1
|
Kerek R, Sawma Awad J, Bassam M, Hajjar C, Ghantous F, Rizk K, Rima M. The multifunctional protein CCN1/CYR61: Bridging physiology and disease. Exp Mol Pathol 2025; 142:104969. [PMID: 40286773 DOI: 10.1016/j.yexmp.2025.104969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
The matricellular protein CYR61/CCN1 is a member of the CCN protein family that plays significant roles in a broad range of physiological processes, including development, tissue repair, and inflammation, among others. CCN1 is also implicated in pathological conditions such as cancer and fibrosis. The diverse functions of CCN1 arise from its ability to bind different receptors located on many cell types, thereby activating diverse signaling pathways. The diverse, yet contradictory, functions mediated by CCN1 makes it a compelling target for investigation, as it offers the prospect of understanding fundamental cellular topics and their possible implications in various diseases. Recently, new cellular functions were attributed to CCN1, including senescence, pro-/anti- fibrosis, and rejuvenation. In this review, we discuss all these new findings along with the basic knowledge about CCN1 to provide an overall understanding of its conflicting roles and their potential corresponding mechanisms of action.
Collapse
Affiliation(s)
- Racha Kerek
- Department of Biological Sciences, Lebanese American University, Byblos, P.O. Box 36, Lebanon
| | - Joe Sawma Awad
- Department of Biological Sciences, Lebanese American University, Byblos, P.O. Box 36, Lebanon
| | - Mariam Bassam
- Department of Biological Sciences, Lebanese American University, Byblos, P.O. Box 36, Lebanon
| | - Carla Hajjar
- Department of Biological Sciences, Lebanese American University, Byblos, P.O. Box 36, Lebanon
| | - Fouad Ghantous
- Department of Biological Sciences, Lebanese American University, Byblos, P.O. Box 36, Lebanon
| | - Karelle Rizk
- Department of Biological Sciences, Lebanese American University, Byblos, P.O. Box 36, Lebanon
| | - Mohamad Rima
- Department of Biological Sciences, Lebanese American University, Byblos, P.O. Box 36, Lebanon.
| |
Collapse
|
2
|
Nakata T, Kirita Y, Umehara M, Nakamura M, Sawai S, Minamida A, Yamauchi-Sawada H, Sunahara Y, Matoba Y, Okuno-Ozeki N, Nakamura I, Nakai K, Yagi-Tomita A, Yamashita N, Tamagaki K, Humphreys BD, Matoba S, Kusaba T. Injured tubular epithelia-derived CCN1 promotes the mobilization of fibroblasts toward injury sites after kidney injury. iScience 2025; 28:112176. [PMID: 40224016 PMCID: PMC11987671 DOI: 10.1016/j.isci.2025.112176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/28/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025] Open
Abstract
Humoral factors that prompt fibroblasts to migrate to an injury site at an appropriate time point are deemed indispensable for repair after kidney injury. We herein demonstrated the pivotal roles of injured tubule-derived cellular communication network factor 1 (CCN1) in the mobilization of fibroblasts to the injury site after kidney injury. Based on analyses of ligand-receptor interactions in vitro and tubular epithelial-specific transcriptomics in vivo, we identified the up-regulation of CCN1 during the early phases of kidney injury. CCN1 promotes fibroblast chemotaxis through focal adhesion kinase-extracellular signal-regulated kinase (ERK) signaling. In vivo analyses utilizing tubular-specific CCN1 knockout (KO) mice demonstrated the sparse accumulation of fibroblasts around injured sites after injury, resulting in ameliorated tissue fibrosis in CCN1-KO mice. These results reveal an epithelial-fibroblast CCN1 signaling axis that mobilizes fibroblasts to injured tubule early after acute injury but that promotes interstitial fibrosis at late time points.
