1
|
Xu J, Zhao C, Kang Y. The Formation and Renewal of Photoreceptor Outer Segments. Cells 2024; 13:1357. [PMID: 39195247 PMCID: PMC11352558 DOI: 10.3390/cells13161357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
The visual system is essential for humans to perceive the environment. In the retina, rod and cone photoreceptor neurons are the initial sites where vision forms. The apical region of both cone and rod photoreceptors contains a light-sensing organelle known as the outer segment (OS), which houses tens of thousands of light-sensitive opsins. The OSs of photoreceptors are not static; they require rhythmic renewal to maintain normal physiological functions. Disruptions in OS renewal can lead to various genetic disorders, such as retinitis pigmentosa (RP). Understanding the patterns and molecular mechanisms of photoreceptor OS renewal remains one of the most intriguing topics in visual biology. This review aims to elucidate the structure of photoreceptor OSs, the molecular mechanisms underlying photoreceptor OS renewal, and the retinal diseases resulting from defects in this renewal process. Additionally, we will explore retinal diseases related to photoreceptor OS renewal and potential therapeutic strategies, concluding with a discussion on future research directions for OS renewal.
Collapse
Affiliation(s)
- Jingjin Xu
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (J.X.); (C.Z.)
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - Chengtian Zhao
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (J.X.); (C.Z.)
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - Yunsi Kang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (J.X.); (C.Z.)
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
2
|
Tamilselvan E, Sotomayor M. CELSR1, a core planar cell polarity protein, features a weakly adhesive and flexible cadherin ectodomain. Structure 2024; 32:476-491.e5. [PMID: 38307021 DOI: 10.1016/j.str.2024.01.003] [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: 05/14/2023] [Revised: 09/30/2023] [Accepted: 01/08/2024] [Indexed: 02/04/2024]
Abstract
Planar cell polarity (PCP), essential to multicellular developmental processes, arises when cells polarize and align across tissues. Central to PCP is CELSR1, an atypical cadherin featuring a long ectodomain with nine extracellular cadherin (EC) repeats, a membrane adjacent domain (MAD10), and several characteristic adhesion GPCR domains. Cell-based aggregation assays have demonstrated CELSR1's homophilic adhesive nature, but mechanistic details are missing. Here, we investigate the possible adhesive properties and structures of CELSR1 EC repeats. Our bead aggregation assays do not support strong adhesion by EC repeats alone. Consistently, EC1-4 only dimerizes at high concentration in solution. Crystal structures of human CELSR1 EC1-4 and EC4-7 reveal typical folds and a non-canonical linker between EC5 and EC6. Simulations and experiments using EC4-7 indicate flexibility at EC5-6, and solution experiments show EC7-MAD10-mediated dimerization. Our results suggest weak homophilic adhesion by CELSR1 cadherin repeats and provide mechanistic insights into the structural determinants of CELSR1 function.
Collapse
Affiliation(s)
- Elakkiya Tamilselvan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
3
|
Chess MM, Douglas W, Saunders J, Ettensohn CA. Genome-wide identification and spatiotemporal expression analysis of cadherin superfamily members in echinoderms. EvoDevo 2023; 14:15. [PMID: 38124068 PMCID: PMC10734073 DOI: 10.1186/s13227-023-00219-7] [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: 10/09/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Cadherins are calcium-dependent transmembrane cell-cell adhesion proteins that are essential for metazoan development. They consist of three subfamilies: classical cadherins, which bind catenin, protocadherins, which contain 6-7 calcium-binding repeat domains, and atypical cadherins. Their functions include forming adherens junctions, establishing planar cell polarity (PCP), and regulating cell shape, proliferation, and migration. Because they are basal deuterostomes, echinoderms provide important insights into bilaterian evolution, but their only well-characterized cadherin is G-cadherin, a classical cadherin that is expressed by many embryonic epithelia. We aimed to better characterize echinoderm cadherins by conducting phylogenetic analyses and examining the spatiotemporal expression patterns of cadherin-encoding genes during Strongylocentrotus purpuratus development. RESULTS Our phylogenetic analyses conducted on two echinoid, three asteroid, and one crinoid species identified ten echinoderm cadherins, including one deuterostome-specific ortholog, cadherin-23, and an echinoderm-specific atypical cadherin that possibly arose in an echinoid-asteroid ancestor. Catenin-binding domains in dachsous-2 orthologs were found to be a deuterostome-specific innovation that was selectively lost in mouse, while those in Fat4 orthologs appeared to be Ambulacraria-specific and were selectively lost in non-crinoid echinoderms. The identified suite of echinoderm cadherins lacks vertebrate-specific innovations but contains two proteins that are present in protostomes and absent from mouse. The spatiotemporal expression patterns of four embryonically expressed cadherins (fat atypical cadherins 1 and 4, dachsous-2, and protocadherin-9) were dynamic and mirrored the expression pattern of Frizzled 5/8, a non-canonical Wnt PCP pathway receptor protein essential for archenteron morphogenesis. CONCLUSIONS The echinoderm cadherin toolkit is more similar to that of an ancient bilaterian predating protostomes and deuterostomes than it is to the suite of cadherins found in extant vertebrates. However, it also appears that deuterostomes underwent several cadherin-related innovations. Based on their similar spatiotemporal expression patterns and orthologous relationships to PCP-related and tumor-suppressing proteins, we hypothesize that sea urchin cadherins may play a role in regulating the shape and growth of embryonic epithelia and organs. Future experiments will examine cadherin expression in non-echinoid echinoderms and explore the functions of cadherins during echinoderm development.
Collapse
Affiliation(s)
- Macie M Chess
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - William Douglas
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Josiah Saunders
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Charles A Ettensohn
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
4
|
Klumpe HE, Garcia-Ojalvo J, Elowitz MB, Antebi YE. The computational capabilities of many-to-many protein interaction networks. Cell Syst 2023; 14:430-446. [PMID: 37348461 PMCID: PMC10318606 DOI: 10.1016/j.cels.2023.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/14/2023] [Accepted: 05/11/2023] [Indexed: 06/24/2023]
Abstract
Many biological circuits comprise sets of protein variants that interact with one another in a many-to-many, or promiscuous, fashion. These architectures can provide powerful computational capabilities that are especially critical in multicellular organisms. Understanding the principles of biochemical computations in these circuits could allow more precise control of cellular behaviors. However, these systems are inherently difficult to analyze, due to their large number of interacting molecular components, partial redundancies, and cell context dependence. Here, we discuss recent experimental and theoretical advances that are beginning to reveal how promiscuous circuits compute, what roles those computations play in natural biological contexts, and how promiscuous architectures can be applied for the design of synthetic multicellular behaviors.
Collapse
Affiliation(s)
- Heidi E Klumpe
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Biological Design Center, Boston University, Boston, MA 02215, USA
| | - Jordi Garcia-Ojalvo
- Department of Medicine and Life Sciences, Pompeu Fabra University, 08003 Barcelona, Spain.
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Yaron E Antebi
- Department of Molecular Genetics, Weizmann Institute of Science 76100, Rehovot, Israel.
| |
Collapse
|
5
|
Identification of tumor antigens and immune subtypes of acute myeloid leukemia for mRNA vaccine development. Clin Transl Oncol 2023:10.1007/s12094-023-03108-6. [PMID: 36781600 PMCID: PMC9924891 DOI: 10.1007/s12094-023-03108-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 01/28/2023] [Indexed: 02/15/2023]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a highly aggressive hematological malignancy, and there has not been any significant improvement in therapy of AML over the past several decades. The mRNA vaccines have become a promising strategy against multiple cancers, however, its application on AML remains undefined. In this study, we aimed to identify novel antigens for developing mRNA vaccines against AML and explore the immune landscape of AML to select appropriate patients for vaccination. METHODS Genomic data and gene mutation data were retrieved from TCGA, GEO and cBioPortal, respectively. GEPIA2 was used to analyze differentially expressed genes. The single cell RNA-seq database Tumor Immune Single-cell Hub (TISCH) was used to explore the association between the potential tumor antigens and the infiltrating immune cells in the bone marrow. Consensus clustering analysis was applied to identify distinct immune subtypes. The correlation between the abundance of antigen presenting cells and the expression level of antigens was evaluated using Spearman correlation analysis. The characteristics of the tumor immune microenvironment in each subtype were investigated based on single-sample gene set enrichment analysis. RESULTS Five potential tumor antigens were identified for mRNA vaccine from the pool of overexpressed and mutated genes, including CDH23, LRP1, MEFV, MYOF and SLC9A9, which were associated with infiltration of antigen-presenting immune cells (APCs). AML patients were stratified into two immune subtypes Cluster1 (C1) and Cluster2 (C2), which were characterized by distinct molecular and clinical features. C1 subtype demonstrated an immune-hot and immunosuppressive phenotype, while the C1 subtype had an immune-cold phenotype. Furthermore, the two immune subtype showed remarkably different expression of immune checkpoints, immunogenic cell death modulators and human leukocyte antigens. CONCLUSION CDH23, LRP1, MEFV, MYOF and SLC9A9 were potential antigens for developing AML mRNA vaccine, and AML patients in immune subtype 1 were suitable for vaccination.
Collapse
|
6
|
Suzuki H, Iwamoto H, Yamamoto K, Tsukaguchi M, Nakamura T, Masuda A, Sakaue T, Tanaka T, Niizeki T, Okamura S, Shimose S, Shirono T, Noda Y, Kamachi N, Kuromatsu R, Hisaka T, Yano H, Koga H, Torimura T. DNA Methylation in Noncancerous Liver Tissues as Biomarker for Multicentric Occurrence of Hepatitis C Virus-Related Hepatocellular Carcinoma. GASTRO HEP ADVANCES 2022; 1:555-562. [PMID: 39132059 PMCID: PMC11307517 DOI: 10.1016/j.gastha.2022.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/16/2022] [Indexed: 08/13/2024]
Abstract
Background and Aims Hepatitis C virus (HCV)-related hepatocellular carcinoma (HCC) progresses with a highly multicentric occurrence (MO) even after radical hepatectomy. Despite several efforts to clarify the pathogenesis of MO, the underlying molecular mechanism remains elusive. The aim of this study was to evaluate alterations in DNA methylation in noncancerous liver tissues in the MO of HCC. Methods A total of 203 patients with HCV-related HCC who underwent radical hepatectomy at our hospital between January 2008 and January 2012 were recruited. We defined a group of nonearly recurrence of HCC (NR) for ≥3 years after radical hepatectomy and a group of early recurrence of HCC (ER) with MO within 2 years after radical hepatectomy. Results Three patients each were selected in the NR and ER groups in the first set, and 13 patients in the NR group and 17 patients in the ER group were selected in the second set. Genome-wide DNA methylation profiles were obtained from noncancerous liver tissues using a Human Methylation 450 BeadChip, and the differences between the groups were analyzed for each set. After excluding single nucleotide polymorphism-associated methylation sites and low-call sites, 401,282 sites were assessed using a generalized linear model without any adjustments. Nine gene regions, APBB1P, CLSTN3, DLG5, IRX5, OAS1, SOX12, SNX19, TENM2, and TRIM54, exhibiting a significant difference (P < .001) in DNA methylation levels were identified in the common direction between the 2 analysis sets. Conclusion Alterations in DNA methylation of 9 genes in noncancerous liver tissues appear to be involved in MO after radical hepatectomy for HCV-related HCC.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hideki Iwamoto
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Ken Yamamoto
- Department of Medical Biochemistry, Kurume University School of Medicine, Kurume, Japan
| | - Mai Tsukaguchi
- Department of Medical Biochemistry, Kurume University School of Medicine, Kurume, Japan
| | - Toru Nakamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Atsutaka Masuda
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takahiko Sakaue
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Toshimitsu Tanaka
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takashi Niizeki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Shusuke Okamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Shigeo Shimose
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tomotake Shirono
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Yu Noda
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Naoki Kamachi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Ryoko Kuromatsu
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Toru Hisaka
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Hirohisa Yano
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Hironori Koga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
7
|
Neel BL, Nisler CR, Walujkar S, Araya-Secchi R, Sotomayor M. Elastic versus brittle mechanical responses predicted for dimeric cadherin complexes. Biophys J 2022; 121:1013-1028. [PMID: 35151631 PMCID: PMC8943749 DOI: 10.1016/j.bpj.2022.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/02/2022] [Accepted: 02/07/2022] [Indexed: 12/15/2022] Open
Abstract
Cadherins are a superfamily of adhesion proteins involved in a variety of biological processes that include the formation of intercellular contacts, the maintenance of tissue integrity, and the development of neuronal circuits. These transmembrane proteins are characterized by ectodomains composed of a variable number of extracellular cadherin (EC) repeats that are similar but not identical in sequence and fold. E-cadherin, along with desmoglein and desmocollin proteins, are three classical-type cadherins that have slightly curved ectodomains and engage in homophilic and heterophilic interactions through an exchange of conserved tryptophan residues in their N-terminal EC1 repeat. In contrast, clustered protocadherins are straighter than classical cadherins and interact through an antiparallel homophilic binding interface that involves overlapped EC1 to EC4 repeats. Here we present molecular dynamics simulations that model the adhesive domains of these cadherins using available crystal structures, with systems encompassing up to 2.8 million atoms. Simulations of complete classical cadherin ectodomain dimers predict a two-phased elastic response to force in which these complexes first softly unbend and then stiffen to unbind without unfolding. Simulated α, β, and γ clustered protocadherin homodimers lack a two-phased elastic response, are brittle and stiffer than classical cadherins and exhibit complex unbinding pathways that in some cases involve transient intermediates. We propose that these distinct mechanical responses are important for function, with classical cadherin ectodomains acting as molecular shock absorbers and with stiffer clustered protocadherin ectodomains facilitating overlap that favors binding specificity over mechanical resilience. Overall, our simulations provide insights into the molecular mechanics of single cadherin dimers relevant in the formation of cellular junctions essential for tissue function.
