cells and muscle interstitial fibroblasts from limb muscle of neonatal mice. Immunofluorescence staining and Western blot analysis revealed secretion and matrix deposition of collagen VI by interstitial fibroblasts but not by myogenic cells in vitro. Using Northern blot and real-time reverse-transcriptase-polymerase chain reaction analysis for the collagen VI genes col6a1, col6a2, col6a3, transcript levels for the 3 mRNAs were high in interstitial fibroblasts, whereas in primary myogenic cells, they were indistinguishable from background. Furthermore, retention of mutant collagen VI in muscle from 3 patients with collagen VI mutation was identified in interstitial fibroblastic cells but not in their myofibers. These results suggest that interstitial fibroblasts but not myogenic cells contribute significantly to the deposition of collagen VI in the extracellular matrix in skeletal muscle and imply major roles of this cell type and the extracellular matrix in the pathogenesis of these diseases.


Subject(s)
Collagen Type VI/genetics , Collagen Type VI/metabolism , Fibroblasts/metabolism , Muscle, Skeletal/cytology , Muscular Dystrophies/pathology , Mutation , Animals , Animals, Newborn , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques/methods , Gene Expression Regulation/physiology , Humans , Mice , Mice, Inbred C57BL , Microscopy, Confocal/methods , Muscle, Skeletal/enzymology , Skin/cytology
13.
Mol Cell Biol ; 28(3): 1061-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18070922

ABSTRACT

Fibulin-2 is an extracellular matrix protein belonging to the five-member fibulin family, of which two members have been shown to play essential roles in elastic fiber formation during development. Fibulin-2 interacts with two major constituents of elastic fibers, tropoelastin and fibrillin-1, in vitro and localizes to elastic fibers in many tissues in vivo. The protein is prominently expressed during morphogenesis of the heart and aortic arch vessels and at early stages of cartilage development. To examine its role in vivo, we generated mice that do not express the fibulin-2 gene (Fbln2) through homologous recombination of embryonic stem cells. Unexpectedly, the fibulin-2-null mice were viable and fertile and did not display gross and anatomical abnormalities. Histological and ultrastructural analyses revealed that elastic fibers assembled normally in the absence of fibulin-2. No compensatory up-regulation of mRNAs for other fibulin members was detected in the aorta and skin tissue. However, in the fibulin-2 null aortae, fibulin-1 immunostaining was increased in the inner elastic lamina, where fibulin-2 preferentially localizes. The results demonstrate that fibulin-2 is not required for mouse development and elastic fiber formation and suggest possible functional redundancy between fibulin-1 and fibulin-2.


Subject(s)
Calcium-Binding Proteins/physiology , Elastic Tissue/growth & development , Extracellular Matrix Proteins/physiology , Animals , Aorta/chemistry , Aorta/cytology , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/deficiency , Extracellular Matrix Proteins/analysis , Extracellular Matrix Proteins/deficiency , Growth and Development , Mice , Mice, Knockout , Phenotype
14.
Mol Cancer Res ; 5(10): 1041-51, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17951404

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant familial cancer syndrome characterized primarily by endocrine tumors of the parathyroids, anterior pituitary, and enteropancreatic endocrine tissues. Affected individuals carry a germ-line loss-of-function mutation of the MEN1 gene, and tumors arise after loss of the second allele. Homozygous loss of Men1 in the germ line of mice results in early embryonic lethality, with defective development of neural tube, heart, liver, and craniofacial structures. We generated immortalized wild-type (WT) and menin-null mouse embryo fibroblast (MEF) cell lines and evaluated their characteristics, including global expression patterns. The WT and menin-null cell lines were aneuploid, and the nulls did not display tumorigenic characteristics in soft agar assay. Expression arrays in menin-null MEFs revealed altered expression of several extracellular matrix proteins that are critical in organogenesis. Specifically, transcripts for fibulin 2 (Fbln2), periostin (Postn), and versican [chondroitin sulfate proteoglycan (Cspg2)], genes critical for the developing heart and known to be induced by transforming growth factor-beta (TGF-beta), were decreased in their expression in menin-null MEFs. Fbln2 expression was the most affected, and the reduction in menin-null MEFs for Fbln2, Postn, and Cspg2 was 16.18-, 5.37-, and 2.15-fold, respectively. Menin-null MEFs also showed poor response to TGF-beta-induced Smad3-mediated transcription in a reporter assay, supporting a role for menin in this pathway. Postn and Cspg2 expression in WT, unlike in null MEFs, increased on TGF-beta treatment. The expression changes associated with the loss of the tumor suppressor menin provide insights into the defective organogenesis observed during early embryonic development in Men1-null mouse embryos.