Collapse
Affiliation(s)
- Tomohiro Nakata
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuhei Kirita
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Minato Umehara
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masashi Nakamura
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shinji Sawai
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Atsushi Minamida
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroko Yamauchi-Sawada
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuto Sunahara
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yayoi Matoba
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Natsuko Okuno-Ozeki
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Itaru Nakamura
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kunihiro Nakai
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Aya Yagi-Tomita
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Noriyuki Yamashita
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keiichi Tamagaki
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Benjamin D. Humphreys
- Division of Nephrology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuro Kusaba
- Department of Nephrolog, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
3
|
Kim S, Yang K, Kim K, Kim HJ, Kim DY, Chae J, Ahn YH, Kang JL. The interplay of cancer-associated fibroblasts and apoptotic cancer cells suppresses lung cancer cell growth through WISP-1-integrin ανβ3-STAT1 signaling pathway. Cell Commun Signal 2025; 23:98. [PMID: 39966869 PMCID: PMC11837402 DOI: 10.1186/s12964-025-02094-2] [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: 08/30/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Cell death within the tumor microenvironment (TME) plays a crucial role in controlling cancer by influencing the balance of tumor-specific immunity. Cancer-associated fibroblasts (CAFs) significantly contribute to tumor progression through paracrine mechanisms. We found that reprogramming of CAFs by apoptotic cancer cells suppresses tumor volume and lung metastasis. Here, we investigated the mechanisms by which the interaction between apoptotic lung cancer cells and CAFs hinders tumor growth. METHODS Experimental methods including CCK assay, colony formation assay, immunoblotting, co-immunoprecipitation, qRT-PCR analysis, qRT-PCR array, apoptosis assay, ELISA, and immunofluorescent staining were used in this study. Additionally, CAFs were isolated from lung tumors of Kras-mutant (KrasLA1) mice and human lung adenocarcinoma samples using magnetic-activated cell sorting. Murine lung cancer cells (344SQ cells) along with various human cancer cell lines (A549, HCT116, and LoVo) were cultured. In animal study, conditioned medium (CM) derived from CAFs (undiluted or 50% diluted) with or without neutralizing anti-WISP-1 antibody was administered into syngeneic mice to study anti-tumoral effects. To confirm the paracrine role of WISP-1, recombinant WISP-1 (rWISP-1) was administered via intratumoral injection. RESULTS We demonstrate that treatment with CM from lung CAFs exposed to apoptotic cancer cells suppresses proliferation and promotes apoptosis in lung cancer cells through STAT1 signaling. Pharmacologic inhibition of Notch1 activation or siRNA-mediated Notch1 silencing in CAFs reversed the antiproliferative and proapoptotic effects. Similarly, knockdown of Wnt-induced signaling protein 1 (WISP-1) in CAFs or neutralizing the CM with anti-WISP-1 antibodies reversed the antiproliferative and proapoptotic effects. WISP-1 signaled through integrin ανβ3-STAT1 signaling pathway to inhibit cancer cell growth and promote apoptosis. The in vivo introduction of CM derived from apoptotic 344SQ-exposed CAFs (ApoSQ-CAF CM) potently decelerated tumor growth. This effect was observed alongside the downregulation of proliferative and anti-apoptotic markers, while simultaneously boosting the activation of phosphorylated STAT1 and pro-apoptotic markers in CD326+ tumor cells within syngeneic immunocompetent mice. rWISP-1 effectively replicates the in vivo effects of ApoSQ-CAF CM. CONCLUSIONS These findings suggest that CM from apoptotic cancer cell-exposed CAFs may offer a promising therapeutic approach by lung cancer suppression.
Collapse
Affiliation(s)
- Shinyoung Kim
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Kyungwon Yang
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Kiyoon Kim
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Hee Ja Kim
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Korea
| | - Da Young Kim
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Jeesoo Chae
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, 07985, Korea
| | - Young-Ho Ahn
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, 07804, Korea
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, 07985, Korea
| | - Jihee Lee Kang
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Korea.
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, 07804, Korea.
| |
Collapse
|
4
|
Guo X, Guo S, Tian F, Gao Z, Fan Y, Wang C, Xu S. CCN1 Promotes Mesenchymal Phenotype Transition Through Activating NF-κB Signaling Pathway Regulated by S100A8 in Glioma Stem Cells. CNS Neurosci Ther 2024; 30:e70128. [PMID: 39659236 PMCID: PMC11632201 DOI: 10.1111/cns.70128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/28/2024] [Accepted: 11/03/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND The presence of glioma stem cells (GSCs) and the occurrence of mesenchymal phenotype transition contribute to the miserable prognosis of glioblastoma (GBM). Cellular communication network factor 1 (CCN1) is upregulated within various malignancies and associated with cancer development and progression, while the implications of CCN1 in the phenotype transition and tumorigenicity of GSCs remain unclear. METHODS Data for bioinformatic analysis were obtained from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) databases. A range of primary GBM and GSC cell models were then used to demonstrate the regulatory role of CCN1 via the phenotype validation, tumor sphere formation assays, extreme limiting dilution assays (ELDA), and transwell assays. To screen out the downstream signaling pathway, we employed high-throughput RNA-seq. Intracranial xenograft GSC mouse models were used to investigate the role of CCN1 in vivo. RESULTS Among the CCN family members, CCN1 was highly expressed in MES-GBM/GSCs and was correlated with a poor prognosis. Both in vitro and in vivo assays indicated that knockdown of CCN1 in MES-GSCs reduced the tumor stemness, proliferation, invasion, and tumorigenicity, whereas CCN1 overexpression in PN-GSCs exhibited the opposite effects. Mechanistically, CCN1 triggered the FAK/STAT3 signaling in autocrine and paracrine manners to upregulate the expression of S100A8. Knockdown of S100A8 inactivated NF-κB/p65 pathway and significantly suppressed the tumorigenesis of MES-GSCs. CONCLUSION Our findings reveal that CCN1 may be an important factor in the enhanced invasiveness and MES phenotype transition of GSCs and highlight the potential to target CCN1 for treating GBM.