Collapse
Affiliation(s)
- Brandon L Neel
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; The Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio
| | - Collin R Nisler
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; Biophysics Graduate Program, The Ohio State University, Columbus, Ohio
| | - Sanket Walujkar
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; Chemical Physics Graduate Program, The Ohio State University, Columbus, Ohio
| | - Raul Araya-Secchi
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago, Chile
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; The Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio; Biophysics Graduate Program, The Ohio State University, Columbus, Ohio; Chemical Physics Graduate Program, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
8
|
Neel BL, Nisler CR, Walujkar S, Araya-Secchi R, Sotomayor M. Collective mechanical responses of cadherin-based adhesive junctions as predicted by simulations. Biophys J 2022; 121:991-1012. [PMID: 35150618 PMCID: PMC8943820 DOI: 10.1016/j.bpj.2022.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/02/2022] [Accepted: 02/07/2022] [Indexed: 12/13/2022] Open
Abstract
Cadherin-based adherens junctions and desmosomes help stabilize cell-cell contacts with additional function in mechano-signaling, while clustered protocadherin junctions are responsible for directing neuronal circuits assembly. Structural models for adherens junctions formed by epithelial cadherin (CDH1) proteins indicate that their long, curved ectodomains arrange to form a periodic, two-dimensional lattice stabilized by tip-to-tip trans interactions (across junction) and lateral cis contacts. Less is known about the exact architecture of desmosomes, but desmoglein (DSG) and desmocollin (DSC) cadherin proteins are also thought to form ordered junctions. In contrast, clustered protocadherin (PCDH)-based cell-cell contacts in neuronal tissues are thought to be responsible for self-recognition and avoidance, and structural models for clustered PCDH junctions show a linear arrangement in which their long and straight ectodomains form antiparallel overlapped trans complexes. Here, we report all-atom molecular dynamics simulations testing the mechanics of minimalistic adhesive junctions formed by CDH1, DSG2 coupled to DSC1, and PCDHγB4, with systems encompassing up to 3.7 million atoms. Simulations generally predict a favored shearing pathway for the adherens junction model and a two-phased elastic response to tensile forces for the adhesive adherens junction and the desmosome models. Complexes within these junctions first unbend at low tensile force and then become stiff to unbind without unfolding. However, cis interactions in both the CDH1 and DSG2-DSC1 systems dictate varied mechanical responses of individual dimers within the junctions. Conversely, the clustered protocadherin PCDHγB4 junction lacks a distinct two-phased elastic response. Instead, applied tensile force strains trans interactions directly, as there is little unbending of monomers within the junction. Transient intermediates, influenced by new cis interactions, are observed after the main rupture event. We suggest that these collective, complex mechanical responses mediated by cis contacts facilitate distinct functions in robust cell-cell adhesion for classical cadherins and in self-avoidance signaling for clustered PCDHs.
Collapse
Affiliation(s)
- Brandon L Neel
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; The Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio
| | - Collin R Nisler
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; Biophysics Graduate Program, The Ohio State University, Columbus, Ohio
| | - Sanket Walujkar
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; Chemical Physics Graduate Program, The Ohio State University, Columbus, Ohio
| | - Raul Araya-Secchi
- Facultad de Ingenieria y Tecnologia, Universidad San Sebastian, Santiago, Chile
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; The Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio; Biophysics Graduate Program, The Ohio State University, Columbus, Ohio; Chemical Physics Graduate Program, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
9
|
Yusuf IH, Garrett A, MacLaren RE, Issa PC. Retinal cadherins and the retinal cadherinopathies: Current concepts and future directions. Prog Retin Eye Res 2022; 90:101038. [DOI: 10.1016/j.preteyeres.2021.101038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022]
|
10
|
Gray ME, Johnson ZR, Modak D, Tamilselvan E, Tyska MJ, Sotomayor M. Heterophilic and homophilic cadherin interactions in intestinal intermicrovillar links are species dependent. PLoS Biol 2021; 19:e3001463. [PMID: 34871294 PMCID: PMC8691648 DOI: 10.1371/journal.pbio.3001463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/21/2021] [Accepted: 10/30/2021] [Indexed: 11/19/2022] Open
Abstract
Enterocytes are specialized epithelial cells lining the luminal surface of the small intestine that build densely packed arrays of microvilli known as brush borders. These microvilli drive nutrient absorption and are arranged in a hexagonal pattern maintained by intermicrovillar links formed by 2 nonclassical members of the cadherin superfamily of calcium-dependent cell adhesion proteins: protocadherin-24 (PCDH24, also known as CDHR2) and the mucin-like protocadherin (CDHR5). The extracellular domains of these proteins are involved in heterophilic and homophilic interactions important for intermicrovillar function, yet the structural determinants of these interactions remain unresolved. Here, we present X-ray crystal structures of the PCDH24 and CDHR5 extracellular tips and analyze their species-specific features relevant for adhesive interactions. In parallel, we use binding assays to identify the PCDH24 and CDHR5 domains involved in both heterophilic and homophilic adhesion for human and mouse proteins. Our results suggest that homophilic and heterophilic interactions involving PCDH24 and CDHR5 are species dependent with unique and distinct minimal adhesive units.
Collapse
Affiliation(s)
- Michelle E. Gray
- Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio, United States of America
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Zachary R. Johnson
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Debadrita Modak
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Elakkiya Tamilselvan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Matthew J. Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Marcos Sotomayor
- Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio, United States of America
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
11
|
Passanha FR, Geuens T, LaPointe VLS. Sticking together: Harnessing cadherin biology for tissue engineering. Acta Biomater 2021; 134:107-115. [PMID: 34358698 DOI: 10.1016/j.actbio.2021.07.070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 12/30/2022]
Abstract
Directing cell behavior and building a tissue for therapeutic impact is the main goal of regenerative medicine, for which scientists need to modulate the interaction of cells with biomaterials. The focus of the field thus far has been on the incorporation of cues from the extracellular matrix but we propose that scientists take lessons from cell-cell adhesion proteins, more specifically cadherin biology, as these proteins make multicellularity possible. In this perspective, we re-examine cadherins through the lens of a tissue engineer for the purpose of advancing regenerative medicine. Furthermore, we summarize exciting developments in biomaterials inspired by cadherins and discuss some challenges and opportunities for the future. STATEMENT OF SIGNIFICANCE: Tissue engineers need tools to direct cell behavior. To date, tissue engineers have designed many sophisticated materials to positively influence cell behavior but are faced with the challenge where these materials sometimes work and sometimes fail. This uncertainty is a big unanswered question that challenges the community. We propose that tissue engineering could be more successful if they would take lessons from cell-cell adhesion proteins, more specifically cadherin biology. In the article, we discuss key structural and functional characteristics that make cadherins ideal for tissue engineering approaches. Furthermore, by providing a state-of-the-art overview of exemplary studies that have used cadherins to influence cell behavior, we show tissue engineers that they already have the tools necessary to incorporate this knowledge.
Collapse
Affiliation(s)
- Fiona R Passanha
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands.
| | - Thomas Geuens
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
| | - Vanessa L S LaPointe
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands.
| |
Collapse
|
12
|
Zhao Z, Li S, Li S, Wang J, Lin H, Fu W. High expression of oncogene cadherin-6 correlates with tumor progression and a poor prognosis in gastric cancer. Cancer Cell Int 2021; 21:493. [PMID: 34530820 PMCID: PMC8447617 DOI: 10.1186/s12935-021-02071-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/30/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common and fatal cancers worldwide. Effective biomarkers to aid the early diagnosis of GC, as well as predict the course of disease, are urgently needed. Hence, we explored the role and function of cadherin-6 (CDH6) in the diagnosis and prognosis of gastric cancer. METHODS The expression levels of CDH6 in cancerous and normal gastric tissue were analyzed using multiple public databases. Gene set enrichment analysis (GSEA) was performed using The Cancer Genome Atlas (TCGA) dataset. The diagnostic efficiency of CDH6 expression in GC patients was determined through receiver operating characteristic (ROC) curve analysis. The associations between clinical variables and CDH6 expression were evaluated statistically, and the prognostic factors for overall survival were analyzed by univariate and multivariate Cox regression. 44 GC tissue samples, 20 donor-matched adjacent normal tissue samples, and associated detailed clinical information, were collected from the Tianjin Medical University General Hospital. CDH6 expression levels were determined for further validation. RESULTS CDH6 was upregulated in GC samples compared to normal gastric tissue. Furthermore, GSEA identified the tricarboxylic acid (TCA) cycle, extracellular matrix (ECM) receptor interaction, glyoxylate and dicarboxylate metabolism, oxidative phosphorylation, and the pentose phosphate pathway as differentially enriched in GC tissue samples. According to the area under the ROC curve (AUC) values (AUC = 0.829 in the TCGA and 0.966 in the GSE54129 dataset), CDH6 expression was associated with high diagnostic efficacy. Patients with high CDH6 levels in their GC tissues had a higher T number (according to the TNM classification) and a worse prognosis than those with low CDH6 expression. Univariate and multivariate Cox regression analysis showed that CDH6 was an independent risk factor for overall survival (univariate: HR = 1.305, P = 0.002, multivariate: HR = 1.481, P < 0.001). CONCLUSION CDH6 was upregulated in GC, and high CDH6 expression was indicative of a higher T number and a worse prognosis. Therefore, CDH6 represents a potentially independent molecular biomarker for the diagnostic and prognostic prediction of GC.
Collapse
Affiliation(s)
- Zongxian Zhao
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, China.
| | - Shuliang Li
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Liaocheng, Shangdong, China.
| | - Shilong Li
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, China
| | - Jun Wang
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, China
| | - Hai Lin
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, China
| | - Weihua Fu
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, China.
| |
Collapse
|
13
|
Zhang YM. Orosomucoid-like protein 3, rhinovirus and asthma. World J Crit Care Med 2021; 10:170-182. [PMID: 34616654 PMCID: PMC8462028 DOI: 10.5492/wjccm.v10.i5.170] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/16/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
The genetic variants of orosomucoid-like protein 3 (ORMDL3) gene are associated with highly significant increases in the number of human rhinovirus (HRV)-induced wheezing episodes in children. Recent investigations have been focused on the mechanisms of ORMDL3 in rhinovirus infection for asthma and asthma exacerbations. ORMDL3 not only regulates major human rhinovirus receptor intercellular adhesion molecule 1 expression, but also plays pivotal roles in viral infection through metabolisms of ceramide and sphingosine-1-phosphate, endoplasmic reticulum (ER) stress, ER-Golgi interface and glycolysis. Research on the roles of ORMDL3 in HRV infection will lead us to identify new biomarkers and novel therapeutic targets in childhood asthma and viral induced asthma exacerbations.