Subject(s)
Embryo, Mammalian/metabolism , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Developmental , Proto-Oncogene Proteins/physiology , Tumor Suppressor Proteins/physiology , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Mice , Organogenesis/genetics , Proto-Oncogene Proteins/genetics , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins/genetics
15.
Ann Neurol ; 59(1): 190-5, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16278855

ABSTRACT

We have identified highly similar heterozygous COL6A1 genomic deletions, spanning from intron 8 to exon 13 or intron 13, in two patients with Ullrich congenital muscular dystrophy and the milder Bethlem myopathy. The 5' breakpoints of both deletions are located within a minisatellite in intron 8. The mutations cause in-frame deletions of 66 and 84 amino acids in the amino terminus of the triple-helical domain, leading to intracellular accumulation of mutant polypeptides and reduced extracellular collagen VI microfibrils. Our studies identify a deletion-prone region in COL6A1 and suggest that similar mutations can lead to congenital muscle disorders of different clinical severity.


Subject(s)
Collagen Type VI/genetics , Gene Deletion , Muscular Diseases/genetics , Muscular Dystrophies/genetics , Mutation , Adult , Amino Acid Sequence , Base Sequence , Cells, Cultured , Child , Child, Preschool , DNA Mutational Analysis , Exons , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Infant , Introns , Male , Molecular Sequence Data
16.
Hum Genet ; 117(5): 460-6, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16075202

ABSTRACT

Ullrich congenital muscular dystrophy (UCMD) is a severe disorder caused, in most cases, by a deficiency in collagen VI microfibrils. Recessive mutations in two of the three collagen VI genes, COL6A2 and COL6A3, have been identified in eight of the nine UCMD patients reported thus far. A heterozygous COL6A1 gene deletion, resulting in a mutant protein that exerts a dominant negative effect, has recently been described in a severely affected UCMD patient. Here we describe a patient in whom reverse transcription-PCR analysis of fibroblast RNA suggested a heterozygous in-frame deletion of exon 13 in the triple-helical domain of COL6A2, which is predicted to be dominantly acting. However, a homozygous A --> G mutation at -10 of intron 12 was found in the genomic DNA. The intron mutation activated numerous cryptic splice acceptor sites, generating normal and exon 13-deleted COL6A2 mRNA, and multiple aberrant transcripts containing frameshifts that were degraded through a nonsense-mediated decay mechanism. Northern analysis indicated diminished COL6A2 mRNA expression as the primary pathogenic mechanism in this UCMD patient. Our results underscore the importance of multifaceted analyses in the accurate molecular diagnosis and interpretation of genotype-phenotype correlations of UCMD.


Subject(s)
Collagen Type VI/genetics , Gene Deletion , Muscular Dystrophies/congenital , Muscular Dystrophies/genetics , Point Mutation , RNA Splicing/genetics , Base Sequence , Child, Preschool , Genotype , Humans , Immunohistochemistry , Introns/genetics , Male , Molecular Sequence Data , Phenotype , Reverse Transcriptase Polymerase Chain Reaction
17.
Am J Hum Genet ; 73(2): 355-69, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12840783

ABSTRACT

Recessive mutations in two of the three collagen VI genes, COL6A2 and COL6A3, have recently been shown to cause Ullrich congenital muscular dystrophy (UCMD), a frequently severe disorder characterized by congenital muscle weakness with joint contractures and coexisting distal joint hyperlaxity. Dominant mutations in all three collagen VI genes had previously been associated with the considerably milder Bethlem myopathy. Here we report that a de novo heterozygous deletion of the COL6A1 gene can also result in a severe phenotype of classical UCMD precluding ambulation. The internal gene deletion occurs near a minisatellite DNA sequence in intron 8 that removes 1.1 kb of genomic DNA encompassing exons 9 and 10. The resulting mutant chain contains a 33-amino acid deletion near the amino-terminus of the triple-helical domain but preserves a unique cysteine in the triple-helical domain important for dimer formation prior to secretion. Thus, dimer formation and secretion of abnormal tetramers can occur and exert a strong dominant negative effect on microfibrillar assembly, leading to a loss of normal localization of collagen VI in the basement membrane surrounding muscle fibers. Consistent with this mechanism was our analysis of a patient with a much milder phenotype, in whom we identified a previously described Bethlem myopathy heterozygous in-frame deletion of 18 amino acids somewhat downstream in the triple-helical domain, a result of exon 14 skipping in the COL6A1 gene. This deletion removes the crucial cysteine, so that dimer formation cannot occur and the abnormal molecule is not secreted, preventing the strong dominant negative effect. Our studies provide a biochemical insight into genotype-phenotype correlations in this group of disorders and establish that UCMD can be caused by dominantly acting mutations.