Collapse
Affiliation(s)
- Xing Guo
- Department of NeurosurgeryQilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain‐Inspired Science, Shandong UniversityJinanShandongChina
| | - Shuhua Guo
- Department of Clinical LaboratoryThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Feng Tian
- Department of Clinical LaboratoryThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Zijie Gao
- Department of NeurosurgeryQilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain‐Inspired Science, Shandong UniversityJinanShandongChina
| | - Yang Fan
- Department of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University &Shandong Provincial Qianfoshan HospitalJinanShandongChina
| | - Chuanxin Wang
- Department of Clinical LaboratoryThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Shuo Xu
- Department of NeurosurgeryQilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain‐Inspired Science, Shandong UniversityJinanShandongChina
| |
Collapse
|
5
|
Fischer AG, Elliott EM, Brittian KR, Garrett L, Sadri G, Aebersold J, Singhal RA, Nong Y, Leask A, Jones SP, Moore Iv JB. Matricellular protein CCN1 promotes collagen alignment and scar integrity after myocardial infarction. Matrix Biol 2024; 133:14-32. [PMID: 39098433 PMCID: PMC11476287 DOI: 10.1016/j.matbio.2024.08.001] [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: 04/13/2024] [Revised: 07/17/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Members of the cellular communication network family (CCN) of matricellular proteins, like CCN1, have long been implicated in the regulation of cellular processes underlying wound healing, tissue fibrogenesis, and collagen dynamics. While many studies suggest antifibrotic actions for CCN1 in the adult heart through the promotion of myofibroblast senescence, they largely relied on exogenous supplementation strategies in in vivo models of cardiac injury where its expression is already induced-which may confound interpretation of its function in this process. The objective of this study was to interrogate the role of the endogenous protein on fibroblast function, collagen structural dynamics, and its associated impact on cardiac fibrosis after myocardial infarction (MI). METHODS/RESULTS Here, we employed CCN1 loss-of-function methodologies, including both in vitro siRNA-mediated depletion and in vivo fibroblast-specific knockout mice to assess the role of the endogenous protein on cardiac fibroblast fibrotic signaling, and its involvement in acute scar formation after MI. In vitro depletion of CCN1 reduced cardiac fibroblast senescence and proliferation. Although depletion of CCN1 decreased the expression of collagen processing and stabilization enzymes (i.e., P4HA1, PLOD1, and PLOD2), it did not inhibit myofibroblast induction or type I collagen synthesis. Alone, fibroblast-specific removal of CCN1 did not negatively impact ventricular performance or myocardial collagen content but did contribute to disorganization of collagen fibrils and increased matrix compliance. Similarly, Ccn1 ablated animals subjected to MI showed no discernible alterations in cardiac structure or function one week after permanent coronary artery ligation, but exhibited marked increases in incidence of mortality and cardiac rupture. Consistent with our findings that CCN1 depletion does not assuage myofibroblast conversion or type I collagen synthesis in vitro, Ccn1 knockout animals revealed no measurable differences in collagen scar width or mass compared to controls; however, detailed structural analyses via SHG and TEM of scar regions revealed marked alterations in their scar collagen topography-exhibiting changes in numerous macro- and micro-level collagen architectural attributes. Specifically, Ccn1 knockout mice displayed heightened ECM structural complexity in post-MI scar regions, including diminished local alignment and heightened tortuosity of collagen fibers, as well as reduced organizational coherency, packing, and size of collagen fibrils. Associated with these changes in ECM topography with the loss of CCN1 were reductions in fibroblast-matrix interactions, as evidenced by reduced fibroblast nuclear and cellular deformation in vivo and reduced focal-adhesion formation in vitro; findings that ultimately suggest CCN1's ability to influence fibroblast-led collagen alignment may in part be credited to its capacity to augment fibroblast-matrix interactions. CONCLUSIONS These findings underscore the pivotal role of endogenous CCN1 in the scar formation process occurring after MI, directing the appropriate arrangement of the extracellular matrix's collagenous components in the maturing scar-shaping the mechanical properties that support its structural stability. While this suggests an adaptive role for CCN1 in regulating collagen structural attributes crucial for supporting scar integrity post MI, the long-term protracted expression of CCN1 holds maladaptive implications, potentially diminishing collagen structural complexity and compliance in non-infarct regions.