Collapse
Affiliation(s)
- You-Ming Zhang
- Section of Genomic and Environmental Medicine, National Heart and Lung Institute, Molecular Genetics Group, Division of Respiratory Sciences, Imperial College London, London SW3 6LY, United Kingdom
| |
Collapse
|
14
|
Rahman MS, Winsvold BS, Chavez Chavez SO, Børte S, Tsepilov YA, Sharapov SZ, Aulchenko YS, Hagen K, Fors EA, Hveem K, Zwart JA, van Meurs JB, Freidin MB, Williams FM. Genome-wide association study identifies RNF123 locus as associated with chronic widespread musculoskeletal pain. Ann Rheum Dis 2021; 80:1227-1235. [PMID: 33926923 PMCID: PMC8372387 DOI: 10.1136/annrheumdis-2020-219624] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND OBJECTIVES Chronic widespread musculoskeletal pain (CWP) is a symptom of fibromyalgia and a complex trait with poorly understood pathogenesis. CWP is heritable (48%-54%), but its genetic architecture is unknown and candidate gene studies have produced inconsistent results. We conducted a genome-wide association study to get insight into the genetic background of CWP. METHODS Northern Europeans from UK Biobank comprising 6914 cases reporting pain all over the body lasting >3 months and 242 929 controls were studied. Replication of three independent genome-wide significant single nucleotide polymorphisms was attempted in six independent European cohorts (n=43 080; cases=14 177). Genetic correlations with risk factors, tissue specificity and colocalisation were examined. RESULTS Three genome-wide significant loci were identified (rs1491985, rs10490825, rs165599) residing within the genes Ring Finger Protein 123 (RNF123), ATPase secretory pathway Ca2+transporting 1 (ATP2C1) and catechol-O-methyltransferase (COMT). The RNF123 locus was replicated (meta-analysis p=0.0002), the ATP2C1 locus showed suggestive association (p=0.0227) and the COMT locus was not replicated. Partial genetic correlation between CWP and depressive symptoms, body mass index, age of first birth and years of schooling were identified. Tissue specificity and colocalisation analysis highlight the relevance of skeletal muscle in CWP. CONCLUSIONS We report a novel association of RNF123 locus and a suggestive association of ATP2C1 locus with CWP. Both loci are consistent with a role of calcium regulation in CWP. The association with COMT, one of the most studied genes in chronic pain field, was not confirmed in the replication analysis.
Collapse
Affiliation(s)
- Md Shafiqur Rahman
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| | - Bendik S Winsvold
- Department of Research, Innovation and Education,Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- Department of Neurology, Oslo universitetssykehus Ullevål, Oslo, Norway
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sergio O Chavez Chavez
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Zuid-Holland, The Netherlands
| | - Sigrid Børte
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Research and Communication Unit for Musculoskeletal Health (FORMI), Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Yakov A Tsepilov
- Laboratory of Theoretical and Applied Functional Genomics, Novosibirsk State University, Novosibirsk, 630090, Novosibirskaâ, Russia
- PolyOmica, 's-Hertogenbosch, PA, The Netherlands
- Laboratory of Recombination and Segregation Analysis, Institute of Cytology and Genetics, 10 Lavrentiev Avenue, Novosibirsk, 630090, Russia
| | - Sodbo Zh Sharapov
- Laboratory of Theoretical and Applied Functional Genomics, Novosibirsk State University, Novosibirsk, 630090, Novosibirskaâ, Russia
- Laboratory of Recombination and Segregation Analysis, Institute of Cytology and Genetics, 10 Lavrentiev Avenue, Novosibirsk, 630090, Russia
| | - Yurii S Aulchenko
- Laboratory of Theoretical and Applied Functional Genomics, Novosibirsk State University, Novosibirsk, 630090, Novosibirskaâ, Russia
- PolyOmica, 's-Hertogenbosch, PA, The Netherlands
| | - Knut Hagen
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Clinical Research Unit Central Norway, St Olavs University Hospital, Trondheim, Norway
| | - Egil A Fors
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kristian Hveem
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Center, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - John Anker Zwart
- Department of Research, Innovation and Education,Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Institute of Clinical Medicine,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Joyce B van Meurs
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Zuid-Holland, The Netherlands
| | - Maxim B Freidin
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| | - Frances Mk Williams
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| |
Collapse
|
15
|
Munteanu CVA, Chirițoiu GN, Chirițoiu M, Ghenea S, Petrescu AJ, Petrescu ȘM. Affinity proteomics and deglycoproteomics uncover novel EDEM2 endogenous substrates and an integrative ERAD network. Mol Cell Proteomics 2021; 20:100125. [PMID: 34332121 PMCID: PMC8455867 DOI: 10.1016/j.mcpro.2021.100125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 07/09/2021] [Accepted: 07/25/2021] [Indexed: 02/08/2023] Open
Abstract
Various pathologies result from disruptions to or stress of endoplasmic reticulum (ER) homeostasis, such as Parkinson's disease and most neurodegenerative illnesses, diabetes, pulmonary fibrosis, viral infections and cancers. A critical process in maintaining ER homeostasis is the selection of misfolded proteins by the ER quality-control system (ERQC) for destruction via ER-associated degradation (ERAD). One key protein proposed to act during the first steps of misfolded glycoprotein degradation is the ER degradation-enhancing α-mannosidase-like protein 2 (EDEM2). Therefore, characterization of the EDEM2 associated proteome is of great interest. We took advantage of using melanoma cells overexpressing EDEM2 as a cancer model system, to start documenting at the deglycoproteome level (N-glycosites identification) the emerging link between ER homeostasis and cancer progression. The dataset created for identifying the EDEM2 glyco-clients carrying high mannose/hybrid N-glycans provides a comprehensive N-glycosites analysis mapping over 1000 N-glycosites on more than 600 melanoma glycoproteins. To identify EDEM2-associated proteins we used affinity-proteomics and proteome-wide analysis of sucrose density fractionation in an integrative workflow. Using intensity and spectral count-based quantification, we identify seven new EDEM2 partners, all of which are involved in ERQC and ERAD. Moreover, we defined novel endogenous candidates for EDEM2-dependent ERAD by combining deglycoproteomics, SILAC-based proteomics, and biochemical methods. These included tumor antigens and several ER-transiting endogenous melanoma proteins, including ITGA1 and PCDH2, the expression of which was negatively correlated with that of EDEM2. Tumor antigens are key in the antigen presentation process, whilst ITGA1 and PCDH2 are involved in melanoma metastasis and invasion. EDEM2 could therefore have a regulatory role in melanoma through the modulation of these glycoproteins degradation and trafficking. The data presented herein suggest that EDEM2 is involved in ER homeostasis to a greater extent than previously suggested.
Collapse
Affiliation(s)
- Cristian V A Munteanu
- Department of Bioinformatics and Structural Biochemistry, Institute of Biochemistry, Splaiul Independenței 296, 060031, Bucharest, Romania
| | - Gabriela N Chirițoiu
- Department of Molecular Cell Biology, Institute of Biochemistry, Splaiul Independenței 296, 060031, Bucharest, Romania
| | - Marioara Chirițoiu
- Department of Molecular Cell Biology, Institute of Biochemistry, Splaiul Independenței 296, 060031, Bucharest, Romania
| | - Simona Ghenea
- Department of Molecular Cell Biology, Institute of Biochemistry, Splaiul Independenței 296, 060031, Bucharest, Romania
| | - Andrei-Jose Petrescu
- Department of Bioinformatics and Structural Biochemistry, Institute of Biochemistry, Splaiul Independenței 296, 060031, Bucharest, Romania
| | - Ștefana M Petrescu
- Department of Molecular Cell Biology, Institute of Biochemistry, Splaiul Independenței 296, 060031, Bucharest, Romania.
| |
Collapse
|
16
|
Cao B, Guo X, Huang L, Wang B, Wang W, Han D, Zhang W, Zhong K. Methylation silencing CDH23 is a poor prognostic marker in diffuse large B-cell lymphoma. Aging (Albany NY) 2021; 13:17768-17788. [PMID: 34252883 PMCID: PMC8312441 DOI: 10.18632/aging.203268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/17/2021] [Indexed: 12/20/2022]
Abstract
Cadherin-23(CDH23) mediates homotypic and heterotypic cell-cell adhesions in cancer cells. However, the epigenetic regulation, the biological functions, the mechanisms and the prognostic value of CDH23 in diffuse large B-cell lymphoma (DLBCL) are still unclear. The Gene Expression Profiling Interactive Analysis (GEPIA) and the Gene Expression Omnibus (GEO) database were employed to analyze the CDH23 expression level in DLBCL. The correlation of CDH23 expression and methylation was analyzed by LinkedOmics database. The prognostic value was analyzed via GEPIA. Correlated genes, target kinase, target miRNA, target transcription factor and biological functions were identified by LinkedOmics and GeneMANIA database. The relationship between CDH23 and the immune cell infiltration was explored by the Tumor Immune Estimation Resource (TIMER). The expression of CDH23 was reduced by DNA methylation significantly in DLBCL tissue. Reduction of CDH23 represented poor outcome of DLBCL patients. Functional enrichment analysis showed that CDH23 mainly enriched in cancer cell growth, cell metastasis, cell adhesion, cell cycle, drug catabolic process, leukocyte mediated immunity and DNA repair by some cancer related kinases, miRNAs and transcription factors. These results indicated that methylated reduction of CDH23 represented poor outcome of DLBCL. CDH23 is associated with essential biological functions and key molecules in DLBCL. CDH23 may play crucial roles in DLBCL tumorigenesis. Our results lay a foundation for further investigation of the role of CDH23 in DLBCL tumorigenesis.
Collapse
Affiliation(s)
- Baoping Cao
- Department of Lymphoma, Beijing Shijitan Hospital, Capital Medical University, Haidian 100038, Beijing, China
| | - Xiaochuan Guo
- Department of Lymphoma, Beijing Shijitan Hospital, Capital Medical University, Haidian 100038, Beijing, China
| | - Lefu Huang
- Department of Lymphoma, Beijing Shijitan Hospital, Capital Medical University, Haidian 100038, Beijing, China
| | - Bin Wang
- Department of Lymphoma, Beijing Shijitan Hospital, Capital Medical University, Haidian 100038, Beijing, China
| | - Weixia Wang
- Department of Lymphoma, Beijing Shijitan Hospital, Capital Medical University, Haidian 100038, Beijing, China
| | - Dong Han
- Department of Lymphoma, Beijing Shijitan Hospital, Capital Medical University, Haidian 100038, Beijing, China
| | - Weijing Zhang
- Department of Lymphoma, Beijing Shijitan Hospital, Capital Medical University, Haidian 100038, Beijing, China
| | - Kaili Zhong
- Department of Lymphoma, Beijing Shijitan Hospital, Capital Medical University, Haidian 100038, Beijing, China
| |
Collapse
|
17
|
Bartolomé RA, Robles J, Martin‐Regalado Á, Pintado‐Berninches L, Burdiel M, Jaén M, Aizpurúa C, Imbaud JI, Casal JI. CDH6-activated αIIbβ3 crosstalks with α2β1 to trigger cellular adhesion and invasion in metastatic ovarian and renal cancers. Mol Oncol 2021; 15:1849-1865. [PMID: 33715292 PMCID: PMC8253092 DOI: 10.1002/1878-0261.12947] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/25/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
Cadherin 6 (CDH6) is significantly overexpressed in advanced ovarian and renal cancers. However, the role of CDH6 in cancer metastasis is largely unclear. Here, we investigated the impact of CDH6 expression on integrin-mediated metastatic progression. CDH6 preferentially bound to αIIbβ3 integrin, a platelet receptor scarcely expressed in cancer cells, and this interaction was mediated through the cadherin Arginine-glycine-aspartic acid (RGD) motif. Furthermore, CDH6 and CDH17 were found to interact with α2β1 in αIIbβ3low cells. Transient silencing of CDH6, ITGA2B, or ITGB3 genes caused a significant loss of proliferation, adhesion, invasion, and lung colonization through the downregulation of SRC, FAK, AKT, and ERK signaling. In ovarian and renal cancer cells, integrin αIIbβ3 activation appears to be a prerequisite for proper α2β1 activation. Interaction of αIIbβ3 with CDH6, and subsequent αIIbβ3 activation, promoted activation of α2β1 and cell adhesion in ovarian and renal cancer cells. Additionally, monoclonal antibodies specific to the cadherin RGD motif and clinically approved αIIbβ3 inhibitors could block pro-metastatic activity in ovarian and renal tumors. In summary, the interaction between CDH6 and αIIbβ3 regulates α2β1-mediated adhesion and invasion of ovarian and renal cancer metastatic cells and constitutes a therapeutic target of broad potential for treating metastatic progression.
