1
|
Fujiwara H. Dynamic duo: Cell-extracellular matrix interactions in hair follicle development and regeneration. Dev Biol 2024; 516:20-34. [PMID: 39059679 DOI: 10.1016/j.ydbio.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 06/20/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Ectodermal organs, such as hair follicles, originate from simple epithelial and mesenchymal sheets through a complex developmental process driven by interactions between these cell types. This process involves dermal condensation, placode formation, bud morphogenesis, and organogenesis, and all of these processes require intricate interactions among various tissues. Recent research has emphasized the crucial role of reciprocal and dynamic interactions between cells and the extracellular matrix (ECM), referred to as the "dynamic duo", in the development of ectodermal organs. These interactions provide spatially and temporally changing biophysical and biochemical cues within tissues. Using the hair follicle as an example, this review highlights two types of cell-ECM adhesion units-focal adhesion-type and hemidesmosome-type adhesion units-that facilitate communication between epithelial and mesenchymal cells. This review further explores how these adhesion units, along with other cell-ECM interactions, evolve during hair follicle development and regeneration, underscoring their importance in guiding both developmental and regenerative processes.
Collapse
|
2
|
Gao G, Zhou Z. Isthmin-1: A critical regulator of branching morphogenesis and metanephric mesenchyme condensation during early kidney development. Bioessays 2024; 46:e2300189. [PMID: 38161234 DOI: 10.1002/bies.202300189] [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: 10/02/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Isthmin-1 (Ism1) was first described to be syn-expressed with Fgf8 in Xenopus. However, its biological role has not been elucidated until recent years. Despite of accumulated evidence that Ism1 participates in angiogenesis, tumor invasion, macrophage apoptosis, and glucose metabolism, the cognate receptors for Ism1 remain largely unknown. Ism1 deficiency in mice results in renal agenesis (RA) with a transient loss of Gdnf transcription and impaired mesenchyme condensation at E11.5. Ism1 binds to and activates Integrin α8β1 to positively regulate Gdnf/Ret signaling, thus promoting mesenchyme condensation and ureteric epithelium branching morphogenesis. Here, we propose the hypothesis underlying the mechanism by which Ism1 regulates branching morphogenesis during early kidney development.
Collapse
Affiliation(s)
- Ge Gao
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhongjun Zhou
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Reproductive Medical Center, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
3
|
Esho T, Kobbe B, Tufa S, Keene D, Paulsson M, Wagener R. The Fraser Complex Proteins (Frem1, Frem2, and Fras1) Can Form Anchoring Cords in the Absence of AMACO at the Dermal–Epidermal Junction of Mouse Skin. Int J Mol Sci 2023; 24:ijms24076782. [PMID: 37047755 PMCID: PMC10095167 DOI: 10.3390/ijms24076782] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/29/2023] [Accepted: 04/01/2023] [Indexed: 04/09/2023] Open
Abstract
AMACO (VWA2 protein), secreted by epithelial cells, is strongly expressed at basement membranes when budding or invagination occurs in embryos. In skin, AMACO associates with proteins of the Fraser complex, which form anchoring cords. These, during development, temporally stabilize the dermal–epidermal junction, pending the formation of collagen VII-containing anchoring fibrils. Fraser syndrome in humans results if any of the core members of the Fraser complex (Fras1, Frem1, Frem2) are mutated. Fraser syndrome is characterized by subepidermal blistering, cryptophthalmos, and syndactyly. In an attempt to determine AMACO function, we generated and characterized AMACO-deficient mice. In contrast to Fraser complex mutant mice, AMACO-deficient animals lack an obvious phenotype. The mutually interdependent basement membrane deposition of the Fraser complex proteins, and the formation of anchoring cords, are not affected. Furthermore, hair follicle development in newborn AMACO-deficient mice showed no gross aberration. Surprisingly, it appears that, while AMACO is a component of the anchoring cords, it is not essential for their formation or function.
Collapse
Affiliation(s)
- Temitope Esho
- Center for Biochemistry, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Birgit Kobbe
- Center for Biochemistry, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Sara Tufa
- Micro-Imaging Center, Shriners Children’s, Portland, OR 97239, USA
| | - Douglas Keene
- Micro-Imaging Center, Shriners Children’s, Portland, OR 97239, USA
| | - Mats Paulsson
- Center for Biochemistry, Medical Faculty, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, 50931 Cologne, Germany
- Cologne Center for Musculoskeletal Biomechanics, 50931 Cologne, Germany
| | - Raimund Wagener
- Center for Biochemistry, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
4
|
Kantaputra PN, Tripuwabhrut K, Anthonappa RP, Chintakanon K, Ngamphiw C, Adisornkanj P, Porntrakulseree N, Olsen B, Intachai W, Hennekam RC, Vieira AR, Tongsima S. Heterozygous Variants in FREM2 Are Associated with Mesiodens, Supernumerary Teeth, Oral Exostoses, and Odontomas. Diagnostics (Basel) 2023; 13:diagnostics13071214. [PMID: 37046432 PMCID: PMC10093539 DOI: 10.3390/diagnostics13071214] [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: 02/13/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Supernumerary teeth refer to extra teeth that exceed the usual number of dentitions. A mesiodens is a particular form of supernumerary tooth, which is located in the premaxilla region. The objective of the study was to investigate the genetic etiology of extra tooth phenotypes, including mesiodens and isolated supernumerary teeth. METHODS Oral and radiographic examinations and whole-exome sequencing were performed on every patient in our cohort of 122 patients, including 27 patients with isolated supernumerary teeth and 94 patients with mesiodens. A patient who had multiple supernumerary teeth also had odontomas. RESULTS We identified a novel (c.8498A>G; p.Asn2833Ser) and six recurrent (c.1603C>T; p.Arg535Cys, c.5852G>A; p.Arg1951His, c.6949A>T; p.Thr2317Ser; c.1549G>A; p.Val517Met, c.1921A>G; p.Thr641Ala, and c.850G>C; p.Val284Leu) heterozygous missense variants in FREM2 in eight patients with extra tooth phenotypes. CONCLUSIONS Biallelic variants in FREM2 are implicated in autosomal recessive Fraser syndrome with or without dental anomalies. Here, we report for the first time that heterozygous carriers of FREM2 variants have phenotypes including oral exostoses, mesiodens, and isolated supernumerary teeth.
Collapse
Affiliation(s)
- Piranit Nik Kantaputra
- Center of Excellence in Medical Genetics Research, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
- Division of Pediatric Dentistry, Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kanich Tripuwabhrut
- Division of Orthodontics, Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Robert P Anthonappa
- Department of Pediatric Dentistry, University of Western Australia Dental School, Nedlands, WA 6009, Australia
| | - Kanoknart Chintakanon
- Division of Orthodontics, Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chumpol Ngamphiw
- National Biobank of Thailand, National Science and Technology Development Agency, Khlong Luang 12120, Thailand
| | - Ploy Adisornkanj
- Center of Excellence in Medical Genetics Research, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
- Division of Pediatric Dentistry, Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
- Dental Department, Sawang Daen Din Crown Prince Hospital, Sakon Nakhon 47110, Thailand
| | | | - Bjorn Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard University, Boston, MA 02115, USA
| | - Worrachet Intachai
- Center of Excellence in Medical Genetics Research, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Raoul C Hennekam
- Department of Pediatrics, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Alexandre R Vieira
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sissades Tongsima
- National Biobank of Thailand, National Science and Technology Development Agency, Khlong Luang 12120, Thailand
| |
Collapse
|
5
|
Wilson CL, Hung CF, Burkel BM, Ponik SM, Gharib SA, Schnapp LM. Nephronectin is required to maintain right lung lobar separation during embryonic development. Am J Physiol Lung Cell Mol Physiol 2023; 324:L335-L344. [PMID: 36719987 PMCID: PMC10027138 DOI: 10.1152/ajplung.00505.2021] [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: 12/15/2021] [Revised: 11/30/2022] [Accepted: 01/25/2023] [Indexed: 02/02/2023] Open
Abstract
Nephronectin (NPNT) is a basement membrane (BM) protein and high-affinity ligand of integrin α8β1 that is required for kidney morphogenesis in mice. In the lung, NPNT also localizes to BMs, but its potential role in pulmonary development has not been investigated. Mice with a floxed Npnt allele were used to generate global knockouts (KOs). Staged embryos were obtained by timed matings of heterozygotes and lungs were isolated for analysis. Although primary and secondary lung bud formation was normal in KO embryos, fusion of right lung lobes, primarily the medial and caudal, was first detected at E13.5 and persisted into adulthood. The lung parenchyma of KO mice was indistinguishable from wild-type (WT) and lobe fusion did not alter respiratory mechanics in adult KO mice. Interrogation of an existing single-cell RNA-seq atlas of embryonic and adult mouse lungs identified Npnt transcripts in mesothelial cells at E12.5 and into the early postnatal period, but not in adult lungs. KO embryonic lungs exhibited increased expression of laminin α5 and deposition of collagen IV in the mesothelial BM, accompanied by abnormalities in collagen fibrils in the adjacent stroma. Cranial and accessory lobes extracted from KO embryonic lungs fused ex vivo when cultured in juxtaposition, with the area of fusion showing loss of the mesothelial marker Wilms tumor 1. Because a similar pattern of lobe fusion was previously observed in integrin α8 KO embryos, our results suggest that NPNT signaling through integrin α8, likely in the visceral pleura, maintains right lung lobe separation during embryogenesis.