Collapse
Affiliation(s)
- Annalara G Fischer
- Center for Cardiometabolic Science, University of Louisville School of Medicine, 580 South Preston Street, Delia Baxter Research Building, Room 304C, Louisville, KY 40202, USA
| | - Erin M Elliott
- Center for Cardiometabolic Science, University of Louisville School of Medicine, 580 South Preston Street, Delia Baxter Research Building, Room 304C, Louisville, KY 40202, USA
| | - Kenneth R Brittian
- Center for Cardiometabolic Science, University of Louisville School of Medicine, 580 South Preston Street, Delia Baxter Research Building, Room 304C, Louisville, KY 40202, USA
| | - Lauren Garrett
- Center for Cardiometabolic Science, University of Louisville School of Medicine, 580 South Preston Street, Delia Baxter Research Building, Room 304C, Louisville, KY 40202, USA
| | - Ghazal Sadri
- Center for Cardiometabolic Science, University of Louisville School of Medicine, 580 South Preston Street, Delia Baxter Research Building, Room 304C, Louisville, KY 40202, USA
| | - Julia Aebersold
- Micro/Nano Technology Center, University of Louisville, Louisville, KY, USA
| | - Richa A Singhal
- Center for Cardiometabolic Science, University of Louisville School of Medicine, 580 South Preston Street, Delia Baxter Research Building, Room 304C, Louisville, KY 40202, USA
| | - Yibing Nong
- Center for Cardiometabolic Science, University of Louisville School of Medicine, 580 South Preston Street, Delia Baxter Research Building, Room 304C, Louisville, KY 40202, USA
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Steven P Jones
- Center for Cardiometabolic Science, University of Louisville School of Medicine, 580 South Preston Street, Delia Baxter Research Building, Room 304C, Louisville, KY 40202, USA
| | - Joseph B Moore Iv
- Center for Cardiometabolic Science, University of Louisville School of Medicine, 580 South Preston Street, Delia Baxter Research Building, Room 304C, Louisville, KY 40202, USA.
| |
Collapse
|
6
|
Christopoulou ME, Aletras AJ, Papakonstantinou E, Stolz D, Skandalis SS. WISP1 and Macrophage Migration Inhibitory Factor in Respiratory Inflammation: Novel Insights and Therapeutic Potentials for Asthma and COPD. Int J Mol Sci 2024; 25:10049. [PMID: 39337534 PMCID: PMC11432718 DOI: 10.3390/ijms251810049] [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: 07/08/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Recent advancements highlight the intricate interplay between the extracellular matrix (ECM) and immune responses, notably in respiratory diseases such as asthma and Chronic Obstructive Pulmonary Disease (COPD). The ECM, a dynamic structural framework within tissues, orches-trates a plethora of cellular processes, including immune cell behavior and tissue repair mecha-nisms. WNT1-inducible-signaling pathway protein 1 (WISP1), a key ECM regulator, controls immune cell behavior, cytokine production, and tissue repair by modulating integrins, PI3K, Akt, β-catenin, and mTOR signaling pathways. WISP1 also induces macrophage migration inhibitory factor (MIF) expression via Src kinases and epidermal growth factor receptor (EGFR) activation. MIF, through its wide range of activities, enhances inflammation and tissue restructuring. Rec-ognized for its versatile roles in regulating the immune system, MIF interacts with multiple immune components, such as the NLRP3 inflammasome, thereby sustaining inflammatory pro-cesses. The WISP1-MIF axis potentially unveils complex molecular mechanisms governing im-mune responses and inflammation. Understanding the intricate roles of WISP1 and MIF in the pathogenesis of chronic respiratory diseases such as asthma and COPD could lead to the identi-fication of novel targets for therapeutic intervention to alleviate disease severity and enhance patient outcomes.