Collapse
Affiliation(s)
- Rubén A. Bartolomé
- Department of Biomolecular MedicineCentro de Investigaciones BiológicasCSICMadridSpain
| | - Javier Robles
- Department of Biomolecular MedicineCentro de Investigaciones BiológicasCSICMadridSpain
| | | | | | - Miranda Burdiel
- Department of Biomolecular MedicineCentro de Investigaciones BiológicasCSICMadridSpain
| | - Marta Jaén
- Department of Biomolecular MedicineCentro de Investigaciones BiológicasCSICMadridSpain
| | | | | | - José Ignacio Casal
- Department of Biomolecular MedicineCentro de Investigaciones BiológicasCSICMadridSpain
| |
Collapse
|
18
|
Gray ME, Sotomayor M. Crystal structure of the nonclassical cadherin-17 N-terminus and implications for its adhesive binding mechanism. Acta Crystallogr F Struct Biol Commun 2021; 77:85-94. [PMID: 33682793 PMCID: PMC7938635 DOI: 10.1107/s2053230x21002247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/25/2021] [Indexed: 12/27/2022] Open
Abstract
The cadherin superfamily of calcium-dependent cell-adhesion proteins has over 100 members in the human genome. All members of the superfamily feature at least a pair of extracellular cadherin (EC) repeats with calcium-binding sites in the EC linker region. The EC repeats across family members form distinct complexes that mediate cellular adhesion. For instance, classical cadherins (five EC repeats) strand-swap their N-termini and exchange tryptophan residues in EC1, while the clustered protocadherins (six EC repeats) use an extended antiparallel `forearm handshake' involving repeats EC1-EC4. The 7D-cadherins, cadherin-16 (CDH16) and cadherin-17 (CDH17), are the most similar to classical cadherins and have seven EC repeats, two of which are likely to have arisen from gene duplication of EC1-2 from a classical ancestor. However, CDH16 and CDH17 lack the EC1 tryptophan residue used by classical cadherins to mediate adhesion. The structure of human CDH17 EC1-2 presented here reveals features that are not seen in classical cadherins and that are incompatible with the EC1 strand-swap mechanism for adhesion. Analyses of crystal contacts, predicted glycosylation and disease-related mutations are presented along with sequence alignments suggesting that the novel features in the CDH17 EC1-2 structure are well conserved. These results hint at distinct adhesive properties for 7D-cadherins.
Collapse
Affiliation(s)
- Michelle E. Gray
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12th Avenue, Columbus, OH 43210, USA
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
19
|
Balan S, Ohnishi T, Watanabe A, Ohba H, Iwayama Y, Toyoshima M, Hara T, Hisano Y, Miyasaka Y, Toyota T, Shimamoto-Mitsuyama C, Maekawa M, Numata S, Ohmori T, Shimogori T, Kikkawa Y, Hayashi T, Yoshikawa T. Role of an Atypical Cadherin Gene, Cdh23 in Prepulse Inhibition, and Implication of CDH23 in Schizophrenia. Schizophr Bull 2021; 47:1190-1200. [PMID: 33595068 PMCID: PMC8266601 DOI: 10.1093/schbul/sbab007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We previously identified quantitative trait loci (QTL) for prepulse inhibition (PPI), an endophenotype of schizophrenia, on mouse chromosome 10 and reported Fabp7 as a candidate gene from an analysis of F2 mice from inbred strains with high (C57BL/6N; B6) and low (C3H/HeN; C3H) PPI levels. Here, we reanalyzed the previously reported QTLs with increased marker density. The highest logarithm of odds score (26.66) peaked at a synonymous coding and splice-site variant, c.753G>A (rs257098870), in the Cdh23 gene on chromosome 10; the c.753G (C3H) allele showed a PPI-lowering effect. Bayesian multiple QTL mapping also supported the same variant with a posterior probability of 1. Thus, we engineered the c.753G (C3H) allele into the B6 genetic background, which led to dampened PPI. We also revealed an e-QTL (expression QTL) effect imparted by the c.753G>A variant for the Cdh23 expression in the brain. In a human study, a homologous variant (c.753G>A; rs769896655) in CDH23 showed a nominally significant enrichment in individuals with schizophrenia. We also identified multiple potentially deleterious CDH23 variants in individuals with schizophrenia. Collectively, the present study reveals a PPI-regulating Cdh23 variant and a possible contribution of CDH23 to schizophrenia susceptibility.
Collapse
Affiliation(s)
- Shabeesh Balan
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan,Neuroscience Research Laboratory, Institute of Mental Health and Neurosciences (IMHANS), Kozhikode, Kerala, India
| | - Tetsuo Ohnishi
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Akiko Watanabe
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Hisako Ohba
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Yoshimi Iwayama
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Manabu Toyoshima
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Tomonori Hara
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan,Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yasuko Hisano
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Yuki Miyasaka
- Deafness Project, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo, Japan,Division of Experimental Animals, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Tomoko Toyota
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | | | - Motoko Maekawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan,Department of Biological Science, Graduate School of Humanities and Science, Ochanomizu University, Tokyo, Japan
| | - Shusuke Numata
- Department of Psychiatry, Institute of Biomedical Science, Tokushima University Graduate School, Tokushima, Japan
| | - Tetsuro Ohmori
- Department of Psychiatry, Institute of Biomedical Science, Tokushima University Graduate School, Tokushima, Japan
| | - Tomomi Shimogori
- Laboratory for Molecular Mechanisms of Brain Development, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Yoshiaki Kikkawa
- Deafness Project, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo, Japan
| | - Takeshi Hayashi
- Agricultural Artificial Intelligence (AI) Research Office, Research Center for Agricultural Information Technology, National Agriculture and Food Research Organization (NARO), Tokyo, Japan
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan,To whom correspondence should be addressed; 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; tel: +81-48-467-5968, fax: +81-48-467-7462, e-mail:
| |
Collapse
|
20
|
Ma C, Cui Z, Wang Y, Zhang L, Wen J, Guo H, Li N, Zhang W. Bioinformatics analysis reveals TSPAN1 as a candidate biomarker of progression and prognosis in pancreatic cancer. Bosn J Basic Med Sci 2021; 21:47-60. [PMID: 33188589 PMCID: PMC7861625 DOI: 10.17305/bjbms.2020.5096] [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: 08/30/2020] [Accepted: 11/08/2020] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer (PCC) is a common malignant tumor of the digestive system that is resistant to traditional treatments and has an overall 5-year survival rate of <7%. Transcriptomics research provides reliable biomarkers for diagnosis, prognosis, and clinical precision treatment, as well as the identification of molecular targets for the development of drugs to improve patient survival. We sought to identify new biomarkers for PCC by combining transcriptomics and clinical data with current knowledge regarding molecular mechanisms. Consequently, we employed weighted gene co-expression network analysis and differentially expressed gene analysis to evaluate genes co-expressed in tumor versus normal tissues using pancreatic adenocarcinoma data from The Cancer Genome Atlas and dataset GSE16515 from the Gene Expression Omnibus. Twenty-one overlapping genes were identified, with enrichment of key Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathways, including epidermal growth factor receptor signaling, cadherin, cell adhesion, ubiquinone, and glycosphingolipid biosynthesis pathways, and retinol metabolism. Protein-protein interaction analysis highlighted 10 hub genes, according to Maximal Clique Centrality. Univariate and multivariate COX analyses indicated that TSPAN1 serves as an independent prognostic factor for PCC patients. Survival analysis distinguished TSPAN1 as an independent prognostic factor among hub genes in PCC. Finally, immunohistochemical staining results suggested that the TSPAN1 protein levels in the Human Protein Atlas were significantly higher in tumor tissue than in normal tissue. Therefore, TSPAN1 may be involved in PCC development and act as a critical biomarker for diagnosing and predicting PCC patient survival.
Collapse
Affiliation(s)
- Chenhui Ma
- Department of Hepatobiliary Medicine, Hebei General Hospital, Shijiazhuang, China. Graduate school of North China University of Science and Technology, Tangshan, China
| | - ZeLong Cui
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - YiChao Wang
- Graduate School of North China University of Science and Technology, Tangshan, China
| | - Lei Zhang
- Department of hepatobiliary, Hebei General Hospital, Shijiazhuang, China
| | - JunYe Wen
- Department of Hepatobiliary diseases, Hebei General Hospital, Shijiazhuang, China
| | - HuaiBin Guo
- Department of Hepatobiliary diseases, Hebei General Hospital, Shijiazhuang, China
| | - Na Li
- Department of Hepatobiliary diseases, Hebei General Hospital, Shijiazhuang, China
| | - WanXing Zhang
- Department of Hepatobiliary diseases, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
21
|
Bouck EG, de la Fuente M, Zunica ER, Li W, Mumaw MM, Nieman MT. Murine cadherin-6 mediates thrombosis in vivo in a platelet-independent manner. Res Pract Thromb Haemost 2021; 5:125-131. [PMID: 33537536 PMCID: PMC7845066 DOI: 10.1002/rth2.12458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Platelet adhesion is the critical process mediating stable thrombus formation. Previous reports of cadherin-6 on human platelets have demonstrated its role in platelet aggregation and thrombus formation. OBJECTIVES We aimed to further characterize the importance of cadherin-6 in thrombosis in vivo. METHODS Cadherin-6 platelet expression was evaluated by western blotting, flow cytometry, and immunoprecipitation. Thrombosis was evaluated using the FeCl3 and Rose Bengal carotid artery models in C57Bl6 mice treated with anti-cadherin-6 or IgG and wild-type or Cdh6-/- mice. Platelet function was compared in wild-type and Cdh6-/- mice using tail-clip assays, aggregometry, and flow cytometry. RESULTS Human platelet expression of cadherin-6 was confirmed at ~3000 copies per platelet. Cdh6-/- mice or those treated with anti-cadherin-6 antibody showed an increased time to occlusion in both thrombosis models. Cadherin-6 was not expressed on mouse platelets, and there were no differences in tail bleeding times, platelet aggregation, or platelet activation in wild-type versus Cdh6-/- mice. CONCLUSIONS Cadherin-6 plays an essential role in thrombosis in vivo. However, cadherin-6 is not expressed on murine platelets. These data are in contrast to human platelets, which express a functional cadherin-6/catenin complex. The essential, platelet-independent role for cadherin-6 in hemostasis may allow it to be an effective and safe therapeutic target.
Collapse
Affiliation(s)
- Emma G. Bouck
- Department of PharmacologyCase Western Reserve UniversityClevelandOHUSA
| | | | | | - Wei Li
- Deparmtent of Biomedical SciencesMarshall University Joan C. Edwards School of MedicineHuntingtonWVUSA
| | - Michele M. Mumaw
- Department of PharmacologyCase Western Reserve UniversityClevelandOHUSA
| | - Marvin T. Nieman
- Department of PharmacologyCase Western Reserve UniversityClevelandOHUSA
| |
Collapse
|
22
|
Martinez-Garay I. Molecular Mechanisms of Cadherin Function During Cortical Migration. Front Cell Dev Biol 2020; 8:588152. [PMID: 33043020 PMCID: PMC7523180 DOI: 10.3389/fcell.2020.588152] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/27/2020] [Indexed: 11/13/2022] Open
Abstract
During development of the cerebral cortex, different types of neurons migrate from distinct origins to create the different cortical layers and settle within them. Along their way, migrating neurons use cell adhesion molecules on their surface to interact with other cells that will play critical roles to ensure that migration is successful. Radially migrating projection neurons interact primarily with radial glia and Cajal-Retzius cells, whereas interneurons originating in the subpallium follow a longer, tangential route and encounter additional cellular substrates before reaching the cortex. Cell-cell adhesion is therefore essential for the correct migration of cortical neurons. Several members of the cadherin superfamily of cell adhesion proteins, which mediate cellular interactions through calcium-dependent, mostly homophilic binding, have been shown to play important roles during neuronal migration of both projection neurons and interneurons. Although several classical cadherins and protocadherins are involved in this process, the most prominent is CDH2. This mini review will explore the cellular and molecular mechanisms underpinning cadherin function during cortical migration, including recent advances in our understanding of the control of adhesive strength through regulation of cadherin surface levels.