Collapse
Affiliation(s)
- Carole L Wilson
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States
| | - Chi F Hung
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Brian M Burkel
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, Wisconsin, United States
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, Wisconsin, United States
| | - Sina A Gharib
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Lynn M Schnapp
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States
| |
Collapse
|
6
|
Ma X, Liu A, Tian S. A meta-analysis of mRNA expression profiling studies in sheep with different FecB genotypes. Anim Genet 2023; 54:225-238. [PMID: 36811249 DOI: 10.1111/age.13304] [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: 01/26/2023] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/24/2023]
Abstract
The FecB mutation in the sheep BMPRIB is strongly correlated with high ovulation traits but its mechanism remains unclear. This study explored differentially expressed genes (DEGs) and their associated molecular mechanisms that may be involved in FecB mutation-induced high ovulation from the perspective of the hypothalamic-pituitary-gonadal (HPG) axis by conducting a systematic review and meta-analysis. The PubMed, EMBASE, CNKI, WanFang, and CBM databases were searched for eligible articles published before August 2022, focusing on mRNA sequencing of different tissues in the HPG axis in sheep with different FecB genotypes. A total of 6555 DEGs were identified from the analysis of six published articles and experimental results from our laboratory. The DEGs were screened by vote-counting rank and robust rank aggregation. Among these, in the follicular phase, FKBP5, CDCA7 and CRABP1 were upregulated in the hypothalamus. INSM2 was upregulated, while LDB3 was downregulated in the pituitary. CLU, SERPINA14, PENK, INHA and STAR were upregulated, while FERMT2 and NPY1R were downregulated in the ovary. On the HPG axis, TAC1 was upregulated and NPNT was downregulated. Many DEGs were found in sheep with different FecB genotypes. The genes FKBP5, CDCA7, CRABP1, INSM2, LDB3, CLU, SERPINA14, PENK, INHA, STAR, FERMT2, NPY1R, TAC1 and NPNT, may be associated with FecB mutation-induced high ovulation in different tissues. These candidate genes will further improve the mechanism of multiple fertility traits induced by the FecB mutation from the perspective of the HPG axis.
Collapse
Affiliation(s)
- Xiaofei Ma
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, Hebei, China
| | - Aiju Liu
- Department of Agricultural and Animal Husbandry Engineering, Cangzhou Technical College, Cangzhou, Hebei, China
| | - Shujun Tian
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, Hebei, China
| |
Collapse
|
7
|
Wang G, Wang Z, Lu H, Zhao Z, Guo L, Kong F, Wang A, Zhao S. Comprehensive analysis of FRAS1/FREM family as potential biomarkers and therapeutic targets in renal clear cell carcinoma. Front Pharmacol 2022; 13:972934. [PMID: 36249757 PMCID: PMC9558830 DOI: 10.3389/fphar.2022.972934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Background: FRAS1 (Fraser syndrome protein 1), together with FREM1 (the Fras1-related extracellular matrix proteins 1) and FREM2, belonging to the FRAS1/FREM extracellular matrix protein family, are considered to play essential roles in renal organogenesis and cancer progression. However, their roles in kidney renal clear cell carcinoma (KIRC) remain to be elucidated. Methods: FRAS1/FREM RNA expression analysis was performed using TCGA/GTEx databases, and valided using GEO databases and real-time PCR. Protein expression was peformed using CPTAC databases. Herein, we employed an array of bioinformatics methods and online databases to explore the potential oncogenic roles of FRAS1/FREM in KIRC. Results: We found that FRAS1, FREM1 and FREM2 genes and proteins expression levels were significantly decreased in KIRC tissues than in normal tissues. Decreased FRAS1/FREM expression levels were significantly associated with advanced clinicopathological parameters (pathological stage, grade and tumor metastasis status). Notably, the patients with decreased FRAS1/FREM2 expression showed a high propensity for metastasis and poor prognosis. FRAS1/FREM were correlated with various immune infiltrating cells, especially CD4+ T cells and its corresponding subsets (Th1, Th2, Tfh and Tregs). FRAS1 and FREM2 had association with DNA methylation and their single CpG methylation levels were associated with prognosis. Moreover, FRAS1/FREM might exert antitumor effects by functioning in key oncogenic signalling pathways and metabolic pathways. Drug sensitivity analysis indicated that high FRAS1 and FREM2 expression can be a reliable predictor of targeted therapeutic drug response, highlighting the potential as anticancer drug targets. Conclusion: Together, our results indicated that FRAS1/FREM family members could be potential therapeutic targets and valuable prognostic biomarkers of KIRC.
Collapse
Affiliation(s)
- Ganggang Wang
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Urology, Maternal and Child Health Care Hospital of Shandong Province, Shandong University, Jinan, Shandong, China
| | - Zheng Wang
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Haiquan Lu
- Advanced Medical Research Institute and Key Laboratory for Experimental Teratology of the Ministry of Education, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhiqun Zhao
- Advanced Medical Research Institute and Key Laboratory for Experimental Teratology of the Ministry of Education, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Liqiang Guo
- Department of Urology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Feng Kong
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Aizhen Wang
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Shengtian Zhao
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Binzhou Medical University, Binzhou, Shandong, China
| |
Collapse
|
8
|
Connaughton DM, Hildebrandt F. Disease mechanisms of monogenic congenital anomalies of the kidney and urinary tract American Journal of Medical Genetics Part C. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:325-343. [PMID: 36208064 PMCID: PMC9618346 DOI: 10.1002/ajmg.c.32006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/14/2022] [Accepted: 09/16/2022] [Indexed: 11/05/2022]
Abstract
Congenital Anomalies of the Kidney and Urinary Tract (CAKUT) is a developmental disorder of the kidney and/or genito-urinary tract that results in end stage kidney disease (ESKD) in up to 50% of children. Despite the congenital nature of the disease, CAKUT accounts for almost 10% of adult onset ESKD. Multiple lines of evidence suggest that CAKUT is a Mendelian disorder, including the observation of familial clustering of CAKUT. Pathogenesis in CAKUT is embryonic in origin, with disturbances of kidney and urinary tract development resulting in a heterogeneous range of disease phenotypes. Despite polygenic and environmental factors being implicated, a significant proportion of CAKUT is monogenic in origin, with studies demonstrating single gene defects in 10%-20% of patients with CAKUT. Here, we review monogenic disease causation with emphasis on the etiological role of gene developmental pathways in CAKUT.
Collapse
Affiliation(s)
- Dervla M Connaughton
- Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Medicine, Division of Nephrology, London Health Sciences Centre, London, Ontario, Canada
| | - Friedhelm Hildebrandt
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Comparative whole-genome transcriptome analysis in renal cell populations reveals high tissue specificity of MAPK/ERK targets in embryonic kidney. BMC Biol 2022; 20:112. [PMID: 35550069 PMCID: PMC9102746 DOI: 10.1186/s12915-022-01309-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 04/25/2022] [Indexed: 12/19/2022] Open
Abstract
Background MAPK/ERK signaling is a well-known mediator of extracellular stimuli controlling intracellular responses to growth factors and mechanical cues. The critical requirement of MAPK/ERK signaling for embryonic stem cell maintenance is demonstrated, but specific functions in progenitor regulation during embryonic development, and in particular kidney development remain largely unexplored. We previously demonstrated MAPK/ERK signaling as a key regulator of kidney growth through branching morphogenesis and normal nephrogenesis where it also regulates progenitor expansion. Here, we performed RNA sequencing-based whole-genome expression analysis to identify transcriptional MAPK/ERK targets in two distinct renal populations: the ureteric bud epithelium and the nephron progenitors. Results Our analysis revealed a large number (5053) of differentially expressed genes (DEGs) in nephron progenitors and significantly less (1004) in ureteric bud epithelium, reflecting likely heterogenicity of cell types. The data analysis identified high tissue-specificity, as only a fraction (362) of MAPK/ERK targets are shared between the two tissues. Tissue-specific MAPK/ERK targets participate in the regulation of mitochondrial energy metabolism in nephron progenitors, which fail to maintain normal mitochondria numbers in the MAPK/ERK-deficient tissue. In the ureteric bud epithelium, a dramatic decline in progenitor-specific gene expression was detected with a simultaneous increase in differentiation-associated genes, which was not observed in nephron progenitors. Our experiments in the genetic model of MAPK/ERK deficiency provide evidence that MAPK/ERK signaling in the ureteric bud maintains epithelial cells in an undifferentiated state. Interestingly, the transcriptional targets shared between the two tissues studied are over-represented by histone genes, suggesting that MAPK/ERK signaling regulates cell cycle progression and stem cell maintenance through chromosome condensation and nucleosome assembly. Conclusions Using tissue-specific MAPK/ERK inactivation and RNA sequencing in combination with experimentation in embryonic kidneys, we demonstrate here that MAPK/ERK signaling maintains ureteric bud tip cells, suggesting a regulatory role in collecting duct progenitors. We additionally deliver new mechanistic information on how MAPK/ERK signaling regulates progenitor maintenance through its effects on chromatin accessibility and energy metabolism. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01309-z.
Collapse
|
10
|
Eraslan G, Drokhlyansky E, Anand S, Fiskin E, Subramanian A, Slyper M, Wang J, Van Wittenberghe N, Rouhana JM, Waldman J, Ashenberg O, Lek M, Dionne D, Win TS, Cuoco MS, Kuksenko O, Tsankov AM, Branton PA, Marshall JL, Greka A, Getz G, Segrè AV, Aguet F, Rozenblatt-Rosen O, Ardlie KG, Regev A. Single-nucleus cross-tissue molecular reference maps toward understanding disease gene function. Science 2022; 376:eabl4290. [PMID: 35549429 PMCID: PMC9383269 DOI: 10.1126/science.abl4290] [Citation(s) in RCA: 167] [Impact Index Per Article: 83.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding gene function and regulation in homeostasis and disease requires knowledge of the cellular and tissue contexts in which genes are expressed. Here, we applied four single-nucleus RNA sequencing methods to eight diverse, archived, frozen tissue types from 16 donors and 25 samples, generating a cross-tissue atlas of 209,126 nuclei profiles, which we integrated across tissues, donors, and laboratory methods with a conditional variational autoencoder. Using the resulting cross-tissue atlas, we highlight shared and tissue-specific features of tissue-resident cell populations; identify cell types that might contribute to neuromuscular, metabolic, and immune components of monogenic diseases and the biological processes involved in their pathology; and determine cell types and gene modules that might underlie disease mechanisms for complex traits analyzed by genome-wide association studies.