Collapse
Affiliation(s)
- Maria-Elpida Christopoulou
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexios J Aletras
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Eleni Papakonstantinou
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Daiana Stolz
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Spyros S Skandalis
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
7
|
Zeng X, Tang J, Zhang Q, Wang C, Qi J, Wei Y, Xu J, Yang K, Zhou Z, Wu H, Luo J, Jiang Y, Song Z, Wu J, Wu J. CircHIPK2 Contributes Cell Growth in Intestinal Epithelial of Colitis and Colorectal Cancer through Promoting TAZ Translation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401588. [PMID: 38981023 PMCID: PMC11425914 DOI: 10.1002/advs.202401588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/13/2024] [Indexed: 07/11/2024]
Abstract
Colorectal cancer (CRC) and inflammatory bowel disease (IBD) are escalating global health concerns. Despite their distinct clinical presentations, both disorders share intricate genetic and molecular interactions. The Hippo signaling pathway plays a crucial role in regulating cell processes and is implicated in the pathogenesis of IBD and CRC. Circular RNAs (circRNAs) have gained attention for their roles in various diseases, including IBD and CRC. However, a comprehensive understanding of specific circRNAs involved in both IBD and CRC, and their functional roles is lacking. Here, it is found that circHIPK2 (hsa_circRNA_104507) is a bona fide circRNA consistently upregulated in both IBD and CRC suggesting its potential as a biomarker. Furthermore, silencing of circHIPK2 suppressed the growth of CRC cells in vitro and in vivo. Interestingly, decreased circHipk2 potentiated dextran sulfate sodium (DSS)-induced colitis but alleviated colitis-associated tumorigenesis. Most significantly, mechanistic investigations further unveil that circHIPK2, mediated by FUS, interacting with EIF4A3 to promote the translation of TAZ, ultimately increasing the transcription of downstream target genes CCN1 and CCN2. Taken together, circHIPK2 emerges as a key player in the shared mechanisms of IBD and CRC, modulating the Hippo signaling pathway. CircHIPK2-EIF4A3 axis contributes to cell growth in intestinal epithelial of colitis and CRC by enhancing TAZ translation.
Collapse
Affiliation(s)
- Xixi Zeng
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
- The Joint Innovation Center for Health and Medicine, Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, Zhejiang, 324000, China
| | - Jielin Tang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, 325035, China
| | - Qian Zhang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Chenxing Wang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children of Wenzhou Medical University, Zhejiang, 325003, China
| | - Ji Qi
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Yusi Wei
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Jiali Xu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Kaiyuan Yang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Zuolin Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Hao Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Jiarong Luo
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Yi Jiang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children of Wenzhou Medical University, Zhejiang, 325003, China
| | - Zengqiang Song
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, 325035, China
| | - Jinyu Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
| | - Jianmin Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315302, China
| |
Collapse
|
8
|
Wang C, Paiva TO, Speziale P, Dufrêne YF. Nanomechanics of CCN1-Mediated Staphylococcus aureus Phagocytosis. NANO LETTERS 2024; 24:8567-8574. [PMID: 38959438 DOI: 10.1021/acs.nanolett.4c01533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Phagocytosis is an essential mechanism of the human immune system where pathogens are eliminated by immune cells. The CCN1 protein plays an important role in the phagocytosis of Staphylococcus aureus by favoring the bridging of the αVβ3 integrin to the bacterial peptidoglycan (PG), through mechanical forces that remain unknown. Here, we employ single-molecule experiments to unravel the nanomechanics of the PG-CCN1-αVβ3 ternary complex. While CCN1 binds αVβ3 integrins with moderate force (∼60 pN), much higher binding strengths (up to ∼800 pN) are observed between CCN1 and PG. Notably, the strength of both CCN1-αVβ3 and CCN1-PG bonds is dramatically enhanced by tensile loading, favoring a model in which mechanical stress induces the exposure of cryptic integrin binding sites in CCN1 and multivalent binding between CCN1 lectin sites and monosaccharides along the PG glycan chains.
Collapse
Affiliation(s)
- Can Wang
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Telmo O Paiva
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Pietro Speziale
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Viale Taramelli 3/b, 27100 Pavia, Italy
| | - Yves F Dufrêne
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
9
|
Singh K, Oladipupo SS. An overview of CCN4 (WISP1) role in human diseases. J Transl Med 2024; 22:601. [PMID: 38937782 PMCID: PMC11212430 DOI: 10.1186/s12967-024-05364-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/01/2024] [Indexed: 06/29/2024] Open
Abstract
CCN4 (cellular communication network factor 4), a highly conserved, secreted cysteine-rich matricellular protein is emerging as a key player in the development and progression of numerous disease pathologies, including cancer, fibrosis, metabolic and inflammatory disorders. Over the past two decades, extensive research on CCN4 and its family members uncovered their diverse cellular mechanisms and biological functions, including but not limited to cell proliferation, migration, invasion, angiogenesis, wound healing, repair, and apoptosis. Recent studies have demonstrated that aberrant CCN4 expression and/or associated downstream signaling is key to a vast array of pathophysiological etiology, suggesting that CCN4 could be utilized not only as a non-invasive diagnostic or prognostic marker, but also as a promising therapeutic target. The cognate receptor of CCN4 remains elusive till date, which limits understanding of the mechanistic insights on CCN4 driven disease pathologies. However, as therapeutic agents directed against CCN4 begin to make their way into the clinic, that may start to change. Also, the pathophysiological significance of CCN4 remains underexplored, hence further research is needed to shed more light on its disease and/or tissue specific functions to better understand its clinical translational benefit. This review highlights the compelling evidence of overlapping and/or diverse functional and mechanisms regulated by CCN4, in addition to addressing the challenges, study limitations and knowledge gaps on CCN4 biology and its therapeutic potential.
