1
|
Zhong X, Yan GG, Chaturvedi A, Li X, Gao Y, Girgenrath M, Corcoran CJ, Diblasio-Smith L, LaVallie ER, de Rham T, Zhou J, Abel M, Riegel L, Lim SK, Bloom L, Lin L, D’Antona AM. Metabolic Engineering of Glycofusion Bispecific Antibodies for α-Dystroglycanopathies. Antibodies (Basel) 2024; 13:83. [PMID: 39449325 PMCID: PMC11503271 DOI: 10.3390/antib13040083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/14/2024] [Accepted: 10/02/2024] [Indexed: 10/26/2024] Open
Abstract
Background: α-dystroglycanopathies are congenital muscular dystrophies in which genetic mutations cause the decrease or absence of a unique and complex O-linked glycan called matriglycan. This hypoglycosylation of O-linked matriglycan on the α-dystroglycan (α-DG) protein subunit abolishes or reduces the protein binding to extracellular ligands such as laminins in skeletal muscles, leading to compromised survival of muscle cells after contraction. Methods: Surrogate molecular linkers reconnecting laminin-211 and the dystroglycan β-subunit through bispecific antibodies can be engineered to improve muscle function in the α-dystroglycanopathies. This study reports the metabolic engineering of a novel glycofusion bispecific (GBi) antibody that fuses the mucin-like domain of the α-DG to the light chain of an anti-β-DG subunit antibody. Results: Transient HEK production with the co-transfection of LARGE1, the glycoenzyme responsible for the matriglycan modification, produced the GBi antibody only with a light matriglycan modification and a weak laminin-211 binding activity. However, when a sugar feed mixture of uridine, galactose, and manganese ion (Mn2+) was added to the culture medium, the GBi antibody produced exhibited a dramatically enhanced matriglycan modification and a much stronger laminin-binding activity. Conclusions: Further investigation has revealed that Mn2+ in the sugar feeds played a critical role in increasing the matriglycan modification of the GBi antibody, key for the function of the resulting bispecific antibody.
Collapse
Affiliation(s)
- Xiaotian Zhong
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Guoying Grace Yan
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Apurva Chaturvedi
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Xiuling Li
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Yijie Gao
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Mahasweta Girgenrath
- Rare Disease Research Unit, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA
| | - Chris J. Corcoran
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Liz Diblasio-Smith
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Edward R. LaVallie
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Teresse de Rham
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Jing Zhou
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Molica Abel
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Logan Riegel
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Sean K.H. Lim
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Laird Bloom
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Laura Lin
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| | - Aaron M. D’Antona
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA (Y.G.); (C.J.C.)
| |
Collapse
|
2
|
Tan RL, Sciandra F, Hübner W, Bozzi M, Reimann J, Schoch S, Brancaccio A, Blaess S. The missense mutation C667F in murine β-dystroglycan causes embryonic lethality, myopathy and blood-brain barrier destabilization. Dis Model Mech 2024; 17:dmm050594. [PMID: 38616731 PMCID: PMC11212641 DOI: 10.1242/dmm.050594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/08/2024] [Indexed: 04/16/2024] Open
Abstract
Dystroglycan (DG) is an extracellular matrix receptor consisting of an α- and a β-DG subunit encoded by the DAG1 gene. The homozygous mutation (c.2006G>T, p.Cys669Phe) in β-DG causes muscle-eye-brain disease with multicystic leukodystrophy in humans. In a mouse model of this primary dystroglycanopathy, approximately two-thirds of homozygous embryos fail to develop to term. Mutant mice that are born undergo a normal postnatal development but show a late-onset myopathy with partially penetrant histopathological changes and an impaired performance on an activity wheel. Their brains and eyes are structurally normal, but the localization of mutant β-DG is altered in the glial perivascular end-feet, resulting in a perturbed protein composition of the blood-brain and blood-retina barrier. In addition, α- and β-DG protein levels are significantly reduced in muscle and brain of mutant mice. Owing to the partially penetrant developmental phenotype of the C669F β-DG mice, they represent a novel and highly valuable mouse model with which to study the molecular effects of β-DG functional alterations both during embryogenesis and in mature muscle, brain and eye, and to gain insight into the pathogenesis of primary dystroglycanopathies.
Collapse
Affiliation(s)
- Rui Lois Tan
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Francesca Sciandra
- Institute of Chemical Sciences and Technologies 'Giulio Natta' (SCITEC)-CNR, 00168 Rome, Italy
| | - Wolfgang Hübner
- Biomolecular Photonics, Faculty of Physics, Bielefeld University, 33615 Bielefeld, Germany
| | - Manuela Bozzi
- Institute of Chemical Sciences and Technologies 'Giulio Natta' (SCITEC)-CNR, 00168 Rome, Italy
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie. Sezione di Biochimica. Università Cattolica del Sacro Cuore di Roma, 00168 Rome, Italy
| | - Jens Reimann
- Department of Neurology, Neuromuscular Diseases Section, University Hospital Bonn, 53127 Bonn, Germany
| | - Susanne Schoch
- Synaptic Neuroscience Team, Institute of Neuropathology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Andrea Brancaccio
- Institute of Chemical Sciences and Technologies 'Giulio Natta' (SCITEC)-CNR, 00168 Rome, Italy
- School of Biochemistry, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
3
|
Canessa EH, Spathis R, Novak JS, Beedle A, Nagaraju K, Bello L, Pegoraro E, Hoffman EP, Hathout Y. Characterization of the dystrophin-associated protein complex by mass spectrometry. MASS SPECTROMETRY REVIEWS 2024; 43:90-105. [PMID: 36420714 DOI: 10.1002/mas.21823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The dystrophin-associated protein complex (DAPC) is a highly organized multiprotein complex that plays a pivotal role in muscle fiber structure integrity and cell signaling. The complex is composed of three distinct interacting subgroups, intracellular peripheral proteins, transmembrane glycoproteins, and extracellular glycoproteins subcomplexes. Dystrophin protein nucleates the DAPC and is important for connecting the intracellular actin cytoskeletal filaments to the sarcolemma glycoprotein complex that is connected to the extracellular matrix via laminin, thus stabilizing the sarcolemma during muscle fiber contraction and relaxation. Genetic mutations that lead to lack of expression or altered expression of any of the DAPC proteins are associated with different types of muscle diseases. Hence characterization of this complex in healthy and dystrophic muscle might bring insights into its role in muscle pathogenesis. This review highlights the role of mass spectrometry in characterizing the DAPC interactome as well as post-translational glycan modifications of some of its components such as α-dystroglycan. Detection and quantification of dystrophin using targeted mass spectrometry are also discussed in the context of healthy versus dystrophic skeletal muscle.
Collapse
Affiliation(s)
- Emily H Canessa
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, SUNY, Binghamton, New York, USA
- Biomedical Engineering Department, Binghamton University, SUNY, Binghamton, New York, USA
| | - Rita Spathis
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, SUNY, Binghamton, New York, USA
| | - James S Novak
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, District of Columbia, USA
- Department of Genomics and Precision Medicine and Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Aaron Beedle
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, SUNY, Binghamton, New York, USA
| | - Kanneboyina Nagaraju
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, SUNY, Binghamton, New York, USA
| | - Luca Bello
- Department of Neuroscience, ERN Neuromuscular Center, University of Padova, Padua, Italy
| | - Elena Pegoraro
- Department of Neuroscience, ERN Neuromuscular Center, University of Padova, Padua, Italy
| | - Eric P Hoffman
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, SUNY, Binghamton, New York, USA
| | - Yetrib Hathout
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, SUNY, Binghamton, New York, USA
| |
Collapse
|
4
|
Yue J, Huang R, Lan Z, Xiao B, Luo Z. Abnormal glycosylation in glioma: related changes in biology, biomarkers and targeted therapy. Biomark Res 2023; 11:54. [PMID: 37231524 DOI: 10.1186/s40364-023-00491-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Glioma is a rapidly growing and aggressive primary malignant tumor of the central nervous system that can diffusely invade the brain tissue around, and the prognosis of patients is not significantly improved by traditional treatments. One of the most general posttranslational modifications of proteins is glycosylation, and the abnormal distribution of this modification in gliomas may shed light on how it affects biological behaviors of glioma cells, including proliferation, migration, and invasion, which may be produced by regulating protein function, cell-matrix and cell‒cell interactions, and affecting receptor downstream pathways. In this paper, from the perspective of regulating protein glycosylation changes and abnormal expression of glycosylation-related proteins (such as glycosyltransferases in gliomas), we summarize how glycosylation may play a crucial role in the discovery of novel biomarkers and new targeted treatment options for gliomas. Overall, the mechanistic basis of abnormal glycosylation affecting glioma progression remains to be more widely and deeply explored, which not only helps to inspire researchers to further explore related diagnostic and prognostic markers but also provides ideas for discovering effective treatment strategies and improving glioma patient survival and prognosis.
Collapse
Affiliation(s)
- Juan Yue
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya road of Kaifu district, 410008, Changsha, Hunan, China
| | - Roujie Huang
- Department of Obstetrics and Gynecology, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Shuaifuyuan No. 1, Dongcheng District, 100730, Beijing, China
| | - Zehao Lan
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya road of Kaifu district, 410008, Changsha, Hunan, China
- Clinical Research Center for Epileptic disease of Hunan Province, Central South University, 410008, Changsha, Hunan, P.R. China
| | - Zhaohui Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya road of Kaifu district, 410008, Changsha, Hunan, China.
- Clinical Research Center for Epileptic disease of Hunan Province, Central South University, 410008, Changsha, Hunan, P.R. China.
| |
Collapse
|
5
|
Differential Urinary Proteomic Analysis of High-Risk Cervical Intraepithelial Neoplasia. Int J Mol Sci 2023; 24:ijms24032531. [PMID: 36768853 PMCID: PMC9916937 DOI: 10.3390/ijms24032531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Human papillomavirus (HPV)-associated lesions and malignancies exhibit alterations in the composition and functionality of the extracellular matrix (ECM) that represent the complex molecular pathways present between infection and disease. A total of 20 urine samples were used, including from 10 patients with cervical intraepithelial neoplasia grade 3 (CIN3) and 10 healthy controls to perform the label-free quantitative analysis using the nano-HPLC and ESI-MS ion trap mass analyzer and matrix-assisted laser desorption ionization-time-of-flight mass spectrometry (MALDI-TOF/MS) fast screening. Among 476 identified/quantified proteins, 48 were significantly changed (log2-fold change ≥1.0 or ≤-1.0, -log10 (bbinominal, p-value ≥ 1.3), of which were 40 proteins (down-regulated) and 8 proteins (up-regulated) in CIN3, in comparison to healthy controls. The biological function and key pathway enrichment of the gene set using gen set enrichment analysis (GSEA) were analyzed. The ECM-receptor interaction pathway (NES = -1.64, p = 0.026) was down-regulated by 13 proteins (HSPG2, COL6A1, COL6A3, SPP1, THBS1, TNC, DAG1, FN1, COMP, GP6, VTN, SDC1, and CD44; log2 FC range from -0.03 to -1.48) for the CIN3 group in the KEGG database. The MALDI-TOF/MS screening showed the difference of protein profiles between the control and CIN3 groups, i.e., using the scatter plot with a well-separated shape, as well as effectively distinguishing both groups (control and CIN3) using genetic algorithms (GA) with cross-validation (51.56%) and recognition capability (95.0%). Decreased levels of ECM-receptor interaction proteins may cause disturbances in the interactions of cells with the ECM and play an important role in the development and progression of cervical cancer.
