1
|
Napolitano F, Fabozzi M, Montuori N. Non-Integrin Laminin Receptors: Shedding New Light and Clarity on Their Involvement in Human Diseases. Int J Mol Sci 2025; 26:3546. [PMID: 40332051 PMCID: PMC12026610 DOI: 10.3390/ijms26083546] [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: 03/14/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
The extracellular matrix (ECM) is a dynamic network of macromolecules that provides structural support for cells and orchestrates cell signaling, functions, and morphology. The basement membrane constitutes a peculiar sheet-like type of ECM located between epithelial tissues and underlying connective tissues. The major constituent of the basement membrane is laminin, which exerts a remarkable repertoire of biological functions such as cell differentiation, migration, adhesion, and wound healing. Laminin performs its functions by interacting with two main classes of receptors, the integrin and the non-integrin laminin receptors, creating a complex network essential for tissue integrity and regeneration. Dysfunctional actions of laminin are the cause of diverse human diseases, including cancer, infectious, and neurodegenerative diseases. This topic has attracted researchers for some time, but the diversity of cell-surface receptors, through which laminin signaling occurs, makes the role of laminin controversial. Moreover, different laminin isoforms were identified, and each specific tissue basement membrane differs from the others in their laminin composition. This review focuses on the structural and molecular basis and pathophysiological relevance of specific interactions between laminins and non-integrin receptors in development, health, and disease.
Collapse
Affiliation(s)
- Filomena Napolitano
- Department of Translational Medical Sciences, University of Naples Federico II, 80135 Naples, Italy; (F.N.); (M.F.)
| | - Maria Fabozzi
- Department of Translational Medical Sciences, University of Naples Federico II, 80135 Naples, Italy; (F.N.); (M.F.)
| | - Nunzia Montuori
- Department of Translational Medical Sciences, University of Naples Federico II, 80135 Naples, Italy; (F.N.); (M.F.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80135 Naples, Italy
| |
Collapse
|
2
|
Pigors M, Goletz S, Wang Y, Emtenani S, Hammers CM, Holtsche MM, Patzelt S, Opelka B, Stang FH, König IR, Radzimski C, Komorowski L, Aumailley M, Has C, Schmidt E. Anti-Laminin β4 IgG Drives Tissue Damage in Anti-p200 Pemphigoid and Shows Interactions with Laminin α3 and γ1/2 Chains. J Invest Dermatol 2025; 145:821-830.e3. [PMID: 39320300 DOI: 10.1016/j.jid.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 09/26/2024]
Abstract
Laminin β4 was recently identified as a structural component of the dermal-epidermal junction and autoantigen of anti-p200 pemphigoid. In this study, we provided further evidence of the pathogenic effect of anti-laminin β4 IgG and identified potential binding partners of laminin β4. We showed that laminin β4 immune complexes led to activation of normal leukocytes and dose-dependent ROS release. Using cryosections of normal skin, we demonstrated that anti-laminin β4 patient serum IgG but not anti-laminin γ1 IgG, which are also detectable in patients with anti-p200 pemphigoid, cause dermal-epidermal separation in the presence of leukocytes. Proximity ligation assay and indirect immunofluorescence staining suggested that laminin β4 localizes closely to laminin α3 and γ2 in primary keratinocytes. Subsequent coimmunoprecipitation experiments using epidermal extracts confirmed the interaction of laminin β4 with the α3 and γ2 chains and indicated additional affinity to laminin γ1. The laminin β4-α3/β4-γ1 protein complexes were also detected using mass spectrometry. In conclusion, this study showed that anti-laminin β4 IgG can exert tissue damage in the skin, supporting their pathogenic role in anti-p200 pemphigoid. Our data further provide strong evidence for an interaction of laminin β4 with laminin α3, whereas its association to the laminin γ1 and γ2 chains is ambiguous.
Collapse
Affiliation(s)
- Manuela Pigors
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany.
| | - Stephanie Goletz
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Yao Wang
- Department of Dermatology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Shirin Emtenani
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Christoph M Hammers
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany; Department of Dermatology, Allergology and Venerology, University of Lübeck, Lübeck, Germany
| | - Maike M Holtsche
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany; Department of Dermatology, Allergology and Venerology, University of Lübeck, Lübeck, Germany
| | - Sabrina Patzelt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Bianca Opelka
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Felix H Stang
- Hand Surgery and Burn Care Unit, Department of Plastic Surgery, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Inke R König
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | | | - Lars Komorowski
- Institute of Experimental Immunology, EUROIMMUN AG, Lübeck, Germany
| | - Monique Aumailley
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Cristina Has
- Department of Dermatology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany; Department of Dermatology, Allergology and Venerology, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
3
|
Abstract
The extracellular matrix (ECM) is the complex meshwork of proteins and glycans that forms the scaffold that surrounds and supports cells. It exerts key roles in all aspects of metazoan physiology, from conferring physical and mechanical properties on tissues and organs to modulating cellular processes such as proliferation, differentiation and migration. Understanding the mechanisms that orchestrate the assembly of the ECM scaffold is thus crucial to understand ECM functions in health and disease. This Review discusses novel insights into the compositional diversity of matrisome components and the mechanisms that lead to tissue-specific assemblies and architectures tailored to support specific functions. The Review then highlights recently discovered mechanisms, including post-translational modifications and metabolic pathways such as amino acid availability and the circadian clock, that modulate ECM secretion, assembly and remodelling in homeostasis and human diseases. Last, the Review explores the potential of 'matritherapies', that is, strategies to normalize ECM composition and architecture to achieve a therapeutic benefit.
Collapse
Affiliation(s)
- Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, USA.
- University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
4
|
Nonnast E, Mira E, Mañes S. Biomechanical properties of laminins and their impact on cancer progression. Biochim Biophys Acta Rev Cancer 2024; 1879:189181. [PMID: 39299492 DOI: 10.1016/j.bbcan.2024.189181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/30/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Laminins (LMs) constitute a family of heterotrimeric glycoproteins essential for the formation of basement membranes (BM). They act as molecular bridges between cells and the extracellular matrix (ECM), thereby transmitting signals influencing cell behavior and tissue organization. In the realm of cancer pathobiology, LMs regulate key processes such as migration, differentiation, or fibrosis. This review critically examines the multifaceted impact of LMs on tumor progression, with a particular focus on the isoform-specific structure-function relationships, and how this structural diversity contributes to the biomechanical properties of BMs. LM interactions with integrin and non-integrin cell surface receptors, as well as with other ECM proteins, modify the response of cancer cells to the ECM stiffness, ultimately influencing the capacity of malignant cells to breach the BM, a limiting step in metastatic dissemination. Comprehension of the mechanisms underlying LM-driven tumor biomechanics holds potential for better understand cancer pathobiology and design new targeted therapeutic strategies.
Collapse
Affiliation(s)
- Elena Nonnast
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), 28049 Madrid, Spain
| | - Emilia Mira
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), 28049 Madrid, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), 28049 Madrid, Spain.
| |
Collapse
|
5
|
Ramdas S, Beeson D, Dong YY. Congenital myasthenic syndromes: increasingly complex. Curr Opin Neurol 2024; 37:493-501. [PMID: 39051439 PMCID: PMC11377046 DOI: 10.1097/wco.0000000000001300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
PURPOSE OF REVIEW Congenital myasthenia syndromes (CMS) are treatable, inherited disorders affecting neuromuscular transmission. We highlight that the involvement of an increasing number of proteins is making the understanding of the disease mechanisms and potential treatments progressively more complex. RECENT FINDINGS Although early studies identified mutations of proteins directly involved in synaptic transmission at the neuromuscular junction, recently, next-generation sequencing has facilitated the identification of many novel mutations in genes that encode proteins that have a far wider expression profile, some even ubiquitously expressed, but whose defective function leads to impaired neuromuscular transmission. Unsurprisingly, mutations in these genes often causes a wider phenotypic disease spectrum where defective neuromuscular transmission forms only one component. This has implications for the management of CMS patients. SUMMARY Given the widening nonneuromuscular junction phenotypes in the newly identified forms of CMS, new therapies need to include disease-modifying approaches that address not only neuromuscular weakness but also the multisystem involvement. Whilst the current treatments for CMS are highly effective for many subtypes there remains, in a proportion of CMS patients, an unmet need for more efficacious therapies.
Collapse
Affiliation(s)
- Sithara Ramdas
- MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford
- Department of Paediatric Neurology, John Radcliffe Hospital
| | - David Beeson
- Neurosciences Group, Weatherall Institute of Molecular Medicine, The John Radcliffe, Oxford OX3 9DS
| | - Yin Yao Dong
- Neurosciences Group, Weatherall Institute of Molecular Medicine, The John Radcliffe, Oxford OX3 9DS
| |
Collapse
|
6
|
Bannykh KS, Fuentes-Fayos AC, Linesch PW, Breunig JJ, Bannykh SI. Laminin Beta 2 Is Localized at the Sites of Blood-Brain Barrier and Its Disruption Is Associated With Increased Vascular Permeability, Histochemical, and Transcriptomic Study. J Histochem Cytochem 2024; 72:641-667. [PMID: 39340425 PMCID: PMC11472343 DOI: 10.1369/00221554241281896] [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: 06/27/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
Heterotrimeric extracellular matrix proteins laminins are mostly deposited at basal membranes and are important in repair and neoplasia. Here, we localize laminin beta 2 (LAMB2) at the sites of blood-brain barrier (BBB). Microvasculature (MV) of normal brain is endowed with complete LAMB2 coverage. In contrast, its cognate protein laminin beta 1 (LAMB1) is absent in MV of normal brain but emerges at the sprouting tip of a growing vessels. Similarly, vascular proliferation in high-grade gliomas (HGG) is accompanied by marked overexpression of LAMB1, whereas LAMB2 shows deficient deposition. We find that many brain pathologies with presence of post-gadolinium enhancement (PGE) on magnetic resonance imaging (MRI) show disruption of LAMB2 vascular ensheathment. Inhibition of vascular endothelial growth factor signaling in HGG blocks angiogenesis, suppresses PGE in HGG, prevents expression of LAMB1, and restores LAMB2 vascular coverage. Analysis of single-cell RNA sequencing (scRNA-seq) databases shows that in quiescent brain LAMB2 is predominantly expressed by BBB-associated pericytes (PCs) and endothelial cells (ECs), whereas neither cell types produce LAMB1. In contrast, in HGG, both LAMB1 and 2 are overexpressed by endothelial precursor cells, a phenotypically unique immature group, specific to proliferating hyperplastic MV.
Collapse
Affiliation(s)
- Katherine S. Bannykh
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Antonio C. Fuentes-Fayos
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Paul W. Linesch
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Joshua J. Breunig
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Serguei I. Bannykh
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
7
|
Finn LS. Nephrotic Syndrome Throughout Childhood: Diagnosing Podocytopathies From the Womb to the Dorm. Pediatr Dev Pathol 2024; 27:426-458. [PMID: 38745407 DOI: 10.1177/10935266241242669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The etiologies of podocyte dysfunction that lead to pediatric nephrotic syndrome (NS) are vast and vary with age at presentation. The discovery of numerous novel genetic podocytopathies and the evolution of diagnostic technologies has transformed the investigation of steroid-resistant NS while simultaneously promoting the replacement of traditional morphology-based disease classifications with a mechanistic approach. Podocytopathies associated with primary and secondary steroid-resistant NS manifest as diffuse mesangial sclerosis, minimal change disease, focal segmental glomerulosclerosis, and collapsing glomerulopathy. Molecular testing, once an ancillary option, has become a vital component of the clinical investigation and when paired with kidney biopsy findings, provides data that can optimize treatment and prognosis. This review focuses on the causes including selected monogenic defects, clinical phenotypes, histopathologic findings, and age-appropriate differential diagnoses of nephrotic syndrome in the pediatric population with an emphasis on podocytopathies.
Collapse
Affiliation(s)
- Laura S Finn
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at The University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
8
|
Yurchenco PD, Kulczyk AW. Polymerizing laminins in development, health, and disease. J Biol Chem 2024; 300:107429. [PMID: 38825010 PMCID: PMC11260871 DOI: 10.1016/j.jbc.2024.107429] [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: 01/11/2024] [Revised: 05/12/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Polymerizing laminins are multi-domain basement membrane (BM) glycoproteins that self-assemble into cell-anchored planar lattices to establish the initial BM scaffold. Nidogens, collagen-IV and proteoglycans then bind to the scaffold at different domain loci to create a mature BM. The LN domains of adjacent laminins bind to each other to form a polymer node, while the LG domains attach to cytoskeletal-anchoring integrins and dystroglycan, as well as to sulfatides and heparan sulfates. The polymer node, the repeating unit of the polymer scaffold, is organized into a near-symmetrical triskelion. The structure, recently solved by cryo-electron microscopy in combination with AlphaFold2 modeling and biochemical studies, reveals how the LN surface residues interact with each other and how mutations cause failures of self-assembly in an emerging group of diseases, the LN-lamininopathies, that include LAMA2-related dystrophy and Pierson syndrome.
Collapse
Affiliation(s)
- Peter D Yurchenco
- Department of Pathology & Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.
| | - Arkadiusz W Kulczyk
- Department of Biochemistry and Microbiology, Institute for Quantitative Biomedicine, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
9
|
Romashin D, Arzumanian V, Poverennaya E, Varshaver A, Luzgina N, Rusanov A. Evaluation of Cd-induced cytotoxicity in primary human keratinocytes. Hum Exp Toxicol 2024; 43:9603271231224458. [PMID: 38174414 DOI: 10.1177/09603271231224458] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
An increasing number of studies have investigated the effects of Cd on human health. Cd-induced dermatotoxicity is an important field of research, but numerous studies have focused on the effects of Cd on the human skin. Moreover, most studies have been performed using HaCaT cells but not primary keratinocytes. In this study, we provide the results describing the cytotoxic effects of Cd exposure on primary human epidermal keratinocytes obtained from different donors. The subtoxic concentration of cadmium chloride was determined via MTT assay, and transcriptomic analysis of the cells exposed to this concentration (25 µM) was performed. As in HaCaT cells, Cd exposure resulted in increased ROS levels, cell cycle arrest, and induction of apoptosis. In addition, we report that exposure to Cd affects zinc and copper homeostasis, induces metallothionein expression, and activates various signaling pathways, including Nrf2, NF-kB, TRAIL, and PI3K. Cd induces the secretion of various cytokines (IL-1, IL-6, IL-10, and PGE2) and upregulates the expression of several cytokeratins, such as KRT6B, KRT6C, KRT16, and KRT17. The results provide a better understanding of the mechanisms of cadmium-induced cytotoxicity and its effect on human epidermal skin cells.
Collapse
|
10
|
Kulczyk AW. Artificial intelligence and the analysis of cryo-EM data provide structural insight into the molecular mechanisms underlying LN-lamininopathies. Sci Rep 2023; 13:17825. [PMID: 37857770 PMCID: PMC10587063 DOI: 10.1038/s41598-023-45200-5] [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: 07/31/2023] [Accepted: 10/17/2023] [Indexed: 10/21/2023] Open
Abstract
Laminins (Lm) are major components of basement membranes (BM), which polymerize to form a planar lattice on cell surface. Genetic alternations of Lm affect their oligomerization patterns and lead to failures in BM assembly manifesting in a group of human disorders collectively defined as Lm N-terminal domain lamininopathies (LN-lamininopathies). We have employed a recently determined cryo-EM structure of the Lm polymer node, the basic repeating unit of the Lm lattice, along with structure prediction and modeling to systematically analyze structures of twenty-three pathogenic Lm polymer nodes implicated in human disease. Our analysis provides the detailed mechanistic explanation how Lm mutations lead to failures in Lm polymerization underlining LN-lamininopathies. We propose the new categorization scheme of LN-lamininopathies based on the insight gained from the structural analysis. Our results can help to facilitate rational drug design aiming in the treatment of Lm deficiencies.
Collapse
Affiliation(s)
- Arkadiusz W Kulczyk
- Institute for Quantitative Biomedicine, Rutgers University, 174 Frelinghuysen Road, Piscataway, NJ, 08854, USA.
- Department of Biochemistry & Microbiology, Rutgers University, 75 Lipman Drive, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
11
|
Balakrishnan B, Altassan R, Budhraja R, Liou W, Lupo A, Bryant S, Mankouski A, Radenkovic S, Preston GJ, Pandey A, Boudina S, Kozicz T, Morava E, Lai K. AAV-based gene therapy prevents and halts the progression of dilated cardiomyopathy in a mouse model of phosphoglucomutase 1 deficiency (PGM1-CDG). Transl Res 2023; 257:1-14. [PMID: 36709920 PMCID: PMC10192047 DOI: 10.1016/j.trsl.2023.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/04/2023] [Accepted: 01/18/2023] [Indexed: 01/27/2023]
Abstract
Phosphoglucomutase 1 (PGM1) deficiency is recognized as the third most common N-linked congenital disorders of glycosylation (CDG) in humans. Affected individuals present with liver, musculoskeletal, endocrine, and coagulation symptoms; however, the most life-threatening complication is the early onset of dilated cardiomyopathy (DCM). Recently, we discovered that oral D-galactose supplementation improved liver disease, endocrine, and coagulation abnormalities, but does not alleviate the fatal cardiomyopathy and the associated myopathy. Here we report on left ventricular ejection fraction (LVEF) in 6 individuals with PGM1-CDG. LVEF was pathologically low in most of these individuals and varied between 10% and 65%. To study the pathobiology of the cardiac disease observed in PGM1-CDG, we constructed a novel cardiomyocyte-specific conditional Pgm2 gene (mouse ortholog of human PGM1) knockout (Pgm2 cKO) mouse model. Echocardiography studies corroborated a DCM phenotype with significantly reduced ejection fraction and left ventricular dilation similar to those seen in individuals with PGM1-CDG. Histological studies demonstrated excess glycogen accumulation and fibrosis, while ultrastructural analysis revealed Z-disk disarray and swollen/fragmented mitochondria, which was similar to the ultrastructural pathology in the cardiac explant of an individual with PGM1-CDG. In addition, we found decreased mitochondrial function in the heart of KO mice. Transcriptomic analysis of hearts from mutant mice demonstrated a gene signature of DCM. Although proteomics revealed only mild changes in global protein expression in left ventricular tissue of mutant mice, a glycoproteomic analysis unveiled broad glycosylation changes with significant alterations in sarcolemmal proteins including different subunits of laminin-211, which was confirmed by immunoblot analyses. Finally, augmentation of PGM1 in KO mice via AAV9-PGM1 gene replacement therapy prevented and halted the progression of the DCM phenotype.
Collapse
Affiliation(s)
- Bijina Balakrishnan
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Ruqaiah Altassan
- Department of Medical Genomics, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Rohit Budhraja
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Willisa Liou
- Electron Microscopy Core Facility, University of Utah, Salt Lake City, Utah, USA
| | - Arielle Lupo
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Sarah Bryant
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Anastasiya Mankouski
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Silvia Radenkovic
- Department of Clinical Genomics, Center of Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Graeme J Preston
- Department of Clinical Genomics, Center of Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA; Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Sihem Boudina
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah, USA
| | - Tamas Kozicz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA; Department of Clinical Genomics, Center of Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Anatomy, University of Pecs School of Medicine, Pecs, Hungary
| | - Eva Morava
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA; Department of Clinical Genomics, Center of Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Medical Genetics, University of Pecs, School of Medicine, Pecs, Hungary
| | - Kent Lai
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA; Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
12
|
Cryo-EM reveals the molecular basis oflaminin polymerization and LN-lamininopathies. Nat Commun 2023; 14:317. [PMID: 36658135 PMCID: PMC9852560 DOI: 10.1038/s41467-023-36077-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
Laminin polymerization is the major step in basement membranes assembly. Its failures cause laminin N-terminal domain lamininopathies including Pierson syndrome. We have employed cryo-electron microscopy to determine a 3.7 Å structure of the trimeric laminin polymer node containing α1, β1 and γ1 subunits. The structure reveals the molecular basis of calcium-dependent formation of laminin lattice, and provides insights into polymerization defects manifesting in human disease.
Collapse
|
13
|
Rousselle P, Laigle C, Rousselet G. The basement membrane in epidermal polarity, stemness, and regeneration. Am J Physiol Cell Physiol 2022; 323:C1807-C1822. [PMID: 36374168 DOI: 10.1152/ajpcell.00069.2022] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The epidermis is a specialized epithelium that constitutes the outermost layer of the skin, and it provides a protective barrier against environmental assaults. Primarily consisting of multilayered keratinocytes, the epidermis is continuously renewed by proliferation of stem cells and the differentiation of their progeny, which undergo terminal differentiation as they leave the basal layer and move upward toward the surface, where they die and slough off. Basal keratinocytes rest on a basement membrane at the dermal-epidermal junction that is composed of specific extracellular matrix proteins organized into interactive and mechanically supportive networks. Firm attachment of basal keratinocytes, and their dynamic regulation via focal adhesions and hemidesmosomes, is essential for maintaining major skin processes, such as self-renewal, barrier function, and resistance to physical and chemical stresses. The adhesive integrin receptors expressed by epidermal cells serve structural, signaling, and mechanosensory roles that are critical for epidermal cell anchorage and tissue homeostasis. More specifically, the basement membrane components play key roles in preserving the stem cell pool, and establishing cell polarity cues enabling asymmetric cell divisions, which result in the transition from a proliferative basal cell layer to suprabasal cells committed to terminal differentiation. Finally, through a well-regulated sequence of synthesis and remodeling, the components of the dermal-epidermal junction play an essential role in regeneration of the epidermis during skin healing. Here too, they provide biological and mechanical signals that are essential to the restoration of barrier function.
Collapse
Affiliation(s)
- Patricia Rousselle
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR 5305, CNRS, Université Lyon 1, Lyon, France
| | - Chloé Laigle
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR 5305, CNRS, Université Lyon 1, Lyon, France
| | - Gaelle Rousselet
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR 5305, CNRS, Université Lyon 1, Lyon, France
| |
Collapse
|
14
|
Kubanov AA, Chikin VV, Karamova AE, Monchakovskaya ES. Junctional epidermolysis bullosa: genotype-phenotype correlations. VESTNIK DERMATOLOGII I VENEROLOGII 2022. [DOI: 10.25208/vdv1391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Junctional epidermolysis bullosa most commonly results from mutations in theLAMA3, LAMB3, LAMC2, COL17A1, ITGA6 and ITGB4genes. Junctional epidermolysis bullosa is characterized by clinical heterogeneity. To date, scientific findings allow to evaluate correlations between the severity of clinical manifestations and genetic defects underlying in the development of the disease. A systematic literature search was performed using PubMed and RSCI, and keywords including junctional epidermolysis bullosa, laminin 332, collagen XVII, 64 integrin. The review includes description of clinical findings of junctional epidermolysis bullosa, mutation location and types, its impact on protein production and functions. To evaluate the impact of gene mutation on protein functions, this review explores the structure and functions of lamina lucida components, including laminin 332, collagen XVII and 64 integrin, which are frequently associated with the development of junctional epidermolysis bullosa. The correlation between severe types of junctional epidermolysis bullosa and mutations resulting in premature stop codon generation and complete absence of protein expression has been described. Although, genotype-phenotype correlations should be analyzed carefully due to mechanisms which enable to improve protein expression.
Collapse
|
15
|
Meyer S, Kaulfuß S, Zechel S, Kummer K, Seif Amir Hosseini A, Ernst MS, Schmidt J, Pauli S, Zschüntzsch J. Evidence of Two Novel LAMA2 Variants in a Patient With Muscular Dystrophy: Facing the Challenges of a Certain Diagnosis. Front Neurol 2022; 13:893605. [PMID: 35928135 PMCID: PMC9344914 DOI: 10.3389/fneur.2022.893605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundBenefits and challenges resulting from advances in genetic diagnostics are two sides of the same coin. Facilitation of a correct and timely diagnosis is paralleled by challenges in interpretation of variants of unknown significance (VUS). Focusing on an individual VUS-re-classification pipeline, this study offers a diagnostic approach for clinically suspected hereditary muscular dystrophy by combining the expertise of an interdisciplinary team.MethodsIn a multi-step approach, a thorough phenotype assessment including clinical examination, laboratory work, muscle MRI and histopathological evaluation of muscle was performed in combination with advanced Next Generation Sequencing (NGS). Different in-silico tools and prediction programs like Alamut, SIFT, Polyphen, MutationTaster and M-Cap as well as 3D- modeling of protein structure and RNA-sequencing were employed to determine clinical significance of the LAMA2 variants.ResultsTwo previously unknown sequence alterations in LAMA2 were detected, a missense variant was classified initially according to ACMG guidelines as a VUS (class 3) whereas a second splice site variant was deemed as likely pathogenic (class 4). Pathogenicity of the splice site variant was confirmed by mRNA sequencing and nonsense mediated decay (NMD) was detected. Combination of the detected variants could be associated to the LGMDR23-phenotype based on the MRI matching and literature research.DiscussionTwo novel variants in LAMA2 associated with LGMDR23-phenotype are described. This study illustrates challenges of the genetic findings due to their VUS classification and elucidates how individualized diagnostic procedure has contributed to the accurate diagnosis in the spectrum of LGMD.
Collapse
Affiliation(s)
- Stefanie Meyer
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Silke Kaulfuß
- Department of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Sabrina Zechel
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Karsten Kummer
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Ali Seif Amir Hosseini
- Department of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Marielle Sophie Ernst
- Department of Neuroradiology, University Medical Center Göttingen, Göttingen, Germany
| | - Jens Schmidt
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Department of Neurology and Pain Treatment, Immanuel Klinik Rüdersdorf, University Hospital of the Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany
| | - Silke Pauli
- Department of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Jana Zschüntzsch
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- *Correspondence: Jana Zschüntzsch
| |
Collapse
|
16
|
Sugden CJ, Iorio V, Troughton LD, Liu K, Morais MRPT, Lennon R, Bou-Gharios G, Hamill KJ. Laminin N-terminus α31 expression during development is lethal and causes widespread tissue-specific defects in a transgenic mouse model. FASEB J 2022; 36:e22318. [PMID: 35648586 PMCID: PMC9328196 DOI: 10.1096/fj.202002588rrr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/28/2022] [Accepted: 04/05/2022] [Indexed: 11/11/2022]
Abstract
Laminins (LMs) are essential components of all basement membranes where they regulate an extensive array of tissue functions. Alternative splicing from the laminin α3 gene produces a non‐laminin but netrin‐like protein, Laminin N terminus α31 (LaNt α31). LaNt α31 is widely expressed in intact tissue and is upregulated in epithelial cancers and during wound healing. In vitro functional studies have shown that LaNt α31 can influence numerous aspects of epithelial cell behavior via modifying matrix organization, suggesting a new model of laminin auto‐regulation. However, the function of this protein has not been established in vivo. Here, a mouse transgenic line was generated using the ubiquitin C promoter to drive inducible expression of LaNt α31. When expression was induced at embryonic day 15.5, LaNt α31 transgenic animals were not viable at birth, exhibiting localized regions of erythema. Histologically, the most striking defect was widespread evidence of extravascular bleeding across multiple tissues. Additionally, LaNt α31 transgene expressing animals exhibited kidney epithelial detachment, tubular dilation, disruption of the epidermal basal cell layer and of the hair follicle outer root sheath, and ~50% reduction of cell numbers in the liver, associated with depletion of hematopoietic erythrocytic foci. These findings provide the first in vivo evidence that LaNt α31 can influence tissue morphogenesis.
Collapse
Affiliation(s)
- Conor J Sugden
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Valentina Iorio
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Lee D Troughton
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| | - Ke Liu
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Mychel R P T Morais
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, The University of Manchester, Manchester, UK
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, The University of Manchester, Manchester, UK
| | - George Bou-Gharios
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Kevin J Hamill
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|