Collapse
Affiliation(s)
- Kirti Singh
- Biotherapeutic Enabling Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46225, USA
| | - Sunday S Oladipupo
- Biotherapeutic Enabling Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46225, USA.
| |
Collapse
|
10
|
Leask A, Nguyen J, Naik A, Chitturi P, Riser BL. The role of yes activated protein (YAP) in melanoma metastasis. iScience 2024; 27:109864. [PMID: 38770136 PMCID: PMC11103372 DOI: 10.1016/j.isci.2024.109864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
Hippo was first identified in a genetic screen as a protein that suppressed proliferation and cell growth. Subsequently, it was shown that hippo acted in a so-called canonical cascade to suppress Yorkie, the Drosophila equivalent of Yes-activated protein (YAP), a mechanosensitive transcriptional cofactor that enhances the activity of the TEAD family of transcription factors. YAP promotes fibrosis, activation of cancer-associated fibroblasts, angiogenesis and cancer cell invasion. YAP activates the expression of the matricellular proteins CCN1 (cyr61) and CCN2 (ctgf), themselves mediators of fibrogenesis and oncogenesis, and coordination of matrix deposition and angiogenesis. This review discusses how therapeutically targeting YAP through YAP inhibitors verteporfin and celastrol and its downstream mediators CCN1 and CCN2 might be useful in treating melanoma.
Collapse
Affiliation(s)
- Andrew Leask
- College of Dentistry, University of Saskatchewan, 105 Wiggins Road, Saskatoon, SK S7N 5E4, Canada
| | - John Nguyen
- College of Dentistry, University of Saskatchewan, 105 Wiggins Road, Saskatoon, SK S7N 5E4, Canada
| | - Angha Naik
- College of Dentistry, University of Saskatchewan, 105 Wiggins Road, Saskatoon, SK S7N 5E4, Canada
| | - Pratyusha Chitturi
- College of Dentistry, University of Saskatchewan, 105 Wiggins Road, Saskatoon, SK S7N 5E4, Canada
| | - Bruce L. Riser
- Department of Physiology & Biophysics, Center for Cancer Cell Biology, Immunology & Infection, Rosalind Franklin University, 3333 N. Green Bay Road, Chicago, IL 60064, USA
- BLR Bio, LLC, Kenosha, WI 53140, USA
| |
Collapse
|
11
|
Sancho L, Boisvert MM, Dawoodtabar T, Burgado J, Wang E, Allen NJ. Astrocyte CCN1 stabilizes neural circuits in the adult brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.585077. [PMID: 38559139 PMCID: PMC10979986 DOI: 10.1101/2024.03.14.585077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Neural circuits in many brain regions are refined by experience. Sensory circuits support higher plasticity at younger ages during critical periods - times of circuit refinement and maturation - and limit plasticity in adulthood for circuit stability. The mechanisms underlying these differing plasticity levels and how they serve to maintain and stabilize the properties of sensory circuits remain largely unclear. By combining a transcriptomic approach with ex vivo electrophysiology and in vivo imaging techniques, we identify that astrocytes release cellular communication network factor 1 (CCN1) to maintain synapse and circuit stability in the visual cortex. By overexpressing CCN1 in critical period astrocytes, we find that it promotes the maturation of inhibitory circuits and limits ocular dominance plasticity. Conversely, by knocking out astrocyte CCN1 in adults, binocular circuits are destabilized. These studies establish CCN1 as a novel astrocyte-secreted factor that stabilizes neuronal circuits. Moreover, they demonstrate that the composition and properties of sensory circuits require ongoing maintenance in adulthood, and that these maintenance cues are provided by astrocytes.
Collapse
|
12
|
Yu D, Lu Z, Nie F, Chong Y. Integrins regulation of wound healing processes: insights for chronic skin wound therapeutics. Front Cell Infect Microbiol 2024; 14:1324441. [PMID: 38505290 PMCID: PMC10949986 DOI: 10.3389/fcimb.2024.1324441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024] Open
Abstract
Integrins are heterodimers composed of non-covalently associated alpha and beta subunits that mediate the dynamic linkage between extracellular adhesion molecules and the intracellular actin cytoskeleton. Integrins are present in various tissues and organs and are involved in different physiological and pathological molecular responses in vivo. Wound healing is an important process in the recovery from traumatic diseases and consists of three overlapping phases: inflammation, proliferation, and remodeling. Integrin regulation acts throughout the wound healing process to promote wound healing. Prolonged inflammation may lead to failure of wound healing, such as wound chronicity. One of the main causes of chronic wound formation is bacterial colonization of the wound. In this review, we review the role of integrins in the regulation of wound healing processes such as angiogenesis and re-epithelialization, as well as the role of integrins in mediating bacterial infections during wound chronicity, and the challenges and prospects of integrins as therapeutic targets for infected wound healing.