Collapse
Affiliation(s)
- Isabel Martinez-Garay
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
23
|
Cell culture dimensionality influences mesenchymal stem cell fate through cadherin-2 and cadherin-11. Biomaterials 2020; 254:120127. [DOI: 10.1016/j.biomaterials.2020.120127] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/16/2020] [Indexed: 12/19/2022]
|
24
|
Kim H, Kim D, Kim J, Lee HY, Park D, Kang H, Matsuda K, Sterky FH, Yuzaki M, Kim JY, Choi SY, Ko J, Um JW. Calsyntenin-3 interacts with both α- and β-neurexins in the regulation of excitatory synaptic innervation in specific Schaffer collateral pathways. J Biol Chem 2020; 295:9244-9262. [PMID: 32434929 PMCID: PMC7335786 DOI: 10.1074/jbc.ra120.013077] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/15/2020] [Indexed: 12/13/2022] Open
Abstract
Calsyntenin-3 (Clstn3) is a postsynaptic adhesion molecule that induces presynaptic differentiation via presynaptic neurexins (Nrxns), but whether Nrxns directly bind to Clstn3 has been a matter of debate. Here, using LC-MS/MS-based protein analysis, confocal microscopy, RNAscope assays, and electrophysiological recordings, we show that β-Nrxns directly interact via their LNS domain with Clstn3 and Clstn3 cadherin domains. Expression of splice site 4 (SS4) insert-positive β-Nrxn variants, but not insert-negative variants, reversed the impaired Clstn3 synaptogenic activity observed in Nrxn-deficient neurons. Consistently, Clstn3 selectively formed complexes with SS4-positive Nrxns in vivo Neuron-specific Clstn3 deletion caused significant reductions in number of excitatory synaptic inputs. Moreover, expression of Clstn3 cadherin domains in CA1 neurons of Clstn3 conditional knockout mice rescued structural deficits in excitatory synapses, especially within the stratum radiatum layer. Collectively, our results suggest that Clstn3 links to SS4-positive Nrxns to induce presynaptic differentiation and orchestrate excitatory synapse development in specific hippocampal neural circuits, including Schaffer collateral afferents.
Collapse
Affiliation(s)
- Hyeonho Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Hyeonpoong-Eup, Dalseong-gun, Daegu, Korea
| | - Dongwook Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Hyeonpoong-Eup, Dalseong-gun, Daegu, Korea
| | - Jinhu Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Hyeonpoong-Eup, Dalseong-gun, Daegu, Korea
| | - Hee-Yoon Lee
- Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Dongseok Park
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Hyeonpoong-Eup, Dalseong-gun, Daegu, Korea
| | - Hyeyeon Kang
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Hyeonpoong-Eup, Dalseong-gun, Daegu, Korea
| | - Keiko Matsuda
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Fredrik H Sterky
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Laboratory Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Michisuke Yuzaki
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Jin Young Kim
- Biomedical Omics Group, Korea Basic Science Institute, Cheongju, Chungbuk, Korea
| | - Se-Young Choi
- Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Hyeonpoong-Eup, Dalseong-gun, Daegu, Korea.
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Hyeonpoong-Eup, Dalseong-gun, Daegu, Korea; Core Protein Resources Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Hyeonpoong-Eup, Dalseong-Gun, Daegu, Korea.
| |
Collapse
|
25
|
Losi L, Zanocco-Marani T, Grande A. Cadherins down-regulation: towards a better understanding of their relevance in colorectal cancer. Histol Histopathol 2020; 35:1391-1402. [PMID: 32567668 DOI: 10.14670/hh-18-236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The down-regulation of cadherin expression in colorectal cancer (CRC) has been widely studied. However, existing data on cadherin expression are highly variable and its relevance to CRC development has not been completely established. This review examines published studies on cadherins whose down-regulation has been already demonstrated in CRC, trying to establish a relationship with promoter methylation, the capacity to influence the Wnt / CTNNB1 (catenin beta 1, beta-catenin) signalling pathway and the clinical implications for disease outcome. Moreover, it also analyses factors that may explain data variability and highlights the importance of considering the altered subcellular localization of the examined cadherins. The results of this survey reveal that thirty of one hundred existing cadherins appear to be down-regulated in CRC. Among these, ten are cadherins, sixteen are protocadherins, equally divided between clustered and non clustered, and four are cadherin - related. These findings suggest that, to better define the role played by cadherin down-regulation in CRC pathogenesis, the expression of multiple rather than individual cadherins should be taken into account and further functional studies are necessary to clarify the relative ability of individual cadherins to inhibit CTNNB1 therefore acting as tumor suppressors.
Collapse
Affiliation(s)
- Lorena Losi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | | | - Alexis Grande
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
26
|
Louvandini H, Corrêa PS, Amorín R, Liu L, Ieda EH, Jimenez CR, Tsai SM, McManus CM, Peñagaricano F. Gestational and lactational exposure to gossypol alters the testis transcriptome. BMC Genomics 2020; 21:59. [PMID: 31952482 PMCID: PMC6969474 DOI: 10.1186/s12864-020-6487-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/13/2020] [Indexed: 11/10/2022] Open
Abstract
Background Reproductive capacity can be altered by challenges experienced during critical periods of development, including fetal development and early neonatal life. Gossypol is a polyphenolic compound, commonly found in cotton seeds, that impairs male reproduction. Here, we investigated whether the exposure to gossypol in utero and during lactation alters male reproductive function in sheep. From conception until 60 days postpartum, ewes were randomly assigned to a control diet or a gossypol-rich diet based on cottonseed. Lamb testicles were removed at 60 days of age and subjected to RNA-sequencing. Results Lambs derived from the maternal cottonseed diet showed significantly lower growth and lower testis weight as a proportion of the total body weight, and reduced testosterone levels. In addition, the testis transcriptome was significantly altered by the maternal cottonseed diet. Most of the altered genes are directly implicated in testis development and sperm biology, cell communication, iron ion metabolism, calcium homeostasis and signaling, among other functions. Interestingly, network analysis revealed that exposure to gossypol significantly disturbed coexpression patterns among spermatogenesis-related genes, suggesting a disruption in coregulation mechanisms. Conclusions Our findings provide evidence that maternal exposure to gossypol alters male reproductive function in the offspring, with potential lasting or lifelong negative consequences.
Collapse
Affiliation(s)
- Helder Louvandini
- Department of Animal Sciences, University of Florida, Gainesville, FL, 32611, USA.,Laboratory of Animal Nutrition, Centre for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba, SP, 13400-970, Brazil
| | - Patricia S Corrêa
- Department of Animal Sciences, University of Florida, Gainesville, FL, 32611, USA.,Laboratory of Animal Nutrition, Centre for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba, SP, 13400-970, Brazil.,Laboratory of Molecular and Cell Biology, Centre for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba, SP, 13400-970, Brazil
| | - Rocío Amorín
- Department of Animal Sciences, University of Florida, Gainesville, FL, 32611, USA
| | - Lihe Liu
- Department of Animal Sciences, University of Florida, Gainesville, FL, 32611, USA
| | - Egon H Ieda
- Laboratory of Animal Nutrition, Centre for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba, SP, 13400-970, Brazil
| | - Carolina R Jimenez
- Laboratory of Animal Nutrition, Centre for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba, SP, 13400-970, Brazil
| | - Siu M Tsai
- Laboratory of Molecular and Cell Biology, Centre for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba, SP, 13400-970, Brazil
| | - Concepta M McManus
- Institute of Biology, University of Brasília, Brasília, Federal District, 70910-900, Brazil
| | - Francisco Peñagaricano
- Department of Animal Sciences, University of Florida, Gainesville, FL, 32611, USA. .,University of Florida Genetics Institute, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
27
|
Homophilic and Heterophilic Interactions of Type II Cadherins Identify Specificity Groups Underlying Cell-Adhesive Behavior. Cell Rep 2019; 23:1840-1852. [PMID: 29742438 PMCID: PMC6029887 DOI: 10.1016/j.celrep.2018.04.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/21/2018] [Accepted: 04/01/2018] [Indexed: 12/04/2022] Open
Abstract
Type II cadherins are cell-cell adhesion proteins critical for tissue patterning and neuronal targeting but whose molecular binding code remains poorly understood. Here, we delineate binding preferences for type II cadherin cell-adhesive regions, revealing extensive heterophilic interactions between specific pairs, in addition to homophilic interactions. Three distinct specificity groups emerge from our analysis with members that share highly similar heterophilic binding patterns and favor binding to one another. Structures of adhesive fragments from each specificity group confirm near-identical dimer topology conserved throughout the family, allowing interface residues whose conservation corresponds to specificity preferences to be identified. We show that targeted mutation of these residues converts binding preferences between specificity groups in biophysical and co-culture assays. Our results provide a detailed understanding of the type II cadherin interaction map and a basis for defining their role in tissue patterning and for the emerging importance of their heterophilic interactions in neural connectivity.
Collapse
|
28
|
Menghini M, Cehajic-Kapetanovic J, Yusuf IH, MacLaren RE. A novel splice-site variant in CDH23 in a patient with Usher syndrome type 1. Ophthalmic Genet 2019; 40:545-548. [PMID: 31755791 DOI: 10.1080/13816810.2019.1692359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: Gene editing has shown huge potential in correcting aberrant splicing and Cas13 has been identified as being particularly suitable for targeting RNA. It has therefore become increasingly important to highlight new splice site mutations that may be correctable, particularly in genes that are too large to be encoded by AAV vectors. About 20% of Usher Type 1 cases are caused by mutations in CDH23.Purpose: To report a novel splice site mutation of CDH23 associated with Usher Type 1D.Materials and Methods: Case report.Results: A 35-year-old Caucasian female who is congenitally deaf with vestibular dysfunction presented with visual acuity of 6/12 in both eyes. Fundus examination revealed findings typical of retinitis pigmentosa with foveal preservation of photoreceptor layer. Next generation sequencing analysis revealed a novel homozygous variant, c.9319 + 1G>T in CDH23 consistent with the diagnosis of Usher Syndrome Type 1D. The c.9319 + 1G>T variant is predicted to affect splicing at the exon 65/intron 65 boundary, which highly likely leads to complete skipping of exon 65.Conclusions: We describe a case of a typical Usher Syndrome Type 1D caused by a novel splice site variant in CDH23. Currently there are no treatments for CDH23 related retinal degeneration, partly because the cDNA size of 10kb is too large for AAV vector gene augmentation therapy. Alternative strategies include CRISPR-Cas9 adenine base editors and RNA editing with CRISPR-Cas13. Single-nucleotide editing represents a promising approach for targeting this variant in CDH23 to restore the wildtype splice donor site at this position.
Collapse
Affiliation(s)
- Moreno Menghini
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, Oxford University, Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jasmina Cehajic-Kapetanovic
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, Oxford University, Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Imran H Yusuf
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, Oxford University, Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, Oxford University, Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
29
|
The Many Roles of Cell Adhesion Molecules in Hepatic Fibrosis. Cells 2019; 8:cells8121503. [PMID: 31771248 PMCID: PMC6952767 DOI: 10.3390/cells8121503] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 01/09/2023] Open
Abstract
Fibrogenesis is a progressive scarring event resulting from disrupted regular wound healing due to repeated tissue injury and can end in organ failure, like in liver cirrhosis. The protagonists in this process, either liver-resident cells or patrolling leukocytes attracted to the site of tissue damage, interact with each other by soluble factors but also by direct cell–cell contact mediated by cell adhesion molecules. Since cell adhesion molecules also support binding to the extracellular matrix, they represent excellent biosensors, which allow cells to modulate their behavior based on changes in the surrounding microenvironment. In this review, we focus on selectins, cadherins, integrins and members of the immunoglobulin superfamily of adhesion molecules as well as some non-classical cell adhesion molecules in the context of hepatic fibrosis. We describe their liver-specific contributions to leukocyte recruitment, cell differentiation and survival, matrix remodeling or angiogenesis and touch on their suitability as targets in antifibrotic therapies.