Collapse
|
6
|
Cubilla M, Papazoglu G, Asteggiano C. Dystroglycanopathies: Genetic Bases of Muscular Dystrophies Due to Alteration in the O-Glycosylation of α-Dystroglycan. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2023; 11. [DOI: 10.1590/2326-4594-jiems-2022-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Affiliation(s)
- M.A. Cubilla
- Hospital de Niños de la Santísima Trinidad, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Argentina
| | - G.M. Papazoglu
- Hospital de Niños de la Santísima Trinidad, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Argentina
| | - C.G. Asteggiano
- Hospital de Niños de la Santísima Trinidad, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Argentina; Universidad Católica de Córdoba, Argentina
| |
Collapse
|
7
|
Quereda C, Pastor À, Martín-Nieto J. Involvement of abnormal dystroglycan expression and matriglycan levels in cancer pathogenesis. Cancer Cell Int 2022; 22:395. [PMID: 36494657 PMCID: PMC9733019 DOI: 10.1186/s12935-022-02812-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Dystroglycan (DG) is a glycoprotein composed of two subunits that remain non-covalently bound at the plasma membrane: α-DG, which is extracellular and heavily O-mannosyl glycosylated, and β-DG, an integral transmembrane polypeptide. α-DG is involved in the maintenance of tissue integrity and function in the adult, providing an O-glycosylation-dependent link for cells to their extracellular matrix. β-DG in turn contacts the cytoskeleton via dystrophin and participates in a variety of pathways transmitting extracellular signals to the nucleus. Increasing evidence exists of a pivotal role of DG in the modulation of normal cellular proliferation. In this context, deficiencies in DG glycosylation levels, in particular those affecting the so-called matriglycan structure, have been found in an ample variety of human tumors and cancer-derived cell lines. This occurs together with an underexpression of the DAG1 mRNA and/or its α-DG (core) polypeptide product or, more frequently, with a downregulation of β-DG protein levels. These changes are in general accompanied in tumor cells by a low expression of genes involved in the last steps of the α-DG O-mannosyl glycosylation pathway, namely POMT1/2, POMGNT2, CRPPA, B4GAT1 and LARGE1/2. On the other hand, a series of other genes acting earlier in this pathway are overexpressed in tumor cells, namely DOLK, DPM1/2/3, POMGNT1, B3GALNT2, POMK and FKTN, hence exerting instead a pro-oncogenic role. Finally, downregulation of β-DG, altered β-DG processing and/or impaired β-DG nuclear levels are increasingly found in human tumors and cell lines. It follows that DG itself, particular genes/proteins involved in its glycosylation and/or their interactors in the cell could be useful as biomarkers of certain types of human cancer, and/or as molecular targets of new therapies addressing these neoplasms.
Collapse
Affiliation(s)
- Cristina Quereda
- grid.5268.90000 0001 2168 1800Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante, Campus Universitario San Vicente, P.O. Box 99, 03080 Alicante, Spain
| | - Àngels Pastor
- grid.5268.90000 0001 2168 1800Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante, Campus Universitario San Vicente, P.O. Box 99, 03080 Alicante, Spain
| | - José Martín-Nieto
- grid.5268.90000 0001 2168 1800Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante, Campus Universitario San Vicente, P.O. Box 99, 03080 Alicante, Spain ,grid.5268.90000 0001 2168 1800Instituto Multidisciplinar para el Estudio del Medio ‘Ramón Margalef’, Universidad de Alicante, 03080 Alicante, Spain
| |
Collapse
|
8
|
Schüler SC, Liu Y, Dumontier S, Grandbois M, Le Moal E, Cornelison DDW, Bentzinger CF. Extracellular matrix: Brick and mortar in the skeletal muscle stem cell niche. Front Cell Dev Biol 2022; 10:1056523. [PMID: 36523505 PMCID: PMC9745096 DOI: 10.3389/fcell.2022.1056523] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/03/2022] [Indexed: 11/30/2022] Open
Abstract
The extracellular matrix (ECM) is an interconnected macromolecular scaffold occupying the space between cells. Amongst other functions, the ECM provides structural support to tissues and serves as a microenvironmental niche that conveys regulatory signals to cells. Cell-matrix adhesions, which link the ECM to the cytoskeleton, are dynamic multi-protein complexes containing surface receptors and intracellular effectors that control various downstream pathways. In skeletal muscle, the most abundant tissue of the body, each individual muscle fiber and its associated muscle stem cells (MuSCs) are surrounded by a layer of ECM referred to as the basal lamina. The core scaffold of the basal lamina consists of self-assembling polymeric laminins and a network of collagens that tether proteoglycans, which provide lateral crosslinking, establish collateral associations with cell surface receptors, and serve as a sink and reservoir for growth factors. Skeletal muscle also contains the fibrillar collagenous interstitial ECM that plays an important role in determining tissue elasticity, connects the basal laminae to each other, and contains matrix secreting mesenchymal fibroblast-like cell types and blood vessels. During skeletal muscle regeneration fibroblast-like cell populations expand and contribute to the transitional fibronectin-rich regenerative matrix that instructs angiogenesis and MuSC function. Here, we provide a comprehensive overview of the role of the skeletal muscle ECM in health and disease and outline its role in orchestrating tissue regeneration and MuSC function.
Collapse
Affiliation(s)
- Svenja C. Schüler
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Yuguo Liu
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Simon Dumontier
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Michel Grandbois
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Emmeran Le Moal
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - DDW Cornelison
- Division of Biological Sciences Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - C. Florian Bentzinger
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
9
|
Kaplan KM, Morgan KG. The importance of dystrophin and the dystrophin associated proteins in vascular smooth muscle. Front Physiol 2022; 13:1059021. [PMID: 36505053 PMCID: PMC9732661 DOI: 10.3389/fphys.2022.1059021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/14/2022] [Indexed: 11/26/2022] Open
Abstract
This review details the role of dystrophin and the dystrophin associated proteins (DAPs) in the vascular smooth muscle. Dystrophin is most comprehensively studied in the skeletal muscle due to serious symptoms found related to the skeletal muscle of patients with muscular dystrophy. Mutations in the dystrophin gene, or DAPs genes, result in a wide range of muscular dystrophies. In skeletal muscle, dystrophin is known to act to as a cytoskeletal stabilization protein and protects cells against contraction-induced damage. In skeletal muscle, dystrophin stabilizes the plasma membrane by transmitting forces generated by sarcomeric contraction to the extracellular matrix (ECM). Dystrophin is a scaffold that binds the dystroglycan complex (DGC) and has many associated proteins (DAPs). These DAPs include sarcoglycans, syntrophins, dystroglycans, dystrobrevin, neuronal nitric oxide synthase, and caveolins. The DAPs provide biomechanical support to the skeletal or cardiac plasma membrane during contraction, and loss of one or several of these DAPs leads to plasma membrane fragility. Dystrophin is expressed near the plasma membrane of all muscles, including cardiac and vascular smooth muscle, and some neurons. Dystrophic mice have noted biomechanical irregularities in the carotid arteries and spontaneous motor activity in portal vein altered when compared to wild type mice. Additionally, some studies suggest the vasculature of patients and animal models with muscular dystrophy is abnormal. Although the function of dystrophin and the DAPs in vascular smooth muscle is not thoroughly established in the field, this review makes the point that these proteins are expressed, and important and further study is warranted.
Collapse
|
10
|
Abstract
The brain harbors a unique ability to, figuratively speaking, shift its gears. During wakefulness, the brain is geared fully toward processing information and behaving, while homeostatic functions predominate during sleep. The blood-brain barrier establishes a stable environment that is optimal for neuronal function, yet the barrier imposes a physiological problem; transcapillary filtration that forms extracellular fluid in other organs is reduced to a minimum in brain. Consequently, the brain depends on a special fluid [the cerebrospinal fluid (CSF)] that is flushed into brain along the unique perivascular spaces created by astrocytic vascular endfeet. We describe this pathway, coined the term glymphatic system, based on its dependency on astrocytic vascular endfeet and their adluminal expression of aquaporin-4 water channels facing toward CSF-filled perivascular spaces. Glymphatic clearance of potentially harmful metabolic or protein waste products, such as amyloid-β, is primarily active during sleep, when its physiological drivers, the cardiac cycle, respiration, and slow vasomotion, together efficiently propel CSF inflow along periarterial spaces. The brain's extracellular space contains an abundance of proteoglycans and hyaluronan, which provide a low-resistance hydraulic conduit that rapidly can expand and shrink during the sleep-wake cycle. We describe this unique fluid system of the brain, which meets the brain's requisites to maintain homeostasis similar to peripheral organs, considering the blood-brain-barrier and the paths for formation and egress of the CSF.
Collapse
Affiliation(s)
- Martin Kaag Rasmussen
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Humberto Mestre
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
11
|
Deficiency of Glycosylated α-Dystroglycan in Ventral Hippocampus Bridges the Destabilization of Gamma-Aminobutyric Acid Type A Receptors With the Depressive-like Behaviors of Male Mice. Biol Psychiatry 2022; 91:593-603. [PMID: 35063187 DOI: 10.1016/j.biopsych.2021.10.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND Depression is a common psychiatric disorder associated with defects in GABAergic (gamma-aminobutyric acidergic) neurotransmission. α-Dystroglycan (α-DG), a cell adhesion molecule known to be essential for skeletal muscle integrity, is also present at inhibitory synapses in the central nervous system and forms a structural element in certain synapses. However, the role of α-DG in the regulation of depressive-like behaviors remains largely unknown. METHODS Depressive-like behaviors were induced by chronic social defeat stress in adult male mice. Surface protein was extracted by a biotin kit, and the expression of protein was detected by Western blotting. Intrahippocampal microinjection of the lentivirus or adeno-associated virus or agrin intervention was carried out using a stereotaxic instrument and followed by behavioral tests. Miniature inhibitory postsynaptic currents were recorded by whole-cell patch-clamp techniques. RESULTS The expression of α-DG and glycosylated α-DG in the ventral hippocampus was significantly lower in chronic social defeat stress-susceptible male mice than in control mice, accompanied by a decreased surface expression of GABAA receptor γ2 subunit and reduced GABAergic neurotransmission. RNA interference-mediated knockdown of Dag1 increased the susceptibility of mice to subthreshold stress. Both in vivo administration of agrin and overexpression of like-acetylglucosaminyltransferase ameliorated depressive-like behaviors and restored the decrease in surface expression of GABAA receptor γ2 subunit and the amplitude of miniature inhibitory postsynaptic currents in chronic social defeat stress-exposed mice. CONCLUSIONS Our findings demonstrate that glycosylated α-DG plays a role in the pathophysiological process of depressive-like behaviors by regulating the surface expression of GABAA receptor γ2 subunit and GABAergic neurotransmission in the ventral hippocampus.