Collapse
Affiliation(s)
- Dong Yu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhaoyu Lu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Fengsong Nie
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yang Chong
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
13
|
Christopoulou ME, Skandalis SS, Papakonstantinou E, Stolz D, Aletras AJ. WISP1 induces the expression of macrophage migration inhibitory factor in human lung fibroblasts through Src kinases and EGFR-activated signaling pathways. Am J Physiol Cell Physiol 2024; 326:C850-C865. [PMID: 38145300 PMCID: PMC11193488 DOI: 10.1152/ajpcell.00410.2023] [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: 08/27/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Wnt1-inducible signaling protein 1 (WISP1/CCN4) is a secreted matricellular protein that is implicated in lung and airway remodeling. The macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that has been associated with chronic lung diseases. In this study, we aimed to investigate the WISP1 signaling pathway and its ability to induce the expression of MIF in primary cultures of fibroblasts from normal human lungs (HLFs). Our results showed that WISP1 significantly stimulated the expression of MIF in a concentration- and time-dependent fashion. In WISP1-induced expression of MIF, αvβ5-integrin and chondroitin sulfate proteoglycans as well as Src tyrosine kinases, MAP kinases, phosphatidylinositol 3-kinase/Akt, PKC, and NF-κB were involved. WISP1-induced expression of MIF was attenuated in the presence of the Src kinase inhibitor PP2 or the MIF tautomerase activity inhibitor ISO-1. Moreover, WISP1 significantly increased the phosphorylation and activation of EGF receptor (EGFR) through transactivation by Src kinases. WISP1 also induced the expression of MIF receptor CD74 and coreceptor CD44, through which MIF exerts its effects on HLFs. In addition, it was found that MIF induced its own expression, as well as its receptors CD74/CD44, acting in an autocrine manner. Finally, WISP1-induced MIF promoted the expression of cyclooxygenase 2, prostaglandin E2, IL-6, and matrix metalloproteinase-2 demonstrating the regulatory role of WISP1-MIF axis in lung inflammation and remodeling involving mainly integrin αvβ5, Src kinases, PKC, NF-κB, and EGFR. The specific signaling pathways involved in WISP1-induced expression of MIF may prove to be excellent candidates for novel targets to control inflammation in chronic lung diseases.NEW & NOTEWORTHY The present study demonstrates for the first time that Wnt1-inducible signaling protein 1 (WISP1) regulates migration inhibitory factor (MIF) expression and activity and identifies the main signaling pathways involved. The newly discovered WISP1-MIF axis may drive lung inflammation and could result in the design of novel targeted therapies in inflammatory lung diseases.
Collapse
Affiliation(s)
- Maria-Elpida Christopoulou
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Spyros S Skandalis
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Eleni Papakonstantinou
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daiana Stolz
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexios J Aletras
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| |
Collapse
|
14
|
Lin JB, Santeford A, Colasanti JJ, Lee Y, Shah AV, Wang TJ, Ruzycki PA, Apte RS. Targeting cell-type-specific, choroid-peripheral immune signaling to treat age-related macular degeneration. Cell Rep Med 2024; 5:101353. [PMID: 38232696 PMCID: PMC10829736 DOI: 10.1016/j.xcrm.2023.101353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/25/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024]
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness featuring pathogenic neovascularization of the choroidal vasculature (CNV). Although systemic immunity plays a role in AMD, the ocular signals that recruit and activate immune cells remain poorly defined. Using single-cell RNA sequencing, we prospectively profile peripheral blood mononuclear cells from 65 individuals including AMD and controls, which we integrate with existing choroid data. We generate a network of choroid-peripheral immune interactions dysregulated in AMD, including known AMD-relevant gene vascular endothelial growth factor (VEGF) receptor 2. Additionally, we find CYR61 is upregulated in choroidal veins and may signal to circulating monocytes. In mice, we validate that CYR61 is abundant in endothelial cells within CNV lesions neighboring monocyte-derived macrophages. Mechanistically, CYR61 activates macrophage anti-angiogenic gene expression, and ocular Cyr61 knockdown increases murine CNV size, indicating CYR61 inhibits CNV. This study highlights the potential of multi-tissue human datasets to identify disease-relevant and potentially therapeutically modifiable targets.