Collapse
|
30
|
Genetics of Pituitary Tumours. EXPERIENTIA. SUPPLEMENTUM 2019. [PMID: 31588533 DOI: 10.1007/978-3-030-25905-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Pituitary tumours are relatively common in the general population. Most often they occur sporadically, with somatic mutations accounting for a significant minority of somatotroph and corticotroph adenomas. Pituitary tumours can also develop secondary to germline mutations as part of a complex syndrome or as familial isolated pituitary adenomas. Tumours occurring in a familial setting may present at a younger age and can behave more aggressively with resistance to treatment. This chapter will focus on the genetics and molecular pathogenesis of pituitary tumours.
Collapse
|
31
|
Modak D, Sotomayor M. Identification of an adhesive interface for the non-clustered δ1 protocadherin-1 involved in respiratory diseases. Commun Biol 2019; 2:354. [PMID: 31583286 PMCID: PMC6769022 DOI: 10.1038/s42003-019-0586-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/21/2019] [Indexed: 12/29/2022] Open
Abstract
Cadherins form a large family of calcium-dependent adhesive proteins involved in morphogenesis, cell differentiation, and neuronal connectivity. Non-clustered δ1 protocadherins form a cadherin subgroup of proteins with seven extracellular cadherin (EC) repeats and cytoplasmic domains distinct from those of classical cadherins. Non-clustered δ1 protocadherins mediate homophilic adhesion and have been implicated in various diseases including asthma, autism, and cancer. Here we present X-ray crystal structures of human Protocadherin-1 (PCDH1), a δ1-protocadherin member essential for New World Hantavirus infection that is typically expressed in the brain, airway epithelium, skin keratinocytes, and lungs. The structures suggest a binding mode that involves antiparallel overlap of repeats EC1 to EC4. Mutagenesis combined with binding assays and biochemical experiments validated this mode of adhesion. Overall, these results reveal the molecular mechanism underlying adhesiveness of PCDH1 and δ1-protocadherins, also shedding light on PCDH1's role in maintaining airway epithelial integrity, the loss of which causes respiratory diseases.
Collapse
Affiliation(s)
- Debadrita Modak
- Department of Chemistry and Biochemistry, The Ohio State University, 484 W 12th Avenue, Columbus, OH 43210 USA
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, 484 W 12th Avenue, Columbus, OH 43210 USA
| |
Collapse
|
32
|
Kim B. Evolutionarily conserved and divergent functions for cell adhesion molecules in neural circuit assembly. J Comp Neurol 2019; 527:2061-2068. [PMID: 30779135 DOI: 10.1002/cne.24666] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022]
Abstract
The developing nervous system generates remarkably precise synaptic connections between neurons and their postsynaptic target cells. Numerous neural cell adhesion proteins have been identified to mediate cell recognition between synaptic partners in several model organisms. Here, I review the role of protein interactions of cell adhesion molecules in neural circuit assembly and address how these interactions are utilized to form different neural circuitries in different species. The emerging evidence suggests that the extracellular trans-interactions of cell adhesion proteins for neural wiring are evolutionarily conserved across taxa, but they are often used in different steps of circuit assembly. I also highlight how these conserved protein interactions work together as a group to specify neural connectivity.
Collapse
Affiliation(s)
- Byunghyuk Kim
- Department of Life Science, Dongguk University Seoul, Goyang, Republic of Korea
| |
Collapse
|
33
|
Mittler E, Dieterle ME, Kleinfelter LM, Slough MM, Chandran K, Jangra RK. Hantavirus entry: Perspectives and recent advances. Adv Virus Res 2019; 104:185-224. [PMID: 31439149 DOI: 10.1016/bs.aivir.2019.07.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Hantaviruses are important zoonotic pathogens of public health importance that are found on all continents except Antarctica and are associated with hemorrhagic fever with renal syndrome (HFRS) in the Old World and hantavirus pulmonary syndrome (HPS) in the New World. Despite the significant disease burden they cause, no FDA-approved specific therapeutics or vaccines exist against these lethal viruses. The lack of available interventions is largely due to an incomplete understanding of hantavirus pathogenesis and molecular mechanisms of virus replication, including cellular entry. Hantavirus Gn/Gc glycoproteins are the only viral proteins exposed on the surface of virions and are necessary and sufficient to orchestrate virus attachment and entry. In vitro studies have implicated integrins (β1-3), DAF/CD55, and gC1qR as candidate receptors that mediate viral attachment for both Old World and New World hantaviruses. Recently, protocadherin-1 (PCDH1) was demonstrated as a requirement for cellular attachment and entry of New World hantaviruses in vitro and lethal HPS in vivo, making it the first clade-specific host factor to be identified. Attachment of hantavirus particles to cellular receptors induces their internalization by clathrin-mediated, dynamin-independent, or macropinocytosis-like mechanisms, followed by particle trafficking to an endosomal compartment where the fusion of viral and endosomal membranes can occur. Following membrane fusion, which requires cholesterol and acid pH, viral nucleocapsids escape into the cytoplasm and launch genome replication. In this review, we discuss the current mechanistic understanding of hantavirus entry, highlight gaps in our existing knowledge, and suggest areas for future inquiry.
Collapse
Affiliation(s)
- Eva Mittler
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Maria Eugenia Dieterle
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Lara M Kleinfelter
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Megan M Slough
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.
| | - Rohit K Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
34
|
De-la-Torre P, Choudhary D, Araya-Secchi R, Narui Y, Sotomayor M. A Mechanically Weak Extracellular Membrane-Adjacent Domain Induces Dimerization of Protocadherin-15. Biophys J 2018; 115:2368-2385. [PMID: 30527337 PMCID: PMC6302040 DOI: 10.1016/j.bpj.2018.11.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/01/2018] [Accepted: 11/05/2018] [Indexed: 10/27/2022] Open
Abstract
The cadherin superfamily of proteins is defined by the presence of extracellular cadherin (EC) "repeats" that engage in protein-protein interactions to mediate cell-cell adhesion, cell signaling, and mechanotransduction. The extracellular domains of nonclassical cadherins often have a large number of EC repeats along with other subdomains of various folds. Protocadherin-15 (PCDH15), a protein component of the inner-ear tip link filament essential for mechanotransduction, has 11 EC repeats and a membrane adjacent domain (MAD12) of atypical fold. Here we report the crystal structure of a pig PCDH15 fragment including EC10, EC11, and MAD12 in a parallel dimeric arrangement. MAD12 has a unique molecular architecture and folds as a ferredoxin-like domain similar to that found in the nucleoporin protein Nup54. Analytical ultracentrifugation experiments along with size-exclusion chromatography coupled to multiangle laser light scattering and small-angle x-ray scattering corroborate the crystallographic dimer and show that MAD12 induces parallel dimerization of PCDH15 near its membrane insertion point. In addition, steered molecular dynamics simulations suggest that MAD12 is mechanically weak and may unfold before tip-link rupture. Sequence analyses and structural modeling predict the existence of similar domains in cadherin-23, protocadherin-24, and the "giant" FAT and CELSR cadherins, indicating that some of them may also exhibit MAD-induced parallel dimerization.
Collapse
Affiliation(s)
- Pedro De-la-Torre
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio
| | - Deepanshu Choudhary
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio
| | - Raul Araya-Secchi
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; Structural Biophysics, Section for Neutron and X-ray Science, Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
| | - Yoshie Narui
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
35
|
Protocadherin-1 is essential for cell entry by New World hantaviruses. Nature 2018; 563:559-563. [PMID: 30464266 DOI: 10.1038/s41586-018-0702-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 09/20/2018] [Indexed: 01/26/2023]
Abstract
The zoonotic transmission of hantaviruses from their rodent hosts to humans in North and South America is associated with a severe and frequently fatal respiratory disease, hantavirus pulmonary syndrome (HPS)1,2. No specific antiviral treatments for HPS are available, and no molecular determinants of in vivo susceptibility to hantavirus infection and HPS are known. Here we identify the human asthma-associated gene protocadherin-1 (PCDH1)3-6 as an essential determinant of entry and infection in pulmonary endothelial cells by two hantaviruses that cause HPS, Andes virus (ANDV) and Sin Nombre virus (SNV). In vitro, we show that the surface glycoproteins of ANDV and SNV directly recognize the outermost extracellular repeat domain of PCDH1-a member of the cadherin superfamily7,8-to exploit PCDH1 for entry. In vivo, genetic ablation of PCDH1 renders Syrian golden hamsters highly resistant to a usually lethal ANDV challenge. Targeting PCDH1 could provide strategies to reduce infection and disease caused by New World hantaviruses.
Collapse
|
36
|
Jaiganesh A, Narui Y, Araya-Secchi R, Sotomayor M. Beyond Cell-Cell Adhesion: Sensational Cadherins for Hearing and Balance. Cold Spring Harb Perspect Biol 2018; 10:a029280. [PMID: 28847902 PMCID: PMC6008173 DOI: 10.1101/cshperspect.a029280] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cadherins form a large family of proteins often involved in calcium-dependent cellular adhesion. Although classical members of the family can provide a physical bond between cells, a subset of special cadherins use their extracellular domains to interlink apical specializations of single epithelial sensory cells. Two of these cadherins, cadherin-23 (CDH23) and protocadherin-15 (PCDH15), form extracellular "tip link" filaments that connect apical bundles of stereocilia on hair cells essential for inner-ear mechanotransduction. As these bundles deflect in response to mechanical stimuli from sound or head movements, tip links gate hair-cell mechanosensitive channels to initiate sensory perception. Here, we review the unusual and diverse structural properties of these tip-link cadherins and the functional significance of their deafness-related missense mutations. Based on the structural features of CDH23 and PCDH15, we discuss the elasticity of tip links and models that bridge the gap between the nanomechanics of cadherins and the micromechanics of hair-cell bundles during inner-ear mechanotransduction.
Collapse
Affiliation(s)
- Avinash Jaiganesh
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210
| | - Yoshie Narui
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210
| | - Raul Araya-Secchi
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
37
|
Cri-du-Chat Syndrome interactome network: Correlating genotypic variations to associated phenotypes. GENE REPORTS 2018. [DOI: 10.1016/j.genrep.2018.03.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
38
|
Casal JI, Bartolomé RA. RGD cadherins and α2β1 integrin in cancer metastasis: A dangerous liaison. Biochim Biophys Acta Rev Cancer 2018; 1869:321-332. [PMID: 29673969 DOI: 10.1016/j.bbcan.2018.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/13/2018] [Accepted: 04/14/2018] [Indexed: 12/24/2022]
Abstract
We propose a new cadherin family classification comprising epithelial cadherins (cadherin 17 [CDH17], cadherin 16, VE-cadherin, cadherin 6 and cadherin 20) containing RGD motifs within their sequences. Expression of some RGD cadherins is associated with aggressive forms of cancer during the late stages of metastasis, and CDH17 and VE-cadherin have emerged as critical actors in cancer metastasis. After binding to α2β1 integrin, these cadherins promote integrin β1 activation, and thereby cell adhesion, invasion and proliferation, in liver and lung metastasis. Activation of α2β1 integrin provokes an affinity increase for type IV collagen, a major component of the basement membrane and a critical partner for cell anchoring in liver and other metastatic organs. Activation of α2β1 integrin by RGD motifs breaks an old paradigm of integrin classification and supports an important role of this integrin in cancer metastasis. Recently, synthetic peptides containing the RGD motif of CDH17 elicited highly specific and selective antibodies that block the ability of CDH17 RGD to activate α2β1 integrin. These monoclonal antibodies inhibit metastatic colonization in orthotopic mouse models of liver and lung metastasis for colorectal cancer and melanoma, respectively. Hopefully, blocking the cadherin RGD ligand capacity will give us control over the integrin activity in solid tumors metastasis, paving the way for development of new agents of cancer treatment.
Collapse
Affiliation(s)
- J Ignacio Casal
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28039 Madrid, Spain.
| | - Rubén A Bartolomé
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28039 Madrid, Spain
| |
Collapse
|
39
|
Vanniya S P, Srisailapathy CRS, Kunka Mohanram R. The tip link protein Cadherin-23: From Hearing Loss to Cancer. Pharmacol Res 2018; 130:25-35. [PMID: 29421162 DOI: 10.1016/j.phrs.2018.01.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 01/24/2018] [Accepted: 01/26/2018] [Indexed: 11/26/2022]
Abstract
Cadherin-23 is an atypical member of the cadherin superfamily, with a distinctly long extracellular domain. It has been known to be a part of the tip links of the inner ear mechanosensory hair cells. Several studies have been carried out to understand the role of Cadherin-23 in the hearing mechanism and defects in the CDH23 have been associated with hearing impairment resulting from defective or absence of tip links. Recent studies have highlighted the role of Cadherin-23 in several pathological conditions, including cancer, suggesting the presence of several unknown functions. Initially, it was proposed that Cadherin-23 represents a yet unspecified subtype of Cadherins; however, no other proteins with similar characteristics have been identified, till date. It has a unique cytoplasmic domain that does not bear a β-catenin binding region, but has been demonstrated to mediate cell-cell adhesions. Several protein interacting partners have been identified for Cadherin-23 and the roles of their interactions in various cellular mechanisms are yet to be explored. This review summarizes the characteristics of Cadherin-23 and its roles in several pathologies including cancer.
Collapse
Affiliation(s)
- Paridhy Vanniya S
- Department of Genetics, Dr. ALM PG Institute of Basic Medical Science, University of Madras, Taramani campus, Chennai, Tamilnadu, India
| | - C R Srikumari Srisailapathy
- Department of Genetics, Dr. ALM PG Institute of Basic Medical Science, University of Madras, Taramani campus, Chennai, Tamilnadu, India
| | - Ramkumar Kunka Mohanram
- SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India.
| |
Collapse
|
40
|
Sannigrahi MK, Srinivas S, Rakshit S. The Prospects of Cadherin-23 as a Mediator of Homophilic Cell-Cell Adhesion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1112:99-105. [PMID: 30637693 DOI: 10.1007/978-981-13-3065-0_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Cadherins (calcium-dependent adhesion proteins) constitute a family of cell surface proteins that mediate cell-cell adhesion and actively participate in tissue morphogenesis and in mediating tissue integrity. The ecto-domains of cadherins from opposing cell surfaces interact with each other to form the load-bearing trans-dimers and mechanically hold cells together. The "classical" cadherins and desmosomes that form separate groups in cadherin superfamily are mostly explored for their roles in cell-cell adhesion. However, majority of cadherins in cells belong to "nonclassical" group which is poorly explored in the context of their cell-binding properties. This review focuses on the role of "nonclassical" cadherin, cadherin-23, in cell-cell adhesion. Overall, this review highlights the need for further investigations on the role of "nonclassical" cadherin-23 in cell-cell adhesion.
Collapse
Affiliation(s)
- Malay Kumar Sannigrahi
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, Punjab, India
| | - Sai Srinivas
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, Punjab, India
| | - Sabyasachi Rakshit
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, Punjab, India.
- Centre for Protein Science Design and Engineering, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, Punjab, India.
| |
Collapse
|
41
|
Choudhary D, Kumar A, Magliery TJ, Sotomayor M. Using thermal scanning assays to test protein-protein interactions of inner-ear cadherins. PLoS One 2017; 12:e0189546. [PMID: 29261728 PMCID: PMC5736220 DOI: 10.1371/journal.pone.0189546] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/27/2017] [Indexed: 12/15/2022] Open
Abstract
Protein-protein interactions play a crucial role in biological processes such as cell-cell adhesion, immune system-pathogen interactions, and sensory perception. Understanding the structural determinants of protein-protein complex formation and obtaining quantitative estimates of their dissociation constant (KD) are essential for the study of these interactions and for the discovery of new therapeutics. At the same time, it is equally important to characterize protein-protein interactions in a high-throughput fashion. Here, we use a modified thermal scanning assay to test interactions of wild type (WT) and mutant variants of N-terminal fragments (EC1+2) of cadherin-23 and protocadherin-15, two proteins essential for inner-ear mechanotransduction. An environmentally sensitive fluorescent dye (SYPRO orange) is used to monitor melting temperature (Tm) shifts of protocadherin-15 EC1+2 (pcdh15) in the presence of increasing concentrations of cadherin-23 EC1+2 (cdh23). These Tm shifts are absent when we use proteins containing deafness-related missense mutations known to disrupt cdh23 binding to pcdh15, and are increased for some rationally designed mutants expected to enhance binding. In addition, surface plasmon resonance binding experiments were used to test if the Tm shifts correlated with changes in binding affinity. We used this approach to find a double mutation (cdh23(T15E)- pcdh15(G16D)) that enhances binding affinity of the cadherin complex by 1.98 kJ/mol, roughly two-fold that of the WT complex. We suggest that the thermal scanning methodology can be used in high-throughput format to quickly compare binding affinities (KD from nM up to 100 μM) for some heterodimeric protein complexes and to screen small molecule libraries to find protein-protein interaction inhibitors and enhancers.
Collapse
Affiliation(s)
- Deepanshu Choudhary
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Anusha Kumar
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Thomas J. Magliery
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
42
|
Cousin H. Cadherins function during the collective cell migration of Xenopus Cranial Neural Crest cells: revisiting the role of E-cadherin. Mech Dev 2017; 148:79-88. [PMID: 28467887 PMCID: PMC5662486 DOI: 10.1016/j.mod.2017.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/21/2017] [Accepted: 04/28/2017] [Indexed: 12/20/2022]
Abstract
Collective cell migration is a process whereby cells move while keeping contact with other cells. The Xenopus Cranial Neural Crest (CNC) is a population of cells that emerge during early embryogenesis and undergo extensive migration from the dorsal to ventral part of the embryo's head. These cells migrate collectively and require cadherin mediated cell-cell contact. In this review, we will describe the key features of Xenopus CNC migration including the key molecules driving their migration. We will also review the role of the various cadherins during Xenopus CNC emergence and migration. Lastly, we will discuss the recent and seemingly controversial findings showing that E-cadherin presence is essential for CNC migration.
Collapse
Affiliation(s)
- Hélène Cousin
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, United States.
| |
Collapse
|
43
|
Corey DP, Ó Maoiléidigh D, Ashmore JF. Mechanical Transduction Processes in the Hair Cell. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/978-3-319-52073-5_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
44
|
Mah KM, Weiner JA. Regulation of Wnt signaling by protocadherins. Semin Cell Dev Biol 2017; 69:158-171. [PMID: 28774578 PMCID: PMC5586504 DOI: 10.1016/j.semcdb.2017.07.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 07/21/2017] [Accepted: 07/28/2017] [Indexed: 12/23/2022]
Abstract
The ∼70 protocadherins comprise the largest group within the cadherin superfamily. Their diversity, the complexity of the mechanisms through which their genes are regulated, and their many critical functions in nervous system development have engendered a growing interest in elucidating the intracellular signaling pathways through which they act. Recently, multiple protocadherins across several subfamilies have been implicated as modulators of Wnt signaling pathways, and through this as potential tumor suppressors. Here, we review the extant data on the regulation by protocadherins of Wnt signaling pathways and components, and highlight some key unanswered questions that could shape future research.
Collapse
Affiliation(s)
- Kar Men Mah
- Department of Biology, The University of Iowa, Iowa City, IA, USA.
| | - Joshua A Weiner
- Department of Biology, The University of Iowa, Iowa City, IA, USA; Department of Psychiatry, The University of Iowa, Iowa City, IA, USA; Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
45
|
Bibollet-Bahena O, Okafuji T, Hokamp K, Tear G, Mitchell KJ. A dual-strategy expression screen for candidate connectivity labels in the developing thalamus. PLoS One 2017; 12:e0177977. [PMID: 28558017 PMCID: PMC5448750 DOI: 10.1371/journal.pone.0177977] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 05/05/2017] [Indexed: 12/13/2022] Open
Abstract
The thalamus or “inner chamber” of the brain is divided into ~30 discrete nuclei, with highly specific patterns of afferent and efferent connectivity. To identify genes that may direct these patterns of connectivity, we used two strategies. First, we used a bioinformatics pipeline to survey the predicted proteomes of nematode, fruitfly, mouse and human for extracellular proteins containing any of a list of motifs found in known guidance or connectivity molecules. Second, we performed clustering analyses on the Allen Developing Mouse Brain Atlas data to identify genes encoding surface proteins expressed with temporal profiles similar to known guidance or connectivity molecules. In both cases, we then screened the resultant genes for selective expression patterns in the developing thalamus. These approaches identified 82 candidate connectivity labels in the developing thalamus. These molecules include many members of the Ephrin, Eph-receptor, cadherin, protocadherin, semaphorin, plexin, Odz/teneurin, Neto, cerebellin, calsyntenin and Netrin-G families, as well as diverse members of the immunoglobulin (Ig) and leucine-rich receptor (LRR) superfamilies, receptor tyrosine kinases and phosphatases, a variety of growth factors and receptors, and a large number of miscellaneous membrane-associated or secreted proteins not previously implicated in axonal guidance or neuronal connectivity. The diversity of their expression patterns indicates that thalamic nuclei are highly differentiated from each other, with each one displaying a unique repertoire of these molecules, consistent with a combinatorial logic to the specification of thalamic connectivity.
Collapse
Affiliation(s)
| | - Tatsuya Okafuji
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Karsten Hokamp
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Guy Tear
- Department of Developmental Neurobiology, New Hunt’s House, Guy’s Campus, King’s College, London, United Kingdom
| | - Kevin J. Mitchell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- * E-mail:
| |
Collapse
|
46
|
Bialucha CU, Collins SD, Li X, Saxena P, Zhang X, Dürr C, Lafont B, Prieur P, Shim Y, Mosher R, Lee D, Ostrom L, Hu T, Bilic S, Rajlic IL, Capka V, Jiang W, Wagner JP, Elliott G, Veloso A, Piel JC, Flaherty MM, Mansfield KG, Meseck EK, Rubic-Schneider T, London AS, Tschantz WR, Kurz M, Nguyen D, Bourret A, Meyer MJ, Faris JE, Janatpour MJ, Chan VW, Yoder NC, Catcott KC, McShea MA, Sun X, Gao H, Williams J, Hofmann F, Engelman JA, Ettenberg SA, Sellers WR, Lees E. Discovery and Optimization of HKT288, a Cadherin-6-Targeting ADC for the Treatment of Ovarian and Renal Cancers. Cancer Discov 2017; 7:1030-1045. [PMID: 28526733 DOI: 10.1158/2159-8290.cd-16-1414] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/11/2017] [Accepted: 05/10/2017] [Indexed: 11/16/2022]
Abstract
Despite an improving therapeutic landscape, significant challenges remain in treating the majority of patients with advanced ovarian or renal cancer. We identified the cell-cell adhesion molecule cadherin-6 (CDH6) as a lineage gene having significant differential expression in ovarian and kidney cancers. HKT288 is an optimized CDH6-targeting DM4-based antibody-drug conjugate (ADC) developed for the treatment of these diseases. Our study provides mechanistic evidence supporting the importance of linker choice for optimal antitumor activity and highlights CDH6 as an antigen for biotherapeutic development. To more robustly predict patient benefit of targeting CDH6, we incorporate a population-based patient-derived xenograft (PDX) clinical trial (PCT) to capture the heterogeneity of response across an unselected cohort of 30 models-a novel preclinical approach in ADC development. HKT288 induces durable tumor regressions of ovarian and renal cancer models in vivo, including 40% of models on the PCT, and features a preclinical safety profile supportive of progression toward clinical evaluation.Significance: We identify CDH6 as a target for biotherapeutics development and demonstrate how an integrated pharmacology strategy that incorporates mechanistic pharmacodynamics and toxicology studies provides a rich dataset for optimizing the therapeutic format. We highlight how a population-based PDX clinical trial and retrospective biomarker analysis can provide correlates of activity and response to guide initial patient selection for first-in-human trials of HKT288. Cancer Discov; 7(9); 1030-45. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 920.
Collapse
Affiliation(s)
- Carl U Bialucha
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts.
| | - Scott D Collins
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Xiao Li
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Parmita Saxena
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Xiamei Zhang
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Clemens Dürr
- Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Bruno Lafont
- Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Pierric Prieur
- Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Yeonju Shim
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Rebecca Mosher
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - David Lee
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Lance Ostrom
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Tiancen Hu
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Sanela Bilic
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | - Vladimir Capka
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Wei Jiang
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Joel P Wagner
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - GiNell Elliott
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Artur Veloso
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Jessica C Piel
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Meghan M Flaherty
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Keith G Mansfield
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Emily K Meseck
- Novartis Institutes for Biomedical Research, East Hanover, New Jersey
| | - Tina Rubic-Schneider
- Novartis Institutes for Biomedical Research, Campus Klybeckstrasse, Basel, Switzerland
| | | | | | - Markus Kurz
- Novartis Pharma AG, Novartis Campus, Basel, Switzerland
| | - Duc Nguyen
- Novartis Pharma, Cambridge, Massachusetts
| | - Aaron Bourret
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Matthew J Meyer
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Jason E Faris
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Mary J Janatpour
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Vivien W Chan
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | | | | | | | - Hui Gao
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Juliet Williams
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Francesco Hofmann
- Novartis Institutes for Biomedical Research, Campus Klybeckstrasse, Basel, Switzerland
| | | | - Seth A Ettenberg
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - William R Sellers
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Emma Lees
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| |
Collapse
|
47
|
Zhang Q, Peng C, Song J, Zhang Y, Chen J, Song Z, Shou X, Ma Z, Peng H, Jian X, He W, Ye Z, Li Z, Wang Y, Ye H, Zhang Z, Shen M, Tang F, Chen H, Shi Z, Chen C, Chen Z, Shen Y, Wang Y, Lu S, Zhang J, Li Y, Li S, Mao Y, Zhou L, Yan H, Shi Y, Huang C, Zhao Y. Germline Mutations in CDH23, Encoding Cadherin-Related 23, Are Associated with Both Familial and Sporadic Pituitary Adenomas. Am J Hum Genet 2017; 100:817-823. [PMID: 28413019 DOI: 10.1016/j.ajhg.2017.03.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/24/2017] [Indexed: 11/19/2022] Open
Abstract
Pituitary adenoma (PA) is one of the most common intracranial neoplasms. Several genetic predisposing factors for PA have been identified, but they account for a small portion of cases. In this study, we sought to identify the PA genetic risk factors by focusing on causative mutations for PAs. Among the 4 affected and 17 asymptomatic members from one family with familial PA, whole-exome sequencing identified cosegregation of the PA phenotype with the heterozygous missense mutation c.4136G>T (p.Arg1379Leu) in cadherin-related 23 (CDH23). This mutation causes an amino acid substitution in the calcium-binding motif of the extracellular cadherin (EC) domains of CDH23 and is predicted to impair cell-cell adhesion. Genomic screening in a total of 12 families with familial PA (20 individuals), 125 individuals with sporadic PA, and 260 control individuals showed that 33% of the families with familial PA (4/12) and 12% of individuals with sporadic PA (15/125) harbored functional CDH23 variants. In contrast, 0.8% of the healthy control individuals (2/260) carried functional CDH23 variants. Gene-based analysis also revealed a significant association between CDH23 genotype and PA (p = 5.54 × 10-7). Moreover, PA individuals who did not harbor functional CDH23 variants displayed tumors that were larger in size (p = 0.005) and more invasive (p < 0.001). Therefore, mutations in CDH23 are linked with familial and sporadic PA and could play important roles in the pathogenesis of PA.
Collapse
Affiliation(s)
- Qilin Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Cheng Peng
- Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jianping Song
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Yichao Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Jianhua Chen
- Bio-X Institutes, Ministry of Education Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhijian Song
- Bio-X Institutes, Ministry of Education Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xuefei Shou
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Zengyi Ma
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Hong Peng
- Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xuemin Jian
- Bio-X Institutes, Ministry of Education Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wenqiang He
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Zhao Ye
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Zhiqiang Li
- Bio-X Institutes, Ministry of Education Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yongfei Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Hongying Ye
- Shanghai Pituitary Tumor Center, Shanghai 200040, China; Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Zhaoyun Zhang
- Shanghai Pituitary Tumor Center, Shanghai 200040, China; Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Ming Shen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Feng Tang
- Shanghai Pituitary Tumor Center, Shanghai 200040, China; Department of Pathology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Hong Chen
- Shanghai Pituitary Tumor Center, Shanghai 200040, China; Department of Pathology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Chunjui Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Zhengyuan Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Yue Shen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Ye Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Shaoyong Lu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jian Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yiming Li
- Shanghai Pituitary Tumor Center, Shanghai 200040, China; Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Shiqi Li
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai 200040, China; Institute of Neurosurgery, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Liangfu Zhou
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Hai Yan
- Department of Pathology, Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Yongyong Shi
- Bio-X Institutes, Ministry of Education Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Psychiatry, First Teaching Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China; Biomedical Sciences Institute of Qingdao University, Qingdao Branch of SJTU Bio-X Institutes and the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Chuanxin Huang
- Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yao Zhao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Pituitary Tumor Center, Shanghai 200040, China; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai 200040, China; Institute of Neurosurgery, Shanghai Medical College, Fudan University, Shanghai 200040, China.
| |
Collapse
|
48
|
Gul IS, Hulpiau P, Saeys Y, van Roy F. Metazoan evolution of the armadillo repeat superfamily. Cell Mol Life Sci 2017; 74:525-541. [PMID: 27497926 PMCID: PMC11107757 DOI: 10.1007/s00018-016-2319-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/05/2016] [Accepted: 07/25/2016] [Indexed: 02/08/2023]
Abstract
The superfamily of armadillo repeat proteins is a fascinating archetype of modular-binding proteins involved in various fundamental cellular processes, including cell-cell adhesion, cytoskeletal organization, nuclear import, and molecular signaling. Despite their diverse functions, they all share tandem armadillo (ARM) repeats, which stack together to form a conserved three-dimensional structure. This superhelical armadillo structure enables them to interact with distinct partners by wrapping around them. Despite the important functional roles of this superfamily, a comprehensive analysis of the composition, classification, and phylogeny of this protein superfamily has not been reported. Furthermore, relatively little is known about a subset of ARM proteins, and some of the current annotations of armadillo repeats are incomplete or incorrect, often due to high similarity with HEAT repeats. We identified the entire armadillo repeat superfamily repertoire in the human genome, annotated each armadillo repeat, and performed an extensive evolutionary analysis of the armadillo repeat proteins in both metazoan and premetazoan species. Phylogenetic analyses of the superfamily classified them into several discrete branches with members showing significant sequence homology, and often also related functions. Interestingly, the phylogenetic structure of the superfamily revealed that about 30 % of the members predate metazoans and represent an ancient subset, which is gradually evolving to acquire complex and highly diverse functions.
Collapse
Affiliation(s)
- Ismail Sahin Gul
- Inflammation Research Center (IRC), VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, FSVM Building, Technologiepark 927, 9052, Ghent, Belgium
| | - Paco Hulpiau
- Inflammation Research Center (IRC), VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, FSVM Building, Technologiepark 927, 9052, Ghent, Belgium
| | - Yvan Saeys
- Inflammation Research Center (IRC), VIB, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Frans van Roy
- Inflammation Research Center (IRC), VIB, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, FSVM Building, Technologiepark 927, 9052, Ghent, Belgium.
| |
Collapse
|
49
|
Zhao T, Jiang W, Wang X, Wang H, Zheng C, Li Y, Sun Y, Huang C, Han ZB, Yang S, Jia Z, Xie K, Ren H, Hao J. ESE3 Inhibits Pancreatic Cancer Metastasis by Upregulating E-Cadherin. Cancer Res 2016; 77:874-885. [PMID: 27923832 DOI: 10.1158/0008-5472.can-16-2170] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/12/2016] [Accepted: 11/19/2016] [Indexed: 12/15/2022]
Abstract
The ETS family transcription factor ESE3 is a crucial element in differentiation and development programs for many epithelial tissues. Here we report its role as a tumor suppressor in pancreatic cancer. We observed drastically lower ESE3 expression in pancreatic ductal adenocarcinomas (PDAC) compared with adjacent normal pancreatic tissue. Reduced expression of ESE3 in PDAC correlated closely with an increase in lymph node metastasis and vessel invasion and a decrease in relapse-free and overall survival in patients. In functional experiments, downregulating the expression of ESE3 promoted PDAC cell motility and invasiveness along with metastasis in an orthotopic mouse model. Mechanistic studies in PDAC cell lines, the orthotopic mouse model, and human PDAC specimens demonstrated that ESE3 inhibited PDAC metastasis by directly upregulating E-cadherin expression at the level of its transcription. Collectively, our results establish ESE3 as a negative regulator of PDAC progression and metastasis by enforcing E-cadherin upregulation. Cancer Res; 77(4); 874-85. ©2016 AACR.
Collapse
Affiliation(s)
- Tiansuo Zhao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin, P.R. China.,Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wenna Jiang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin, P.R. China
| | - Xiuchao Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin, P.R. China
| | - Hongwei Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin, P.R. China
| | - Chen Zheng
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin, P.R. China
| | - Yang Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin, P.R. China
| | - Yan Sun
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P.R. China
| | - Chongbiao Huang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin, P.R. China
| | - Zhi-Bo Han
- Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, P.R. China
| | - Shengyu Yang
- Department of Tumor Biology and Comprehensive Melanoma Research Center, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Zhiliang Jia
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Keping Xie
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - He Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin, P.R. China
| | - Jihui Hao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin, P.R. China.
| |
Collapse
|
50
|
Cooper SR, Jontes JD, Sotomayor M. Structural determinants of adhesion by Protocadherin-19 and implications for its role in epilepsy. eLife 2016; 5. [PMID: 27787195 PMCID: PMC5115871 DOI: 10.7554/elife.18529] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/25/2016] [Indexed: 01/27/2023] Open
Abstract
Non-clustered δ-protocadherins are homophilic cell adhesion molecules essential for the development of the vertebrate nervous system, as several are closely linked to neurodevelopmental disorders. Mutations in protocadherin-19 (PCDH19) result in a female-limited, infant-onset form of epilepsy (PCDH19-FE). Over 100 mutations in PCDH19 have been identified in patients with PCDH19-FE, about half of which are missense mutations in the adhesive extracellular domain. Neither the mechanism of homophilic adhesion by PCDH19, nor the biochemical effects of missense mutations are understood. Here we present a crystallographic structure of the minimal adhesive fragment of the zebrafish Pcdh19 extracellular domain. This structure reveals the adhesive interface for Pcdh19, which is broadly relevant to both non-clustered δ and clustered protocadherin subfamilies. In addition, we show that several PCDH19-FE missense mutations localize to the adhesive interface and abolish Pcdh19 adhesion in in vitro assays, thus revealing the biochemical basis of their pathogenic effects during brain development. DOI:http://dx.doi.org/10.7554/eLife.18529.001 As the brain develops, its basic building blocks – cells called neurons – need to form the correct connections with one another in order to give rise to neural circuits. A mistake that leads to the formation of incorrect connections can result in a number of disorders or brain abnormalities. Proteins called cadherins that are present on the surface of neurons enable them to stick to their correct partners like Velcro. One of these proteins is called Protocadherin-19. However, it was not fully understood how this protein forms an adhesive bond with other Protocadherin-19 molecules, or how some of the proteins within the cadherin family are able to distinguish between one another. Cooper et al. used X-ray crystallography to visualize the molecular structure of Protocadherin-19 taken from zebrafish in order to better understand the adhesive bond that these proteins form with each other. In addition, the new structure showed the sites of the mutations that cause a form of epilepsy in infant females. From this, Cooper et al. could predict how the mutations would disrupt Protocadherin-19’s shape and function. The structures revealed that Protocadherin-19 molecules from adjacent cells engage in a “forearm handshake” to form the bond that connects neurons. Some of the mutations that cause epilepsy occur in the region responsible for this Protocadherin-19 forearm handshake. Laboratory experiments confirmed that these mutations impair the formation of the adhesive bond, revealing the molecular basis for some of the mutations that underlie Protocadherin-19-female-limited epilepsy. Other cadherin molecules may interact via a similar forearm handshake; this could be investigated in future experiments. It also remains to be discovered how brain wiring depends on Protocadherin-19 adhesion in animal development, and how altering these proteins can rewire developing brain circuits. DOI:http://dx.doi.org/10.7554/eLife.18529.002
Collapse
Affiliation(s)
- Sharon R Cooper
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, United States.,Department of Neuroscience, The Ohio State University, Columbus, United States
| | - James D Jontes
- Department of Neuroscience, The Ohio State University, Columbus, United States
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, United States
| |
Collapse
|