Collapse
|
12
|
Sandonà M, Saccone V. Post-translational Modification in Muscular Dystrophies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1382:71-84. [DOI: 10.1007/978-3-031-05460-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
13
|
Brancaccio A. A molecular overview of the primary dystroglycanopathies. J Cell Mol Med 2019; 23:3058-3062. [PMID: 30838779 PMCID: PMC6484290 DOI: 10.1111/jcmm.14218] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 01/17/2023] Open
Abstract
Dystroglycan is a major non-integrin adhesion complex that connects the cytoskeleton to the surrounding basement membranes, thus providing stability to skeletal muscle. In Vertebrates, hypoglycosylation of α-dystroglycan has been strongly linked to muscular dystrophy phenotypes, some of which also show variable degrees of cognitive impairments, collectively termed dystroglycanopathies. Only a small number of mutations in the dystroglycan gene, leading to the so called primary dystroglycanopathies, has been described so far, as opposed to the ever-growing number of identified secondary or tertiary dystroglycanopathies (caused by genetic abnormalities in glycosyltransferases or in enzymes involved in the synthesis of the carbohydrate building blocks). The few mutations found within the autonomous N-terminal domain of α-dystroglycan seem to destabilise it to different degrees, without influencing the overall folding and targeting of the dystroglycan complex. On the contrary other mutations, some located at the α/β interface of the dystroglycan complex, seem to be able to interfere with its maturation, thus compromising its stability and eventually leading to the intracellular engulfment and/or partial or even total degradation of the dystroglycan uncleaved precursor.
Collapse
Affiliation(s)
- Andrea Brancaccio
- School of Biochemistry, University of Bristol, Bristol, UK.,Istituto di Chimica del Riconoscimento Molecolare - CNR c/o Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
14
|
Kálmán M, Lőrincz DL, Sebők OM, Ari C, Oszwald E, Somiya H, Jancsik V. Cerebrovascular β-dystroglycan immunoreactivity in vertebrates: not detected in anurans and in the teleosts Ostariophysi and Euteleostei. Integr Zool 2019; 15:16-31. [PMID: 30811839 DOI: 10.1111/1749-4877.12384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The aim of the present paper was to check for the presence of cerebrovascular dystroglycan in vertebrates, because dystroglycan, which is localized in the vascular astroglial end-feet, has a pivotal function in glio-vascular connections. In mammalian brains, the immunoreactivity of β-dystroglycan subunit delineates the vessels. The results of the present study demonstrate similar patterns in other vertebrates, except for anurans and the teleost groups Ostariophysi and Euteleostei. In this study, we investigated 1 or 2 representative species of the main groups of Chondrichthyes, teleost and non-teleost ray-finned fishes, urodeles, anurans, and reptiles. We also investigated 5 mammalian and 3 bird species. Animals were obtained from breeders or fishermen. The presence of β-dystroglycan was investigated immunohistochemically in free-floating sections. Pre-embedding electron microscopical immunohistochemistry on Heterodontus japonicus shark brains demonstrated that in Elasmobranchii, β-dystroglycan is also localized in the perivascular glial end-feet despite the different construction of their blood-brain barrier. The results indicated that the cerebrovascular β-dystroglycan immunoreactivity disappeared separately in anurans, and in teleosts, in the latter group before its division to Ostariophysi and Euteleostei. Immunohistochemistry in muscles and western blots from brain homogenates, however, detected the presence of β-dystroglycan, even in anurans and all teleosts. A possible explanation is that in the glial end-feet, β-dystroglycan is masked in these animals, or disappeared during adaptation to the freshwater habitat.
Collapse
Affiliation(s)
- Mihály Kálmán
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - David L Lőrincz
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary.,University of Leicester, Dept. of Neuroscience, Psychology and Behaviour, Leicester, United Kingdom
| | - Olivér M Sebők
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Csilla Ari
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary.,Hyperbaric Neuroscience Research Lab., Dept of Psychology, University of South Florida, Tampa, Florida, USA
| | - Erzsébet Oszwald
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Hiroaki Somiya
- Department of Environmental Biology, Chubu University, Chubu, Japan
| | | |
Collapse
|
15
|
Souttou S, Benabdesselam R, Siqueiros-Marquez L, Sifi M, Deliba M, Vacca O, Charles-Messance H, Vaillend C, Rendon A, Guillonneau X, Dorbani-Mamine L. Expression and localization of dystrophins and β-dystroglycan in the hypothalamic supraoptic nuclei of rat from birth to adulthood. Acta Histochem 2019; 121:218-226. [PMID: 30595391 DOI: 10.1016/j.acthis.2018.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 11/19/2018] [Accepted: 12/10/2018] [Indexed: 10/27/2022]
Abstract
Dystrophins (Dps) are the sub-membranous proteins that work via the dystrophin-associated proteins complex, which comprises β-dystroglycan (β-DG), a cell surface receptor for extracellular matrix. Recently, we have revealed β-DG decrease and central function impairment of supraoptic nucleus (SON) in Dp71 deficient adult mice, opening the question on the profiles of Dps and β-DG during SON development. At birth and the age of 10, 20 and 60 days, we examined the expression by RT-PCR and Western-blotting, and the distribution by immunohistochemistry of Dps and β-DG. Also, we analyzed, by immunohistochemistry and Western-blotting, the neuropeptide, arginine vasopressin (AVP), in the SON at the different ages. At birth, Dp71 and to a lesser extends, Dp140 and Dp427, and also β-DG are revealed in the SON. They are localized in the magnocellular neurons (MCNs), astrocytes and vessels. From birth to adulthood, the AVP raise in the SON coincides with the progressive increase of Dp71 level while the level of Dp140 and Dp427 increased only at D20, D10 post-natal development, respectively, and β-DG expression did not change. Moreover, the location of Dps or/and β-DG in the cell compartments was modified during development: at D10, Dps appeared in the astrocytes end-feet surrounding MCNs, and at D20, Dps and β-DG codistributed in the astrocytes end-feet, surrounding MCNs and vessels. Such a distribution marks the first steps of post-natal SON development and may be considered essential in the establishment of structural plasticity mechanisms in SON, where astrocyte end-feet, vessels, magnocellular neurons, are physiologically associated. The disappearance of β-DG in the MCNs nucleus marks the adulthood SON and suggests that the complex of Dps associating β-DG is required for the nucleoskeleton function in the post-natal development.
Collapse
|
16
|
Nirwane A, Yao Y. Laminins and their receptors in the CNS. Biol Rev Camb Philos Soc 2019; 94:283-306. [PMID: 30073746 DOI: 10.1111/brv.12454] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/05/2018] [Accepted: 07/09/2018] [Indexed: 01/24/2023]
Abstract
Laminin, an extracellular matrix protein, is widely expressed in the central nervous system (CNS). By interacting with integrin and non-integrin receptors, laminin exerts a large variety of important functions in the CNS in both physiological and pathological conditions. Due to the existence of many laminin isoforms and their differential expression in various cell types in the CNS, the exact functions of each individual laminin molecule in CNS development and homeostasis remain largely unclear. In this review, we first briefly introduce the structure and biochemistry of laminins and their receptors. Next, the dynamic expression of laminins and their receptors in the CNS during both development and in adulthood is summarized in a cell-type-specific manner, which allows appreciation of their functional redundancy/compensation. Furthermore, we discuss the biological functions of laminins and their receptors in CNS development, blood-brain barrier (BBB) maintenance, neurodegeneration, stroke, and neuroinflammation. Last, key challenges and potential future research directions are summarized and discussed. Our goals are to provide a synthetic review to stimulate future studies and promote the formation of new ideas/hypotheses and new lines of research in this field.
Collapse
Affiliation(s)
- Abhijit Nirwane
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W Green Street, Athens, GA 30602, U.S.A
| | - Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W Green Street, Athens, GA 30602, U.S.A
| |
Collapse
|
17
|
Reduction of Cellular Nucleic Acid Binding Protein Encoded by a Myotonic Dystrophy Type 2 Gene Causes Muscle Atrophy. Mol Cell Biol 2018; 38:MCB.00649-17. [PMID: 29735719 DOI: 10.1128/mcb.00649-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/26/2018] [Indexed: 12/17/2022] Open
Abstract
Myotonic dystrophy type 2 (DM2) is a neuromuscular disease caused by an expansion of intronic CCTG repeats in the CNBP gene, which encodes a protein regulating translation and transcription. To better understand the role of cellular nucleic acid binding protein (CNBP) in DM2 pathology, we examined skeletal muscle in a new model of Cnbp knockout (KO) mice. This study showed that a loss of Cnbp disturbs myofibrillar sarcomeric organization at birth. Surviving homozygous Cnbp KO mice develop muscle atrophy at a young age. The skeletal muscle phenotype in heterozygous Cnbp KO mice was milder, but they developed severe muscle wasting at an advanced age. Several proteins that control global translation and muscle contraction are altered in muscle of Cnbp KO mice. A search for CNBP binding proteins showed that CNBP interacts with the α subunit of the dystroglycan complex, a core component of the multimeric dystrophin-glycoprotein complex, which regulates membrane stability. Whereas CNBP is reduced in cytoplasm of DM2 human fibers, it is a predominantly membrane protein in DM2 fibers, and its interaction with α-dystroglycan is increased in DM2. These findings suggest that alterations of CNBP in DM2 might cause muscle atrophy via CNBP-mediated translation and via protein-protein interactions affecting myofiber membrane function.
Collapse
|
18
|
Kálmán M, Oszwald E, Adorján I. Appearance of β-dystroglycan precedes the formation of glio-vascular end-feet in developing rat brain. Eur J Histochem 2018; 62:2908. [PMID: 29943956 PMCID: PMC5966711 DOI: 10.4081/ejh.2018.2908] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/29/2018] [Accepted: 05/03/2018] [Indexed: 11/23/2022] Open
Abstract
Dystroglycan has an important role in binding of perivascular glial end-feet tothe basal lamina. Its β-subunit is localized in the glial end-feet. The investigation period lasted from E(embryonic day)12 to E20. Laminin and β-dystroglycan were detected by immunohistochemistry, the glial localization of the latter one was supported by electron microscopy. The immatureglial structures were visualized by the immunostaining of nestin. The β-dystroglycan immunoreactivity appeared at E16 following the laminin of basal lamina but preceding the perivascular processes of radial glia (E18) and astrocyte-like cells (E20). It occurred in cell bodies which attached to the vessels directly but not with vascular processes and end-feet. The presence of β-dystroglycan in such immature cells may promote their differentiation to perivascular astrocytes and influence the formation of the glio-vascular processes.