Collapse
Affiliation(s)
- Gökcen Eraslan
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Eugene Drokhlyansky
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shankara Anand
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Evgenij Fiskin
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ayshwarya Subramanian
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michal Slyper
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jiali Wang
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - John M. Rouhana
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Julia Waldman
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Orr Ashenberg
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Monkol Lek
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Thet Su Win
- Department of Dermatology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Michael S. Cuoco
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Olena Kuksenko
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Philip A. Branton
- The Joint Pathology Center Gynecologic/Breast Pathology, Silver Spring, MD 20910, USA
| | | | - Anna Greka
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Gad Getz
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Cancer Research and Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Ayellet V. Segrè
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - François Aguet
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Orit Rozenblatt-Rosen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
11
|
Wilson CL, Hung CF, Schnapp LM. Endotoxin-induced acute lung injury in mice with postnatal deletion of nephronectin. PLoS One 2022; 17:e0268398. [PMID: 35552565 PMCID: PMC9097991 DOI: 10.1371/journal.pone.0268398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/28/2022] [Indexed: 11/18/2022] Open
Abstract
Acute injury of the lung involves damage to the epithelium and its underlying extracellular matrix (ECM), the basement membrane (BM). How BMs contribute to injury resolution is poorly understood. Nephronectin (NPNT) is a high-affinity ligand for integrin α8β1 and, although first identified in the mouse kidney, is prominently expressed in the lung, where it localizes to BMs in the alveoli. To determine if NPNT plays a role in acute injury and inflammation of the lung, we developed a model for postnatal deletion of NPNT using mice with a floxed allele of Npnt in combination with a tamoxifen-inducible Cre recombinase expressed at the ROSA locus. Expression of NPNT was substantially reduced in lungs from tamoxifen-treated Cre+ animals. Cre+ mice and Cre- controls were given E. coli LPS by oropharyngeal aspiration to induce injury and inflammation. In Cre- lungs, although both Npnt and Itga8 (integrin α8) transcripts were downregulated at the peak of inflammation, NPNT protein was still detectable. While the onset of inflammation was similar for Cre+ and Cre-, NPNT-deficient lungs still had thickened alveolar septa and there were increased macrophages in the bronchoalveolar lavage fluid (BALF) in the resolution phase. BALF from Cre+ lungs was more chemotactic for bone marrow-derived macrophages than Cre- in in vitro experiments, but there were no differences in the elaboration of chemokines in vivo. We speculate that absence of NPNT in BMs of the alveoli impairs or delays inflammatory and injury resolution in this model, but further studies are needed to establish the precise role of NPNT in tissue repair.
Collapse
Affiliation(s)
- Carole L. Wilson
- Division of Pulmonary, Critical Care, Allergy, Sleep Medicine, Dept of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| | - Chi F. Hung
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of Washington, Seattle, Washington, United States of America
| | - Lynn M. Schnapp
- Division of Pulmonary, Critical Care, Allergy, Sleep Medicine, Dept of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
12
|
Esho T, Tufa SF, Kobbe B, Wohl AP, Sengle G, Paulsson M, Keene DR, Wagener R. Anchoring cords, a distinct suprastructure in the developing skin. J Invest Dermatol 2022; 142:2940-2948.e2. [DOI: 10.1016/j.jid.2022.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 11/17/2022]
|
13
|
Al-Hamed MH, Altuwaijri N, Alsahan N, Ali W, Abdulwahab F, Alzahrani F, Majrashi N, Alkuraya FS. A null founder variant in NPNT, encoding nephronectin, causes autosomal recessive renal agenesis. Clin Genet 2022; 102:61-65. [PMID: 35246978 DOI: 10.1111/cge.14128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 11/27/2022]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are a spectrum of abnormalities affecting morphogenesis of the kidneys and other structures of the urinary tract. Bilateral renal agenesis (BRA) is the most severe presentation of CAKUT. Loss of either nephronectin (NPNT) or its receptor ITGA8 leads to failure of metanephric kidney development with resulting renal agenesis in murine models. Very recently, a single family with renal agenesis and a homozygous truncating variant in NPNT was reported. We report two families in whom genome-wide linkage analysis showed an autozygous locus linked to BRA (at least one member with unilateral renal agenesis) at 4q24, with an LOD score of ~3. Exome sequencing detected a nonsense variant in NPNT in both families within the linkage interval. The pathogenicity of this variant was supported by RT-PCR data showing complete nonsense-mediated decay of the NPNT transcript. Our report confirms the candidacy of NPNT in renal agenesis in humans and shows that even complete loss of function can be compatible with the formation of a single kidney. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mohamed H Al-Hamed
- Department of Clinical Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Norah Altuwaijri
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nada Alsahan
- Department of Obstetrics and Genecology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Wafaa Ali
- Department of Clinical Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Firdous Abdulwahab
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fatma Alzahrani
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nada Majrashi
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Ma J, Bi L, Spurlin J, Lwigale P. Nephronectin-Integrin α8 signaling is required for proper migration of periocular neural crest cells during chick corneal development. eLife 2022; 11:74307. [PMID: 35238772 PMCID: PMC8916771 DOI: 10.7554/elife.74307] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/02/2022] [Indexed: 11/19/2022] Open
Abstract
During development, cells aggregate at tissue boundaries to form normal tissue architecture of organs. However, how cells are segregated into tissue precursors remains largely unknown. Cornea development is a perfect example of this process whereby neural crest cells aggregate in the periocular region prior to their migration and differentiation into corneal cells. Our recent RNA-seq analysis identified upregulation of nephronectin (Npnt) transcripts during early stages of corneal development where its function has not been investigated. We found that Npnt mRNA and protein are expressed by various ocular tissues, including the migratory periocular neural crest (pNC), which also express the integrin alpha 8 (Itgα8) receptor. Knockdown of either Npnt or Itgα8 attenuated cornea development, whereas overexpression of Npnt resulted in cornea thickening. Moreover, overexpression of Npnt variants lacking RGD-binding sites did not affect corneal thickness. Neither the knockdown nor augmentation of Npnt caused significant changes in cell proliferation, suggesting that Npnt directs pNC migration into the cornea. In vitro analyses showed that Npnt promotes pNC migration from explanted periocular mesenchyme, which requires Itgα8, focal adhesion kinase, and Rho kinase. Combined, these data suggest that Npnt augments cell migration into the presumptive cornea extracellular matrix by functioning as a substrate for Itgα8-positive pNC cells.
Collapse
Affiliation(s)
- Justin Ma
- Department of Biosciences, Rice University, Houston, United States
| | - Lian Bi
- Department of Biosciences, Rice University, Houston, United States
| | - James Spurlin
- Department of Biosciences, Rice University, Houston, United States
| | - Peter Lwigale
- Department of Biosciences, Rice University, Houston, United States
| |
Collapse
|
15
|
Cryptophthalmos, dental anomalies, oral vestibule defect, and a novel FREM2 mutation. J Hum Genet 2021; 67:115-118. [PMID: 34408272 DOI: 10.1038/s10038-021-00972-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/04/2021] [Accepted: 08/10/2021] [Indexed: 11/08/2022]
Abstract
FREM2 is a member of the FREM2-FRAS1-FREM1 protein complex which contributes to epithelial-mesenchymal coupling. We report a Thai woman with cryptophthalmos, dental anomalies, and oral vestibule defect. A compound heterozygous mutation (c.6499C>T; p.Arg2167Trp and c.641_642del; p.Glu214GlyfsTer135) in the FREM2 gene was identified. The frameshift variant p.Glu214GlyfsTer135 is de novo and novel. It is predicted to result in the loss of most of the functional domains. The p.Arg2167Trp mutation was predicted to disrupt both Ca2+ binding and conformational change. The Arg2167Trp mutant protein has been shown to cause partial loss of function, decrease its interaction with FREM1 and result in impaired function of the FRAS1-FREM2-FREM1 complex. Frem2 was shown to be expressed in the developing tooth and vestibular lamina. It is hypothesized that these mutations resulted in aberration of the FRAS1-FREM2-FREM1 protein complex, resulting in loss of nephronectin, basement membrane disruption, and abnormal epithelial-mesenchymal interactions leading to dental and oral vestibule malformations.
Collapse
|
16
|
Kiyozumi D, Yaguchi S, Yaguchi J, Yamazaki A, Sekiguchi K. Human disease-associated extracellular matrix orthologs ECM3 and QBRICK regulate primary mesenchymal cell migration in sea urchin embryos. Exp Anim 2021; 70:378-386. [PMID: 33828019 PMCID: PMC8390315 DOI: 10.1538/expanim.21-0001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/11/2021] [Indexed: 10/31/2022] Open
Abstract
Sea urchin embryos have been one of model organisms to investigate cellular behaviors because of their simple cell composition and transparent body. They also give us an opportunity to investigate molecular functions of human proteins of interest that are conserved in sea urchin. Here we report that human disease-associated extracellular matrix orthologues ECM3 and QBRICK are necessary for mesenchymal cell migration during sea urchin embryogenesis. Immunofluorescence has visualized the colocalization of QBRICK and ECM3 on both apical and basal surface of ectoderm. On the basal surface, QBRICK and ECM3 constitute together a mesh-like fibrillar structure along the blastocoel wall. When the expression of ECM3 was knocked down by antisense-morpholino oligonucleotides, the ECM3-QBRICK fibrillar structure completely disappeared. When QBRICK was knocked down, the ECM3 was still present, but the basally localized fibers became fragmented. The ingression and migration of primary mesenchymal cells were not critically affected, but their migration at later stages was severely affected in both knock-down embryos. As a consequence of impaired primary mesenchymal cell migration, improper spicule formation was observed. These results indicate that ECM3 and QBRICK are components of extracellular matrix, which play important role in primary mesenchymal cell migration, and that sea urchin is a useful experimental animal model to investigate human disease-associated extracellular matrix proteins.
Collapse
Affiliation(s)
- Daiji Kiyozumi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shunsuke Yaguchi
- Shimoda Marine Research Center, University of Tsukuba, 5-10-1 Shimoda, Shizuoka 415-0025, Japan
- PRESTO, JST, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Junko Yaguchi
- Shimoda Marine Research Center, University of Tsukuba, 5-10-1 Shimoda, Shizuoka 415-0025, Japan
| | - Atsuko Yamazaki
- Shimoda Marine Research Center, University of Tsukuba, 5-10-1 Shimoda, Shizuoka 415-0025, Japan
| | - Kiyotoshi Sekiguchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
17
|
Mapping the molecular and structural specialization of the skin basement membrane for inter-tissue interactions. Nat Commun 2021; 12:2577. [PMID: 33972551 PMCID: PMC8110968 DOI: 10.1038/s41467-021-22881-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 04/01/2021] [Indexed: 12/11/2022] Open
Abstract
Inter-tissue interaction is fundamental to multicellularity. Although the basement membrane (BM) is located at tissue interfaces, its mode of action in inter-tissue interactions remains poorly understood, mainly because the molecular and structural details of the BM at distinct inter-tissue interfaces remain unclear. By combining quantitative transcriptomics and immunohistochemistry, we systematically identify the cellular origin, molecular identity and tissue distribution of extracellular matrix molecules in mouse hair follicles, and reveal that BM composition and architecture are exquisitely specialized for distinct inter-tissue interactions, including epithelial–fibroblast, epithelial–muscle and epithelial–nerve interactions. The epithelial–fibroblast interface, namely, hair germ–dermal papilla interface, makes asymmetrically organized side-specific heterogeneity in the BM, defined by the newly characterized interface, hook and mesh BMs. One component of these BMs, laminin α5, is required for hair cycle regulation and hair germ–dermal papilla anchoring. Our study highlights the significance of BM heterogeneity in distinct inter-tissue interactions. The basement membrane is located at tissue interfaces, but how it mediates distinct inter-tissue interactions is unclear. Here, the authors systematically define the spatial heterogeneity of skin basement membrane composition and show its functional importance in inter-tissue interactions.
Collapse
|
18
|
Nephronectin as a Matrix Effector in Cancer. Cancers (Basel) 2021; 13:cancers13050959. [PMID: 33668838 PMCID: PMC7956348 DOI: 10.3390/cancers13050959] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary The extracellular matrix provides an important scaffold for cells and tissues of multicellular organisms. The scaffold not only provides a secure anchorage point, but also functions as a reservoir for signalling molecules, sequestered and released when necessary. A dysregulated extracellular matrix may therefore modulate cellular behaviour, as seen during cancer progression. The extracellular matrix protein nephronectin was discovered two decades ago and found to regulate important embryonic developmental processes. Loss of either nephronectin or its receptor, integrin α8β1, leads to underdeveloped kidneys. Recent findings show that nephronectin is also dysregulated in breast cancer and plays a role in promoting metastasis. To enable therapeutic intervention, it is important to fully understand the role of nephronectin and its receptors in cancer progression. In this review, we summarise the literature on nephronectin, analyse the structure and domain-related functions of nephronectin and link these functions to potential roles in cancer progression. Abstract The extracellular matrix protein nephronectin plays an important regulatory role during embryonic development, controlling renal organogenesis through integrin α8β1 association. Nephronectin has three main domains: five N-terminal epidermal growth factor-like domains, a linker region harbouring two integrin-binding motifs (RGD and LFEIFEIER), and a C-terminal MAM domain. In this review, we look into the domain-related functions of nephronectin, and tissue distribution and expression. During the last two decades it has become evident that nephronectin also plays a role during cancer progression and in particular metastasis. Nephronectin is overexpressed in both human and mouse breast cancer compared to normal breast tissue where the protein is absent. Cancer cells expressing elevated levels of nephronectin acquire increased ability to colonise distant organs. In particular, the enhancer-motif (LFEIFEIER) which is specific to the integrin α8β1 association induces viability via p38 MAPK and plays a role in colonization. Integrins have long been desired as therapeutic targets, where low efficiency and receptor redundancy have been major issues. Based on the summarised publications, the enhancer-motif of nephronectin could present a novel therapeutic target.
Collapse
|
19
|
Mészáros B, Sámano-Sánchez H, Alvarado-Valverde J, Čalyševa J, Martínez-Pérez E, Alves R, Shields DC, Kumar M, Rippmann F, Chemes LB, Gibson TJ. Short linear motif candidates in the cell entry system used by SARS-CoV-2 and their potential therapeutic implications. Sci Signal 2021; 14:eabd0334. [PMID: 33436497 PMCID: PMC7928535 DOI: 10.1126/scisignal.abd0334] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022]
Abstract
The first reported receptor for SARS-CoV-2 on host cells was the angiotensin-converting enzyme 2 (ACE2). However, the viral spike protein also has an RGD motif, suggesting that cell surface integrins may be co-receptors. We examined the sequences of ACE2 and integrins with the Eukaryotic Linear Motif (ELM) resource and identified candidate short linear motifs (SLiMs) in their short, unstructured, cytosolic tails with potential roles in endocytosis, membrane dynamics, autophagy, cytoskeleton, and cell signaling. These SLiM candidates are highly conserved in vertebrates and may interact with the μ2 subunit of the endocytosis-associated AP2 adaptor complex, as well as with various protein domains (namely, I-BAR, LC3, PDZ, PTB, and SH2) found in human signaling and regulatory proteins. Several motifs overlap in the tail sequences, suggesting that they may act as molecular switches, such as in response to tyrosine phosphorylation status. Candidate LC3-interacting region (LIR) motifs are present in the tails of integrin β3 and ACE2, suggesting that these proteins could directly recruit autophagy components. Our findings identify several molecular links and testable hypotheses that could uncover mechanisms of SARS-CoV-2 attachment, entry, and replication against which it may be possible to develop host-directed therapies that dampen viral infection and disease progression. Several of these SLiMs have now been validated to mediate the predicted peptide interactions.
Collapse
Affiliation(s)
- Bálint Mészáros
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.
| | - Hugo Sámano-Sánchez
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Jesús Alvarado-Valverde
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences
| | - Jelena Čalyševa
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences
| | - Elizabeth Martínez-Pérez
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Laboratorio de bioinformática estructural, Fundación Instituto Leloir, C1405BWE Buenos Aires, Argentina
| | - Renato Alves
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Denis C Shields
- School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Manjeet Kumar
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.
| | - Friedrich Rippmann
- Computational Chemistry & Biology, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Lucía B Chemes
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", IIB-UNSAM, IIBIO-CONICET, Universidad Nacional de San Martín, CP1650 San Martín, Buenos Aires, Argentina.
| | - Toby J Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.
| |
Collapse
|
20
|
Nephronectin promotes breast cancer brain metastatic colonization via its integrin-binding domains. Sci Rep 2020; 10:12237. [PMID: 32699247 PMCID: PMC7376038 DOI: 10.1038/s41598-020-69242-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 07/07/2020] [Indexed: 12/18/2022] Open
Abstract
This study demonstrates a role for the extracellular matrix protein nephronectin (NPNT) in promoting experimental breast cancer brain metastasis, possibly through enhanced binding to- and migration through brain endothelial cells. With the introduction of more targeted breast cancer treatments, a prolonged survival has resulted during the last decade. Consequently, an increased number of patients develop metastasis in the brain, a challenging organ to treat. We recently reported that NPNT was highly expressed in primary breast cancer and associated with unfavourable prognosis. The current study addresses our hypothesis that NPNT promotes brain metastases through its integrin-binding motifs. SAGE-sequencing revealed that NPNT was significantly up-regulated in human breast cancer tissue compared to pair-matched normal breast tissue. Human brain metastatic breast cancers expressed both NPNT and its receptor, integrin α8β1. Using an open access repository; BreastMark, we found a correlation between high NPNT mRNA levels and poor prognosis for patients with the luminal B subtype. The 66cl4 mouse cell line was used for expression of wild-type and mutant NPNT, which is unable to bind α8β1. Using an in vivo model of brain metastatic colonization, 66cl4-NPNT cells showed an increased ability to form metastatic lesions compared to cells with mutant NPNT, possibly through reduced endothelial adhesion and transmigration.
Collapse
|
21
|
Kunz F, Kayserili H, Midro A, Silva D, Basnayake S, Güven Y, Borys J, Schanze D, Stellzig‐Eisenhauer A, Bloch‐Zupan A, Zenker M. Characteristic dental pattern with hypodontia and short roots in Fraser syndrome. Am J Med Genet A 2020; 182:1681-1689. [DOI: 10.1002/ajmg.a.61610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Felix Kunz
- Department of Orthodontics University Hospital of Würzburg Würzburg Germany
| | - Hülya Kayserili
- KOÇ University School of Medicine (KUSoM) Medical Genetics Department Topkapi Zeytinburnu Istanbul Turkey
| | - Alina Midro
- Department of Clinical Genetics Medical University Białystok Poland
| | - Deepthi Silva
- Department of Physiology, Faculty of Medicine University of Kelaniya Ragama Sri Lanka
| | | | - Yeliz Güven
- Department of Pedodontics, Faculty of Dentistry Istanbul University Istanbul Turkey
| | - Jan Borys
- Department of Maxillofacial and Plastic Surgery Medical University of Bialystok Poland
| | - Denny Schanze
- Institute of Human Genetics, University Hospital Magdeburg Germany
| | | | - Agnes Bloch‐Zupan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS‐UMR7104, Université de Strasbourg Illkirch‐Graffenstaden France
- Hôpitaux Universitaires de Strasbourg (HUS), Pôle de Médecine et Chirurgie Bucco‐Dentaires, Hôpital Civil, Centre de référence des maladies rares orales et dentaires, O‐Rares, Filière Santé Maladies rares TETE COU, European Reference Network ERN CRANIO Strasbourg France
- Faculté de Chirurgie Dentaire Université de Strasbourg Strasbourg France
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg Germany
| |
Collapse
|
22
|
Kiyozumi D, Nakano I, Sato-Nishiuchi R, Tanaka S, Sekiguchi K. Laminin is the ECM niche for trophoblast stem cells. Life Sci Alliance 2020; 3:3/2/e201900515. [PMID: 31937556 PMCID: PMC6977391 DOI: 10.26508/lsa.201900515] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 01/07/2023] Open
Abstract
Laminin functions as an ECM niche factor for trophoblast stem cells and secures trophoblast stem cell expansion through its interactions with integrin. The niche is a specialized microenvironment for tissue stem cells in vivo. It has long been emphasized that niche ECM molecules act on tissue stem cells to regulate their behavior, but the molecular entities of these interactions remain to be fully elucidated. Here, we report that laminin forms the in vivo ECM niche for trophoblast stem cells (TSCs), the tissue stem cells of the placenta. TSCs expressed fibronectin-binding, vitronectin-binding, and laminin-binding integrins, whereas the integrin ligands present in the TSC niche were collagen and laminin. Therefore, the only niche integrin ligand available for TSCs in vivo was laminin. Laminin promoted TSC adhesion and proliferation in vitro in an integrin binding–dependent manner. Importantly, when the integrin-binding ability of laminin was genetically ablated in mice, the size of the TSC population was significantly reduced compared with that in control mice. The present findings underscore an ECM niche function of laminin to support tissue stem cell maintenance in vivo.
Collapse
Affiliation(s)
- Daiji Kiyozumi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Itsuko Nakano
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Ryoko Sato-Nishiuchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Satoshi Tanaka
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kiyotoshi Sekiguchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| |
Collapse
|
23
|
Theisen DJ, Ferris ST, Briseño CG, Kretzer N, Iwata A, Murphy KM, Murphy TL. Batf3-Dependent Genes Control Tumor Rejection Induced by Dendritic Cells Independently of Cross-Presentation. Cancer Immunol Res 2019; 7:29-39. [PMID: 30482745 DOI: 10.1158/2326-6066.cir-18-0138] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 09/12/2018] [Accepted: 11/21/2018] [Indexed: 11/16/2022]
Abstract
The BATF3-dependent cDC1 lineage of conventional dendritic cells (cDC) is required for rejection of immunogenic sarcomas and for rejection of progressive sarcomas during checkpoint blockade therapy. One unique function of the cDC1 lineage is the efficient cross-presentation of tumor-derived neoantigens to CD8+ T cells, but it is not clear that this is the only unique function of cDC1 required for tumor rejection. We previously showed that BATF3 functions during cDC1 lineage commitment to maintain IRF8 expression in the specified cDC1 progenitor. However, since cDC1 progenitors do not develop into mature cDC1s in Batf3 -/- mice, it is still unclear whether BATF3 has additional functions in mature cDC1 cells. A transgenic Irf8-Venus reporter allele increases IRF8 protein concentration sufficiently to allow autonomous cDC1 development in spleens of Batf3 -/- mice. These restored Batf3 -/- cDC1s are transcriptionally similar to control wild-type cDC1s but have reduced expression of a restricted set of cDC1-specific genes. Restored Batf3 -/- cDC1s are able to cross-present cell-associated antigens both in vitro and in vivo However, Batf3 -/- cDC1 exhibit altered characteristics in vivo and are unable to mediate tumor rejection. These results show that BATF3, in addition to regulating Irf8 expression to stabilize cDC1 lineage commitment, also controls expression of a small set of genes required for cDC1-mediated tumor rejection. These BATF3-regulated genes may be useful targets in immunotherapies aimed at promoting tumor rejection.
Collapse
Affiliation(s)
- Derek J Theisen
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Stephen T Ferris
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Carlos G Briseño
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Nicole Kretzer
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Arifumi Iwata
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
- Howard Hughes Medical Institute, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Theresa L Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri.
| |
Collapse
|
24
|
Sato Y, Kiyozumi D, Futaki S, Nakano I, Shimono C, Kaneko N, Ikawa M, Okabe M, Sawamoto K, Sekiguchi K. Ventricular-subventricular zone fractones are speckled basement membranes that function as a neural stem cell niche. Mol Biol Cell 2018; 30:56-68. [PMID: 30379609 PMCID: PMC6337917 DOI: 10.1091/mbc.e18-05-0286] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neural stem cells (NSCs) are retained in the adult ventricular–subventricular zone (V-SVZ), a specialized neurogenic niche with a unique cellular architecture. It currently remains unclear whether or how NSCs utilize basement membranes (BMs) in this niche. Here, we examine the molecular compositions and functions of BMs in the adult mouse V-SVZ. Whole-mount V-SVZ immunostaining revealed that fractones, which are fingerlike processes of extravascular BMs, are speckled BMs unconnected to the vasculature, and differ in their molecular composition from vascular BMs. Glial fibrillary acidic protein (GFAP)-positive astrocytes and NSCs produce and adhere to speckled BMs. Furthermore, Gfap-Cre-mediated Lamc1flox(E1605Q) knockin mice, in which integrin-binding activities of laminins are specifically nullified in GFAP-positive cells, exhibit a decreased number and size of speckled BMs and reduced in vitro neurosphere-forming activity. Our results reveal niche activities of fractones/speckled BMs for NSCs and provide molecular insights into how laminin–integrin interactions regulate NSCs in vivo.
Collapse
Affiliation(s)
- Yuya Sato
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Daiji Kiyozumi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Sugiko Futaki
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Itsuko Nakano
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Chisei Shimono
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Naoko Kaneko
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8610, Japan
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masaru Okabe
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8610, Japan.,Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
| | - Kiyotoshi Sekiguchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan.,Laboratory of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
25
|
Kiyozumi D, Taniguchi Y, Nakano I, Toga J, Yagi E, Hasuwa H, Ikawa M, Sekiguchi K. Laminin γ1 C-terminal Glu to Gln mutation induces early postimplantation lethality. Life Sci Alliance 2018; 1:e201800064. [PMID: 30456378 PMCID: PMC6238537 DOI: 10.26508/lsa.201800064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 11/24/2022] Open
Abstract
Mouse embryos with an ablated ability of integrins to bind laminins are still able to form basement membranes, but die just after implantation because of deficient extraembryonic development. Laminin–integrin interactions regulate various adhesion-dependent cellular processes. γ1C-Glu, the Glu residue in the laminin γ1 chain C-terminal tail, is crucial for the binding of γ1-laminins to several integrin isoforms. Here, we investigated the impact of γ1C Glu to Gln mutation on γ1-laminin binding to all possible integrin partners in vitro, and found that the mutation specifically ablated binding to α3, α6, and α7 integrins. To examine the physiological significance of γ1C-Glu, we generated a knock-in allele, Lamc1EQ, in which the γ1C Glu to Gln mutation was introduced. Although Lamc1EQ/EQ homozygotes developed into blastocysts and deposited laminins in their basement membranes, they died just after implantation because of disordered extraembryonic development. Given the impact of the Lamc1EQ allele on embryonic development, we developed a knock-in mouse strain enabling on-demand introduction of the γ1C Glu to Gln mutation by the Cre-loxP system. The present study has revealed a crucial role of γ1C-Glu–mediated integrin binding in postimplantation development and provides useful animal models for investigating the physiological roles of laminin–integrin interactions in vivo.
Collapse
Affiliation(s)
- Daiji Kiyozumi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Yukimasa Taniguchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Itsuko Nakano
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Junko Toga
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Emiko Yagi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Hidetoshi Hasuwa
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kiyotoshi Sekiguchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| |
Collapse
|
26
|
Abstract
The basement membrane is a thin but dense, sheet-like specialized type of extracellular matrix that has remarkably diverse functions tailored to individual tissues and organs. Tightly controlled spatial and temporal changes in its composition and structure contribute to the diversity of basement membrane functions. These different basement membranes undergo dynamic transformations throughout animal life, most notably during development. Numerous developmental mechanisms are regulated or mediated by basement membranes, often by a combination of molecular and mechanical processes. A particularly important process involves cell transmigration through a basement membrane because of its link to cell invasion in disease. While developmental and disease processes share some similarities, what clearly distinguishes the two is dysregulation of cells and extracellular matrices in disease. With its relevance to many developmental and disease processes, the basement membrane is a vitally important area of research that may provide novel insights into biological mechanisms and development of innovative therapeutic approaches. Here we present a review of developmental and disease dynamics of basement membranes in Caenorhabditis elegans, Drosophila, and vertebrates.
Collapse
|
27
|
van der Ven AT, Kobbe B, Kohl S, Shril S, Pogoda HM, Imhof T, Ityel H, Vivante A, Chen J, Hwang DY, Connaughton DM, Mann N, Widmeier E, Taglienti M, Schmidt JM, Nakayama M, Senguttuvan P, Kumar S, Tasic V, Kehinde EO, Mane SM, Lifton RP, Soliman N, Lu W, Bauer SB, Hammerschmidt M, Wagener R, Hildebrandt F. A homozygous missense variant in VWA2, encoding an interactor of the Fraser-complex, in a patient with vesicoureteral reflux. PLoS One 2018; 13:e0191224. [PMID: 29351342 PMCID: PMC5774751 DOI: 10.1371/journal.pone.0191224] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 12/29/2017] [Indexed: 11/18/2022] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause (40-50%) of chronic kidney disease (CKD) in children. About 40 monogenic causes of CAKUT have so far been discovered. To date less than 20% of CAKUT cases can be explained by mutations in these 40 genes. To identify additional monogenic causes of CAKUT, we performed whole exome sequencing (WES) and homozygosity mapping (HM) in a patient with CAKUT from Indian origin and consanguineous descent. We identified a homozygous missense mutation (c.1336C>T, p.Arg446Cys) in the gene Von Willebrand factor A domain containing 2 (VWA2). With immunohistochemistry studies on kidneys of newborn (P1) mice, we show that Vwa2 and Fraser extracellular matrix complex subunit 1 (Fras1) co-localize in the nephrogenic zone of the renal cortex. We identified a pronounced expression of Vwa2 in the basement membrane of the ureteric bud (UB) and derivatives of the metanephric mesenchyme (MM). By applying in vitro assays, we demonstrate that the Arg446Cys mutation decreases translocation of monomeric VWA2 protein and increases translocation of aggregated VWA2 protein into the extracellular space. This is potentially due to the additional, unpaired cysteine residue in the mutated protein that is used for intermolecular disulfide bond formation. VWA2 is a known, direct interactor of FRAS1 of the Fraser-Complex (FC). FC-encoding genes and interacting proteins have previously been implicated in the pathogenesis of syndromic and/or isolated CAKUT phenotypes in humans. VWA2 therefore constitutes a very strong candidate in the search for novel CAKUT-causing genes. Our results from in vitro experiments indicate a dose-dependent neomorphic effect of the Arg446Cys homozygous mutation in VWA2.
Collapse
Affiliation(s)
- Amelie T. van der Ven
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Birgit Kobbe
- Center for Biochemistry, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Stefan Kohl
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Pediatrics, Cologne Children’s Hospital, Cologne, Germany
| | - Shirlee Shril
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hans-Martin Pogoda
- Institute of Zoology-Developmental Biology, Biocenter Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Thomas Imhof
- Institute for Dental Research and Oral Musculoskeletal Biology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Hadas Ityel
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Asaf Vivante
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel
| | - Jing Chen
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Nephrology, Children’s Hospital of Fudan University, Shanghai, China
| | - Daw-Yang Hwang
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Nephrology, Department of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Dervla M. Connaughton
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Nina Mann
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Eugen Widmeier
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mary Taglienti
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Johanna Magdalena Schmidt
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Makiko Nakayama
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Prabha Senguttuvan
- Department of Pediatric Nephrology, Dr. Mehta’s Multispeciality Hospital, Chennai, India
| | - Selvin Kumar
- Department of Pediatric Nephrology, Institute of Child Health and Hospital for Children, the Tamil Nadu Dr. M.G.R. Medical University, Chennai, Tamil Nadu, India
| | - Velibor Tasic
- Medical Faculty Skopje, University Children’s Hospital, Skopje, Macedonia
| | - Elijah O. Kehinde
- Division of Urology, Department of Surgery, Nazarbayev University, Astana, Kazakhstan
| | - Shrikant M. Mane
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Richard P. Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Neveen Soliman
- Department of Pediatrics, Center of Pediatric Nephrology & Transplantation, Cairo University, Egyptian Group for Orphan Renal Diseases, Cairo, Egypt
| | - Weining Lu
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, United States of America
| | - Stuart B. Bauer
- Department of Urology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Matthias Hammerschmidt
- Institute of Zoology-Developmental Biology, Biocenter Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Raimund Wagener
- Center for Biochemistry, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- * E-mail: (RW); (FH)
| | - Friedhelm Hildebrandt
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (RW); (FH)
| |
Collapse
|
28
|
Zimmerman SE, Hiremath C, Tsunezumi J, Yang Z, Finney B, Marciano DK. Nephronectin Regulates Mesangial Cell Adhesion and Behavior in Glomeruli. J Am Soc Nephrol 2018; 29:1128-1140. [PMID: 29335243 DOI: 10.1681/asn.2017070752] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/13/2017] [Indexed: 01/03/2023] Open
Abstract
A critical aspect of kidney function occurs at the glomerulus, the capillary network that filters the blood. The glomerular basement membrane (GBM) is a key component of filtration, yet our understanding of GBM interactions with mesangial cells, specialized pericytes that provide structural stability to glomeruli, is limited. We investigated the role of nephronectin (Npnt), a GBM component and known ligand of α8β1 integrin. Immunolocalization and in situ hybridization studies in kidneys of adult mice revealed that nephronectin is produced by podocytes and deposited into the GBM. Conditional deletion of Npnt from nephron progenitors caused a pronounced increase in mesangial cell number and mesangial sclerosis. Nephronectin colocalized with α8β1 integrin to novel, specialized adhesion structures that occurred at sites of mesangial cell protrusion at the base of the capillary loops. Absence of nephronectin disrupted these adhesion structures, leading to mislocalization of α8β1. Podocyte-specific deletion of Npnt also led to mesangial sclerosis in mice. These results demonstrate a novel role for nephronectin and α8β1 integrin in a newly described adhesion complex and begin to uncover the molecular interactions between the GBM and mesangial cells, which govern mesangial cell behavior and may have a role in pathologic states.
Collapse
Affiliation(s)
- Susan E Zimmerman
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chitkale Hiremath
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jun Tsunezumi
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhufeng Yang
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bronwyn Finney
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Denise K Marciano
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
29
|
van der Ven AT, Vivante A, Hildebrandt F. Novel Insights into the Pathogenesis of Monogenic Congenital Anomalies of the Kidney and Urinary Tract. J Am Soc Nephrol 2017; 29:36-50. [PMID: 29079659 DOI: 10.1681/asn.2017050561] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Congenital anomalies of the kidneys and urinary tract (CAKUT) comprise a large spectrum of congenital malformations ranging from severe manifestations, such as renal agenesis, to potentially milder conditions, such as vesicoureteral reflux. CAKUT causes approximately 40% of ESRD that manifests within the first three decades of life. Several lines of evidence indicate that CAKUT is often caused by recessive or dominant mutations in single (monogenic) genes. To date, approximately 40 monogenic genes are known to cause CAKUT if mutated, explaining 5%-20% of patients. However, hundreds of different monogenic CAKUT genes probably exist. The discovery of novel CAKUT-causing genes remains challenging because of this pronounced heterogeneity, variable expressivity, and incomplete penetrance. We here give an overview of known genetic causes for human CAKUT and shed light on distinct renal morphogenetic pathways that were identified as relevant for CAKUT in mice and humans.
Collapse
Affiliation(s)
- Amelie T van der Ven
- Divison of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Asaf Vivante
- Divison of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Friedhelm Hildebrandt
- Divison of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
30
|
Sun Y, Kuek V, Qiu H, Tickner J, Chen L, Wang H, He W, Xu J. The emerging role of NPNT in tissue injury repair and bone homeostasis. J Cell Physiol 2017; 233:1887-1894. [DOI: 10.1002/jcp.26013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 05/15/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Youqiang Sun
- The National Key Discipline and the Orthopedic Laboratory; Guangzhou University of Chinese Medicine; Guangzhou Guangdong P. R. China
- Department of Orthopedics, First Affiliated Hospital; Guangzhou University of Chinese Medicine; Guangzhou Guangdong P. R. China
- School of Pathology and Laboratory Medicine; The University of Western Australia; Perth WA Australia
| | - Vincent Kuek
- School of Pathology and Laboratory Medicine; The University of Western Australia; Perth WA Australia
| | - Heng Qiu
- School of Pathology and Laboratory Medicine; The University of Western Australia; Perth WA Australia
| | - Jennifer Tickner
- School of Pathology and Laboratory Medicine; The University of Western Australia; Perth WA Australia
| | - Leilei Chen
- The National Key Discipline and the Orthopedic Laboratory; Guangzhou University of Chinese Medicine; Guangzhou Guangdong P. R. China
- Department of Orthopedics, First Affiliated Hospital; Guangzhou University of Chinese Medicine; Guangzhou Guangdong P. R. China
| | - Haibin Wang
- The National Key Discipline and the Orthopedic Laboratory; Guangzhou University of Chinese Medicine; Guangzhou Guangdong P. R. China
- Department of Orthopedics, First Affiliated Hospital; Guangzhou University of Chinese Medicine; Guangzhou Guangdong P. R. China
| | - Wei He
- The National Key Discipline and the Orthopedic Laboratory; Guangzhou University of Chinese Medicine; Guangzhou Guangdong P. R. China
- Department of Orthopedics, First Affiliated Hospital; Guangzhou University of Chinese Medicine; Guangzhou Guangdong P. R. China
| | - Jiake Xu
- The National Key Discipline and the Orthopedic Laboratory; Guangzhou University of Chinese Medicine; Guangzhou Guangdong P. R. China
- School of Pathology and Laboratory Medicine; The University of Western Australia; Perth WA Australia
| |
Collapse
|
31
|
Nephronectin plays critical roles in Sox2 expression and proliferation in dental epithelial stem cells via EGF-like repeat domains. Sci Rep 2017; 7:45181. [PMID: 28345658 PMCID: PMC5366923 DOI: 10.1038/srep45181] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 02/17/2017] [Indexed: 12/31/2022] Open
Abstract
Tooth development is initiated by epithelial-mesenchymal interactions via basement membrane (BM) and growth factors. In the present study, we found that nephronectin (Npnt), a component of the BM, is highly expressed in the developing tooth. Npnt localizes in the BM on the buccal side of the tooth germ and shows an expression pattern opposite that of the dental epithelial stem cell marker Sox2. To identify the roles of Npnt during tooth development, we performed knockdown and overexpression experiments using ex vivo organ and dental epithelial cell cultures. Our findings showed that loss of Npnt induced ectopic Sox2-positive cells and reduced tooth germ size. Over expression of Npnt showed increased proliferation, whereas the number of Sox2-positive cells was decreased in dental epithelial cells. Npnt contains 5 EGF-like repeat domains, as well as an RGD sequence and MAM domain. We found that the EGF-like repeats are critical for Sox2 expression and cell proliferation. Furthermore, Npnt activated the EGF receptor (EGFR) via the EGF-like repeat domains and induced the PI3K-Akt signaling pathway. Our results indicate that Npnt plays a critical scaffold role in dental epithelial stem cell differentiation and proliferation, and regulates Sox2 expression during tooth development.
Collapse
|
32
|
Talbot JC, Nichols JT, Yan YL, Leonard IF, BreMiller RA, Amacher SL, Postlethwait JH, Kimmel CB. Pharyngeal morphogenesis requires fras1-itga8-dependent epithelial-mesenchymal interaction. Dev Biol 2016; 416:136-148. [PMID: 27265864 PMCID: PMC4967372 DOI: 10.1016/j.ydbio.2016.05.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/31/2016] [Indexed: 01/08/2023]
Abstract
Both Fras1 and Itga8 connect mesenchymal cells to epithelia by way of an extracellular 'Fraser protein complex' that functions in signaling and adhesion; these proteins are vital to the development of several vertebrate organs. We previously found that zebrafish fras1 mutants have craniofacial defects, specifically, shortened symplectic cartilages and cartilage fusions that spare joint elements. During a forward mutagenesis screen, we identified a new zebrafish mutation, b1161, that we show here disrupts itga8, as confirmed using CRISPR-generated itga8 alleles. fras1 and itga8 single mutants and double mutants have similar craniofacial phenotypes, a result expected if loss of either gene disrupts function of the Fraser protein complex. Unlike fras1 mutants or other Fraser-related mutants, itga8 mutants do not show blistered tail fins. Thus, the function of the Fraser complex differs in the craniofacial skeleton and the tail fin. Focusing on the face, we find that itga8 mutants consistently show defective outpocketing of a late-forming portion of the first pharyngeal pouch, and variably express skeletal defects, matching previously characterized fras1 mutant phenotypes. In itga8 and fras1 mutants, skeletal severity varies markedly between sides, indicating that both mutants have increased developmental instability. Whereas fras1 is expressed in epithelia, we show that itga8 is expressed complementarily in facial mesenchyme. Paired with the observed phenotypic similarity, this expression indicates that the genes function in epithelial-mesenchymal interactions. Similar interactions between Fras1 and Itga8 have previously been found in mouse kidney, where these genes both regulate Nephronectin (Npnt) protein abundance. We find that zebrafish facial tissues express both npnt and the Fraser gene fibrillin2b (fbn2b), but their transcript levels do not depend on fras1 or itga8 function. Using a revertible fras1 allele, we find that the critical window for fras1 function in the craniofacial skeleton is between 1.5 and 3 days post fertilization, which coincides with the onset of fras1-dependent and itga8-dependent morphogenesis. We propose a model wherein Fras1 and Itga8 interact during late pharyngeal pouch morphogenesis to sculpt pharyngeal arches through epithelial-mesenchymal interactions, thereby stabilizing the developing craniofacial skeleton.
Collapse
Affiliation(s)
- Jared Coffin Talbot
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA; Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA.
| | - James T Nichols
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Yi-Lin Yan
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Isaac F Leonard
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Ruth A BreMiller
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Sharon L Amacher
- Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | | | - Charles B Kimmel
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA.
| |
Collapse
|
33
|
Khalifeh-Soltani A, Ha A, Podolsky MJ, McCarthy DA, McKleroy W, Azary S, Sakuma S, Tharp KM, Wu N, Yokosaki Y, Hart D, Stahl A, Atabai K. α8β1 integrin regulates nutrient absorption through an Mfge8-PTEN dependent mechanism. eLife 2016; 5. [PMID: 27092791 PMCID: PMC4868538 DOI: 10.7554/elife.13063] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 04/18/2016] [Indexed: 12/25/2022] Open
Abstract
Coordinated gastrointestinal smooth muscle contraction is critical for proper nutrient absorption and is altered in a number of medical disorders. In this work, we demonstrate a critical role for the RGD-binding integrin α8β1 in promoting nutrient absorption through regulation of gastrointestinal motility. Smooth muscle-specific deletion and antibody blockade of α8 in mice result in enhanced gastric antral smooth muscle contraction, more rapid gastric emptying, and more rapid transit of food through the small intestine leading to malabsorption of dietary fats and carbohydrates as well as protection from weight gain in a diet-induced model of obesity. Mechanistically, ligation of α8β1 by the milk protein Mfge8 reduces antral smooth muscle contractile force by preventing RhoA activation through a PTEN-dependent mechanism. Collectively, our results identify a role for α8β1 in regulating gastrointestinal motility and identify α8 as a potential target for disorders characterized by hypo- or hyper-motility. DOI:http://dx.doi.org/10.7554/eLife.13063.001 Animals absorb nutrients from the food they eat in a complicated process that involves multiple steps. In the mouth, teeth break down the food into smaller chunks. Then the food passes through the stomach and small intestine, where enzymes break it down into individual molecules that are small enough to be absorbed by cells that line the small intestine. These cells then package the molecules and release them into the bloodstream so that they can be distributed to the rest of the body. Muscles in the wall of the small intestine control how quickly food travels through this part of the gut. If food moves too quickly, the cells that line the intestine have less time to absorb the food molecules and may fail to absorb enough nutrients. If the food moves too slowly, an individual may experience nausea or vomiting, or the contents of their stomach may spill into their lungs. In 2014, researchers reported that a protein in breast milk called Mfge8 helps to boost the number of fat molecules absorbed from food. Now, Khalifeh-Soltani et al. – including some of the same researchers involved in the earlier work – show that Mfge8 also slows the rate at which food travels through the small intestine in mice. Mfge8 binds to another protein called integrin α8β1 to control how often the intestine muscles contract. Genetically engineered mice that lacked integrin α8β1 developed diarrhea and food passed through their intestines more quickly than in normal mice. Furthermore, these mice did not gain as much weight as normal mice when they were fed a high-fat diet. Khalifeh-Soltani et al.’s findings show that Mfge8 has a dual role in controlling the absorption of food molecules in the small intestine. The next challenge is to find out whether drugs that alter the activity of integrin α8β1 could be used to help treat patients with diseases in which food moves too quickly, or too slowly, through the gut. DOI:http://dx.doi.org/10.7554/eLife.13063.002
Collapse
Affiliation(s)
- Amin Khalifeh-Soltani
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States.,Department of Medicine, University of California, San Francisco, San Francisco, United States
| | - Arnold Ha
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Michael J Podolsky
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States.,Department of Medicine, University of California, San Francisco, San Francisco, United States
| | - Donald A McCarthy
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - William McKleroy
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Saeedeh Azary
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Stephen Sakuma
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Kevin M Tharp
- Metabolic Biology, University of California, Berkeley, Berkeley, United States.,Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, United States
| | - Nanyan Wu
- Lung Biology Center, University of California, San Francisco, San Francisco, United States
| | - Yasuyuki Yokosaki
- Cell-Matrix Frontier Laboratory, Biomedical Research Unit, Health Administration Center, Hiroshima University, Hiroshima, Japan
| | - Daniel Hart
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Andreas Stahl
- Metabolic Biology, University of California, Berkeley, Berkeley, United States.,Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, United States
| | - Kamran Atabai
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States.,Department of Medicine, University of California, San Francisco, San Francisco, United States.,Lung Biology Center, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
34
|
Has C, Nyström A. Epidermal Basement Membrane in Health and Disease. CURRENT TOPICS IN MEMBRANES 2015; 76:117-70. [PMID: 26610913 DOI: 10.1016/bs.ctm.2015.05.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Skin, as the organ protecting the individual from environmental aggressions, constantly meets external insults and is dependent on mechanical toughness for its preserved function. Accordingly, the epidermal basement membrane (BM) zone has adapted to enforce tissue integrity. It harbors anchoring structures created through unique organization of common BM components and expression of proteins exclusive to the epidermal BM zone. Evidence for the importance of its correct assembly and the nonredundancy of its components for skin integrity is apparent from the multiple skin blistering disorders caused by mutations in genes coding for proteins associated with the epidermal BM and from autoimmune disorders in which autoantibodies target these molecules. However, it has become clear that these proteins not only provide mechanical support but are also critically involved in tissue homeostasis, repair, and regeneration. In this chapter, we provide an overview of the unique organization and components of the epidermal BM. A special focus will be given to its function during regeneration, and in inherited and acquired diseases.
Collapse
Affiliation(s)
- Cristina Has
- Department of Dermatology, University Medical Center Freiburg, Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, University Medical Center Freiburg, Freiburg, Germany
| |
Collapse
|
35
|
Kiyozumi D, Sato-Nishiuchi R, Sekiguchi K. In Situ Detection of Integrin Ligands. CURRENT PROTOCOLS IN CELL BIOLOGY 2014; 65:9.7.1-17. [PMID: 26061156 DOI: 10.1002/0471143030.cb0907s65] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Integrins are cell surface receptors for cell adhesion. Integrin-mediated cell adhesion regulates various cellular processes, including cell survival, migration, proliferation, and differentiation. In vivo, ligands for integrins are immobilized within extracellular matrices, insoluble sheet-like or fibrous supramolecular complexes that associate with or surround cells. To better understand the molecular basis of integrin-mediated regulation of cellular behavior in vivo, it is of critical importance to collect information regarding the activities as well as spatial distributions of integrin ligands in situ. This unit describes a protocol for detecting the spatial distribution of the complement of integrin ligands in situ by overlaying soluble recombinant integrins.
Collapse
Affiliation(s)
- Daiji Kiyozumi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Ryoko Sato-Nishiuchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Kiyotoshi Sekiguchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| |
Collapse
|
36
|
Kiyozumi D, Sato-Nishiuchi R, Sekiguchi K. In situ detection of integrin ligands. CURRENT PROTOCOLS IN CELL BIOLOGY 2014; 65:10.19.1-17. [PMID: 25447073 DOI: 10.1002/0471143030.cb1019s65] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Integrins are cell surface receptors for cell adhesion. Integrin-mediated cell adhesion regulates various cellular processes, including cell survival, migration, proliferation, and differentiation. In vivo, ligands for integrins are immobilized within extracellular matrices, insoluble sheet-like or fibrous supramolecular complexes that associate with or surround cells. To better understand the molecular basis of integrin-mediated regulation of cellular behavior in vivo, it is of critical importance to collect information regarding the activities as well as spatial distributions of integrin ligands in situ. This unit describes a protocol for detecting the spatial distribution of the complement of integrin ligands in situ by overlaying soluble recombinant integrins.
Collapse
Affiliation(s)
- Daiji Kiyozumi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | | | | |
Collapse
|
37
|
Abstract
In embryos, the Fraser Complex (FC) mediates epithelial-connective tissue interactions. Loss of expression of FC components leads to Fraser Syndrome (FS) in which cohesion of epithelial tissues and stroma is perturbed. Using zebrafish, Richardson et al (in this issue) identified the protein AMACO in the FC. We discuss the utility of zebrafish in determining FC functions and identifying FS targets.
Collapse
|
38
|
Rafiq K, Shashikant T, McManus CJ, Ettensohn CA. Genome-wide analysis of the skeletogenic gene regulatory network of sea urchins. Development 2014; 141:950-61. [PMID: 24496631 DOI: 10.1242/dev.105585] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A central challenge of developmental and evolutionary biology is to understand the transformation of genetic information into morphology. Elucidating the connections between genes and anatomy will require model morphogenetic processes that are amenable to detailed analysis of cell/tissue behaviors and to systems-level approaches to gene regulation. The formation of the calcified endoskeleton of the sea urchin embryo is a valuable experimental system for developing such an integrated view of the genomic regulatory control of morphogenesis. A transcriptional gene regulatory network (GRN) that underlies the specification of skeletogenic cells (primary mesenchyme cells, or PMCs) has recently been elucidated. In this study, we carried out a genome-wide analysis of mRNAs encoded by effector genes in the network and uncovered transcriptional inputs into many of these genes. We used RNA-seq to identify >400 transcripts differentially expressed by PMCs during gastrulation, when these cells undergo a striking sequence of behaviors that drives skeletal morphogenesis. Our analysis expanded by almost an order of magnitude the number of known (and candidate) downstream effectors that directly mediate skeletal morphogenesis. We carried out genome-wide analysis of (1) functional targets of Ets1 and Alx1, two pivotal, early transcription factors in the PMC GRN, and (2) functional targets of MAPK signaling, a pathway that plays an essential role in PMC specification. These studies identified transcriptional inputs into >200 PMC effector genes. Our work establishes a framework for understanding the genomic regulatory control of a major morphogenetic process and has important implications for reconstructing the evolution of biomineralization in metazoans.
Collapse
Affiliation(s)
- Kiran Rafiq
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
39
|
Kohl S, Hwang DY, Dworschak GC, Hilger AC, Saisawat P, Vivante A, Stajic N, Bogdanovic R, Reutter HM, Kehinde EO, Tasic V, Hildebrandt F. Mild recessive mutations in six Fraser syndrome-related genes cause isolated congenital anomalies of the kidney and urinary tract. J Am Soc Nephrol 2014; 25:1917-22. [PMID: 24700879 DOI: 10.1681/asn.2013101103] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) account for approximately 40% of children with ESRD in the United States. Hitherto, mutations in 23 genes have been described as causing autosomal dominant isolated CAKUT in humans. However, >90% of cases of isolated CAKUT still remain without a molecular diagnosis. Here, we hypothesized that genes mutated in recessive mouse models with the specific CAKUT phenotype of unilateral renal agenesis may also be mutated in humans with isolated CAKUT. We applied next-generation sequencing technology for targeted exon sequencing of 12 recessive murine candidate genes in 574 individuals with isolated CAKUT from 590 families. In 15 of 590 families, we identified recessive mutations in the genes FRAS1, FREM2, GRIP1, FREM1, ITGA8, and GREM1, all of which function in the interaction of the ureteric bud and the metanephric mesenchyme. We show that isolated CAKUT may be caused partially by mutations in recessive genes. Our results also indicate that biallelic missense mutations in the Fraser/MOTA/BNAR spectrum genes cause isolated CAKUT, whereas truncating mutations are found in the multiorgan form of Fraser syndrome. The newly identified recessive biallelic mutations in these six genes represent the molecular cause of isolated CAKUT in 2.5% of the 590 affected families in this study.
Collapse
Affiliation(s)
- Stefan Kohl
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daw-Yang Hwang
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Division of Nephrology, Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Gabriel C Dworschak
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Institute of Human Genetics, and
| | - Alina C Hilger
- Institute of Human Genetics, and Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Pawaree Saisawat
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Asaf Vivante
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Natasa Stajic
- Medical Faculty, University of Belgrade, Belgrade, Serbia; Institute of Mother and Child Healthcare of Serbia, Belgrade, Serbia
| | - Radovan Bogdanovic
- Medical Faculty, University of Belgrade, Belgrade, Serbia; Institute of Mother and Child Healthcare of Serbia, Belgrade, Serbia
| | - Heiko M Reutter
- Institute of Human Genetics, and Department of Neonatology, Children's Hospital, University of Bonn, Bonn, Germany
| | | | - Velibor Tasic
- Department of Pediatric Nephrology, University Children's Hospital, Skopje, Macedonia; and
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Howard Hughes Medical Institute, Chevy Chase, Maryland
| |
Collapse
|
40
|
Miller KA, Gordon CT, Welfare MF, Caruana G, Bertram JF, Bateman JF, Farlie PG. bfb, a novel ENU-induced blebs mutant resulting from a missense mutation in Fras1. PLoS One 2013; 8:e76342. [PMID: 24143185 PMCID: PMC3797057 DOI: 10.1371/journal.pone.0076342] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 08/23/2013] [Indexed: 12/28/2022] Open
Abstract
Fras1 is an extracellular matrix associated protein with essential roles in adhesion of epithelia and mesenchyme during early embryonic development. The adhesive function of Fras1 is achieved through interaction with a group of related proteins, Frem 1–3, and a cytoplasmic adaptor protein Grip1. Mutation of each of these proteins results in characteristic epithelial blistering and have therefore become known as “blebs” proteins. Human Fraser syndrome presents with a similar phenotype and the blebs mice have been instrumental in identification of the genetic basis of Fraser syndrome. We have identified a new ENU-induced blebs allele resulting from a novel missense mutation in Fras1. The resulting mouse strain, blood filled blisters (bfb), presents with a classic blebs phenotype but does not exhibit embryonic lethality typical of other blebs mutants and in addition, we report novel palate and sternal defects. Analysis of the bfb phenotype confirms the presence of epithelial-mesenchymal adhesion defects but also supports the emerging role of blebs proteins in regulating signalling during organogenesis. The bfb strain provides new opportunities to investigate the role of Fras1 in development.
Collapse
Affiliation(s)
- Kerry A. Miller
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Christopher T. Gordon
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Megan F. Welfare
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Georgina Caruana
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - John F. Bertram
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - John F. Bateman
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Departments of Paediatrics, Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Peter G. Farlie
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- * E-mail:
| |
Collapse
|
41
|
Beck TF, Shchelochkov OA, Yu Z, Kim BJ, Hernández-García A, Zaveri HP, Bishop C, Overbeek PA, Stockton DW, Justice MJ, Scott DA. Novel frem1-related mouse phenotypes and evidence of genetic interactions with gata4 and slit3. PLoS One 2013; 8:e58830. [PMID: 23536828 PMCID: PMC3594180 DOI: 10.1371/journal.pone.0058830] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 02/07/2013] [Indexed: 11/27/2022] Open
Abstract
The FRAS1-related extracellular matrix 1 (FREM1) gene encodes an extracellular matrix protein that plays a critical role in the development of multiple organ systems. In humans, recessive mutations in FREM1 cause eye defects, congenital diaphragmatic hernia, renal anomalies and anorectal malformations including anteriorly placed anus. A similar constellation of findings-microphthalmia, cryptophthalmos, congenital diaphragmatic hernia, renal agenesis and rectal prolapse-have been described in FREM1-deficient mice. In this paper, we identify a homozygous Frem1 missense mutation (c.1687A>T, p.Ile563Phe) in an N-ethyl-N-nitrosourea (ENU)-derived mouse strain, crf11, with microphthalmia, cryptophthalmos, renal agenesis and rectal prolapse. This mutation affects a highly conserved residue in FREM1's third CSPG domain. The p.Ile563Phe change is predicted to be deleterious and to cause decreased FREM1 protein stability. The crf11 allele also fails to complement the previously described eyes2 allele of Frem1 (p.Lys826*) providing further evidence that the crf11 phenotype is due to changes affecting Frem1 function. We then use mice bearing the crf11 and eyes2 alleles to identify lung lobulation defects and decreased anogenital distance in males as novel phenotypes associated with FREM1 deficiency in mice. Due to phenotypic overlaps between FREM1-deficient mice and mice that are deficient for the retinoic acid-responsive transcription factor GATA4 and the extracellular matrix protein SLIT3, we also perform experiments to look for in vivo genetic interactions between the genes that encode these proteins. These experiments reveal that Frem1 interacts genetically with Gata4 in the development of lung lobulation defects and with Slit3 in the development of renal agenesis. These results demonstrate that FREM1-deficient mice faithfully recapitulate many of the phenotypes seen in individuals with FREM1 deficiency and that variations in GATA4 and SLIT3 expression modulate some FREM1-related phenotypes in mice.
Collapse
Affiliation(s)
- Tyler F. Beck
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Oleg A. Shchelochkov
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa, United States of America
| | - Zhiyin Yu
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Bum Jun Kim
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Andrés Hernández-García
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hitisha P. Zaveri
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Colin Bishop
- The Wake Forest Institute for Regenerative Medicine, Winston Salem, North Carolina, United States of America
| | - Paul A. Overbeek
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - David W. Stockton
- Departments of Pediatrics and Internal Medicine, Wayne State University, Detroit, Michigan, United States of America
| | - Monica J. Justice
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Daryl A. Scott
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
42
|
Sato Y, Shimono C, Li S, Nakano I, Norioka N, Sugiura N, Kimata K, Yamada M, Sekiguchi K. Nephronectin binds to heparan sulfate proteoglycans via its MAM domain. Matrix Biol 2013; 32:188-95. [PMID: 23357641 DOI: 10.1016/j.matbio.2013.01.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 12/25/2012] [Accepted: 01/02/2013] [Indexed: 01/28/2023]
Abstract
Nephronectin is a basement membrane protein comprising five N-terminal epidermal growth factor (EGF)-like repeats, a central linker segment containing an Arg-Gly-Asp (RGD) motif and a C-terminal meprin-A5 protein-receptor protein tyrosine phosphatase μ (MAM) domain. Nephronectin has been shown to interact with α8β1 integrin through the central linker segment, but its interactions with other molecules remain to be elucidated. Here, we examined the binding of nephronectin to a panel of glycosaminoglycan (GAG) chains. Nephronectin bound strongly to heparin and chondroitin sulfate (CS)-E and moderately to heparan sulfate (HS), but failed to bind to CS-A, CS-C, CS-D, dermatan sulfate and hyaluronic acid. Deletion of the MAM domain severely impaired the binding of nephronectin to heparin but not CS-E, whereas deletion of the EGF-like repeats reduced its binding to CS-E but not heparin, suggesting that nephronectin interacts with CS-E and heparin through the EGF-like repeats and MAM domain, respectively. Consistent with these results, nephronectin bound to agrin and perlecan, which are heparan sulfate proteoglycans (HSPGs) in basement membranes, in HS-dependent manners. Site-directed mutagenesis of the MAM domain revealed that multiple basic amino acid residues in the putative loop regions were involved in the binding of the MAM domain to agrin. The binding of nephronectin to basement membrane HSPGs was further confirmed by in situ nephronectin overlay assays using mouse frozen tissue sections. Taken together, these findings indicate that nephronectin is capable of binding to HSPGs in basement membranes via the MAM domain, and thereby raise the possibility that interactions with basement membrane HSPGs may be involved in the deposition of nephronectin onto basement membranes.
Collapse
Affiliation(s)
- Yuya Sato
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Leslie M. Unfinished basement membrane spurs developmental disorder. J Biophys Biochem Cytol 2012. [PMCID: PMC3365492 DOI: 10.1083/jcb.1975iti3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|