Subject(s)
Collagen Type VI/genetics , Muscular Dystrophies/congenital , Muscular Dystrophies/genetics , Amino Acid Sequence , Base Sequence , Child , Collagen Type VI/chemistry , DNA, Complementary/genetics , Dimerization , Exons , Extracellular Matrix/chemistry , Fibroblasts/chemistry , Genes, Dominant , Genotype , Heterozygote , Humans , Introns , Male , Molecular Sequence Data , Muscles/metabolism , Muscles/pathology , Muscular Dystrophies/pathology , Phenotype , Protein Structure, Tertiary , RNA, Messenger/genetics , Sequence Deletion , Sequence Homology, Nucleic Acid
18.
J Biol Chem ; 277(46): 43557-64, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12218063

ABSTRACT

We recently reported a severe deficiency in collagen type VI, resulting from recessive mutations of the COL6A2 gene, in patients with Ullrich congenital muscular dystrophy. Their parents, who are all carriers of one mutant allele, are unaffected, although heterozygous mutations in collagen VI caused Bethlem myopathy. Here we investigated the consequences of three COL6A2 mutations in fibroblasts from patients and their parents in two Ullrich families. All three mutations lead to nonsense-mediated mRNA decay. However, very low levels of undegraded mutant mRNA remained in patient B with compound heterozygous mutations at the distal part of the triple-helical domain, resulting in deposition of abnormal microfibrils that cannot form extensive networks. This observation suggests that the C-terminal globular domain is not essential for triple-helix formation but is critical for microfibrillar assembly. In all parents, the COL6A2 mRNA levels are reduced to 57-73% of the control, but long term collagen VI matrix depositions are comparable with that of the control. The almost complete absence of abnormal protein and near-normal accumulation of microfibrils in the parents may account for their lack of myopathic symptoms.


Subject(s)
Collagen Type VI/chemistry , Collagen Type VI/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , RNA, Messenger/metabolism , Alleles , Amino Acid Sequence , Blotting, Northern , Cells, Cultured , Exons , Extracellular Matrix/metabolism , Family Health , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Humans , Microscopy, Electron , Microscopy, Fluorescence , Microscopy, Immunoelectron , Models, Genetic , Molecular Sequence Data , Mutation , Precipitin Tests , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
19.
Matrix Biol ; 21(3): 227-41, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12009329

ABSTRACT

Three distinct alpha chains form the collagen VI monomer, the alpha 3(VI) chain being much larger than the alpha 1(VI) and alpha 2(VI) chains. The alpha 3(VI) chain has 10 von Willebrand Factor type A domains of approximately 200 amino acids at the N-terminus (N1-N10) compared with only one such domain in the alpha 1(VI) and alpha 2(VI) chains. Domains N10, N9, N7 and N3 of the alpha 3(VI) chain are subject to alternative splicing in chick and/or human tissues, indicating the possibility of isoforms that have different functions depending on which N-terminal domains are included or excluded. In this study we have PCR amplified and sequenced mouse alpha 3(VI) cDNA encoding the N2-N10 domains. By reverse transcription-PCR using oligonucleotides spanning different regions of the cDNA we have undertaken a comprehensive analysis of alternative splicing of the alpha 3(VI) mRNA in embryonic and adult mouse tissues. We demonstrate that domains N10, N9 and N7 are also subject to alternative splicing in mouse tissues and in addition identify an abundant novel variant transcript that lacks all four N-terminal domains (N7-N10) in mouse tissues and human cells. We also identify less abundant transcripts that lack a large part of the N3 domain, and transcripts lacking the entire N5 domain. Using specific RNase protection assays we show that the shorter transcripts containing domains (N8+N7+N6), (N8+N6) and N6 are present at higher levels than transcripts containing the N10 and/or N9 domains, with tissue-specific variation in the levels of variant transcripts. These studies demonstrate a larger range of collagen VI protein variants than previously described.


Subject(s)
Alternative Splicing , Collagen Type VI/genetics , 5' Untranslated Regions , Amino Acid Sequence , Animals , Base Sequence , Chickens , Cloning, Molecular , DNA, Complementary , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Molecular Sequence Data , Protein Isoforms/genetics , Sequence Homology, Amino Acid
20.
Muscle Nerve ; 25(4): 513-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11932968

ABSTRACT

Bethlem myopathy is an early-onset benign myopathy characterized by proximal muscular weakness and multiple flexion contractures. It is a dominantly inherited disorder associated with mutations in the three COL6 genes encoding type VI collagen. We detected a g-->a substitution at +1 position of COL6A1 intron 3 in a four-generation Italian family affected by a mild form of Bethlem myopathy. The mutation results in the activation of a cryptic splice donor site at the 3' end of exon 3, leading to the loss of 66 nucleotides and an "in-frame" deletion of 22 amino acids in the NH2-domain. Molecular analysis on fibroblasts of the propositus showed that the mutated mRNA was present and stable, but the mutated protein could not be detected. Western blot and immunofluorescence analyses showed a decreased level of collagen VI synthesis and deposition in fibroblasts of the propositus. Together, the results suggest that the mutated protein was highly unstable and rapidly degraded, and that the mild phenotype was caused by a reduced amount of normal collagen VI microfibrils. In addition, we demonstrated that lymphocytes can be used for the first mutation screening analysis of patients with Bethlem myopathy.


Subject(s)
Alternative Splicing/genetics , Collagen Type VI/genetics , Muscle, Skeletal/metabolism , Muscular Diseases/genetics , Point Mutation/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Collagen Type VI/biosynthesis , DNA Mutational Analysis , Female , Gene Expression Regulation/genetics , Genetic Testing , Humans , Male , Middle Aged , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Pedigree , RNA, Messenger/analysis , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL