1
|
Srinivasan S, Ramos-Lewis W, Morais MRPT, Chi Q, Soh AWJ, Williams E, Lennon R, Sherwood DR. A collagen IV fluorophore knock-in toolkit reveals trimer diversity in C. elegans basement membranes. J Cell Biol 2025; 224:e202412118. [PMID: 40100062 PMCID: PMC11917169 DOI: 10.1083/jcb.202412118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025] Open
Abstract
The type IV collagen triple helix, composed of three ⍺-chains, is a core basement membrane (BM) component that assembles into a network within BMs. Endogenous tagging of all ⍺-chains with genetically encoded fluorophores has remained elusive, limiting our understanding of this crucial BM component. Through genome editing, we show that the C termini of the C. elegans type IV collagen ⍺-chains EMB-9 and LET-2 can be fused to a variety of fluorophores to create a strain toolkit with wild-type health. Using quantitative imaging, our results suggest a preference for LET-2-LET-2-EMB-9 trimer construction, but also tissue-specific flexibility in trimers assembled driven by differences in ⍺-chain expression levels. By tagging emb-9 and let-2 mutants that model human Gould syndrome, a complex multitissue disorder, we further discover defects in extracellular accumulation and turnover that might help explain disease pathology. Together, our findings identify a permissive tagging site in C. elegans that will allow diverse studies on type IV collagen regulation and function in animals.
Collapse
Affiliation(s)
| | | | - Mychel R P T Morais
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Qiuyi Chi
- Department of Biology, Duke University, Durham, NC, USA
| | - Adam W J Soh
- Department of Biology, Duke University, Durham, NC, USA
| | - Emily Williams
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Rachel Lennon
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | | |
Collapse
|
2
|
Malcor JD, Ferruz N, Romero-Romero S, Dhingra S, Sagar V, Jalan AA. Deciphering the folding code of collagens. Nat Commun 2025; 16:2702. [PMID: 40108160 PMCID: PMC11923368 DOI: 10.1038/s41467-024-54046-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 10/30/2024] [Indexed: 03/22/2025] Open
Abstract
Collagen proteins contain a characteristic structural motif called a triple helix. During the self-assembly of this motif, three polypeptides form a folding nucleus at the C-termini and then propagate towards the N-termini like a zip-chain. While polypeptides from human collagens contain up to a 1000 amino acids, those found in bacteria can contain up to 6000 amino acids. Additionally, the collagen polypeptides are also frequently interrupted by non-helical sequences that disrupt folding and reduce stability. Given the length of polypeptides and the disruptive interruptions, compensating mechanisms that stabilize against local unfolding during propagation and offset the entropic cost of folding are not fully understood. Here, we show that the information for the correct folding of collagen triple helices is encoded in their sequence as interchain electrostatic interactions, which likely act as molecular clamps that prevent local unfolding. In the case of humans, disrupting these electrostatic interactions is associated with severe to lethal diseases.
Collapse
Affiliation(s)
- Jean-Daniel Malcor
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305 University of Lyon, Lyon, France
| | - Noelia Ferruz
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
- Centre for Genomic Regulation, Barcelona, Spain
| | - Sergio Romero-Romero
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
- Department of Biochemistry and Structural Biology. Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Surbhi Dhingra
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
| | - Vamika Sagar
- Department of Biomaterials, University of Bayreuth, Bayreuth, Germany
| | - Abhishek A Jalan
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany.
- Department of Biomaterials, University of Bayreuth, Bayreuth, Germany.
| |
Collapse
|
3
|
Caparali EB, De Gregorio V, Barua M. Genotype-Based Molecular Mechanisms in Alport Syndrome. J Am Soc Nephrol 2025:00001751-990000000-00551. [PMID: 39899372 DOI: 10.1681/asn.0000000647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/29/2025] [Indexed: 02/05/2025] Open
Abstract
Alport syndrome is an inherited disorder characterized by kidney disease, sensorineural hearing loss, and ocular abnormalities. Alport syndrome is caused by pathogenic variants in COL4A3 , COL4A4 , or COL4A5 , which encode the α 3, α 4, and α 5 chains of type 4 collagen that forms a heterotrimer expressed in the glomerular basement membrane. Knowledge of its genetic basis has informed the development of different models in dogs, mice, and rats that reflect its autosomal and X-linked inheritance patterns as well as different mutation types, including protein-truncating and missense variants. A key difference between these two types is the synthesis of α 3 α 4 α 5(IV), which is not made in autosomal Alport syndrome (two pathogenic variants in trans or biallelic) or male patients with X-linked Alport syndrome due to protein-truncating variants. By contrast, α 3 α 4 α 5(IV) is synthesized in Alport syndrome because of missense variants. For missense variants, in vitro studies suggest that these cause impaired type 4 collagen trafficking and endoplasmic reticulum stress. For protein-truncating variants, knockout models suggest that persistence of an immature α 1 α 1 α 2(IV) network is associated with biomechanical strain, which activates endothelin-A receptors leading to mesangial filopodia formation. Moreover, studies suggest that activation of collagen receptors, integrins and discoidin domain receptor 1, play a role in disease propagation. In this review, we provide an overview of how these genotype-phenotype mechanisms are key for a precision medicine-based approach in the future.
Collapse
Affiliation(s)
- Emine Bilge Caparali
- Department of Internal Medicine, University of Texas Southwestern, Dallas, Texas
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Division of Nephrology, University Health Network, Toronto, Ontario, Canada
| | | | - Moumita Barua
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Division of Nephrology, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Uemura M, Tanaka N, Ando S, Yanagihara T, Onodera O. Missense Variants in COL4A1/2 Are Associated with Cerebral Aneurysms: A Case Report and Literature Review. Neurol Int 2024; 16:226-238. [PMID: 38392956 PMCID: PMC10892350 DOI: 10.3390/neurolint16010015] [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: 12/09/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Although cerebral aneurysm (CA) is a defining complication of COL4A1/2-related vasculopathy, the specific factors influencing its onset remain uncertain. This study aimed to identify and analyze these factors. METHODS We described a family presenting with a novel variant of the COL4A1 gene complicated with CA. Concurrently, an exhaustive review of previously documented patients with COL4A1/2-related vasculopathy was conducted by sourcing data from PubMed, Web of Science, Google Scholar, and Ichushi databases. We compared the variant types and locations between patients with CA (positive group) and those without CA (negative group). RESULTS This study included 53 COL4A1/2 variants from 76 patients. Except for one start codon variant, all the identified variants in CA were missense variants. Otherwise, CA was not associated with other clinical manifestations, such as small-vessel disease or other large-vessel abnormalities. A higher frequency of missense variants (95.5% vs. 58.1%, p = 0.0035) was identified in the CA-positive group. CONCLUSIONS CA development appears to necessitate qualitative alterations in COL4A1/2, and the underlying mechanism seems independent of small-vessel disease or other large-vessel anomalies. Our findings suggest that a meticulous evaluation of CA is necessary when missense variants in COL4A1/2 are identified.
Collapse
Affiliation(s)
- Masahiro Uemura
- Department of Neurology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan (O.O.)
| | - Natsuki Tanaka
- Department of Neurology, Tane General Hospital, Osaka 550-0025, Japan
| | - Shoichiro Ando
- Department of Neurology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan (O.O.)
| | | | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan (O.O.)
| |
Collapse
|
5
|
Summers JA, Yarbrough M, Liu M, McDonald WH, Hudson BG, Pastor-Pareja JC, Boudko SP. Collagen IV of basement membranes: IV. Adaptive mechanism of collagen IV scaffold assembly in Drosophila. J Biol Chem 2023; 299:105394. [PMID: 37890775 PMCID: PMC10694668 DOI: 10.1016/j.jbc.2023.105394] [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: 09/10/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Collagen IV is an essential structural protein in all metazoans. It provides a scaffold for the assembly of basement membranes, a specialized form of extracellular matrix, which anchors and signals cells and provides microscale tensile strength. Defective scaffolds cause basement membrane destabilization and tissue dysfunction. Scaffolds are composed of α-chains that coassemble into triple-helical protomers of distinct chain compositions, which in turn oligomerize into supramolecular scaffolds. Chloride ions mediate the oligomerization via NC1 trimeric domains, forming an NC1 hexamer at the protomer-protomer interface. The chloride concentration-"chloride pressure"-on the outside of cells is a primordial innovation that drives the assembly and dynamic stabilization of collagen IV scaffolds. However, a Cl-independent mechanism is operative in Ctenophora, Ecdysozoa, and Rotifera, which suggests evolutionary adaptations to environmental or tissue conditions. An understanding of these exceptions, such as the example of Drosophila, could shed light on the fundamentals of how NC1 trimers direct the oligomerization of protomers into scaffolds. Here, we investigated the NC1 assembly of Drosophila. We solved the crystal structure of the NC1 hexamer, determined the chain composition of protomers, and found that Drosophila adapted an evolutionarily unique mechanism of scaffold assembly that requires divalent cations. By studying the Drosophila case we highlighted the mechanistic role of chloride pressure for maintaining functionality of the NC1 domain in humans. Moreover, we discovered that the NC1 trimers encode information for homing protomers to distant tissue locations, providing clues for the development of protein replacement therapy for collagen IV genetic diseases.
Collapse
Affiliation(s)
- Jacob A Summers
- Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Madison Yarbrough
- Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Min Liu
- School of Life Sciences, Tsinghua University, Beijing, China
| | - W Hayes McDonald
- Proteomics Laboratory, Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Billy G Hudson
- Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - José C Pastor-Pareja
- School of Life Sciences, Tsinghua University, Beijing, China; Tsinghua-Peking Center for Life Sciences, Beijing, China; Institute of Neurosciences, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Sergei P Boudko
- Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
6
|
Guimaraes TACD, Arram E, Shakarchi AF, Georgiou M, Michaelides M. Inherited causes of combined vision and hearing loss: clinical features and molecular genetics. Br J Ophthalmol 2023; 107:1403-1414. [PMID: 36162969 DOI: 10.1136/bjo-2022-321790] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/07/2022] [Indexed: 11/04/2022]
Abstract
Combined vision and hearing loss, also known as dual sensory impairment, can occur in several genetic conditions, including ciliopathies such as Usher and Bardet-Biedl syndrome, mitochondrial DNA disorders and systemic diseases, such as CHARGE, Stickler, Waardenburg, Alport and Alstrom syndrome. The retinal phenotype may point to the diagnosis of such disorders. Herein, we aim to provide a comprehensive review of the molecular genetics and clinical features of the most common non-chromosomal inherited disorders to cause dual sensory impairment.
Collapse
Affiliation(s)
| | - Elizabeth Arram
- Moorfields Eye Hospital NHS Foundation Trust, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Ahmed F Shakarchi
- Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Michalis Georgiou
- Moorfields Eye Hospital NHS Foundation Trust, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
- Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Michel Michaelides
- Moorfields Eye Hospital NHS Foundation Trust, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
7
|
Eckersley A, Morais MR, Ozols M, Lennon R. Peptide location fingerprinting identifies structural alterations within basement membrane components in ageing kidney. Matrix Biol 2023; 121:167-178. [PMID: 37437747 DOI: 10.1016/j.matbio.2023.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/04/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
During ageing, the glomerular and tubular basement membranes (BM) of the kidney undergo a progressive decline in function that is underpinned by histological changes, including glomerulosclerosis and tubular interstitial fibrosis and atrophy. This BM-specific ageing is thought to result from damage accumulation to long-lived extracellular matrix (ECM) protein structures. Determining which BM proteins are susceptible to these structure-associated changes, and the possible mechanisms and downstream consequences, is critical to understand age-related kidney degeneration and to identify markers for therapeutic intervention. Peptide location fingerprinting (PLF) is an emerging proteomic mass spectrometry analysis technique capable of identifying ECM proteins with structure-associated differences that may occur by damage modifications in ageing. Here, we apply PLF as a bioinformatic screening tool to identify BM proteins with structure-associated differences between young and aged human glomerular and tubulointerstitial compartments. Several functional regions within key BM components displayed alterations in tryptic peptide yield, reflecting potential age-dependent shifts in molecular (e.g. laminin-binding regions in agrin) and cellular (e.g. integrin-binding regions in laminins 521 and 511) interactions, oxidation (e.g. collagen IV) and the fragmentation and release of matrikines (e.g. canstatin and endostatin from collagens IV and XVIII). Furthermore, we found that periostin and the collagen IV α2 chain exhibited structure-associated differences in ageing that were conserved between human kidney and previously analysed mouse lung, revealing BM components that harbour shared susceptibilities across species and organs.
Collapse
Affiliation(s)
- Alexander Eckersley
- Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| | - Mychel Rpt Morais
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Matiss Ozols
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Department of Human Genetics, Wellcome Sanger Institute, Genome Campus, Hinxton, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
| | - Rachel Lennon
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
8
|
Boudko SP, Konopka EH, Kim W, Taga Y, Mizuno K, Springer TA, Hudson BG, Moy TI, Lin FY. A recombinant technique for mapping functional sites of heterotrimeric collagen helices: Collagen IV CB3 fragment as a prototype for integrin binding. J Biol Chem 2023; 299:104901. [PMID: 37302550 PMCID: PMC10404678 DOI: 10.1016/j.jbc.2023.104901] [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: 05/08/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023] Open
Abstract
Collagen superfamily of proteins is a major component of the extracellular matrix. Defects in collagens underlie the cause of nearly 40 human genetic diseases in millions of people worldwide. Pathogenesis typically involves genetic alterations of the triple helix, a hallmark structural feature that bestows exceptional mechanical resistance to tensile forces and a capacity to bind a plethora of macromolecules. Yet, there is a paramount knowledge gap in understanding the functionality of distinct sites along the triple helix. Here, we present a recombinant technique to produce triple helical fragments for functional studies. The experimental strategy utilizes the unique capacity of the NC2 heterotrimerization domain of collagen IX to drive three α-chain selection and registering the triple helix stagger. For proof of principle, we produced and characterized long triple helical fragments of collagen IV that were expressed in a mammalian system. The heterotrimeric fragments encompassed the CB3 trimeric peptide of collagen IV, which harbors the binding motifs for α1β1 and α2β1 integrins. Fragments were characterized and shown to have a stable triple helix, post-translational modifications, and high affinity and specific binding of integrins. The NC2 technique is a universal tool for the high-yield production of heterotrimeric fragments of collagens. Fragments are suitable for mapping functional sites, determining coding sequences of binding sites, elucidating pathogenicity and pathogenic mechanisms of genetic mutations, and production of fragments for protein replacement therapy.
Collapse
Affiliation(s)
- Sergei P Boudko
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA.
| | | | - Woojin Kim
- Morphic Therapeutic, Inc, Waltham, Massachusetts, USA
| | - Yuki Taga
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Timothy A Springer
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Billy G Hudson
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Terence I Moy
- Morphic Therapeutic, Inc, Waltham, Massachusetts, USA
| | - Fu-Yang Lin
- Morphic Therapeutic, Inc, Waltham, Massachusetts, USA.
| |
Collapse
|
9
|
Ma P, Wang S, Geng R, Gong Y, Li M, Xie D, Dong Y, Zheng T, Li B, Zhao T, Zheng Q. MiR-29a-deficiency causes thickening of the basilar membrane and age-related hearing loss by upregulating collagen IV and laminin. Front Cell Neurosci 2023; 17:1191740. [PMID: 37275774 PMCID: PMC10232818 DOI: 10.3389/fncel.2023.1191740] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/03/2023] [Indexed: 06/07/2023] Open
Abstract
Age-related hearing loss (ARHL) is the most common sensory degenerative disease and can significantly impact the quality of life in elderly people. A previous study using GeneChip miRNA microarray assays showed that the expression of miR-29a changes with age, however, its role in hearing loss is still unclear. In this study, we characterized the cochlear phenotype of miR-29a knockout (miR-29a-/-) mice and found that miR-29a-deficient mice had a rapid progressive elevation of the hearing threshold from 2 to 5 months of age compared with littermate controls as measured by the auditory brainstem response. Stereocilia degeneration, hair cell loss and abnormal stria vascularis (SV) were observed in miR-29a-/- mice at 4 months of age. Transcriptome sequencing results showed elevated extracellular matrix (ECM) gene expression in miR-29a-/- mice. Both Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that the key differences were closely related to ECM. Further examination with a transmission electron microscope showed thickening of the basilar membrane in the cochlea of miR-29a-/- mice. Five Col4a genes (Col4a1-a5) and two laminin genes (Lamb2 and Lamc1) were validated as miR-29a direct targets by dual luciferase assays and miR-29a inhibition assays with a miR-29a inhibitor. Consistent with the target gene validation results, the expression of these genes was significantly increased in the cochlea of miR-29a-/- mice, as shown by RT-PCR and Western blot. These findings suggest that miR-29a plays an important role in maintaining cochlear structure and function by regulating the expression of collagen and laminin and that the disturbance of its expression could be a cause of progressive hearing loss.
Collapse
Affiliation(s)
- Peng Ma
- School of Basic Medicine, Qingdao University, Qingdao, China
- School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Shuli Wang
- Department of Hearing and Speech Rehabilitation, School of Special Education, Binzhou Medical University, Yantai, China
| | - Ruishuang Geng
- Department of Hearing and Speech Rehabilitation, School of Special Education, Binzhou Medical University, Yantai, China
| | - Yongfeng Gong
- School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Mulan Li
- Department of Hearing and Speech Rehabilitation, School of Special Education, Binzhou Medical University, Yantai, China
| | - Daoli Xie
- Department of Hearing and Speech Rehabilitation, School of Special Education, Binzhou Medical University, Yantai, China
| | - Yaning Dong
- Department of Hearing and Speech Rehabilitation, School of Special Education, Binzhou Medical University, Yantai, China
| | - Tihua Zheng
- Department of Hearing and Speech Rehabilitation, School of Special Education, Binzhou Medical University, Yantai, China
| | - Bo Li
- Department of Hearing and Speech Rehabilitation, School of Special Education, Binzhou Medical University, Yantai, China
| | - Tong Zhao
- Department of Hearing and Speech Rehabilitation, School of Special Education, Binzhou Medical University, Yantai, China
| | - Qingyin Zheng
- School of Basic Medicine, Qingdao University, Qingdao, China
- Department of Otolaryngology, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
10
|
Adeva-Andany MM, Carneiro-Freire N. Biochemical composition of the glomerular extracellular matrix in patients with diabetic kidney disease. World J Diabetes 2022; 13:498-520. [PMID: 36051430 PMCID: PMC9329837 DOI: 10.4239/wjd.v13.i7.498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/19/2022] [Accepted: 06/26/2022] [Indexed: 02/06/2023] Open
Abstract
In the glomeruli, mesangial cells produce mesangial matrix while podocytes wrap glomerular capillaries with cellular extensions named foot processes and tether the glomerular basement membrane (GBM). The turnover of the mature GBM and the ability of adult podocytes to repair injured GBM are unclear. The actin cytoskeleton is a major cytoplasmic component of podocyte foot processes and links the cell to the GBM. Predominant components of the normal glomerular extracellular matrix (ECM) include glycosaminoglycans, proteoglycans, laminins, fibronectin-1, and several types of collagen. In patients with diabetes, multiorgan composition of extracellular tissues is anomalous, including the kidney, so that the constitution and arrangement of glomerular ECM is profoundly altered. In patients with diabetic kidney disease (DKD), the global quantity of glomerular ECM is increased. The level of sulfated proteoglycans is reduced while hyaluronic acid is augmented, compared to control subjects. The concentration of mesangial fibronectin-1 varies depending on the stage of DKD. Mesangial type III collagen is abundant in patients with DKD, unlike normal kidneys. The amount of type V and type VI collagens is higher in DKD and increases with the progression of the disease. The GBM contains lower amount of type IV collagen in DKD compared to normal tissue. Further, genetic variants in the α3 chain of type IV collagen may modulate susceptibility to DKD and end-stage kidney disease. Human cellular models of glomerular cells, analyses of human glomerular proteome, and improved microscopy procedures have been developed to investigate the molecular composition and organization of the human glomerular ECM.
Collapse
|
11
|
Gibson JT, Sadeghi-Alavijeh O, Gale DP, Rothe H, Savige J. Pathogenicity of missense variants affecting the collagen IV α5 carboxy non-collagenous domain in X-linked Alport syndrome. Sci Rep 2022; 12:11257. [PMID: 35789182 PMCID: PMC9253329 DOI: 10.1038/s41598-022-14928-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/15/2022] [Indexed: 12/05/2022] Open
Abstract
X-linked Alport syndrome is a genetic kidney disease caused by pathogenic COL4A5 variants, but little is known of the consequences of missense variants affecting the NC1 domain of the corresponding collagen IV α5 chain. This study examined these variants in a normal (gnomAD) and other databases (LOVD, Clin Var and 100,000 Genomes Project) to determine their pathogenicity and clinical significance. Males with Cys substitutions in the collagen IV α5 NC1 domain reported in LOVD (n = 25) were examined for typical Alport features, including age at kidney failure. All NC1 variants in LOVD (n = 86) were then assessed for structural damage using an online computational tool, Missense3D. Variants in the ClinVar, gnomAD and 100,000 Genomes Project databases were also examined for structural effects. Predicted damage associated with NC1 substitutions was then correlated with the level of conservation of the affected residues. Cys substitutions in males were associated with the typical features of X-linked Alport syndrome, with a median age at kidney failure of 31 years. NC1 substitutions predicted to cause structural damage were overrepresented in LOVD (p < 0.001), and those affecting Cys residues or 'buried' Gly residues were more common than expected (both p < 0.001). Most NC1 substitutions in gnomAD (88%) were predicted to be structurally-neutral. Substitutions affecting conserved residues resulted in more structural damage than those affecting non-conserved residues (p < 0.001). Many pathogenic missense variants affecting the collagen IV α5 NC1 domain have their effect through molecular structural damage and 3D modelling is a useful tool in their assessment.
Collapse
Affiliation(s)
- Joel T Gibson
- Department of Medicine (Melbourne Health and Northern Health), The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Omid Sadeghi-Alavijeh
- Department of Renal Medicine, University College London, London, UK
- Genomics England, Queen Mary University of London, London, UK
| | - Daniel P Gale
- Department of Renal Medicine, University College London, London, UK
- Genomics England, Queen Mary University of London, London, UK
| | - Hansjörg Rothe
- Centre for Nephrology and Metabolic Disorders, 02943, Weisswasser, Germany
| | - Judy Savige
- Department of Medicine (Melbourne Health and Northern Health), The University of Melbourne, Parkville, VIC, 3050, Australia.
- Genomics England, Queen Mary University of London, London, UK.
| |
Collapse
|
12
|
Xu F, Zheng Z, Yao M, Zhu F, Shen T, Li J, Zhu C, Yang T, Shao M, Wan Z, Fang C. A regulatory mechanism of a stepwise osteogenesis-mimicking decellularized extracellular matrix on the osteogenic differentiation of bone marrow-derived mesenchymal stem cells. J Mater Chem B 2022; 10:6171-6180. [PMID: 35766339 DOI: 10.1039/d2tb00721e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A cell-derived decellularized extracellular matrix (dECM) plays a vital role in controlling cell functions because of its similarity to the in vivo microenvironment. In the process of stem cell differentiation, the composition of the dECM is not constant but is dynamically remolded. However, there is little information regarding the dynamic regulation by the dECM of the osteogenic differentiation of stem cells. Herein, four types of stepwise dECMs (0, 7, 14, and 21 d-ECM) were prepared from bone marrow-derived mesenchymal stem cells (BMSCs) undergoing osteogenic differentiation for 0, 7, 14, and 21 days after decellularization. In vitro experiments were designed to study the regulation of BMSC osteogenesis by dECMs. The results showed that all the dECMs could support the activity and proliferation of BMSCs but had different effects on their osteogenic differentiation. The 14d-ECM promoted the osteogenesis of BMSCs significantly compared with the other dECMs. Proteomic analysis demonstrated that the composition of dECMs changed over time. The 14d ECM had higher amounts of collagen type IV alpha 2 chain (COL4A2) than the other dECMs. Furthermore, COL4A2 was obviously enriched in the activated focal adhesion kinase (FAK)/phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT) signaling pathways. Thus, the 14d-ECM could promote the osteogenic differentiation of BMSCs, which might be related to the high content of COL4A2 in the 14d-ECM by activating the FAK/PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Fei Xu
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Ziran Zheng
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Mianfeng Yao
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Feiya Zhu
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
| | - Ting Shen
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
| | - Jiang Li
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Chao Zhu
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Tianru Yang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
| | - Mengying Shao
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
| | - Zicheng Wan
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Changyun Fang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| |
Collapse
|
13
|
Mao M, Labelle-Dumais C, Tufa SF, Keene DR, Gould DB. Elevated TGFβ signaling contributes to ocular anterior segment dysgenesis in Col4a1 mutant mice. Matrix Biol 2022; 110:151-173. [PMID: 35525525 PMCID: PMC10410753 DOI: 10.1016/j.matbio.2022.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/08/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
Ocular anterior segment dysgenesis (ASD) refers to a collection of developmental disorders affecting the anterior structures of the eye. Although a number of genes have been implicated in the etiology of ASD, the underlying pathogenetic mechanisms remain unclear. Mutations in genes encoding collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) cause Gould syndrome, a multi-system disorder that often includes ocular manifestations such as ASD and glaucoma. COL4A1 and COL4A2 are abundant basement membrane proteins that provide structural support to tissues and modulate signaling through interactions with other extracellular matrix proteins, growth factors, and cell surface receptors. In this study, we used a combination of histological, molecular, genetic and pharmacological approaches to demonstrate that altered TGFβ signaling contributes to ASD in mouse models of Gould syndrome. We show that TGFβ signaling was elevated in anterior segments from Col4a1 mutant mice and that genetically reducing TGFβ signaling partially prevented ASD. Notably, we identified distinct roles for TGFβ1 and TGFβ2 in ocular defects observed in Col4a1 mutant mice. Importantly, we show that pharmacologically promoting type IV collagen secretion or reducing TGFβ signaling ameliorated ocular pathology in Col4a1 mutant mice. Overall, our findings demonstrate that altered TGFβ signaling contributes to COL4A1-related ocular dysgenesis and implicate this pathway as a potential therapeutic target for the treatment of Gould syndrome.
Collapse
Affiliation(s)
- Mao Mao
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, United States
| | - Cassandre Labelle-Dumais
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, United States
| | - Sara F Tufa
- Shriners Children's, Micro-Imaging Center, Portland, Oregon 97239, United States
| | - Douglas R Keene
- Shriners Children's, Micro-Imaging Center, Portland, Oregon 97239, United States
| | - Douglas B Gould
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, United States; Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, United States; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, United States; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, United States; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94143, United States.
| |
Collapse
|
14
|
Analysis of missense variants in the human genome reveals widespread gene-specific clustering and improves prediction of pathogenicity. Am J Hum Genet 2022; 109:457-470. [PMID: 35120630 PMCID: PMC8948164 DOI: 10.1016/j.ajhg.2022.01.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
We used a machine learning approach to analyze the within-gene distribution of missense variants observed in hereditary conditions and cancer. When applied to 840 genes from the ClinVar database, this approach detected a significant non-random distribution of pathogenic and benign variants in 387 (46%) and 172 (20%) genes, respectively, revealing that variant clustering is widespread across the human exome. This clustering likely occurs as a consequence of mechanisms shaping pathogenicity at the protein level, as illustrated by the overlap of some clusters with known functional domains. We then took advantage of these findings to develop a pathogenicity predictor, MutScore, that integrates qualitative features of DNA substitutions with the new additional information derived from this positional clustering. Using a random forest approach, MutScore was able to identify pathogenic missense mutations with very high accuracy, outperforming existing predictive tools, especially for variants associated with autosomal-dominant disease and cancer. Thus, the within-gene clustering of pathogenic and benign DNA changes is an important and previously underappreciated feature of the human exome, which can be harnessed to improve the prediction of pathogenicity and disambiguation of DNA variants of uncertain significance.
Collapse
|
15
|
Gibson JT, Huang M, Shenelli Croos Dabrera M, Shukla K, Rothe H, Hilbert P, Deltas C, Storey H, Lipska-Ziętkiewicz BS, Chan MMY, Sadeghi-Alavijeh O, Gale DP, Cerkauskaite A, Savige J. Genotype-phenotype correlations for COL4A3-COL4A5 variants resulting in Gly substitutions in Alport syndrome. Sci Rep 2022; 12:2722. [PMID: 35177655 PMCID: PMC8854626 DOI: 10.1038/s41598-022-06525-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/24/2022] [Indexed: 12/21/2022] Open
Abstract
Alport syndrome is the commonest inherited kidney disease and nearly half the pathogenic variants in the COL4A3-COL4A5 genes that cause Alport syndrome result in Gly substitutions. This study examined the molecular characteristics of Gly substitutions that determine the severity of clinical features. Pathogenic COL4A5 variants affecting Gly in the Leiden Open Variation Database in males with X-linked Alport syndrome were correlated with age at kidney failure (n = 157) and hearing loss diagnosis (n = 80). Heterozygous pathogenic COL4A3 and COL4A4 variants affecting Gly (n = 304) in autosomal dominant Alport syndrome were correlated with the risk of haematuria in the UK 100,000 Genomes Project. Gly substitutions were stratified by exon location (1 to 20 or 21 to carboxyl terminus), being adjacent to a non-collagenous region (interruption or terminus), and the degree of instability caused by the replacement residue. Pathogenic COL4A5 variants that resulted in a Gly substitution with a highly destabilising residue reduced the median age at kidney failure by 7 years (p = 0.002), and age at hearing loss diagnosis by 21 years (p = 0.004). Substitutions adjacent to a non-collagenous region delayed kidney failure by 19 years (p = 0.014). Heterozygous pathogenic COL4A3 and COL4A4 variants that resulted in a Gly substitution with a highly destabilising residue (Arg, Val, Glu, Asp, Trp) were associated with an increased risk of haematuria (p = 0.018), and those adjacent to a non-collagenous region were associated with a reduced risk (p = 0.046). Exon location had no effect. In addition, COL4A5 variants adjacent to non-collagenous regions were over-represented in the normal population in gnomAD (p < 0.001). The nature of the substitution and of nearby residues determine the risk of haematuria, early onset kidney failure and hearing loss for Gly substitutions in X-linked and autosomal dominant Alport syndrome.
Collapse
Affiliation(s)
- Joel T Gibson
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Mary Huang
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Marina Shenelli Croos Dabrera
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Krushnam Shukla
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Hansjörg Rothe
- Centre for Nephrology and Metabolic Disorders, 02943, Weisswasser, Germany
| | - Pascale Hilbert
- Departement de Biologie Moleculaire, Institute de Pathologie et de Genetique ASBL, Gosselies, Belgium
| | - Constantinos Deltas
- Center of Excellence in Biobanking and Biomedical Research, University of Cyprus Medical School, Nicosia, Cyprus
| | - Helen Storey
- Molecular Genetics, Viapath Laboratories, 5th Floor Tower Wing, Guy's Hospital, London, SE1 9RT, UK
| | | | - Melanie M Y Chan
- Department of Renal Medicine, University College London, London, UK
| | | | - Daniel P Gale
- Department of Renal Medicine, University College London, London, UK
| | - Agne Cerkauskaite
- Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Judy Savige
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia.
| |
Collapse
|
16
|
Dekkers BG, Saad SI, van Spelde LJ, Burgess JK. Basement membranes in obstructive pulmonary diseases. Matrix Biol Plus 2021; 12:100092. [PMID: 34877523 PMCID: PMC8632995 DOI: 10.1016/j.mbplus.2021.100092] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 11/04/2021] [Accepted: 11/07/2021] [Indexed: 12/24/2022] Open
Abstract
Basement membrane composition is changed in the airways of patients with obstructive airway diseases. Basement membrane changes are linked to disease characteristics in patients. Mechanisms behind the altered BM composition remain to be elucidated. Laminin and collagen IV affect key pathological processes in obstructive airway diseases.
Increased and changed deposition of extracellular matrix proteins is a key feature of airway wall remodeling in obstructive pulmonary diseases, including asthma and chronic obstructive pulmonary disease. Studies have highlighted that the deposition of various basement membrane proteins in the lung tissue is altered and that these changes reflect tissue compartment specificity. Inflammatory responses in both diseases may result in the deregulation of production and degradation of these proteins. In addition to their role in tissue development and integrity, emerging evidence indicates that basement membrane proteins also actively modulate cellular processes in obstructive airway diseases, contributing to disease development, progression and maintenance. In this review, we summarize the changes in basement membrane composition in airway remodeling in obstructive airway diseases and explore their potential application as innovative targets for treatment development.
Collapse
Key Words
- ADAM9, a metalloproteinase domain 9
- ASM, airway smooth muscle
- Airway inflammation
- Airway remodeling
- Asthma
- BM, basement membrane
- COPD, chronic obstructive pulmonary disease
- Chronic obstructive pulmonary disease
- Col IV, collagen IV
- Collagen IV
- ECM, extracellular matrix
- LN, laminin
- Laminin
- MMP, matrix metalloproteinase
- TIMP, tissue inhibitors of metalloproteinase
- Th2, T helper 2
- VSM, vascular smooth muscle
Collapse
Affiliation(s)
- Bart G.J. Dekkers
- University of Groningen, University Medical Center Groningen, Department of Clinical Pharmacy and Pharmacology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Corresponding author at: Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Shehab I. Saad
- University of Groningen, University Medical Centre Groningen, Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research, Groningen, The Netherlands
| | - Leah J. van Spelde
- University of Groningen, University Medical Centre Groningen, Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research, Groningen, The Netherlands
| | - Janette K. Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- University of Groningen, University Medical Centre Groningen, Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research, Groningen, The Netherlands
| |
Collapse
|
17
|
Ambade AS, Hassoun PM, Damico RL. Basement Membrane Extracellular Matrix Proteins in Pulmonary Vascular and Right Ventricular Remodeling in Pulmonary Hypertension. Am J Respir Cell Mol Biol 2021; 65:245-258. [PMID: 34129804 PMCID: PMC8485997 DOI: 10.1165/rcmb.2021-0091tr] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
The extracellular matrix (ECM), a highly organized network of structural and nonstructural proteins, plays a pivotal role in cellular and tissue homeostasis. Changes in the ECM are critical for normal tissue repair, whereas dysregulation contributes to aberrant tissue remodeling. Pulmonary arterial hypertension is a severe disorder of the pulmonary vasculature characterized by pathologic remodeling of the pulmonary vasculature and right ventricle, increased production and deposition of structural and nonstructural proteins, and altered expression of ECM growth factors and proteases. Furthermore, ECM remodeling plays a significant role in disease progression, as several dynamic changes in its composition, quantity, and organization are documented in both humans and animal models of disease. These ECM changes impact vascular cell biology and affect proliferation of resident cells. Furthermore, ECM components determine the tissue architecture of the pulmonary and myocardial vasculature as well as the myocardium itself and provide mechanical stability crucial for tissue homeostasis. However, little is known about the basement membrane (BM), a specialized, self-assembled conglomerate of ECM proteins, during remodeling. In the vasculature, the BM is in close physical association with the vascular endothelium and smooth muscle cells. While in the myocardium, each cardiomyocyte is enclosed by a BM that serves as the interface between cardiomyocytes and the surrounding interstitial matrix. In this review, we provide a brief overview on the current state of knowledge of the BM and its ECM composition and their impact on pulmonary vascular remodeling and right ventricle dysfunction and failure in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Anjira S Ambade
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Rachel L Damico
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
18
|
Cunha AM, Teixeira D, Cabral D, Estrela-Silva S, Falcão-Reis F, Carneiro Â. Quantitative Optical Coherence Tomography Angiography Biomarkers for Alport Syndrome. Ophthalmologica 2021; 245:41-48. [PMID: 34469897 DOI: 10.1159/000519242] [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: 03/28/2021] [Accepted: 08/12/2021] [Indexed: 11/19/2022]
Abstract
PURPOSE The aim of this study was to evaluate microvascular abnormalities of patients with Alport syndrome (AS) using optical coherence tomography angiography (OCT-A) quantitative biomarkers. METHODS This was cross sectional, prospective evaluation of consecutive patients with AS and healthy subjects. AS diagnosis was performed by the genetic test. All participants underwent a retinal vasculature evaluation by spectral-domain optical coherence tomography (SD-OCT) and OCT-A of the macula. Quantitative analysis included whole vascular density, foveal avascular zone area, fractal dimension (FD), and lacunarity (LAC). RESULTS Ninety-four eyes were included in this study, 45 eyes from patients with AS and 49 eyes from healthy subjects. The pathogenic mutation in the COL4A5 gene on the chromosome X was found in 14 patients; the pathogenic autosomal recessive mutations in the COL4A3 gene were found in 9 patients. Quantitative evaluation demonstrated a significant difference between AS and healthy subjects on LAC of the superficial capillary plexus and deep capillary plexus (DCP) (p < 0.001 and p < 0.001, respectively) and on FD in the DCP (p < 0.001). CONCLUSION The DCP Alport patients have a higher vessel nonuniformity than DCP of healthy subjects. We hypothesize that endothelial cell lesion in the setting of low resistance at the DCP circuit could lead to long-term structural disorganization.
Collapse
Affiliation(s)
- Ana Maria Cunha
- Department of Ophthalmology, Centro Hospitalar Universitário de São João, Porto, Portugal
| | | | - Diogo Cabral
- Instituto de Oftalmologia Dr. Gama Pinto, Lisboa, Portugal
| | - Sérgio Estrela-Silva
- Department of Ophthalmology, Centro Hospitalar Universitário de São João, Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine of University of Porto, Porto, Portugal
| | - Fernando Falcão-Reis
- Department of Ophthalmology, Centro Hospitalar Universitário de São João, Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine of University of Porto, Porto, Portugal
| | - Ângela Carneiro
- Department of Ophthalmology, Centro Hospitalar Universitário de São João, Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine of University of Porto, Porto, Portugal
| |
Collapse
|
19
|
Al-Shaer A, Lyons A, Ishikawa Y, Hudson BG, Boudko SP, Forde NR. Sequence-dependent mechanics of collagen reflect its structural and functional organization. Biophys J 2021; 120:4013-4028. [PMID: 34390685 DOI: 10.1016/j.bpj.2021.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/25/2021] [Accepted: 08/06/2021] [Indexed: 01/06/2023] Open
Abstract
Extracellular matrix mechanics influence diverse cellular functions, yet surprisingly little is known about the mechanical properties of their constituent collagen proteins. In particular, network-forming collagen IV, an integral component of basement membranes, has been far less studied than fibril-forming collagens. A key feature of collagen IV is the presence of interruptions in the triple-helix-defining (Gly-X-Y) sequence along its collagenous domain. Here, we used atomic force microscopy to determine the impact of sequence heterogeneity on the local flexibility of collagen IV and of the fibril-forming collagen III. Our extracted flexibility profile of collagen IV reveals that it possesses highly heterogeneous mechanics, ranging from semiflexible regions as found for fibril-forming collagens to a lengthy region of high flexibility toward its N-terminus. A simple model in which flexibility is dictated only by the presence of interruptions fit the extracted profile reasonably well, providing insight into the alignment of chains and demonstrating that interruptions, particularly when coinciding in multiple chains, significantly enhance local flexibility. To a lesser extent, sequence variations within the triple helix lead to variable flexibility, as seen along the continuously triple-helical collagen III. We found this fibril-forming collagen to possess a high-flexibility region around its matrix-metalloprotease binding site, suggesting a unique mechanical fingerprint of this region that is key for matrix remodeling. Surprisingly, proline content did not correlate with local flexibility in either collagen type. We also found that physiologically relevant changes in pH and chloride concentration did not alter the flexibility of collagen IV, indicating such environmental changes are unlikely to control its compaction during secretion. Although extracellular chloride ions play a role in triggering collagen IV network formation, they do not appear to modulate the structure of its collagenous domain.
Collapse
Affiliation(s)
- Alaa Al-Shaer
- Department of Molecular Biology and Biochemistry, Burnaby, British Columbia, Canada
| | - Aaron Lyons
- Department of Physics, Burnaby, British Columbia, Canada
| | - Yoshihiro Ishikawa
- Department of Ophthalmology, University of California San Francisco, School of Medicine, San Francisco, California
| | - Billy G Hudson
- Department of Medicine, Division of Nephrology and Hypertension, Nashville, Tennessee; Vanderbilt Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Biochemistry, Nashville, Tennessee; Department of Pathology, Microbiology, and Immunology, Nashville, Tennessee; Department of Cell and Developmental Biology, Nashville, Tennessee; Vanderbilt-Ingram Cancer Center, Nashville, Tennessee; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee
| | - Sergei P Boudko
- Department of Medicine, Division of Nephrology and Hypertension, Nashville, Tennessee; Vanderbilt Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Biochemistry, Nashville, Tennessee
| | - Nancy R Forde
- Department of Molecular Biology and Biochemistry, Burnaby, British Columbia, Canada; Department of Physics, Burnaby, British Columbia, Canada; Department of Chemistry, Burnaby, British Columbia, Canada; Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, British Columbia, Canada.
| |
Collapse
|
20
|
Savige J, Storey H, Watson E, Hertz JM, Deltas C, Renieri A, Mari F, Hilbert P, Plevova P, Byers P, Cerkauskaite A, Gregory M, Cerkauskiene R, Ljubanovic DG, Becherucci F, Errichiello C, Massella L, Aiello V, Lennon R, Hopkinson L, Koziell A, Lungu A, Rothe HM, Hoefele J, Zacchia M, Martic TN, Gupta A, van Eerde A, Gear S, Landini S, Palazzo V, al-Rabadi L, Claes K, Corveleyn A, Van Hoof E, van Geel M, Williams M, Ashton E, Belge H, Ars E, Bierzynska A, Gangemi C, Lipska-Ziętkiewicz BS. Consensus statement on standards and guidelines for the molecular diagnostics of Alport syndrome: refining the ACMG criteria. Eur J Hum Genet 2021; 29:1186-1197. [PMID: 33854215 PMCID: PMC8384871 DOI: 10.1038/s41431-021-00858-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 02/13/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
The recent Chandos House meeting of the Alport Variant Collaborative extended the indications for screening for pathogenic variants in the COL4A5, COL4A3 and COL4A4 genes beyond the classical Alport phenotype (haematuria, renal failure; family history of haematuria or renal failure) to include persistent proteinuria, steroid-resistant nephrotic syndrome, focal and segmental glomerulosclerosis (FSGS), familial IgA glomerulonephritis and end-stage kidney failure without an obvious cause. The meeting refined the ACMG criteria for variant assessment for the Alport genes (COL4A3-5). It identified 'mutational hotspots' (PM1) in the collagen IV α5, α3 and α4 chains including position 1 Glycine residues in the Gly-X-Y repeats in the intermediate collagenous domains; and Cysteine residues in the carboxy non-collagenous domain (PP3). It considered that 'well-established' functional assays (PS3, BS3) were still mainly research tools but sequencing and minigene assays were commonly used to confirm splicing variants. It was not possible to define the Minor Allele Frequency (MAF) threshold above which variants were considered Benign (BA1, BS1), because of the different modes of inheritances of Alport syndrome, and the occurrence of hypomorphic variants (often Glycine adjacent to a non-collagenous interruption) and local founder effects. Heterozygous COL4A3 and COL4A4 variants were common 'incidental' findings also present in normal reference databases. The recognition and interpretation of hypomorphic variants in the COL4A3-COL4A5 genes remains a challenge.
Collapse
Affiliation(s)
- Judy Savige
- grid.1008.90000 0001 2179 088XDepartment of Medicine (MH and NH), The University of Melbourne, Parkville, VIC Australia
| | - Helen Storey
- grid.239826.40000 0004 0391 895XMolecular Genetics, Viapath Laboratories, Guy’s Hospital, London, UK
| | - Elizabeth Watson
- Elizabeth Watson, South West Genomic Laboratory Hub, North Bristol Trust, Bristol, UK
| | - Jens Michael Hertz
- grid.7143.10000 0004 0512 5013Jens Michael Hertz, Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Constantinos Deltas
- grid.6603.30000000121167908Center of Excellence in Biobanking and Biomedical Research and Molecule Medicine Center, University of Cyprus, Nicosia, Cyprus
| | - Alessandra Renieri
- grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Francesca Mari
- Institute de Pathologie et de Genetique ASBL, Departement de Biologie Moleculaire, Gosselies, Belgium
| | - Pascale Hilbert
- Institute de Pathologie et de Genetique ASBL, Departement de Biologie Moleculaire, Gosselies, Belgium
| | - Pavlina Plevova
- grid.412727.50000 0004 0609 0692Department of Medical Genetics, and Department of Biomedical Sciences, University Hospital of Ostrava, Ostrava, Czech Republic
| | - Peter Byers
- grid.34477.330000000122986657Departments of Pathology and Medicine (Medical Genetics), University of Washington, Seattle, WA USA
| | - Agne Cerkauskaite
- grid.6441.70000 0001 2243 2806Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Martin Gregory
- grid.223827.e0000 0001 2193 0096Division of Nephrology, Department of Medicine, University of Utah Health, Salt Lake City, UT USA
| | - Rimante Cerkauskiene
- grid.6441.70000 0001 2243 2806Clinic of Pediatrics, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Danica Galesic Ljubanovic
- grid.412095.b0000 0004 0631 385XDepartment of Pathology, University of Zagreb, School of Medicine, Dubrava University Hospital, Zagreb, Croatia
| | - Francesca Becherucci
- grid.411477.00000 0004 1759 0844Nephrology Unit and Meyer Children’s University Hospital, Firenze, Italy
| | - Carmela Errichiello
- grid.411477.00000 0004 1759 0844Nephrology Unit and Meyer Children’s University Hospital, Firenze, Italy
| | - Laura Massella
- grid.414125.70000 0001 0727 6809Division of Nephrology and Dialysis, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Valeria Aiello
- grid.6292.f0000 0004 1757 1758Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, University of Bologna, Bologna, Italy
| | - Rachel Lennon
- grid.5379.80000000121662407Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - Louise Hopkinson
- grid.5379.80000000121662407Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - Ania Koziell
- grid.13097.3c0000 0001 2322 6764School of Immunology and Microbial Sciences, Faculty of Life Sciences, King’s College London, London, UK
| | - Adrian Lungu
- grid.415180.90000 0004 0540 9980Fundeni Clinical Institute, Pediatric Nephrology Department, Bucharest, Romania
| | | | - Julia Hoefele
- grid.6936.a0000000123222966Institute of Human Genetics, Technical University of Munich, München, Germany
| | | | - Tamara Nikuseva Martic
- grid.4808.40000 0001 0657 4636Department of Biology, School of Medicine University of Zagreb, Zagreb, Croatia
| | - Asheeta Gupta
- grid.415246.00000 0004 0399 7272Birmingham Children’s Hospital, Birmingham, UK
| | - Albertien van Eerde
- grid.5477.10000000120346234Departments of Genetics and Center for Molecular Medicine, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | | | - Samuela Landini
- grid.8404.80000 0004 1757 2304Medical Genetics Unit, Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Viviana Palazzo
- grid.411477.00000 0004 1759 0844Medical Genetics Unit, Meyer Children’s University Hospital, Florence, Italy
| | - Laith al-Rabadi
- grid.223827.e0000 0001 2193 0096Health Sciences Centre, University of UTAH, Salt Lake City, UT USA
| | - Kathleen Claes
- grid.410569.f0000 0004 0626 3338Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Anniek Corveleyn
- grid.410569.f0000 0004 0626 3338Center for Human Genetics, University Hospitals and KU Leuven, Leuven, Belgium
| | - Evelien Van Hoof
- grid.410569.f0000 0004 0626 3338Center for Human Genetics, University Hospitals and KU Leuven, Leuven, Belgium
| | - Micheel van Geel
- grid.412966.e0000 0004 0480 1382Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Maggie Williams
- grid.416201.00000 0004 0417 1173Bristol Genetics Laboratory Pathology Sciences, Southmead Hospital, Bristol, UK
| | - Emma Ashton
- grid.420468.cNorth East Thames Regional Genetics Laboratory, Great Ormond Street Hospital, London, UK
| | - Hendica Belge
- grid.10417.330000 0004 0444 9382Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elisabeth Ars
- grid.7080.f0000 0001 2296 0625Inherited Kidney Disorders, Fundacio Puigvert, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Agnieszka Bierzynska
- grid.5337.20000 0004 1936 7603Bristol Renal Unit, Bristol Medical School, University of Bristol, Bristol, UK
| | - Concetta Gangemi
- grid.411475.20000 0004 1756 948XDivision of Nephrology and Dialysis, University Hospital of Verona, Verona, Italy
| | - Beata S. Lipska-Ziętkiewicz
- grid.11451.300000 0001 0531 3426Centre for Rare Diseases, and Clinical Genetics Unit, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
21
|
Kaushik BK, Singh L, Singh R, Zhu G, Zhang B, Wang Q, Kumar S. Detection of Collagen-IV Using Highly Reflective Metal Nanoparticles-Immobilized Photosensitive Optical Fiber-Based MZI Structure. IEEE Trans Nanobioscience 2021; 19:477-484. [PMID: 32603296 DOI: 10.1109/tnb.2020.2998520] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In this work, a photosensitive (PS) optical fiber-based Mach-Zehnder interferometer (MZI) structure is developed to diagnose the presence of collagen-IV in human bodies. The MZI is fabricated by sequentially splicing the single mode-multimode-photosensitive-multimode-single mode (SMPMS) fiber segments. The sensing region in MZI structure is created by partially removing the cladding of photosensitive fiber by using 40% hydrofluoric (HF) acid and depositing the layers of highly reflective metal nanoparticles (NPs) over it. The used NPs are polyvinyl alcohol stabilized silver nanoparticles (PVA-AgNPs), gold nanoparticles (AuNPs), and zinc oxide nanoparticles (ZnO-NPs). The size of AuNPs, PVA-AgNPs, and ZnO-NPs are 10 ± 0.2 nm, ∼ 4 -5 nm, and < 50 nm, respectively. In order to avoid the interference of other biomolecules in the detection of collagen-IV, the sensing region is functionalized with a collagenase enzyme. The sensing ability of the probe is ascertained by sensing a wide concentration of collagen solution ranging from 0 ng/ml to [Formula: see text]/ml. It is observed that sensing performance of probe is much better on immobilizing it with PVA-AgNPs and ZnO-NPs.
Collapse
|
22
|
Ma Y, Wang Z, Gao M, Liu X, Sun W, Gong Y, Sun G, Liu G. Generation of two induced pluripotent stem cell lines from patients with X-linked Alport syndrome. Stem Cell Res 2021; 53:102343. [PMID: 33878711 DOI: 10.1016/j.scr.2021.102343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/27/2021] [Accepted: 04/05/2021] [Indexed: 11/19/2022] Open
Abstract
Mutations in COL4A5 on chromosome Xq22 cause X-linked Alport syndrome (XLAS). In this study, we generated two human induced pluripotent stem cell (iPSC) lines from two male patients carrying mutation c.796C > T (p.R266X) in COL4A5 gene. The two iPSC lines retain the original mutation, possess normal karyotypes, express pluripotency markers and bear differentiation potential.
Collapse
Affiliation(s)
- Yanyan Ma
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhendong Wang
- Department of Nephrology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Min Gao
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Jinan, Shandong 250022, China
| | - Xiaolin Liu
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wenjie Sun
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yaoqin Gong
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Gongping Sun
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Guangyi Liu
- Department of Nephrology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
23
|
Mao M, Popli T, Jeanne M, Hoff K, Sen S, Gould DB. Identification of fibronectin 1 as a candidate genetic modifier in a Col4a1 mutant mouse model of Gould syndrome. Dis Model Mech 2021; 14:dmm048231. [PMID: 34424299 PMCID: PMC8106953 DOI: 10.1242/dmm.048231] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Collagen type IV alpha 1 and alpha 2 (COL4A1 and COL4A2) are major components of almost all basement membranes. COL4A1 and COL4A2 mutations cause a multisystem disorder that can affect any organ but typically involves the cerebral vasculature, eyes, kidneys and skeletal muscles. In recent years, patient advocacy and family support groups have united under the name of Gould syndrome. The manifestations of Gould syndrome are highly variable, and animal studies suggest that allelic heterogeneity and genetic context contribute to the clinical variability. We previously characterized a mouse model of Gould syndrome caused by a Col4a1 mutation in which the severities of ocular anterior segment dysgenesis (ASD), myopathy and intracerebral hemorrhage (ICH) were dependent on genetic background. Here, we performed a genetic modifier screen to provide insight into the mechanisms contributing to Gould syndrome pathogenesis and identified a single locus [modifier of Gould syndrome 1 (MoGS1)] on Chromosome 1 that suppressed ASD. A separate screen showed that the same locus ameliorated myopathy. Interestingly, MoGS1 had no effect on ICH, suggesting that this phenotype could be mechanistically distinct. We refined the MoGS1 locus to a 4.3 Mb interval containing 18 protein-coding genes, including Fn1, which encodes the extracellular matrix component fibronectin 1. Molecular analysis showed that the MoGS1 locus increased Fn1 expression, raising the possibility that suppression is achieved through a compensatory extracellular mechanism. Furthermore, we found evidence of increased integrin-linked kinase levels and focal adhesion kinase phosphorylation in Col4a1 mutant mice that is partially restored by the MoGS1 locus, implicating the involvement of integrin signaling. Taken together, our results suggest that tissue-specific mechanistic heterogeneity contributes to the variable expressivity of Gould syndrome and that perturbations in integrin signaling may play a role in ocular and muscular manifestations.
Collapse
Affiliation(s)
- Mao Mao
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Tanav Popli
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Marion Jeanne
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kendall Hoff
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Saunak Sen
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94143, USA
- Institute of Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Preventive Medicine, University of Tennessee Health Science Center, 66 North Pauline St, Memphis, TN 38163, USA
| | - Douglas B. Gould
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
- Institute of Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
24
|
Xie X, He H, Zhang N, Wang X, Rui W, Xu D, Zhu Y. Overexpression of DDR1 Promotes Migration, Invasion, Though EMT-Related Molecule Expression and COL4A1/DDR1/MMP-2 Signaling Axis. Technol Cancer Res Treat 2020; 19:1533033820973277. [PMID: 33234027 PMCID: PMC7705183 DOI: 10.1177/1533033820973277] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose: Discoidin domain receptor 1 (DDR1) belongs to a novel class of receptor tyrosine kinases. Previous evidence indicates that DDR1 overexpression promotes the aggressive growth of bladder cancer (BC) cells. This study aimed to investigate the molecular mechanisms by which DDR1 influences BC. Methods: DDR1 was transfected into human BC RT4 cells. DDR1, COL4A1, and MMP-2 expression in 30 BC tissues and paired adjacent tissues were examined by real-time polymerase chain reaction (RT-PCR) and immunohistochemistry. Transwell assays were conducted to determine cell migration and invasion. RT-PCR and western blot (WB) were also used to measure the DDR1, COL4A1, MMP-2, and EMT-related gene (ZEB1 and SLUG) expression in RT4 cells after DDR1 overexpression. Results: COL4A1 and MMP-2 interacted with DDR1 in the PPI network. RT-PCR and immunohistochemistry results showed that both mRNA and protein levels of DDR1 and COL4A1 were significantly increased in BC tissue, while the expression of MMP-2 was increased only at the mRNA level (P < 0.05). Overexpression of DDR1 in RT4 cells significantly promoted their migratory and invasive capabilities in vitro (P < 0.05). Moreover, overexpression of DDR1 in RT4 cells increased the mRNA and protein expression of ZEB1, SLUG, COL4A1, and MMP-2 (P < 0.01). DDR1-mediated migration and invasion of RT4 cells were reversed after COL4A1-siRNA treatment. Conclusion: DDR1 may be a potential therapeutic target in BC patients.
Collapse
Affiliation(s)
- Xin Xie
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongchao He
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ning Zhang
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaojing Wang
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenbin Rui
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Danfeng Xu
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu Zhu
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Roy S, Kim D. Retinal capillary basement membrane thickening: Role in the pathogenesis of diabetic retinopathy. Prog Retin Eye Res 2020; 82:100903. [PMID: 32950677 DOI: 10.1016/j.preteyeres.2020.100903] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023]
Abstract
Vascular basement membrane (BM) thickening has been hailed over half a century as the most prominent histological lesion in diabetic microangiopathy, and represents an early ultrastructural change in diabetic retinopathy (DR). Although vascular complications of DR have been clinically well established, specific cellular and molecular mechanisms underlying dysfunction of small vessels are not well understood. In DR, small vessels develop insidiously as BM thickening occurs. Studies examining high resolution imaging data have established BM thickening as one of the foremost structural abnormalities of retinal capillaries. This fundamental structural change develops, at least in part, from excess accumulation of BM components. Although BM thickening is closely associated with the development of DR, its contributory role in the pathogenesis of DR is coming to light recently. DR develops over several years before clinical manifestations appear, and it is during this clinically silent period that hyperglycemia induces excess synthesis of BM components, contributes to vascular BM thickening, and promotes structural and functional lesions including cell death and vascular leakage in the diabetic retina. Studies using animal models show promising results in preventing BM thickening with subsequent beneficial effects. Several gene regulatory approaches are being developed to prevent excess synthesis of vascular BM components in an effort to reduce BM thickening. This review highlights current understanding of capillary BM thickening development, role of BM thickening in retinal vascular lesions, and strategies for preventing vascular BM thickening as a potential therapeutic strategy in alleviating characteristic lesions associated with DR.
Collapse
Affiliation(s)
- Sayon Roy
- Boston University School of Medicine, Boston, MA, USA.
| | - Dongjoon Kim
- Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
26
|
Mutgan AC, Jandl K, Kwapiszewska G. Endothelial Basement Membrane Components and Their Products, Matrikines: Active Drivers of Pulmonary Hypertension? Cells 2020; 9:cells9092029. [PMID: 32899187 PMCID: PMC7563239 DOI: 10.3390/cells9092029] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a vascular disease that is characterized by elevated pulmonary arterial pressure (PAP) due to progressive vascular remodeling. Extracellular matrix (ECM) deposition in pulmonary arteries (PA) is one of the key features of vascular remodeling. Emerging evidence indicates that the basement membrane (BM), a specialized cluster of ECM proteins underlying the endothelium, may be actively involved in the progression of vascular remodeling. The BM and its steady turnover are pivotal for maintaining appropriate vascular functions. However, the pathologically elevated turnover of BM components leads to an increased release of biologically active short fragments, which are called matrikines. Both BM components and their matrikines can interfere with pivotal biological processes, such as survival, proliferation, adhesion, and migration and thus may actively contribute to endothelial dysfunction. Therefore, in this review, we summarize the emerging role of the BM and its matrikines on the vascular endothelium and further discuss its implications on lung vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Ayse Ceren Mutgan
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, 8010 Graz, Austria;
| | - Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, 8010 Graz, Austria;
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
- Correspondence:
| |
Collapse
|
27
|
Isolation of Epidermal Keratinocytes from Human Skin: The Scratch-Wound Assay for Assessment of Epidermal Keratinocyte Migration. Methods Mol Biol 2020. [PMID: 32314203 DOI: 10.1007/978-1-0716-0648-3_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
The migration of epidermal keratinocytes is the basis for skin reepithelialization during wound healing. The in vitro scratch-wound assay using monolayers of primary human epidermal keratinocytes is a straightforward and effective method to assess their migratory capacity. The mechanical scratch of a confluent monolayer directly disrupts the adhesion of the keratinocytes to one another and to the underlying matrix, resembling the physical trauma of a wound in an in vitro assay. The keratinocytes will undergo an epithelial-to-mesenchymal transition, which will confer an ability to migrate toward each other to cover the gap by restructuring cell-cell and cell-extracellular matrix connections. However, a good scratch-wound method and protocol to ensure scratch reproducibility is essential, particularly when using primary cell cultures where donor variability may also impact on results.
Collapse
|
28
|
Yeo J, Qiu Y, Jung GS, Zhang YW, Buehler MJ, Kaplan DL. Adverse effects of Alport syndrome-related Gly missense mutations on collagen type IV: Insights from molecular simulations and experiments. Biomaterials 2020; 240:119857. [PMID: 32085975 DOI: 10.1016/j.biomaterials.2020.119857] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 01/28/2020] [Accepted: 02/08/2020] [Indexed: 12/13/2022]
Abstract
Patients with Alport syndrome (AS) exhibit blood and elevated protein levels in their urine, inflamed kidneys, and many other abnormalities. AS is attributed to mutations in type IV collagen genes, particularly glycine missense mutations in the collagenous domain of COL4A5 that disrupt common structural motifs in collagen from the repeat (Gly-Xaa-Yaa)n amino acid sequence. To characterize and elucidate the molecular mechanisms underlying how AS-related mutations perturb the structure and function of type IV collagen, experimental studies and molecular simulations were integrated to investigate the structure, stability, protease sensitivity, and integrin binding affinity of collagen-like proteins containing amino acid sequences from the α5(IV) chain and AS-related Gly missense mutations. We show adverse effects where (i) three AS-related Gly missense mutations significantly reduced the structural stability of the collagen in terms of decreased melting temperatures and calorimetric enthalpies, in conjunction with a collective drop in the external work needed to unfold the peptides containing mutation sequences; (ii) due to local unwinding around the sites of mutations, these triple helical peptides were also degraded more rapidly by trypsin and chymotrypsin, as these enzymes could access the collagenous triple helix more easily and increase the number of contacts; (iii) the mutations further abolished the ability of the recombinant collagens to bind to integrins and greatly reduced the binding affinities between collagen and integrins, thus preventing cells from adhering to these mutants. Our unified experimental and computational approach provided underlying insights needed to guide potential therapies for AS that ameliorate the adverse effects from AS disease onset and progression.
Collapse
Affiliation(s)
- Jingjie Yeo
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA; Laboratory for Atomistic and Molecular Mechanics (LAMM), Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Institute of High Performance Computing, A*STAR, 1 Fusionopolis Way, Singapore 138632, Singapore; Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yimin Qiu
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA; National Biopesticide Engineering Technology Research Center, Hubei Biopesticide Engineering Research Center, Hubei Academy of Agricultural Sciences, Biopesticide Branch of Hubei Innovation Centre of Agricultural Science and Technology, Wuhan, 430064, PR China
| | - Gang Seob Jung
- Laboratory for Atomistic and Molecular Mechanics (LAMM), Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yong-Wei Zhang
- Institute of High Performance Computing, A*STAR, 1 Fusionopolis Way, Singapore 138632, Singapore
| | - Markus J Buehler
- Laboratory for Atomistic and Molecular Mechanics (LAMM), Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA.
| |
Collapse
|
29
|
|
30
|
Falcone S, Wisby L, Nicol T, Blease A, Starbuck B, Parker A, Sanderson J, Brown SDM, Scudamore CL, Pusey CD, Tam FWK, Potter PK. Modification of an aggressive model of Alport Syndrome reveals early differences in disease pathogenesis due to genetic background. Sci Rep 2019; 9:20398. [PMID: 31892712 PMCID: PMC6938516 DOI: 10.1038/s41598-019-56837-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/05/2019] [Indexed: 12/13/2022] Open
Abstract
The link between mutations in collagen genes and the development of Alport Syndrome has been clearly established and a number of animal models, including knock-out mouse lines, have been developed that mirror disease observed in patients. However, it is clear from both patients and animal models that the progression of disease can vary greatly and can be modified genetically. We have identified a point mutation in Col4a4 in mice where disease is modified by strain background, providing further evidence of the genetic modification of disease symptoms. Our results indicate that C57BL/6J is a protective background and postpones end stage renal failure from 7 weeks, as seen on a C3H background, to several months. We have identified early differences in disease progression, including expression of podocyte-specific genes and podocyte morphology. In C57BL/6J mice podocyte effacement is delayed, prolonging normal renal function. The slower disease progression has allowed us to begin dissecting the pathogenesis of murine Alport Syndrome in detail. We find that there is evidence of differential gene expression during disease on the two genetic backgrounds, and that disease diverges by 4 weeks of age. We also show that an inflammatory response with increasing MCP-1 and KIM-1 levels precedes loss of renal function.
Collapse
Affiliation(s)
- Sara Falcone
- Mammalian Genetics Unit, Medical Research Council, Harwell science and innovation campus, Oxford, OX11 0RD, UK
| | - Laura Wisby
- Mammalian Genetics Unit, Medical Research Council, Harwell science and innovation campus, Oxford, OX11 0RD, UK
| | - Thomas Nicol
- Mammalian Genetics Unit, Medical Research Council, Harwell science and innovation campus, Oxford, OX11 0RD, UK
| | - Andrew Blease
- Mammalian Genetics Unit, Medical Research Council, Harwell science and innovation campus, Oxford, OX11 0RD, UK
| | - Becky Starbuck
- Mammalian Genetics Unit, Medical Research Council, Harwell science and innovation campus, Oxford, OX11 0RD, UK
| | - Andrew Parker
- Mammalian Genetics Unit, Medical Research Council, Harwell science and innovation campus, Oxford, OX11 0RD, UK
| | - Jeremy Sanderson
- Mammalian Genetics Unit, Medical Research Council, Harwell science and innovation campus, Oxford, OX11 0RD, UK
| | - Steve D M Brown
- Mammalian Genetics Unit, Medical Research Council, Harwell science and innovation campus, Oxford, OX11 0RD, UK
| | - Cheryl L Scudamore
- Mary Lyon Centre, Medical Research Council, Harwell science and innovation campus, Oxford, OX11 0RD, UK
| | - Charles D Pusey
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College, London, W12 0N, UK
| | - Frederick W K Tam
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College, London, W12 0N, UK
| | - Paul K Potter
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College, London, W12 0N, UK.
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK.
| |
Collapse
|
31
|
Kirkness MWH, Lehmann K, Forde NR. Mechanics and structural stability of the collagen triple helix. Curr Opin Chem Biol 2019; 53:98-105. [DOI: 10.1016/j.cbpa.2019.08.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/24/2019] [Accepted: 08/12/2019] [Indexed: 01/18/2023]
|
32
|
Barreiro KA, Tulamo R, Jouppila A, Albäck A, Lassila R. Novel Locally Acting Dual Antiplatelet and Anticoagulant (APAC) Targets Multiple Sites of Vascular Injury in an Experimental Porcine Model. Eur J Vasc Endovasc Surg 2019; 58:903-911. [PMID: 31708337 DOI: 10.1016/j.ejvs.2019.05.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 05/16/2019] [Accepted: 05/23/2019] [Indexed: 01/18/2023]
Abstract
OBJECTIVES Vascular binding of dual antiplatelet and anticoagulant (APAC) was assessed in surgically created femoral arteriovenous fistula (AVF) and iliac and carotid artery injury in porcine models. METHODS Three models of collagen exposing injury were used: 1) femoral AVF, 2) in vivo iliac and carotid artery balloon angioplasty injury, and 3) in vitro femoral artery endothelial denudation injury. Biotinylated APAC (0.5 mg/mL) was incubated with the injury site before releasing blood flow. APAC, von Willebrand factor (vWF), laminin, platelet endothelial cell adhesion molecule 1 (PECAM-1), and podocalyxin were detected in histological sections using immunofluorescence and confocal microscopy and Manders' co-localisation coefficient (M1). RESULTS APAC bound to AVF at anastomosis and to both in vivo and in vitro injured arteries. APAC co-localised with matrix vWF (M1 ≥ 0.66) and laminin (M1 ≥ 0.60), but less so if endothelial PECAM-1 or podocalyxin was present (M1 ≤ 0.25). APAC targeted and penetrated the injured vessel wall, especially the AVF vein. CONCLUSIONS APAC, compatible with its high negative charge, rapidly targets injured vessels co-localizing with matrix vWF and laminin, but not with endothelial PECAM-1 and podocalyxin. This localising feature may have potential antithrombotic implications for vascular interventions.
Collapse
Affiliation(s)
- Karina A Barreiro
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland; Aplagon Oy, Helsinki, Finland
| | - Riikka Tulamo
- Department of Vascular Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Annukka Jouppila
- Helsinki University Hospital Clinical Research Institute, Helsinki, Finland
| | - Anders Albäck
- Department of Vascular Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Riitta Lassila
- Coagulation Disorders Unit, University of Helsinki, Departments of Haematology and Clinical Chemistry (HUSLAB Laboratory Services), Comprehensive Cancer Center, Helsinki University Hospital and Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Helsinki University, Faculty of Medicine, Research Program in Oncology, Helsinki, Finland; Aplagon Oy, Helsinki, Finland.
| |
Collapse
|
33
|
Type IV Collagen Is Essential for Proper Function of Integrin-Mediated Adhesion in Drosophila Muscle Fibers. Int J Mol Sci 2019; 20:ijms20205124. [PMID: 31623094 PMCID: PMC6829409 DOI: 10.3390/ijms20205124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/09/2019] [Accepted: 10/13/2019] [Indexed: 01/18/2023] Open
Abstract
Congenital muscular dystrophy (CMD), a subgroup of myopathies is a genetically and clinically heterogeneous group of inherited muscle disorders and is characterized by progressive muscle weakness, fiber size variability, fibrosis, clustered necrotic fibers, and central myonuclei present in regenerating muscle. Type IV collagen (COL4A1) mutations have recently been identified in patients with intracerebral, vascular, renal, ophthalmologic pathologies and congenital muscular dystrophy, consistent with diagnoses of Walker–Warburg Syndrome or Muscle–Eye–Brain disease. Morphological characteristics of muscular dystrophy have also been demonstrated Col4a1 mutant mice. Yet, several aspects of the pathomechanism of COL4A1-associated muscle defects remained largely uncharacterized. Based on the results of genetic, histological, molecular, and biochemical analyses in an allelic series of Drosophila col4a1 mutants, we provide evidence that col4a1 mutations arise by transitions in glycine triplets, associate with severely compromised muscle fibers within the single-layer striated muscle of the common oviduct, characterized by loss of sarcomere structure, disintegration and streaming of Z-discs, indicating an essential role for the COL4A1 protein. Features of altered cytoskeletal phenotype include actin bundles traversing over sarcomere units, amorphous actin aggregates, atrophy, and aberrant fiber size. The mutant COL4A1-associated defects appear to recapitulate integrin-mediated adhesion phenotypes observed in RNA-inhibitory Drosophila. Our results provide insight into the mechanistic details of COL4A1-associated muscle disorders and suggest a role for integrin-collagen interaction in the maintenance of sarcomeres.
Collapse
|
34
|
Davis-Hall D, Nguyen V, D'Ovidio TJ, Tsai E, Bilousova G, Magin CM. Peptide-Functionalized Hydrogels Modulate Integrin Expression and Stemness in Adult Human Epidermal Keratinocytes. ADVANCED BIOSYSTEMS 2019; 3:e1900022. [PMID: 32648724 PMCID: PMC11949078 DOI: 10.1002/adbi.201900022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/20/2019] [Indexed: 01/18/2023]
Abstract
The extracellular matrix (ECM) controls keratinocyte proliferation, migration, and differentiation through β-integrin signaling. Wound-healing research requires expanding cells in vitro while maintaining replicative capacity; however, early terminal differentiation under traditional culture conditions limits expansion. Here, a design of experiments approach identifies poly(ethylene glycol)-based hydrogel formulations with mechanical properties (elastic modulus, E = 20.9 ± 0.56 kPa) and bioactive peptide sequences that mimic the epidermal ECM. These hydrogels enable systematic investigation of the influence of cell-binding domains from fibronectin (RGDS), laminin (YIGSR), and collagen IV (HepIII) on keratinocyte stemness and β1 integrin expression. Quantification of 14-day keratin protein expression shows four hydrogels improve stemness compared to standard techniques. Three hydrogels increase β1 integrin expression, demonstrating a positive linear relationship between stemness and β1 integrin expression. Multifactorial statistical analysis predicts an optimal peptide combination ([RGDS] = 0.67 mm, [YIGSR] = 0.13 mm, and [HepIII] = 0.02 mm) for maintaining stemness in vitro. Best-performing hydrogels exhibit no decrease in Ki-67-positive cells compared to standards (15% decrease, day 7 to 14; p < 0.05, Tukey Test). These data demonstrate that precisely designed hydrogel biomaterials direct integrin expression and promote proliferation, improving the regenerative capability of cultured keratinocytes for basic science and translational work.
Collapse
Affiliation(s)
- Duncan Davis-Hall
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine and Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, 12700 E 19th Ave, MS C272, Aurora, CO, 80045, USA
| | - Vy Nguyen
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine and Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, 12700 E 19th Ave, MS C272, Aurora, CO, 80045, USA
| | - Tyler J D'Ovidio
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine and Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, 12700 E 19th Ave, MS C272, Aurora, CO, 80045, USA
| | - Ethan Tsai
- Metropolitan State University of Denver, Chemistry and Biochemistry Department, P.O. Box 173362, Campus Box 52, Denver, CO, 80217-3362, USA
| | - Ganna Bilousova
- University of Colorado Anschutz Medical Campus, School of Medicine, Department of Dermatology and Charles C. Gates Center for Regenerative Medicine, 12800 E. 19th Ave, P18-8125, Aurora, CO, 80045, USA
| | - Chelsea M Magin
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine and Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, 12700 E 19th Ave, MS C272, Aurora, CO, 80045, USA
| |
Collapse
|
35
|
Salem RM, Todd JN, Sandholm N, Cole JB, Chen WM, Andrews D, Pezzolesi MG, McKeigue PM, Hiraki LT, Qiu C, Nair V, Di Liao C, Cao JJ, Valo E, Onengut-Gumuscu S, Smiles AM, McGurnaghan SJ, Haukka JK, Harjutsalo V, Brennan EP, van Zuydam N, Ahlqvist E, Doyle R, Ahluwalia TS, Lajer M, Hughes MF, Park J, Skupien J, Spiliopoulou A, Liu A, Menon R, Boustany-Kari CM, Kang HM, Nelson RG, Klein R, Klein BE, Lee KE, Gao X, Mauer M, Maestroni S, Caramori ML, de Boer IH, Miller RG, Guo J, Boright AP, Tregouet D, Gyorgy B, Snell-Bergeon JK, Maahs DM, Bull SB, Canty AJ, Palmer CNA, Stechemesser L, Paulweber B, Weitgasser R, Sokolovska J, Rovīte V, Pīrāgs V, Prakapiene E, Radzeviciene L, Verkauskiene R, Panduru NM, Groop LC, McCarthy MI, Gu HF, Möllsten A, Falhammar H, Brismar K, Martin F, Rossing P, Costacou T, Zerbini G, Marre M, Hadjadj S, McKnight AJ, Forsblom C, McKay G, Godson C, Maxwell AP, Kretzler M, Susztak K, Colhoun HM, Krolewski A, Paterson AD, Groop PH, Rich SS, Hirschhorn JN, Florez JC. Genome-Wide Association Study of Diabetic Kidney Disease Highlights Biology Involved in Glomerular Basement Membrane Collagen. J Am Soc Nephrol 2019; 30:2000-2016. [PMID: 31537649 PMCID: PMC6779358 DOI: 10.1681/asn.2019030218] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 07/08/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Although diabetic kidney disease demonstrates both familial clustering and single nucleotide polymorphism heritability, the specific genetic factors influencing risk remain largely unknown. METHODS To identify genetic variants predisposing to diabetic kidney disease, we performed genome-wide association study (GWAS) analyses. Through collaboration with the Diabetes Nephropathy Collaborative Research Initiative, we assembled a large collection of type 1 diabetes cohorts with harmonized diabetic kidney disease phenotypes. We used a spectrum of ten diabetic kidney disease definitions based on albuminuria and renal function. RESULTS Our GWAS meta-analysis included association results for up to 19,406 individuals of European descent with type 1 diabetes. We identified 16 genome-wide significant risk loci. The variant with the strongest association (rs55703767) is a common missense mutation in the collagen type IV alpha 3 chain (COL4A3) gene, which encodes a major structural component of the glomerular basement membrane (GBM). Mutations in COL4A3 are implicated in heritable nephropathies, including the progressive inherited nephropathy Alport syndrome. The rs55703767 minor allele (Asp326Tyr) is protective against several definitions of diabetic kidney disease, including albuminuria and ESKD, and demonstrated a significant association with GBM width; protective allele carriers had thinner GBM before any signs of kidney disease, and its effect was dependent on glycemia. Three other loci are in or near genes with known or suggestive involvement in this condition (BMP7) or renal biology (COLEC11 and DDR1). CONCLUSIONS The 16 diabetic kidney disease-associated loci may provide novel insights into the pathogenesis of this condition and help identify potential biologic targets for prevention and treatment.
Collapse
Affiliation(s)
- Rany M Salem
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California
| | - Jennifer N Todd
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts
- Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
- Center for Genomic Medicine and
| | - Niina Sandholm
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
| | - Joanne B Cole
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts
- Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
- Center for Genomic Medicine and
| | - Wei-Min Chen
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, Virginia
| | - Darrell Andrews
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Marcus G Pezzolesi
- Division of Nephrology and Hypertension, Diabetes and Metabolism Center, University of Utah, Salt Lake City, Utah
| | - Paul M McKeigue
- Usher Institute of Population Health Sciences and Informatics and
| | - Linda T Hiraki
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Chengxiang Qiu
- Departments of Medicine and Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine and
| | - Chen Di Liao
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jing Jing Cao
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Erkka Valo
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
| | - Suna Onengut-Gumuscu
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, Virginia
| | | | - Stuart J McGurnaghan
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Jani K Haukka
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
| | - Valma Harjutsalo
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
- The Chronic Disease Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Eoin P Brennan
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Natalie van Zuydam
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Emma Ahlqvist
- Department of Genomics, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Ross Doyle
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | | | - Maria Lajer
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Maria F Hughes
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Jihwan Park
- Departments of Medicine and Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jan Skupien
- Joslin Diabetes Center, Boston, Massachusetts
| | | | | | - Rajasree Menon
- Division of Nephrology, Department of Internal Medicine and
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | | | - Hyun M Kang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Robert G Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona
| | - Ronald Klein
- University of Wisconsin-Madison, Madison, Wisconsin
| | | | | | - Xiaoyu Gao
- The George Washington University, Washington, DC
| | | | - Silvia Maestroni
- Complications of Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | | | | | - Rachel G Miller
- University of Pittsburgh Public Health, Pittsburgh, Pennsylvania
| | - Jingchuan Guo
- University of Pittsburgh Public Health, Pittsburgh, Pennsylvania
| | | | - David Tregouet
- INSERM UMR_S 1166, Sorbonne Université, UPMC Univ Paris 06, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Beata Gyorgy
- INSERM UMR_S 1166, Sorbonne Université, UPMC Univ Paris 06, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | | | - David M Maahs
- Department of Pediatrics-Endocrinology, Stanford University, Stanford, California
| | - Shelley B Bull
- The Lunenfeld-Tanenbaum Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Angelo J Canty
- Department of Mathematics and Statistics, McMaster University, Hamilton, Ontario, Canada
| | - Colin N A Palmer
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Lars Stechemesser
- First Department of Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Bernhard Paulweber
- First Department of Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Raimund Weitgasser
- First Department of Medicine, Paracelsus Medical University, Salzburg, Austria
- Department of Medicine, Diakonissen-Wehrle Hospital, Salzburg, Austria
| | | | - Vita Rovīte
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Valdis Pīrāgs
- University of Latvia, Riga, Latvia
- Pauls Stradins University Hospital, Riga, Latvia
| | | | - Lina Radzeviciene
- Institute of Endocrinology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rasa Verkauskiene
- Institute of Endocrinology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Nicolae Mircea Panduru
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- 2nd Clinical Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Leif C Groop
- Department of Genomics, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- Genentech, 1 DNA Way, South San Francisco, California
| | - Harvest F Gu
- Department of Clinical Science, Intervention and Technology and
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Anna Möllsten
- Division of Pediatrics, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Rolf Luft Center for Diabetes Research and Endocrinology, Karolinska Institutet, Stockholm, Sweden
- Department of Endocrinology, Diabetes and Metabolism, Karolinska University Hospital, Stockholm, Sweden
| | - Kerstin Brismar
- Department of Molecular Medicine and Surgery, Rolf Luft Center for Diabetes Research and Endocrinology, Karolinska Institutet, Stockholm, Sweden
- Department of Endocrinology, Diabetes and Metabolism, Karolinska University Hospital, Stockholm, Sweden
| | - Finian Martin
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | - Tina Costacou
- University of Pittsburgh Public Health, Pittsburgh, Pennsylvania
| | - Gianpaolo Zerbini
- Complications of Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Michel Marre
- Department of Diabetology, Endocrinology and Nutrition, Bichat Hospital, DHU FIRE, Assistance Publique-Hôpitaux de Paris, Paris, France
- UFR de Médecine, Paris Diderot University, Sorbonne Paris Cité, Paris, France
- INSERM UMRS 1138, Cordeliers Research Center, Paris, France
- Fondation Ophtalmologique Adolphe de Rothschild, Paris, France
| | - Samy Hadjadj
- Department of Endocrinology and Diabetology, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
- INSERM CIC 1402, Poitiers, France
- L'institut du thorax, INSERM, CNRS, CHU Nantes, Nantes, France
| | - Amy J McKnight
- Centre for Public Health, Queens University of Belfast, Northern Ireland, UK
| | - Carol Forsblom
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
| | - Gareth McKay
- Centre for Public Health, Queens University of Belfast, Northern Ireland, UK
| | - Catherine Godson
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - A Peter Maxwell
- Centre for Public Health, Queens University of Belfast, Northern Ireland, UK
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine and
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Katalin Susztak
- Departments of Medicine and Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Helen M Colhoun
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Per-Henrik Groop
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia; and
| | - Stephen S Rich
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, Virginia
| | - Joel N Hirschhorn
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts
- Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Jose C Florez
- Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts;
- Center for Genomic Medicine and
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
36
|
Clauder F, Czerniak AS, Friebe S, Mayr SG, Scheinert D, Beck-Sickinger AG. Endothelialization of Titanium Surfaces by Bioinspired Cell Adhesion Peptide Coatings. Bioconjug Chem 2019; 30:2664-2674. [DOI: 10.1021/acs.bioconjchem.9b00573] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Franziska Clauder
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Anne Sophie Czerniak
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Sabrina Friebe
- Leibniz-Institute of Surface Engineering (IOM), Permoserstrasse 15, 04318 Leipzig, Germany
| | - Stefan G. Mayr
- Leibniz-Institute of Surface Engineering (IOM), Permoserstrasse 15, 04318 Leipzig, Germany
| | - Dierk Scheinert
- Department of Angiology, University Hospital Leipzig, Liebigstrasse 20, 04103 Leipzig, Germany
| | - Annette G. Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| |
Collapse
|
37
|
Jayadev R, Chi Q, Keeley DP, Hastie EL, Kelley LC, Sherwood DR. α-Integrins dictate distinct modes of type IV collagen recruitment to basement membranes. J Cell Biol 2019; 218:3098-3116. [PMID: 31387941 PMCID: PMC6719451 DOI: 10.1083/jcb.201903124] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/16/2019] [Accepted: 07/10/2019] [Indexed: 01/04/2023] Open
Abstract
Basement membranes (BMs) are cell-associated extracellular matrices that support tissue integrity, signaling, and barrier properties. Type IV collagen is critical for BM function, yet how it is directed into BMs in vivo is unclear. Through live-cell imaging of endogenous localization, conditional knockdown, and misexpression experiments, we uncovered distinct mechanisms of integrin-mediated collagen recruitment to Caenorhabditis elegans postembryonic gonadal and pharyngeal BMs. The putative laminin-binding αINA-1/βPAT-3 integrin was selectively activated in the gonad and recruited laminin, which directed moderate collagen incorporation. In contrast, the putative Arg-Gly-Asp (RGD)-binding αPAT-2/βPAT-3 integrin was activated in the pharynx and recruited high levels of collagen in an apparently laminin-independent manner. Through an RNAi screen, we further identified the small GTPase RAP-3 (Rap1) as a pharyngeal-specific PAT-2/PAT-3 activator that modulates collagen levels. Together, these studies demonstrate that tissues can use distinct mechanisms to direct collagen incorporation into BMs to precisely control collagen levels and construct diverse BMs.
Collapse
Affiliation(s)
- Ranjay Jayadev
- Department of Biology, Regeneration Next, Duke University, Durham, NC
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - Qiuyi Chi
- Department of Biology, Regeneration Next, Duke University, Durham, NC
| | - Daniel P Keeley
- Department of Biology, Regeneration Next, Duke University, Durham, NC
| | - Eric L Hastie
- Department of Biology, Regeneration Next, Duke University, Durham, NC
| | - Laura C Kelley
- Department of Biology, Regeneration Next, Duke University, Durham, NC
| | - David R Sherwood
- Department of Biology, Regeneration Next, Duke University, Durham, NC
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| |
Collapse
|
38
|
Savige J, Ariani F, Mari F, Bruttini M, Renieri A, Gross O, Deltas C, Flinter F, Ding J, Gale DP, Nagel M, Yau M, Shagam L, Torra R, Ars E, Hoefele J, Garosi G, Storey H. Expert consensus guidelines for the genetic diagnosis of Alport syndrome. Pediatr Nephrol 2019; 34:1175-1189. [PMID: 29987460 DOI: 10.1007/s00467-018-3985-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/22/2018] [Accepted: 05/10/2018] [Indexed: 12/16/2022]
Abstract
Recent expert guidelines recommend genetic testing for the diagnosis of Alport syndrome. Here, we describe current best practice and likely future developments. In individuals with suspected Alport syndrome, all three COL4A5, COL4A3 and COL4A4 genes should be examined for pathogenic variants, probably by high throughput-targeted next generation sequencing (NGS) technologies, with a customised panel for simultaneous testing of the three Alport genes. These techniques identify up to 95% of pathogenic COL4A variants. Where causative pathogenic variants cannot be demonstrated, the DNA should be examined for deletions or insertions by re-examining the NGS sequencing data or with multiplex ligation-dependent probe amplification (MLPA). These techniques identify a further 5% of variants, and the remaining few changes include deep intronic splicing variants or cases of somatic mosaicism. Where no pathogenic variants are found, the basis for the clinical diagnosis should be reviewed. Genes in which mutations produce similar clinical features to Alport syndrome (resulting in focal and segmental glomerulosclerosis, complement pathway disorders, MYH9-related disorders, etc.) should be examined. NGS approaches have identified novel combinations of pathogenic variants in Alport syndrome. Two variants, with one in COL4A3 and another in COL4A4, produce a more severe phenotype than an uncomplicated heterozygous change. NGS may also identify further coincidental pathogenic variants in genes for podocyte-expressed proteins that also modify the phenotype. Our understanding of the genetics of Alport syndrome is evolving rapidly, and both genetic and non-genetic factors are likely to contribute to the observed phenotypic variability.
Collapse
Affiliation(s)
- Judy Savige
- Department of Medicine, Melbourne and Northern Health, The University of Melbourne, Parkville, VIC, 3050, Australia.
| | | | | | | | | | - Oliver Gross
- Clinic of Nephrology and Rheumatology, University of Gottingen, Gottingen, Germany
| | | | - Frances Flinter
- Department of Clinical Genetics, Guys' and St Thomas' NHS Foundation Trust, London, UK
| | - Jie Ding
- Peking University First Hospital, Beijing, China
| | - Daniel P Gale
- Centre for Nephrology, Royal Free Hospital, University College London, London, UK
| | - Mato Nagel
- Centre for Nephrology and Metabolic Disorders, Weisswasser, Germany
| | - Michael Yau
- Genetics, Guy's Hospital, Viapath, London, UK
| | - Lev Shagam
- Institute of Pediatrics, Pirogov Russian Medical University, Moscow, Russia
| | - Roser Torra
- Inherited Kidney Disorders, Nephrology Department, Fundacio Puigvert, Instituto de Investigacion Carlos III, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Elisabet Ars
- Molecular Biology Laboratory, Fundacio Puigvert, Instituto de Investigacion Carlos III, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Julia Hoefele
- Institute of Human Genetics, Technical University Munich, Munich, Germany
| | - Guido Garosi
- Nephrology, Dialysis and Transplantation, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | |
Collapse
|
39
|
Wen Y, Yang H, Wu J, Wang A, Chen X, Hu S, Zhang Y, Bai D, Jin Z. COL4A2 in the tissue-specific extracellular matrix plays important role on osteogenic differentiation of periodontal ligament stem cells. Am J Cancer Res 2019; 9:4265-4286. [PMID: 31285761 PMCID: PMC6599665 DOI: 10.7150/thno.35914] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 04/26/2019] [Indexed: 02/05/2023] Open
Abstract
Periodontal ligament stem cells (PDLSCs) can repair alveolar bone defects in periodontitis in a microenvironment context-dependent manner. This study aimed to determine whether different extracellular matrices (ECMs) exert diverse effects on osteogenic differentiation of PDLSCs and accurately control alveolar bone defect repair. Methods: The characteristics of PDLSCs and bone marrow mesenchymal stem cells (BMSCs) with respect to surface markers and multi-differentiation ability were determined. Then, we prepared periodontal ligament cells (PDLCs)-derived and bone marrow cells (BMCs)-derived ECMs (P-ECM and B-ECM) and the related decellularized ECMs (dECMs). Transmission electron microscopy (TEM), scanning electron microscopy (SEM), atomic force microscopy (AFM), and protein mass spectrometry were used to distinguish the ECMs. The expression of Type IV collagen A2 (COL4A2) in the ECMs was inhibited by siRNA or activated by lentiviral transduction of relevant cells. The stemness, proliferation, and differentiation of PDLSCs were determined in vitro in different dECMs. For the in vivo analysis, different dECMs under the regulation of COL4A2 mixed with PDLSCs and Bio-Oss bone powder were subcutaneously implanted into immunocompromised mice or in defects in rat alveolar bone. The repair effects were identified by histological or immunohistochemical staining and micro-CT. Results: B-dECM exhibited more compact fibers than P-dECM, as revealed by TEM, SEM, and AFM. Protein mass spectrometry showed that COL4A2 was significantly increased in B-dECM compared with P-dECM. PDLSCs displayed stronger proliferation, stemness, and osteogenic differentiation ability when cultured on B-dECM than P-dECM. Interestingly, B-dECM enhanced the osteogenic differentiation of PDLSCs to a greater extent than P-dECM both in vitro and in vivo, whereas downregulation of COL4A2 in B-dECM showed the opposite results. Furthermore, the classical Wnt/β-catenin pathway was found to play an important role in the negative regulation of osteogenesis through COL4A2, confirmed by experiments with the Wnt inhibitor DKK-1 and the Wnt activator Wnt3a. Conclusion: These findings indicate that COL4A2 in the ECM promotes osteogenic differentiation of PDLSCs through negative regulation of the Wnt/β-catenin pathway, which can be used as a potential therapeutic strategy to repair bone defects.
Collapse
|
40
|
Harjumäki R, Nugroho RWN, Zhang X, Lou YR, Yliperttula M, Valle-Delgado JJ, Österberg M. Quantified forces between HepG2 hepatocarcinoma and WA07 pluripotent stem cells with natural biomaterials correlate with in vitro cell behavior. Sci Rep 2019; 9:7354. [PMID: 31089156 PMCID: PMC6517585 DOI: 10.1038/s41598-019-43669-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/16/2019] [Indexed: 12/20/2022] Open
Abstract
In vitro cell culture or tissue models that mimic in vivo cellular response have potential in tissue engineering and regenerative medicine, and are a more economical and accurate option for drug toxicity tests than animal experimentation. The design of in vivo-like cell culture models should take into account how the cells interact with the surrounding materials and how these interactions affect the cell behavior. Cell-material interactions are furthermore important in cancer metastasis and tumor progression, so deeper understanding of them can support the development of new cancer treatments. Herein, the colloidal probe microscopy technique was used to quantify the interactions of two cell lines (human pluripotent stem cell line WA07 and human hepatocellular carcinoma cell line HepG2) with natural, xeno-free biomaterials of different chemistry, morphology, and origin. Key components of extracellular matrices -human collagens I and IV, and human recombinant laminin-521-, as well as wood-derived, cellulose nanofibrils -with evidenced potential for 3D cell culture and tissue engineering- were analysed. Both strength of adhesion and force curve profiles depended on biomaterial nature and cell characteristics. The successful growth of the cells on a particular biomaterial required cell-biomaterial adhesion energies above 0.23 nJ/m. The information obtained in this work supports the development of new materials or hybrid scaffolds with tuned cell adhesion properties for tissue engineering, and provides a better understanding of the interactions of normal and cancerous cells with biomaterials in the human body.
Collapse
Affiliation(s)
- Riina Harjumäki
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Robertus Wahyu N Nugroho
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| | - Xue Zhang
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| | - Yan-Ru Lou
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Marjo Yliperttula
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, I-35131, Padova, Italy
| | - Juan José Valle-Delgado
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland.
| | - Monika Österberg
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland.
| |
Collapse
|
41
|
Labelle-Dumais C, Schuitema V, Hayashi G, Hoff K, Gong W, Dao DQ, Ullian EM, Oishi P, Margeta M, Gould DB. COL4A1 Mutations Cause Neuromuscular Disease with Tissue-Specific Mechanistic Heterogeneity. Am J Hum Genet 2019; 104:847-860. [PMID: 31051113 DOI: 10.1016/j.ajhg.2019.03.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/05/2019] [Indexed: 01/18/2023] Open
Abstract
Collagen type IV alpha 1 and alpha 2 chains form heterotrimers ([α1(IV)]2α2(IV)) that represent a fundamental basement membrane constituent. Dominant COL4A1 and COL4A2 mutations cause a multisystem disorder that is marked by clinical heterogeneity and variable expressivity and that is generally characterized by the presence of cerebrovascular disease with ocular, renal, and muscular involvement. Despite the fact that muscle pathology is reported in up to one-third of individuals with COL4A1 and COL4A2 mutations and in animal models with mutations in COL4A1 and COL4A2 orthologs, the pathophysiological mechanisms underlying COL4A1-related myopathy are unknown. In general, mutations are thought to impair [α1(IV)]2α2(IV) secretion. Whether pathogenesis results from intracellular retention, extracellular deficiency, or the presence of mutant proteins in basement membranes represents an important gap in knowledge and a major obstacle for developing targeted interventions. We report that Col4a1 mutant mice develop progressive neuromuscular pathology that models human disease. We demonstrate that independent muscular, neural, and vascular insults contribute to neuromyopathy and that there is mechanistic heterogeneity among tissues. Importantly, we provide evidence of a COL4A1 functional subdomain with disproportionate significance for tissue-specific pathology and demonstrate that a potential therapeutic strategy aimed at promoting [α1(IV)]2α2(IV) secretion can ameliorate or exacerbate myopathy in a mutation-dependent manner. These data have important translational implications for prediction of clinical outcomes based on genotype, development of mechanism-based interventions, and genetic stratification for clinical trials. Collectively, our data underscore the importance of the [α1(IV)]2α2(IV) network as a multifunctional signaling platform and show that allelic and tissue-specific mechanistic heterogeneities contribute to the variable expressivity of COL4A1 and COL4A2 mutations.
Collapse
|
42
|
Advances in molecular diagnosis and therapeutics in nephrotic syndrome and focal and segmental glomerulosclerosis. Curr Opin Nephrol Hypertens 2019; 27:194-200. [PMID: 29465426 DOI: 10.1097/mnh.0000000000000408] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW The widespread adoption of next-generation sequencing by research and clinical laboratories has begun to uncover the previously unknown genetic basis of many diseases. In nephrology, one of the best examples of this is seen in focal and segmental glomerulosclerosis (FSGS) and nephrotic syndrome. We review advances made in 2017 as a result of human and molecular genetic studies as it relates to FSGS and nephrotic syndrome. RECENT FINDINGS There are more than 50 monogenic genes described in steroid-resistant nephrotic syndrome and FSGS, with seven reported in 2017. In individuals presenting with FSGS or nephrotic syndrome before or at the age of 18 years, the commonest genes in which a mutation is found continues to be limited to only a few including NPHS1 and NPHS2 based on multiple studies. For FSGS or nephrotic syndrome that presents after 18 years, mutations in COl4A3/4/5, traditionally associated with Alport syndrome, are increasingly being reported. Despite the extensive genetic heterogeneity in FSGS, there is evidence that some of these genes converge onto common pathways. There are also reports of in-vivo models exploring apolipoprotein 1 biology, variants in which account for part of the increased risk of nondiabetic kidney disease in African-Americans. Finally, genetic testing has several clinical uses including clarification of diagnosis and treatment; identification of suitable young biologic relatives for kidney donation; and preimplantation genetic diagnosis. CRISPR gene editing is currently an experimental tool only, but the recent reports of excising mutations in embryos could be a therapeutic option for individuals with any monogenic disorder in the future. SUMMARY Sequencing efforts are bringing novel variants into investigation and directing the efforts to understand how these lead to disease phenotypes. Expanding our understanding of the genetic basis of health and disease processes is the necessary first step to elaborate the repertoire of therapeutic agents available for patients with FSGS and nephrotic syndrome.
Collapse
|
43
|
Scimone MT, Cramer III HC, Bar-Kochba E, Amezcua R, Estrada JB, Franck C. Modular approach for resolving and mapping complex neural and other cellular structures and their associated deformation fields in three dimensions. Nat Protoc 2018; 13:3042-3064. [DOI: 10.1038/s41596-018-0077-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Zhang W, Yu X, Li Y, Su Z, Jandt KD, Wei G. Protein-mimetic peptide nanofibers: Motif design, self-assembly synthesis, and sequence-specific biomedical applications. Prog Polym Sci 2018. [DOI: 10.1016/j.progpolymsci.2017.12.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
45
|
Fidler AL, Boudko SP, Rokas A, Hudson BG. The triple helix of collagens - an ancient protein structure that enabled animal multicellularity and tissue evolution. J Cell Sci 2018; 131:jcs203950. [PMID: 29632050 PMCID: PMC5963836 DOI: 10.1242/jcs.203950] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The cellular microenvironment, characterized by an extracellular matrix (ECM), played an essential role in the transition from unicellularity to multicellularity in animals (metazoans), and in the subsequent evolution of diverse animal tissues and organs. A major ECM component are members of the collagen superfamily -comprising 28 types in vertebrates - that exist in diverse supramolecular assemblies ranging from networks to fibrils. Each assembly is characterized by a hallmark feature, a protein structure called a triple helix. A current gap in knowledge is understanding the mechanisms of how the triple helix encodes and utilizes information in building scaffolds on the outside of cells. Type IV collagen, recently revealed as the evolutionarily most ancient member of the collagen superfamily, serves as an archetype for a fresh view of fundamental structural features of a triple helix that underlie the diversity of biological activities of collagens. In this Opinion, we argue that the triple helix is a protein structure of fundamental importance in building the extracellular matrix, which enabled animal multicellularity and tissue evolution.
Collapse
Affiliation(s)
- Aaron L Fidler
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sergei P Boudko
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Antonis Rokas
- Department of Biological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Billy G Hudson
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Medical Education and Administration, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| |
Collapse
|
46
|
Montgomery NT, Zientek KD, Pokidysheva EN, Bächinger HP. Post-translational modification of type IV collagen with 3-hydroxyproline affects its interactions with glycoprotein VI and nidogens 1 and 2. J Biol Chem 2018; 293:5987-5999. [PMID: 29491144 DOI: 10.1074/jbc.ra117.000406] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 02/15/2018] [Indexed: 01/18/2023] Open
Abstract
Type IV collagen is a major component of the basement membrane and interacts with numerous other basement membrane proteins. Many of these interactions are poorly characterized. Type IV collagen is abundantly post-translationally modified with 3-hydroxyproline (3-Hyp), but 3-Hyp's biochemical role in type IV collagen's interactions with other proteins is not well established. In this work, we present binding data consistent with a major role of 3-Hyp in interactions of collagen IV with glycoprotein VI and nidogens 1 and 2. The increased binding interaction between type IV collagen without 3-Hyp and glycoprotein VI has been the subject of some controversy, which we sought to explore, whereas the lack of binding of nidogens to type IV collagen without 3-Hyp is novel. Using tandem MS, we show that the putative glycoprotein VI-binding site is 3-Hyp-modified in WT PFHR-9 type IV collagen, but not in PFHR-9 cells in which prolyl-3-hydroxylase 2 (P3H2) has been knocked out (KO). Moreover, we observed altered 3-Hyp occupancy across many other sites. Using amino acid analysis of type IV collagen from the WT and P3H2 KO cell lines, we confirm that P3H2 is the major, but not the only 3-Hyp-modifying enzyme of type IV collagen. These findings underscore the importance of post-translational modifications of type IV collagen for interactions with other proteins.
Collapse
Affiliation(s)
- Nathan T Montgomery
- From the Research Department, Shriners Hospital for Children, Portland, Oregon 97239.,the Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon 97239, and
| | - Keith D Zientek
- From the Research Department, Shriners Hospital for Children, Portland, Oregon 97239
| | - Elena N Pokidysheva
- the Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University, Nashville, Tennessee 37232
| | - Hans Peter Bächinger
- From the Research Department, Shriners Hospital for Children, Portland, Oregon 97239, .,the Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon 97239, and
| |
Collapse
|
47
|
JingSong H, Hong G, Yang J, Duo Z, Li F, WeiCai C, XueYing L, YouSheng M, YiWen O, Yue P, Zou C. siRNA-mediated suppression of collagen type iv alpha 2 (COL4A2) mRNA inhibits triple-negative breast cancer cell proliferation and migration. Oncotarget 2018; 8:2585-2593. [PMID: 27906681 PMCID: PMC5356825 DOI: 10.18632/oncotarget.13716] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 11/21/2016] [Indexed: 01/18/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is more aggressive than other breast cancer subtypes. Collagen type IV alpha 2 (COL4A2), a major component of the basement membrane, dynamically influences a wide range of biological processes, including cancer pathogenesis and progression. This study evaluated the effects of COL4A2 siRNA delivered by lentiviral vector to TNBC cells. COL4A2 siRNA lenti-viral vector was constructed and transfected into MDA-MB-231 and MDA-MB-468 cells. The COL4A2 mRNA levels were quantified by RT-PCR. CCK8 assay was performed to evaluate cell proliferation and migration. Cell migration and invasion assays were carried out using Transwell. Cell apoptosis and cell cycle analyses were conducted using flow cytometric approach. We found that COL4A2 mRNA levels were significantly down-regulated in MDA-MB-231 and MDA-MB-468 cells after transfection with COL4A2 siRNA. Furthermore, cell migration and proliferation were significantly decreased and the cell cycle was arrested. Our results indicated that COL4A2 siRNA significantly suppresses the migration and proliferation of TNBC cells. Inhibition of COL4A2 may be a new target for the prevention and treatment of TNBC.
Collapse
Affiliation(s)
- He JingSong
- Department of Breast Surgery, The First Affiliated Hospital of Shenzhen University, Second People's Hospital of Shen Zhen, Shen Zhen, 518035, China
| | - Guan Hong
- Department of Pathology, The First Affiliated Hospital of Shenzhen University, Second People's Hospital of Shen Zhen, Shen Zhen 518035, China
| | - Jianbo Yang
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, University of Minnesota, UMN Twin Cities, MN 55455, USA
| | - Zheng Duo
- Shenzhen Key Laboratory of Translational Medicine of Tumor, Department of Cell Biology and Genetics, School of Medicine, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Fu Li
- Department of Pharmacology, Shenzhen Key Laboratory of Translational Medicine of Tumor and Cancer Research Centre, School of Medicine, Shenzhen University, Shenzhen, 518060 China
| | - Chen WeiCai
- Department of Breast Surgery, The First Affiliated Hospital of Shenzhen University, Second People's Hospital of Shen Zhen, Shen Zhen, 518035, China
| | - Luo XueYing
- Department of Breast Surgery, The First Affiliated Hospital of Shenzhen University, Second People's Hospital of Shen Zhen, Shen Zhen, 518035, China
| | - Mao YouSheng
- Department of Breast Surgery, The First Affiliated Hospital of Shenzhen University, Second People's Hospital of Shen Zhen, Shen Zhen, 518035, China
| | - OuYang YiWen
- Department of Breast Surgery, The First Affiliated Hospital of Shenzhen University, Second People's Hospital of Shen Zhen, Shen Zhen, 518035, China
| | - Pan Yue
- Department of Breast Surgery, The First Affiliated Hospital of Shenzhen University, Second People's Hospital of Shen Zhen, Shen Zhen, 518035, China
| | - Chang Zou
- Clinical Medical Research Center, Shen Zhen People's Hospital, The Second Clinical Medical College of Jinan University, 518020, China
| |
Collapse
|
48
|
Barua M, John R, Stella L, Li W, Roslin NM, Sharif B, Hack S, Lajoie-Starkell G, Schwaderer AL, Becknell B, Wuttke M, Köttgen A, Cattran D, Paterson AD, Pei Y. X-Linked Glomerulopathy Due to COL4A5 Founder Variant. Am J Kidney Dis 2017; 71:441-445. [PMID: 29198386 DOI: 10.1053/j.ajkd.2017.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 09/01/2017] [Indexed: 12/30/2022]
Abstract
Alport syndrome is a rare hereditary disorder caused by rare variants in 1 of 3 genes encoding for type IV collagen. Rare variants in COL4A5 on chromosome Xq22 cause X-linked Alport syndrome, which accounts for ∼80% of the cases. Alport syndrome has a variable clinical presentation, including progressive kidney failure, hearing loss, and ocular defects. Exome sequencing performed in 2 affected related males with an undefined X-linked glomerulopathy characterized by global and segmental glomerulosclerosis, mesangial hypercellularity, and vague basement membrane immune complex deposition revealed a COL4A5 sequence variant, a substitution of a thymine by a guanine at nucleotide 665 (c.T665G; rs281874761) of the coding DNA predicted to lead to a cysteine to phenylalanine substitution at amino acid 222, which was not seen in databases cataloguing natural human genetic variation, including dbSNP138, 1000 Genomes Project release version 01-11-2004, Exome Sequencing Project 21-06-2014, or ExAC 01-11-2014. Review of the literature identified 2 additional families with the same COL4A5 variant leading to similar atypical histopathologic features, suggesting a unique pathologic mechanism initiated by this specific rare variant. Homology modeling suggests that the substitution alters the structural and dynamic properties of the type IV collagen trimer. Genetic analysis comparing members of the 3 families indicated a distant relationship with a shared haplotype, implying a founder effect.
Collapse
Affiliation(s)
- Moumita Barua
- Division of Nephrology, University Health Network, Toronto, Canada; Toronto General Research Institute, Toronto General Hospital, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Canada.
| | - Rohan John
- Department of Pathology, University Health Network, Toronto, Canada
| | - Lorenzo Stella
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Weili Li
- The Centre for Applied Genomics, Hospital for Sick Children's, Brampton, Canada
| | - Nicole M Roslin
- The Centre for Applied Genomics, Hospital for Sick Children's, Brampton, Canada
| | - Bedra Sharif
- Division of Nephrology, University Health Network, Toronto, Canada
| | - Saidah Hack
- Division of Nephrology, University Health Network, Toronto, Canada
| | - Ginette Lajoie-Starkell
- William Osler Health System, Brampton, Canada; Department of Pathobiology and Laboratory Medicine, University of Toronto, Toronto, Canada
| | - Andrew L Schwaderer
- Nephrology Section, Nationwide Children's Hospital, Columbus, OH; Department of Pediatrics, Ohio State University Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Brian Becknell
- Nephrology Section, Nationwide Children's Hospital, Columbus, OH; Department of Pediatrics, Ohio State University Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Medical Center and Faculty of Medicine - University of Freiburg, Freiburg, Germany; Division of Nephrology, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Medical Center and Faculty of Medicine - University of Freiburg, Freiburg, Germany; Division of Nephrology, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Daniel Cattran
- Division of Nephrology, University Health Network, Toronto, Canada; Toronto General Research Institute, Toronto General Hospital, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada
| | - Andrew D Paterson
- Institute of Medical Sciences, University of Toronto, Toronto, Canada; The Centre for Applied Genomics, Hospital for Sick Children's, Brampton, Canada; Epidemiology & Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - York Pei
- Division of Nephrology, University Health Network, Toronto, Canada; Toronto General Research Institute, Toronto General Hospital, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Canada
| |
Collapse
|
49
|
Brown KL, Cummings CF, Vanacore RM, Hudson BG. Building collagen IV smart scaffolds on the outside of cells. Protein Sci 2017; 26:2151-2161. [PMID: 28845540 DOI: 10.1002/pro.3283] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/22/2022]
Abstract
Collagen IV scaffolds assemble through an intricate pathway that begins intracellularly and is completed extracellularly. Multiple intracellular enzymes act in concert to assemble collagen IV protomers, the building blocks of collagen IV scaffolds. After being secreted from cells, protomers are activated to initiate oligomerization, forming insoluble networks that are structurally reinforced with covalent crosslinks. Within these networks, embedded binding sites along the length of the protomer lead to the "decoration" of collagen IV triple helix with numerous functional molecules. We refer to these networks as "smart" scaffolds, which as a component of the basement membrane enable the development and function of multicellular tissues in all animal phyla. In this review, we present key molecular mechanisms that drive the assembly of collagen IV smart scaffolds.
Collapse
Affiliation(s)
- Kyle L Brown
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232.,Center for Structural Biology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232.,Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232
| | | | - Roberto M Vanacore
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232.,Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232
| | - Billy G Hudson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232.,Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232.,Department of Biochemistry, Vanderbilt University Medical Center, Nashville, Tennessee, 37232.,Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232
| |
Collapse
|
50
|
Tong Z, Martyn K, Yang A, Yin X, Mead BE, Joshi N, Sherman NE, Langer RS, Karp JM. Towards a defined ECM and small molecule based monolayer culture system for the expansion of mouse and human intestinal stem cells. Biomaterials 2017; 154:60-73. [PMID: 29120819 DOI: 10.1016/j.biomaterials.2017.10.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 10/21/2017] [Accepted: 10/22/2017] [Indexed: 01/04/2023]
Abstract
Current ISC culture systems face significant challenges such as animal-derived or undefined matrix compositions, batch-to-batch variability (e.g. Matrigel-based organoid culture), and complexity of assaying cell aggregates such as organoids which renders the research and clinical translation of ISCs challenging. Here, through screening for suitable ECM components, we report a defined, collagen based monolayer culture system that supports the growth of mouse and human intestinal epithelial cells (IECs) enriched for an Lgr5+ population comparable or higher to the levels found in a standard Matrigel-based organoid culture. The system, referred to as the Bolstering Lgr5 Transformational (BLT) Sandwich culture, comprises a collagen IV-coated porous substrate and a collagen I gel overlay which sandwich an IEC monolayer in between. The distinct collagen cues synergistically regulate IEC attachment, proliferation, and Lgr5 expression through maximizing the engagement of distinct cell surface adhesion receptors (i.e. integrin α2β1, integrin β4) and cell polarity. Further, we apply our BLT Sandwich system to identify that the addition of a bone morphogenetic protein (BMP) receptor inhibitor (LDN-193189) improves the expansion of Lgr5-GFP+ cells from mouse small intestinal crypts by nearly 2.5-fold. Notably, the BLT Sandwich culture is capable of expanding human-derived IECs with higher LGR5 mRNA levels than conventional Matrigel culture, providing superior expansion of human LGR5+ ISCs. Considering the key roles Lgr5+ ISCs play in intestinal epithelial homeostasis and regeneration, we envision that our BLT Sandwich culture system holds great potential for understanding and manipulating ISC biology in vitro (e.g. for modeling ISC-mediated gut diseases) or for expanding a large number of ISCs for clinical utility (e.g. for stem cell therapy).
Collapse
Affiliation(s)
- Zhixiang Tong
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States
| | - Keir Martyn
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States
| | - Andy Yang
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States
| | - Xiaolei Yin
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States; David H. Koch Institute for Integrative Cancer Research at MIT, United States
| | - Benjamin E Mead
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States; Broad Institute of Harvard and MIT, United States; David H. Koch Institute for Integrative Cancer Research at MIT, United States
| | - Nitin Joshi
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States
| | - Nicholas E Sherman
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States
| | - Robert S Langer
- Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States; Department of Chemical Engineering at MIT, United States; David H. Koch Institute for Integrative Cancer Research at MIT, United States
| | - Jeffrey M Karp
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States.
| |
Collapse
|