Collapse
Affiliation(s)
- Mihály Kálmán
- Semmelweis University, Department of Anatomy, Histology and Embryology.
| | | | | |
Collapse
|
19
|
Nagy N, Barad C, Hotta R, Bhave S, Arciero E, Dora D, Goldstein AM. Collagen 18 and agrin are secreted by neural crest cells to remodel their microenvironment and regulate their migration during enteric nervous system development. Development 2018; 145:dev.160317. [PMID: 29678817 DOI: 10.1242/dev.160317] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 04/03/2018] [Indexed: 12/12/2022]
Abstract
The enteric nervous system (ENS) arises from neural crest cells that migrate, proliferate, and differentiate into enteric neurons and glia within the intestinal wall. Many extracellular matrix (ECM) components are present in the embryonic gut, but their role in regulating ENS development is largely unknown. Here, we identify heparan sulfate proteoglycan proteins, including collagen XVIII (Col18) and agrin, as important regulators of enteric neural crest-derived cell (ENCDC) development. In developing avian hindgut, Col18 is expressed at the ENCDC wavefront, while agrin expression occurs later. Both proteins are normally present around enteric ganglia, but are absent in aganglionic gut. Using chick-mouse intestinal chimeras and enteric neurospheres, we show that vagal- and sacral-derived ENCDCs from both species secrete Col18 and agrin. Whereas glia express Col18 and agrin, enteric neurons only express the latter. Functional studies demonstrate that Col18 is permissive whereas agrin is strongly inhibitory to ENCDC migration, consistent with the timing of their expression during ENS development. We conclude that ENCDCs govern their own migration by actively remodeling their microenvironment through secretion of ECM proteins.
Collapse
Affiliation(s)
- Nandor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094 Hungary
| | - Csilla Barad
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094 Hungary
| | - Ryo Hotta
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sukhada Bhave
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Emily Arciero
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - David Dora
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094 Hungary
| | - Allan M Goldstein
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
20
|
Thomsen MS, Routhe LJ, Moos T. The vascular basement membrane in the healthy and pathological brain. J Cereb Blood Flow Metab 2017; 37:3300-3317. [PMID: 28753105 PMCID: PMC5624399 DOI: 10.1177/0271678x17722436] [Citation(s) in RCA: 328] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 06/21/2017] [Accepted: 06/28/2017] [Indexed: 12/24/2022]
Abstract
The vascular basement membrane contributes to the integrity of the blood-brain barrier (BBB), which is formed by brain capillary endothelial cells (BCECs). The BCECs receive support from pericytes embedded in the vascular basement membrane and from astrocyte endfeet. The vascular basement membrane forms a three-dimensional protein network predominantly composed of laminin, collagen IV, nidogen, and heparan sulfate proteoglycans that mutually support interactions between BCECs, pericytes, and astrocytes. Major changes in the molecular composition of the vascular basement membrane are observed in acute and chronic neuropathological settings. In the present review, we cover the significance of the vascular basement membrane in the healthy and pathological brain. In stroke, loss of BBB integrity is accompanied by upregulation of proteolytic enzymes and degradation of vascular basement membrane proteins. There is yet no causal relationship between expression or activity of matrix proteases and the degradation of vascular matrix proteins in vivo. In Alzheimer's disease, changes in the vascular basement membrane include accumulation of Aβ, composite changes, and thickening. The physical properties of the vascular basement membrane carry the potential of obstructing drug delivery to the brain, e.g. thickening of the basement membrane can affect drug delivery to the brain, especially the delivery of nanoparticles.
Collapse
Affiliation(s)
- Maj S Thomsen
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Lisa J Routhe
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Torben Moos
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
21
|
Bhat HF, Mir SS, Dar KB, Bhat ZF, Shah RA, Ganai NA. ABC of multifaceted dystrophin glycoprotein complex (DGC). J Cell Physiol 2017; 233:5142-5159. [DOI: 10.1002/jcp.25982] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/01/2017] [Indexed: 01/16/2023]
Affiliation(s)
- Hina F. Bhat
- Division of BiotechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of Kashmir SKUAST‐KShuhama, SrinagarJammu and KashmirIndia
| | - Saima S. Mir
- Department of BiotechnologyUniversity of KashmirHazratbal, SrinagarJammu and KashmirIndia
| | - Khalid B. Dar
- Department of BiochemistryUniversity of KashmirHazratbal, SrinagarJammu and KashmirIndia
| | - Zuhaib F. Bhat
- Division of Livestock Products and TechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of Jammu (SKUAST‐J), R.S. PoraJammuJammu and KashmirIndia
| | - Riaz A. Shah
- Division of BiotechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of Kashmir SKUAST‐KShuhama, SrinagarJammu and KashmirIndia
| | - Nazir A. Ganai
- Division of BiotechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of Kashmir SKUAST‐KShuhama, SrinagarJammu and KashmirIndia
| |
Collapse
|
22
|
Brancaccio A, Adams JC. An evaluation of the evolution of the gene structure of dystroglycan. BMC Res Notes 2017; 10:19. [PMID: 28057052 PMCID: PMC5216574 DOI: 10.1186/s13104-016-2322-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 12/06/2016] [Indexed: 11/10/2022] Open
Abstract
Background Dystroglycan (DG) is an adhesion receptor complex composed of two non-covalently associated subunits, transcribed from a single gene. The extracellular α-DG is highly and heterogeneously glycosylated and binds with high affinity to laminins, and the transmembrane β-DG binds intracellular dystrophin. Multiple cellular functions have been proposed for DG, notwithstanding that its role in skeletal muscle appears central as demonstrated by both primary and secondary severe muscular dystrophic phenotypes collectively known as dystroglycanopathies. We recently analysed the molecular phylogeny of the DG core protein and identified the α/β interface, transmembrane and cytoplasmic domains of β-DG as the most conserved region. It was also identified that the IG2_MAT_NU region has been independently duplicated in multiple lineages. Results To understand the evolution of dystroglycan in more depth, we investigated dystroglycan gene structure in 35 species representative of the phyla in which dystroglycan has been identified (i.e., all metazoan phyla except Ctenophora). The gene structure of three exons and two introns is remarkably conserved. However, additional lineage-specific introns were identified, which interrupt the coding sequence at distinct points, were identified in multiple metazoan groups, most prominently in ecdysozoans. Conclusions A coding DNA sequence (CDS) intron that interrupts the encoding of the IG1 domain is universally conserved and this intron is longer in gnathostomes (jawed vertebrates) than in other metazoans. Lineage-specific gain of additional introns has occurred notably in ecdysozoans, where multiple introns interrupt the large 3′ exon. More limited intron gain has also occurred in placozoa, cnidarians, urochordates and the DG paralogues of lamprey and teleost fish. Electronic supplementary material The online version of this article (doi:10.1186/s13104-016-2322-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrea Brancaccio
- Istituto di Chimica del Riconoscimento Molecolare, CNR, Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy. .,School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| | - Josephine C Adams
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| |
Collapse
|
23
|
Ragni E, Lommel M, Moro M, Crosti M, Lavazza C, Parazzi V, Saredi S, Strahl S, Lazzari L. Protein O-mannosylation is crucial for human mesencyhmal stem cells fate. Cell Mol Life Sci 2016; 73:445-58. [PMID: 26245304 PMCID: PMC11108538 DOI: 10.1007/s00018-015-2007-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 07/27/2015] [Accepted: 07/29/2015] [Indexed: 10/23/2022]
Abstract
Human mesenchymal stem cells (MSC) are promising cell types in the field of regenerative medicine. Although many pathways have been dissected in the effort to better understand and characterize MSC potential, the impact of protein N- or O-glycosylation has been neglected. Deficient protein O-mannosylation is a pathomechanism underlying severe congenital muscular dystrophies (CMD) that start to develop at the embryonic developmental stage and progress in the adult, often in tissues where MSC exert their function. Here we show that O-mannosylation genes, many of which are putative or verified glycosyltransferases (GTs), are expressed in a similar pattern in MSC from adipose tissue, bone marrow, and umbilical cord blood and that their expression levels are retained constant during mesengenic differentiation. Inhibition of the first players of the enzymatic cascade, POMT1/2, resulted in complete abolishment of chondrogenesis and alterations of adipogenic and osteogenic potential together with a lethal effect during myogenic induction. Since to date, no therapy for CMD is available, we explored the possibility of using MSC extracellular vesicles (EVs) as molecular source of functional GTs mRNA. All MSC secrete POMT1 mRNA-containing EVs that are able to efficiently fuse with myoblasts which are among the most affected cells by CMD. Intriguingly, in a pomt1 patient myoblast line EVs were able to partially revert O-mannosylation deficiency and contribute to a morphology recovery. Altogether, these results emphasize the crucial role of protein O-mannosylation in stem cell fate and properties and open the possibility of using MSC vesicles as a novel therapeutic approach to CMD.
Collapse
Affiliation(s)
- E Ragni
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - M Lommel
- Centre for Organismal Studies, Cell Chemistry and Center for Molecular Biology, University of Heidelberg, 69120, Heidelberg, Germany
| | - M Moro
- Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi" (INGM), Milan, Italy
| | - M Crosti
- Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi" (INGM), Milan, Italy
| | - C Lavazza
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - V Parazzi
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - S Saredi
- Division of Neuromuscular Diseases and Neuroimmunology, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - S Strahl
- Centre for Organismal Studies, Cell Chemistry and Center for Molecular Biology, University of Heidelberg, 69120, Heidelberg, Germany
| | - L Lazzari
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
24
|
Dystroglycan Depletion Impairs Actin-Dependent Functions of Differentiated Kasumi-1 Cells. PLoS One 2015; 10:e0144078. [PMID: 26630171 PMCID: PMC4668107 DOI: 10.1371/journal.pone.0144078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 11/12/2015] [Indexed: 01/20/2023] Open
Abstract
Background Dystroglycan has recently been characterised in blood tissue cells, as part of the dystrophin glycoprotein complex involved in the differentiation process of neutrophils. Purpose In the present study we have investigated the role of dystroglycan in the human promyelocytic leukemic cell line Kasumi-1 differentiated to macrophage-like cells. Methods We characterised the pattern expression and subcellular distribution of dystroglycans in non-differentiated and differentiated Kasumi-1 cells. Results Our results demonstrated by WB and flow cytometer assays that during the differentiation process to macrophages, dystroglycans were down-regulated; these results were confirmed with qRT-PCR assays. Additionally, depletion of dystroglycan by RNAi resulted in altered morphology and reduced properties of differentiated Kasumi-1 cells, including morphology, migration and phagocytic activities although secretion of IL-1β and expression of markers of differentiation are not altered. Conclusion Our findings strongly implicate dystroglycan as a key membrane adhesion protein involved in actin-based structures during the differentiation process in Kasumi-1 cells.
Collapse
|
25
|
Adams JC, Brancaccio A. The evolution of the dystroglycan complex, a major mediator of muscle integrity. Biol Open 2015; 4:1163-79. [PMID: 26319583 PMCID: PMC4582122 DOI: 10.1242/bio.012468] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Basement membrane (BM) extracellular matrices are crucial for the coordination of different tissue layers. A matrix adhesion receptor that is important for BM function and stability in many mammalian tissues is the dystroglycan (DG) complex. This comprises the non-covalently-associated extracellular α-DG, that interacts with laminin in the BM, and the transmembrane β-DG, that interacts principally with dystrophin to connect to the actin cytoskeleton. Mutations in dystrophin, DG, or several enzymes that glycosylate α-DG underlie severe forms of human muscular dystrophy. Nonwithstanding the pathophysiological importance of the DG complex and its fundamental interest as a non-integrin system of cell-ECM adhesion, the evolution of DG and its interacting proteins is not understood. We analysed the phylogenetic distribution of DG, its proximal binding partners and key processing enzymes in extant metazoan and relevant outgroups. We identify that DG originated after the divergence of ctenophores from porifera and eumetazoa. The C-terminal half of the DG core protein is highly-conserved, yet the N-terminal region, that includes the laminin-binding region, has undergone major lineage-specific divergences. Phylogenetic analysis based on the C-terminal IG2_MAT_NU region identified three distinct clades corresponding to deuterostomes, arthropods, and mollusks/early-diverging metazoans. Whereas the glycosyltransferases that modify α-DG are also present in choanoflagellates, the DG-binding proteins dystrophin and laminin originated at the base of the metazoa, and DG-associated sarcoglycan is restricted to cnidarians and bilaterians. These findings implicate extensive functional diversification of DG within invertebrate lineages and identify the laminin-DG-dystrophin axis as a conserved adhesion system that evolved subsequent to integrin-ECM adhesion, likely to enhance the functional complexity of cell-BM interactions in early metazoans.
Collapse
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Andrea Brancaccio
- Istituto di Chimica del Riconoscimento Molecolare, CNR, Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Roma 00168, Italy
| |
Collapse
|
26
|
Humphrey EL, Lacey E, Le LT, Feng L, Sciandra F, Morris CR, Hewitt JE, Holt I, Brancaccio A, Barresi R, Sewry CA, Brown SC, Morris GE. A new monoclonal antibody DAG-6F4 against human alpha-dystroglycan reveals reduced core protein in some, but not all, dystroglycanopathy patients. Neuromuscul Disord 2015; 25:32-42. [DOI: 10.1016/j.nmd.2014.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 09/01/2014] [Accepted: 09/08/2014] [Indexed: 11/28/2022]
|
27
|
Zhang X, Dong XH, Ma Y, Li LF, Wu H, Zhou M, Gu YH, Li GZ, Wang DS, Zhang XF, Mou J, Qi JP. Reduction of α-dystroglycan expression is correlated with poor prognosis in glioma. Tumour Biol 2014; 35:11621-9. [PMID: 25139094 DOI: 10.1007/s13277-014-2418-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/29/2014] [Indexed: 01/12/2023] Open
Abstract
Dystroglycan (DG), a multifunctional protein dimer of non-covalently linked α and β subunits, is best known as an adhesion and transduction molecule linking the cytoskeleton and intracellular signaling pathways to extracellular matrix proteins. Loss of DG binding, possibly by degradation or disturbed glycosylation, has been reported in a variety of cancers. DG is abundant at astroglial endfeet forming the blood-brain barrier (BBB) and glia limitans; so, we examined if loss of expression is associated with glioma. Expression levels of α-DG and β-DG were assessed by immunohistochemistry in a series of 78 glioma specimens to determine the relationship with tumor grade and possible prognostic significance. α-DG immunostaining was undetectable in 44 of 49 high-grade specimens (89.8%) compared to 15 of 29 low-grade specimens (51.72%) (P<0.05). Moreover, loss of α-DG expression was an independent predictor of shorter disease-free survival (DFS) (hazards ratio (HR) = 0.142, 95% confidence interval (CI) 0.033-0.611, P=0.0088). Reduced expression of both α-DG and β-DG was also a powerful negative prognostic factor for DFS (HR=2.556, 95% CI 1.403-4.654, P=0.0022) and overall survival (OS) (HR=2.193, 95% CI 1.031-4.666, P=0.0414). Lack of α-DG immunoreactivity is more frequent in high-grade glioma and is an independent predictor of poor clinical outcome. Similarly, lack of both α-DG and β-DG immunoreactivity is a strong independent predictor of clinical outcome.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, No.23 Youzheng Street, NanGang District, Harbin, 150001, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Zhang HZ, Xia XY, Zhu F, Shen H, Song K, Shang ZJ. Correlation of deregulated like-acetylglucosaminyl transferase and aberrant α-dystroglycan expression with human tongue cancer metastasis. J Oral Maxillofac Surg 2014; 72:1106-18. [PMID: 24629698 DOI: 10.1016/j.joms.2013.12.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 12/20/2013] [Accepted: 12/26/2013] [Indexed: 11/16/2022]
Abstract
PURPOSE The present study examined the correlation of α-dystroglycan (α-DG) expression and like-acetylglucosaminyl transferase (LARGE) with metastasis of human tongue cancer. MATERIALS AND METHODS Fifty human tongue cancer tissues and 2 tongue squamous cell carcinoma cell lines (CAL27 and SCC4) were involved. Immunohistochemistry was used to detect the expression of α-DG and LARGE. Methylation-specific polymerase chain reaction was performed to assess the methylation status of the LARGE gene promoter. CAL27 and SCC4 cells were transfected with exogenous LARGE and treated with 5-aza-2'-deoxycytidine (Aza-dC), respectively. Glycol sites of α-DG were detected by western blotting. In addition, the laminin overlay assay, cell adhesion assay, and invasion assay were performed. RESULTS Immunohistochemical results showed that decreased expression of VIA4-1 and IIH6 (antibodies that recognize the glycol sites of α-DG) were correlated with the lymph node metastasis of tongue cancer (n = 50; P = .016 and .025, respectively). Decreased LARGE expression and hypermethylation of the LARGE gene promoter were correlated with lymph node metastasis and α-DG glycosylation in human tongue cancer (n = 50; P = .043 and .015 respectively). In addition, LARGE overexpression and Aza-dC treatment actively led to restoration of functional α-DG expression, elevation of laminin binding, and decrease of migratory ability in cancer cells. CONCLUSION The results suggested that absent α-DG expression and LARGE deregulation were closely associated with nodal metastasis of tongue cancer. Aberrant α-DG expression and glycosylation were attributed at least in part to the abnormal epigenetic modification of LARGE, especially the hypermethylation of its promoter.
Collapse
Affiliation(s)
- Han-Zhong Zhang
- PhD Student, State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Xiao-Yan Xia
- PhD Student, State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Fei Zhu
- PhD Student, State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Hui Shen
- PhD Student, State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Kai Song
- PhD Student, State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Zheng-Jun Shang
- Professor, State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial and Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
29
|
C-terminal region of teneurin-1 co-localizes with the dystroglycan complex in adult mouse testes and regulates testicular size and testosterone production. Histochem Cell Biol 2013; 141:191-211. [PMID: 24154551 DOI: 10.1007/s00418-013-1154-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2013] [Indexed: 01/10/2023]
Abstract
Testicular size is directly proportional to fertility potential and is dependent on the integration of developmental proteins, trophic factors, and sex steroids. The teneurins are transmembrane glycoproteins that function as signaling and cell adhesion molecules in the establishment and maintenance of the somatic gonad, gametogenesis, and basement membrane. Moreover, teneurins are thought to function redundantly to the extracellular matrix protein, dystroglycan. Encoded on the last exon of the teneurin genes is a family of bioactive peptides termed the teneurin C-terminal-associated peptides (TCAPs). One of these peptides, TCAP-1, functionally interacts with β-dystroglycan to act as a neuromodulatory peptide with trophic characteristics independent from the teneurins. However, little is known about the localization and relationship between the teneurin-TCAP-1 system and the dystroglycans in the gonad. In the adult mouse testis, immunoreactive TCAP-1 was localized to spermatogonia and spermatocytes and co-localized with β-dystroglycan. However, teneurin-1 was localized to the peritubular myoid cell layer of seminiferous tubules and tubules within the epididymis, and co-localized with α-dystroglycan and α-smooth muscle actin. TCAP-1-binding sites were identified in the germ cell layers and adluminal compartment of the seminiferous tubules, and epithelial cells of the epididymis. In vivo, TCAP-1 administration to adult mice for 9 days increased testicular size, seminiferous and epididymal tubule short-diameter and elevated testosterone levels. TCAP-1-treated mice also showed increased TCAP-1 immunoreactivity in the caput and corpa epididymis. Our data provide novel evidence of TCAP-1 localization in the testes that is distinct from teneurin-1, but is integrated through an association with the dystroglycan complex.
Collapse
|
30
|
Flow cytometry for the analysis of α-dystroglycan glycosylation in fibroblasts from patients with dystroglycanopathies. PLoS One 2013; 8:e68958. [PMID: 23894383 PMCID: PMC3718821 DOI: 10.1371/journal.pone.0068958] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 06/10/2013] [Indexed: 01/26/2023] Open
Abstract
α-dystroglycan (α-DG) is a peripheral membrane protein that is an integral component of the dystrophin-glycoprotein complex. In an inherited subset of muscular dystrophies known as dystroglycanopathies, α-DG has reduced glycosylation which results in lower affinity binding to several extracellular matrix proteins including laminins. The glycosylation status of α-DG is normally assessed by the binding of the α-DG antibody IIH6 to a specific glycan epitope on α-DG involved in laminin binding. Immunocytochemistry and immunoblotting are two of the most widely used methods to detect the amount of α-DG glycosylation in muscle. While the interpretation of the presence or absence of the epitope on muscle using these techniques is straightforward, the assessment of a mild defect can be challenging. In this study, flow cytometry was used to compare the amount of IIH6-reactive glycans in fibroblasts from dystroglycanopathy patients with defects in genes known to cause α-DG hypoglycosylation to the amount in fibroblasts from healthy and pathological control subjects. A total of twenty one dystroglycanopathy patient fibroblasts were assessed, as well as fibroblasts from three healthy controls and seven pathological controls. Control fibroblasts have clearly detectable amounts of IIH6-reactive glycans, and there is a significant difference in the amount of this glycosylation, as measured by the mean fluorescence intensity of an antibody recognising the epitope and the percentage of cells positive for the epitope, between these controls and dystroglycanopathy patient fibroblasts (p<0.0001 for both). Our results indicate that the amount of α-DG glycosylation in patient fibroblasts is comparable to that in patient skeletal muscle. This method could complement existing immunohistochemical assays in skeletal muscle as it is quantitative and simple to perform, and could be used when a muscle biopsy is not available. This test could also be used to assess the pathogenicity of variants of unknown significance in genes involved in dystroglycanopathies.
Collapse
|
31
|
Mitchell A, Mathew G, Jiang T, Hamdy FC, Cross SS, Eaton C, Winder SJ. Dystroglycan function is a novel determinant of tumor growth and behavior in prostate cancer. Prostate 2013; 73:398-408. [PMID: 22996647 DOI: 10.1002/pros.22581] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 08/16/2012] [Indexed: 12/16/2022]
Abstract
BACKGROUND Dystroglycan is a ubiquitously expressed cell adhesion molecule frequently found to be altered or reduced in adenocarcinomas, however the mechanisms or consequences of dystroglycan loss have not been studied extensively. METHODS We examined the consequence of overexpression or RNAi depletion of dystroglycan on properties of in vitro growth migration and invasion of LNCaP, PC3, and DU145 prostate cancer cell lines. RESULTS Using LNCaP cells we observed cell density-dependent changes in β-dystroglycan with the appearance of several lower molecular weight species ranging in size from 43 to 26 kDa. The bands of 31 and 26 kDa were attributed to proteolysis, whereas bands between 43 and 38 kDa were a consequence of mis-glycosylation. The localization of β-dystroglycan in LNCaP colonies in culture also varied, cells with a mesenchymal appearance at the periphery of the colony had more pronounced membrane localization of dystroglycan. Whereas some cells demonstrated nuclear dystroglycan. Increased dystroglycan levels were inhibitory to growth in soft agar but promoted Matrigel invasion, whereas reduced dystroglycan levels promoted growth in soft agar but inhibited invasion. Similar results were also obtained for PC3 and DU145 cells. CONCLUSIONS This study suggests that changes in β-dystroglycan distribution within the cell and/or the loss of dystroglycan during tumorigenesis, through a combination of proteolysis and altered glycosylation, leads to an increased ability to grow in an anchorage independent manner, however dystroglycan may need to be re-expressed for cell invasion and metastasis to occur.
Collapse
Affiliation(s)
- A Mitchell
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | |
Collapse
|
32
|
Martínez-Vieyra IA, Vásquez-Limeta A, González-Ramírez R, Morales-Lázaro SL, Mondragón M, Mondragón R, Ortega A, Winder SJ, Cisneros B. A role for β-dystroglycan in the organization and structure of the nucleus in myoblasts. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1833:698-711. [PMID: 23220011 DOI: 10.1016/j.bbamcr.2012.11.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 11/17/2012] [Accepted: 11/24/2012] [Indexed: 12/19/2022]
Abstract
We recently characterized a nuclear import pathway for β-dystroglycan; however, its nuclear role remains unknown. In this study, we demonstrate for the first time, the interaction of β-dystroglycan with distinct proteins from different nuclear compartments, including the nuclear envelope (NE) (emerin and lamins A/C and B1), splicing speckles (SC35), Cajal bodies (p80-coilin), and nucleoli (Nopp140). Electron microscopy analysis revealed that β-dystroglycan localized in the inner nuclear membrane, nucleoplasm, and nucleoli. Interestingly, downregulation of β-dystroglycan resulted in both mislocalization and decreased expression of emerin and lamin B1, but not lamin A/C, as well in disorganization of nucleoli, Cajal bodies, and splicing speckles with the concomitant decrease in the levels of Nopp140, and p80-coilin, but not SC35. Quantitative reverse transcription PCR and cycloheximide-mediated protein arrest assays revealed that β-dystroglycan deficiency did not change mRNA expression of NE proteins emerin and lamin B1 bud did alter their stability, accelerating protein turnover. Furthermore, knockdown of β-dystroglycan disrupted NE-mediated processes including nuclear morphology and centrosome-nucleus linkage, which provides evidence that β-dystroglycan association with NE proteins is biologically relevant. Unexpectedly, β-dystroglycan-depleted cells exhibited multiple centrosomes, a characteristic of cancerous cells. Overall, these findings imply that β-dystroglycan is a nuclear scaffolding protein involved in nuclear organization and NE structure and function, and that might be a contributor to the biogenesis of nuclear envelopathies.
Collapse
Affiliation(s)
- Ivette A Martínez-Vieyra
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del IPN, México, DF 07360, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Role of perlecan, a basement membrane-type heparan sulfate proteoglycan, in enamel organ morphogenesis. J Oral Biosci 2013. [DOI: 10.1016/j.job.2012.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
34
|
Sarkozy A, Bushby K, Mercuri E. Muscular Dystrophies. EMERY AND RIMOIN'S PRINCIPLES AND PRACTICE OF MEDICAL GENETICS 2013:1-58. [DOI: 10.1016/b978-0-12-383834-6.00134-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
35
|
Dobson CM, Hempel SJ, Stalnaker SH, Stuart R, Wells L. O-Mannosylation and human disease. Cell Mol Life Sci 2012; 70:2849-57. [PMID: 23115008 DOI: 10.1007/s00018-012-1193-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 10/02/2012] [Accepted: 10/08/2012] [Indexed: 12/21/2022]
Abstract
Glycosylation of proteins is arguably the most prevalent co- and post-translational modification. It is responsible for increased heterogeneity and functional diversity of proteins. Here we discuss the importance of one type of glycosylation, specifically O-mannosylation and its relationship to a number of human diseases. The most widely studied O-mannose modified protein is alpha-dystroglycan (α-DG). Recent studies have focused intensely on α-DG due to the severity of diseases associated with its improper glycosylation. O-mannosylation of α-DG is involved in cancer metastasis, arenavirus entry, and multiple forms of congenital muscular dystrophy [1, 2]. In this review, we discuss the structural and functional characteristics of O-mannose-initiated glycan structures on α-DG, enzymes involved in the O-mannosylation pathway, and the diseases that are a direct result of disruptions within this pathway.
Collapse
Affiliation(s)
- Christina M Dobson
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | | | | | | | | |
Collapse
|
36
|
Coco C, Zannoni GF, Caredda E, Sioletic S, Boninsegna A, Migaldi M, Rizzo G, Bonetti LR, Genovese G, Stigliano E, Cittadini A, Sgambato A. Increased expression of CD133 and reduced dystroglycan expression are strong predictors of poor outcome in colon cancer patients. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:71. [PMID: 22964035 PMCID: PMC3541988 DOI: 10.1186/1756-9966-31-71] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 08/28/2012] [Indexed: 02/08/2023]
Abstract
Background Expression levels of CD133, a cancer stem cell marker, and of the α-subunit of the dystroglycan (α-DG) complex, have been previously reported to be altered in colorectal cancers. Methods Expression levels of CD133 and α-DG were assessed by immunohistochemistry in a series of colon cancers and their prognostic significance was evaluated. Results Scattered cells positive for CD133 were rarely detected at the bases of the crypts in normal colonic mucosa while in cancer cells the median percentage of positive cells was 5% (range 0–80). A significant correlation was observed with pT parameter and tumor stage but not with tumor grade and N status. Recurrence and death from disease were significantly more frequent in CD133-high expressing tumors and Kaplan-Meier curves showed a significant separation between high vs low expressor groups for both disease-free (p = 0.002) and overall (p = 0.008) survival. Expression of α-DG was reduced in a significant fraction of tumors but low α-DG staining did not correlate with any of the classical clinical-pathological parameters. Recurrence and death from the disease were significantly more frequent in α-DG-low expressing tumors and Kaplan-Meier curves showed a significant separation between high vs low expressor tumors for both disease-free (p = 0.02) and overall (p = 0.02) survival. Increased expression of CD133, but not loss of α-DG, confirmed to be an independent prognostic parameters at a multivariate analysis associated with an increased risk of recurrence (RR = 2.4; p = 0.002) and death (RR = 2.3; p = 0.003). Conclusions Loss of α-DG and increased CD133 expression are frequent events in human colon cancer and evaluation of CD133 expression could help to identify high-risk colon cancer patients.
Collapse
Affiliation(s)
- Claudio Coco
- Dipartimento di Scienze Chirurgiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Miller G, Moore CJ, Terry R, La Riviere T, Mitchell A, Piggott R, Dear TN, Wells DJ, Winder SJ. Preventing phosphorylation of dystroglycan ameliorates the dystrophic phenotype in mdx mouse. Hum Mol Genet 2012; 21:4508-20. [PMID: 22810924 DOI: 10.1093/hmg/dds293] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Loss of dystrophin protein due to mutations in the DMD gene causes Duchenne muscular dystrophy. Dystrophin loss also leads to the loss of the dystrophin glycoprotein complex (DGC) from the sarcolemma which contributes to the dystrophic phenotype. Tyrosine phosphorylation of dystroglycan has been identified as a possible signal to promote the proteasomal degradation of the DGC. In order to test the role of tyrosine phosphorylation of dystroglycan in the aetiology of DMD, we generated a knock-in mouse with a phenylalanine substitution at a key tyrosine phosphorylation site in dystroglycan, Y890. Dystroglycan knock-in mice (Dag1(Y890F/Y890F)) had no overt phenotype. In order to examine the consequence of blocking dystroglycan phosphorylation on the aetiology of dystrophin-deficient muscular dystrophy, the Y890F mice were crossed with mdx mice an established model of muscular dystrophy. Dag1(Y890F/Y890F)/mdx mice showed a significant improvement in several parameters of muscle pathophysiology associated with muscular dystrophy, including a reduction in centrally nucleated fibres, less Evans blue dye infiltration and lower serum creatine kinase levels. With the exception of dystrophin, other DGC components were restored to the sarcolemma including α-sarcoglycan, α-/β-dystroglycan and sarcospan. Furthermore, Dag1(Y890F/Y890F)/mdx showed a significant resistance to muscle damage and force loss following repeated eccentric contractions when compared with mdx mice. While the Y890F substitution may prevent dystroglycan from proteasomal degradation, an increase in sarcolemmal plectin appeared to confer protection on Dag1(Y890F/Y890F)/mdx mouse muscle. This new model confirms dystroglycan phosphorylation as an important pathway in the aetiology of DMD and provides novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Gaynor Miller
- Department of Cardiovascular Science, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Sciandra F, Angelucci E, Altieri F, Ricci D, Hübner W, Petrucci TC, Giardina B, Brancaccio A, Bozzi M. Dystroglycan is associated to the disulfide isomerase ERp57. Exp Cell Res 2012; 318:2460-9. [PMID: 22814252 PMCID: PMC3459099 DOI: 10.1016/j.yexcr.2012.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 06/27/2012] [Accepted: 07/08/2012] [Indexed: 11/26/2022]
Abstract
Dystroglycan (DG) is an extracellular receptor composed of two subunits, α-DG and β-DG, connected through the α-DG C-terminal domain and the β-DG N-terminal domain. We report an alanine scanning of all DG cysteine residues performed on DG-GFP constructs overexpressed in 293-Ebna cells, demonstrating that Cys-669 and Cys-713, both located within the β-DG N-terminal domain, are key residues for the DG precursor cleavage and trafficking, but not for the interaction between the two DG subunits. In addition, we have used immunprecipitation and confocal microscopy showing that ERp57, a member of the disulfide isomerase family involved in glycoprotein folding, is associated and colocalizes immunohistochemically with β-DG in the ER and at the plasma membrane of 293-Ebna cells. The β-DG–ERp57 complex also included α-DG. DG mutants, unable to undergo the precursor cleavage, were still associated to ERp57. β-DG and ERp57 were also co-immunoprecipitated in rat heart and kidney tissues. In vitro, a mutant ERp57, mimicking the reduced form of the wild-type protein, interacts directly with the recombinant N-terminal domain of both α-DG and β-DG with apparent dissociation constant values in the micromolar range. ERp57 is likely to be involved in the DG processing/maturation pathway, but its association to the mature DG complex might also suggest some further functional role that needs to be investigated.
Collapse
Affiliation(s)
- Francesca Sciandra
- Istituto di Chimica del Riconoscimento Molecolare (CNR), c/o Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Roma, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Moore CJ, Winder SJ. The inside and out of dystroglycan post-translational modification. Neuromuscul Disord 2012; 22:959-65. [PMID: 22770978 DOI: 10.1016/j.nmd.2012.05.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 04/27/2012] [Accepted: 05/28/2012] [Indexed: 01/06/2023]
Abstract
In neuromuscular systems dystroglycan provides a vital link between laminin in the extracellular matrix and dystrophin in the membrane cytoskeleton. The integrity of this link is maintained and regulated by post-translational modifications of dystroglycan that have effects both inside and outside the cell. Glycosylation of α-dystroglycan is crucial for its link to laminin and phosphorylation of β-dystroglycan on tyrosine regulates its association with intracellular binding partners. This short review focuses on some of the recent developments in our understanding of the role of these post-translational modification in regulating dystroglycan function, and how new knowledge of signalling through the laminin-dystroglycan axis is leading to hope for treatment for some neuromuscular diseases associated with this adhesion complex.
Collapse
Affiliation(s)
- Chris J Moore
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK.
| | | |
Collapse
|
40
|
Lee JK, Matthews RT, Lim JM, Swanier K, Wells L, Pierce JM. Developmental expression of the neuron-specific N-acetylglucosaminyltransferase Vb (GnT-Vb/IX) and identification of its in vivo glycan products in comparison with those of its paralog, GnT-V. J Biol Chem 2012; 287:28526-36. [PMID: 22715095 DOI: 10.1074/jbc.m112.367565] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The severe phenotypic effects of altered glycosylation in the congenital muscular dystrophies, including Walker-Warburg syndrome, muscle-eye-brain disease, Fukuyama congenital muscular dystrophy, and congenital muscular dystrophy 1D, are caused by mutations resulting in altered glycans linked to proteins through O-linked mannose. A glycosyltransferase that branches O-Man, N-acetylglucosaminyltransferase Vb (GnT-Vb), is highly expressed in neural tissues. To understand the expression and function of GnT-Vb, we studied its expression during neuromorphogenesis and generated GnT-Vb null mice. A paralog of GnT-Vb, N-acetylglucosaminyltransferase (GnT-V), is expressed in many tissues and brain, synthesizing N-linked, β1,6-branched glycans, but its ability to synthesize O-mannosyl-branched glycans is unknown; conversely, although GnT-Vb can synthesize N-linked glycans in vitro, its contribution to their synthesis in vivo is unknown. Our results showed that deleting both GnT-V and GnT-Vb results in the total loss of both N-linked and O-Man-linked β1,6-branched glycans. GnT-V null brains lacked N-linked, β1,6-glycans but had normal levels of O-Man β1,6-branched structures, showing that GnT-Vb could not compensate for the loss of GnT-V. By contrast, GnT-Vb null brains contained normal levels of N-linked β1,6-glycans but low levels of some O-Man β1,6-branched glycans. Therefore, GnT-V could partially compensate for GnT-Vb activity in vivo. We found no apparent change in α-dystroglycan binding of glycan-specific antibody IIH6C4 or binding to laminin in GnT-Vb null mice. These results demonstrate that GnT-V is involved in synthesizing branched O-mannosyl glycans in brain, but the function of these branched O-mannosyl structures is unresolved using mice that lack these glycosyltransferases.
Collapse
Affiliation(s)
- Jin Kyu Lee
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30605, USA
| | | | | | | | | | | |
Collapse
|
41
|
Alpha-Dystrobrevin and its associated proteins in human promyelocytic leukemia cells induced to apoptosis. J Proteomics 2012; 75:3291-303. [PMID: 22507200 DOI: 10.1016/j.jprot.2012.03.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 03/19/2012] [Accepted: 03/25/2012] [Indexed: 12/13/2022]
Abstract
Dystrobrevin is a dystrophin-related component of the dystrophin-associated protein complex (DAPC). Using alpha-dystrobrevin as indicator, we aimed to elucidate the interaction network of the DAPC with other proteins during apoptosis of promyelocytic HL-60 cells. The precise role(s) of DBs are not known, but we and others have shown that they play a role in intracellular signal transduction and cellular organization. Apoptosis was induced with etoposide in the absence or presence of Z-VAD to block caspase activity, and we then followed the cellular distribution of α-DB and its association with other proteins, using confocal imaging and cell fractions analyses after immune-precipitation with anti-α-DB and mass spectrometry. Confocal imaging revealed distinct spatial relocalizations of α-DB between the cell membrane, cytosol and nucleus after induction of apoptosis. The expression levels of the identified proteins were evaluated with computer-assisted image analysis of the gels. We thus identified associations with structural and transport proteins (tropomyosin, myosin), membrane (ADAM21, syntrophin), ER-Golgi (TGN51, eIF38) and nuclear (Lamins, ribonucleoprotein C1/C2) proteins. These results suggest that apoptosis-induction in HL-60 cells involves not only classical markers of apoptosis but also a network α-DB-associated proteins at the cell membrane, the cytoplasm and nucleus, affecting key cellular transport processes and cellular structure.
Collapse
|
42
|
Chand D, Song L, deLannoy L, Barsyte-Lovejoy D, Ackloo S, Boutros PC, Evans K, Belsham DD, Lovejoy DA. C-Terminal region of teneurin-1 co-localizes with dystroglycan and modulates cytoskeletal organization through an extracellular signal-regulated kinase-dependent stathmin- and filamin A-mediated mechanism in hippocampal cells. Neuroscience 2012; 219:255-70. [PMID: 22698694 DOI: 10.1016/j.neuroscience.2012.05.069] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 05/07/2012] [Accepted: 05/30/2012] [Indexed: 10/28/2022]
Abstract
The pyramidal neurons in the hippocampus are extremely neuroplastic, and the complexity of dendritic branches can be dynamically altered in response to a variety of stimuli, including learning and stress. Recently, the teneurin family of proteins has emerged as an interneuronal and extracellular matrix signaling system that plays a significant role in brain development and neuronal communication. Encoded on the last exon of the teneurin genes is a new family of bioactive peptides termed the teneurin C-terminal-associated peptides (TCAPs). Previous studies indicate that TCAP-1 regulates axon fasciculation and dendritic morphology in the hippocampus. This study was aimed at understanding the molecular mechanisms by which TCAP-1 regulates these changes in the mouse hippocampus. Fluoresceinisothiocyanate (FITC)-labeled TCAP-1 binds to the pyramidal neurons of the CA2 and CA3, and dentate gyrus in the hippocampus of the mouse brain. Moreover, FITC-TCAP-1 co-localizes with β-dystroglycan upon binding to the plasma membrane of cultured immortalized mouse E14 hippocampal cells. In culture, TCAP-1 stimulates ERK1/2-dependent phosphorylation of the cytoskeletal regulatory proteins, stathmin at serine-25 and filamin A at serine-2152. In addition, TCAP-1 induces actin polymerization, increases immunoreactivity of tubulin-based cytoskeletal elements and causes a corresponding increase in filopodia formation and mean filopodia length in cultured hippocampal cells. We postulate that the TCAP-1 region of teneurin-1 has a direct action on the cytoskeletal reorganization that precedes neurite and process development in hippocampal neurons. Our data provides novel evidence that functionally links the teneurin and dystroglycan systems and provides new insight into the molecular mechanisms by which TCAP-1 regulates cytoskeletal dynamics in hippocampal neurons. The TCAP-dystroglycan system may represent a novel mechanism associated with the regulation of hippocampal-function.
Collapse
Affiliation(s)
- D Chand
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON, Canada M5S 3G5.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Benabdesselam R, Dorbani-Mamine L, Benmessaoud-Mesbah O, Rendon A, Mhaouty-Kodja S, Hardin-Pouzet H. Dp71 gene disruption alters the composition of the dystrophin-associated protein complex and neuronal nitric oxide synthase expression in the hypothalamic supraoptic and paraventricular nuclei. J Endocrinol 2012; 213:239-49. [PMID: 22493004 DOI: 10.1530/joe-12-0066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
DP71 is the major cerebral dystrophin isoform and exerts its multiple functions via the dystrophin-associated protein complex (DAPC), also comprised of β-dystroglycan (β-DG) and α1-syntrophin (α1-Syn). Since DP71 disruption leads to impairment in the central control of the osmoregulatory axis, we investigated: 1) the DAPC composition in the hypothalamic supraoptic nucleus (SON) and paraventricular nucleus (PVN) of Dp71-null mice; and 2) the expression and activity of neuronal nitric oxide synthase (nNOS), because it is a potential partner of the DAPC and a functional index of osmoregulatory axis activity. In wild-type mice, dystrophins and their autosomal homologs the utrophins, β-DG, and α1-Syn were localized in astrocyte end feet. In Dp71-null mice, the levels of β-DG and α1-Syn were lower and utrophin expression did not change. The location of the DAPC in astrocytic end feet suggests that it could be involved in hypothalamic osmosensitivity, which adapts the osmotic response. The altered composition of the DAPC in Dp71-null mice could thus explain why these mice manifest an hypo-osmolar status. In the SON and PVN neurons of Dp71-null mice, nNOS expression and activity were increased. Although we previously established that DP140 is expressed de novo in these neurons, the DAPC remained incomplete due to the low levels of β-DG and α1-Syn produced in these cells. Our data reveal the importance of DP71 for the constitution of a functional DAPC in the hypothalamus. Such DAPC disorganization may lead to modification of the microenvironment of the SON and PVN neurons and thus may result in a perturbed osmoregulation.
Collapse
Affiliation(s)
- Roza Benabdesselam
- Unité de Recherches, Faculté des Sciences Biologiques/UMMTO, BP 17, Tizi-Ouzou, Algeria
| | | | | | | | | | | |
Collapse
|
44
|
Horie N, Pereira MP, Niizuma K, Sun G, Keren-Gill H, Encarnacion A, Shamloo M, Hamilton SA, Jiang K, Huhn S, Palmer TD, Bliss TM, Steinberg GK. Transplanted stem cell-secreted vascular endothelial growth factor effects poststroke recovery, inflammation, and vascular repair. Stem Cells 2011; 29:274-85. [PMID: 21732485 DOI: 10.1002/stem.584] [Citation(s) in RCA: 196] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cell transplantation offers a novel therapeutic strategy for stroke; however, how transplanted cells function in vivo is poorly understood. We show for the first time that after subacute transplantation into the ischemic brain of human central nervous system stem cells grown as neurospheres (hCNS-SCns), the stem cell-secreted factor, human vascular endothelial growth factor (hVEGF), is necessary for cell-induced functional recovery. We correlate this functional recovery to hVEGF-induced effects on the host brain including multiple facets of vascular repair and its unexpected suppression of the inflammatory response. We found that transplanted hCNS-SCns affected multiple parameters in the brain with different kinetics: early improvement in blood-brain barrier integrity and suppression of inflammation was followed by a delayed spatiotemporal regulated increase in neovascularization. These events coincided with a bimodal pattern of functional recovery, with, an early recovery independent of neovascularization, and a delayed hVEGF-dependent recovery coincident with neovascularization. Therefore, cell transplantation therapy offers an exciting multimodal strategy for brain repair in stroke and potentially other disorders with a vascular or inflammatory component.
Collapse
Affiliation(s)
- Nobutaka Horie
- Department of Neurosurgery and Stanford Stroke Center, Stanford Institute for Neuro-Innovation and Translational Neurosciences, Stanford University School of Medicine, Stanford, California 94305-5487, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Dendritic spines are dynamic structures that accommodate the majority of excitatory synapses in the brain and are influenced by extracellular signals from presynaptic neurons, glial cells, and the extracellular matrix (ECM). The ECM surrounds dendritic spines and extends into the synaptic cleft, maintaining synapse integrity as well as mediating trans-synaptic communications between neurons. Several scaffolding proteins and glycans that compose the ECM form a lattice-like network, which serves as an attractive ground for various secreted glycoproteins, lectins, growth factors, and enzymes. ECM components can control dendritic spines through the interactions with their specific receptors or by influencing the functions of other synaptic proteins. In this review, we focus on ECM components and their receptors that regulate dendritic spine development and plasticity in the normal and diseased brain.
Collapse
Affiliation(s)
- Lorraine E. Dansie
- Division of Biomedical Sciences, Biochemistry and Molecular Biology Program, University of California Riverside, Riverside, California 92521
| | - Iryna M. Ethell
- Division of Biomedical Sciences, Biochemistry and Molecular Biology Program, University of California Riverside, Riverside, California 92521
| |
Collapse
|
46
|
Pawlisz AS, Feng Y. Three-dimensional regulation of radial glial functions by Lis1-Nde1 and dystrophin glycoprotein complexes. PLoS Biol 2011; 9:e1001172. [PMID: 22028625 PMCID: PMC3196477 DOI: 10.1371/journal.pbio.1001172] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 08/30/2011] [Indexed: 01/09/2023] Open
Abstract
Lis1-Nde1 integrates cerebral cortical neurogenesis with neuronal migration by stabilizing the basal-lateral surface of radial glial cells. Radial glial cells (RGCs) are distinctive neural stem cells with an extraordinary slender bipolar morphology and dual functions as precursors and migration scaffolds for cortical neurons. Here we show a novel mechanism by which the Lis1-Nde1 complex maintains RGC functions through stabilizing the dystrophin/dystroglycan glycoprotein complex (DGC). A direct interaction between Nde1 and utrophin/dystrophin allows for the assembly of a multi-protein complex that links the cytoskeleton to the extracellular matrix of RGCs to stabilize their lateral membrane, cell-cell adhesion, and radial morphology. Lis1-Nde1 mutations destabilized the DGC and resulted in deformed, disjointed RGCs and disrupted basal lamina. Besides impaired RGC self-renewal and neuronal migration arrests, Lis1-Nde1 deficiencies also led to neuronal over-migration. Additional to phenotypic resemblances of Lis1-Nde1 with DGC, strong synergistic interactions were found between Nde1 and dystroglycan in RGCs. As functional insufficiencies of LIS1, NDE1, and dystroglycan all cause lissencephaly syndromes, our data demonstrated that a three-dimensional regulation of RGC's cytoarchitecture by the Lis1-Nde1-DGC complex determines the number and spatial organization of cortical neurons as well as the size and shape of the cerebral cortex. The processes of neurogenesis and neuronal migration within the developing cerebral cortex must be tightly orchestrated to enable ordered generation and transportation of neurons to designated cortical layers. The mechanism by which these two processes are integrated remains elusive. Radial glial cells, the major neural stem cells in the developing brain, serve both as progenitors and migration scaffolds for cortical neurons as they migrate. The cortical developmental disease lissencephaly (smooth brain) is a result of defects in neurogenesis and neuronal migration, and is associated with the protein LIS1 and its binding partner NDE1. In this study, we show that several key players in human cerebral cortical development, including LIS1, NDE1, dystrophin, and dystroglycan, form a molecular complex to regulate cortical neurogenesis and neuronal migration in a mouse model. This multi-protein complex is active on the basal-lateral surface of radial glial cells, which is known to provide guidance to migrating neurons. When we depleted NDE1 in mice, dystrophin and dystroglycan were lost from the membrane and radial glial cells were deformed, indicating the importance of the multi-protein complex for proper cell morphology. This effect on morphology resulted in a loss of normal migration and cortical phenotypes similar to lissencephaly. Our findings suggest that genes that regulate the structure and function of the basal-lateral membrane of radial glial cells may integrate the dual functions of these cells and determine the size, shape, and function of the cerebral cortex.
Collapse
Affiliation(s)
- Ashley S. Pawlisz
- Department of Neurology and Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Yuanyi Feng
- Department of Neurology and Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
47
|
Zhang P, Hu H. Differential glycosylation of α-dystroglycan and proteins other than α-dystroglycan by like-glycosyltransferase. Glycobiology 2011; 22:235-47. [PMID: 21930648 DOI: 10.1093/glycob/cwr131] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Genetic defects in like-glycosyltransferase (LARGE) cause congenital muscular dystrophy with central nervous system manifestations. The underlying molecular pathomechanism is the hypoglycosylation of α-dystroglycan (α-DG), which is evidenced by diminished immunoreactivity to IIH6C4 and VIA4-1, antibodies that recognize carbohydrate epitopes. Previous studies indicate that LARGE participates in the formation of a phosphoryl glycan branch on O-linked mannose or it modifies complex N- and mucin O-glycans. In this study, we overexpressed LARGE in neural stem cells deficient in protein O-mannosyltransferase 2 (POMT2), an enzyme required for O-mannosyl glycosylation. The results showed that overexpressing LARGE did not lead to hyperglycosylation of α-DG in POMT2 knockout (KO) cells but did generate IIH6C4 and VIA4-1 immunoreactivity and laminin-binding activity. Additionally, overexpressing LARGE in cells deficient in both POMT2 and α-DG generated laminin-binding IIH6C4 immunoreactivity. These results indicate that LARGE expression resulted in the glycosylation of proteins other than α-DG in the absence of O-mannosyl glycosylation. The IIH6C4 immunoreactivity generated in double-KO cells was largely removed by treatment either with peptide N-glycosidase F or with cold aqueous hydrofluoric acid, suggesting that LARGE expression caused phosphoryl glycosylation of N-glycans. However, the glycosylation of α-DG by LARGE is dependent on POMT2, indicating that LARGE expression only modifies O-linked mannosyl glycans of α-DG. Thus, LARGE expression mediates the phosphoryl glycosylation of not only O-mannosyl glycans including those on α-DG but also N-glycans on proteins other than α-DG.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, 750 E. Adams Street, Syracuse, NY 13210, USA
| | | |
Collapse
|
48
|
Parberry-Clark C, Bury JP, Cross SS, Winder SJ. Loss of dystroglycan function in oesophageal cancer. Histopathology 2011; 59:180-7. [DOI: 10.1111/j.1365-2559.2011.03930.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
49
|
Hesse C, Johansson I, Mattsson N, Bremell D, Andreasson U, Halim A, Anckarsäter R, Blennow K, Anckarsäter H, Zetterberg H, Larson G, Hagberg L, Grahn A. The N-terminal domain of α-dystroglycan, released as a 38 kDa protein, is increased in cerebrospinal fluid in patients with Lyme neuroborreliosis. Biochem Biophys Res Commun 2011; 412:494-9. [PMID: 21843510 DOI: 10.1016/j.bbrc.2011.07.129] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 07/30/2011] [Indexed: 10/17/2022]
Abstract
α-Dystroglycan is an extracellular adhesion protein that is known to interact with different ligands. The interaction is thought to stabilize the integrity of the plasma membrane. The N-terminal part of α-dystroglycan may be proteolytically processed to generate a small 38 kDa protein (α-DG-N). The physiological significance of α-DG-N is unclear but has been suggested to be involved in nerve regeneration and myelination and to function as a potential biomarker for neurodegenerative and neuromuscular diseases. In this report we show that α-DG-N is released into different body fluids, such as lachrimal fluid, cerebrospinal fluid (CSF), urine and plasma. To investigate the significance of α-DG-N in CSF we examined the levels of α-DG-N and known neurodegenerative markers in CSF from patients diagnosed with Lyme neuroborreliosis (LNB) and healthy controls. In untreated acute phase LNB patients, 67% showed a significant increase of CSF α-DG-N compared to healthy controls. After treatment with antibiotics the CSF α-DG-N levels were normalized in the LNB patients.
Collapse
Affiliation(s)
- Camilla Hesse
- Institute of Biomedicine, Department of Clinical Chemistry and Transfusion Medicine, The Sahlgrenska Academy, University of Gothenburg, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hormonal therapy in the senescence: Prostatic microenvironment structure and adhesion molecules. Micron 2011; 42:642-55. [DOI: 10.1016/j.micron.2011.03.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 03/08/2011] [Indexed: 02/07/2023]
|