Collapse
Affiliation(s)
- Joseph B Lin
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Neurosciences Graduate Program, Roy and Diana Vagelos Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrea Santeford
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jason J Colasanti
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Molecular Cell Biology Graduate Program, Roy and Diana Vagelos Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yoon Lee
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Aaditya V Shah
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tzu Jui Wang
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Philip A Ruzycki
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Rajendra S Apte
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
15
|
Leask A, Naik A, Stratton RJ. Back to the future: targeting the extracellular matrix to treat systemic sclerosis. Nat Rev Rheumatol 2023; 19:713-723. [PMID: 37789119 DOI: 10.1038/s41584-023-01032-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 10/05/2023]
Abstract
Fibrosis is the excessive deposition of a stable extracellular matrix (ECM); fibrotic tissue is composed principally of highly crosslinked type I collagen and highly contractile myofibroblasts. Systemic sclerosis (SSc) is a multisystem autoimmune connective tissue disease characterized by skin and organ fibrosis. The fibrotic process has been recognized in SSc for >40 years, but drugs with demonstrable efficacy against SSc fibrosis in ameliorating the lung involvement have only recently been identified. Unfortunately, these treatments are ineffective at improving the skin score in patients with SSc. Previous clinical trials in SSc have largely focused on the cross-purposing of anti-inflammatory drugs and the use of immunosuppressive drugs from the transplantation field, which address inflammatory and/or autoimmune processes. Limited examination has taken place of specific anti-fibrotic agents developed through their ability to directly target the ECM in SSc by, for example, alleviating the persistent matrix stiffness and mechanotransduction that might be required for both the initiation and maintenance of fibrosis, including in SSc. However, because of the importance of the ECM in the SSc phenotype, attempts have now been made to identify drugs that specifically target the ECM, including some drugs that are currently under consideration for the treatment of cancer.
Collapse
Affiliation(s)
- Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | - Angha Naik
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Richard J Stratton
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, London, UK
| |
Collapse
|
16
|
Kato S, Kawata K, Nishida T, Mizukawa T, Takigawa M, Iida S, Kubota S. Expression and function of CCN2-derived circRNAs in chondrocytes. J Cell Commun Signal 2023:10.1007/s12079-023-00782-7. [PMID: 37695440 DOI: 10.1007/s12079-023-00782-7] [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: 07/12/2023] [Accepted: 08/18/2023] [Indexed: 09/12/2023] Open
Abstract
Cellular communication network factor 2 (CCN2) molecules promote endochondral ossification and articular cartilage regeneration, and circular RNAs (circRNAs), which arise from various genes and regulate gene expression by adsorbing miRNAs, are known to be synthesized from CCN2 in human vascular endothelial cells and other types of cells. However, in chondrocytes, not only the function but also the presence of CCN2-derived circRNA remains completely unknown. In the present study, we investigated the expression and function of CCN2-derived circRNAs in chondrocytes. Amplicons smaller than those from known CCN2-derived circRNAs were observed using RT-PCR analysis that could specifically amplify CCN2-derived circRNAs in human chondrocytic HCS-2/8 cells. The nucleotide sequences of the PCR products indicated novel circRNAs in the HCS-2/8 cells that were different from known CCN2-derived circRNAs. Moreover, the expression of several Ccn2-derived circRNAs in murine chondroblastic ATDC5 cells was confirmed and observed to change alongside chondrocytic differentiation. Next, one of these circRNAs was knocked down in HCS-2/8 cells to investigate the function of the human CCN2-derived circRNA. As a result, CCN2-derived circRNA knockdown significantly reduced the expression of aggrecan mRNA and proteoglycan synthesis. Our data suggest that CCN2-derived circRNAs are expressed in chondrocytes and play a role in chondrogenic differentiation. Production and role of CCN2-derived RNAs in chondrocytes.
Collapse
Affiliation(s)
- Soma Kato
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8525, Japan
- Department of Oral Maxillofacial Reconstructive Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazumi Kawata
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8525, Japan
| | - Takashi Nishida
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8525, Japan
| | - Tomomi Mizukawa
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8525, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Seiji Iida
- Department of Oral Maxillofacial Reconstructive Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Kubota
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8525, Japan.
| |
Collapse
|
17
|
Romeo SG, Secco I, Schneider E, Reumiller CM, Santos CXC, Zoccarato A, Musale V, Pooni A, Yin X, Theofilatos K, Trevelin SC, Zeng L, Mann GE, Pathak V, Harkin K, Stitt AW, Medina RJ, Margariti A, Mayr M, Shah AM, Giacca M, Zampetaki A. Human blood vessel organoids reveal a critical role for CTGF in maintaining microvascular integrity. Nat Commun 2023; 14:5552. [PMID: 37689702 PMCID: PMC10492781 DOI: 10.1038/s41467-023-41326-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
The microvasculature plays a key role in tissue perfusion and exchange of gases and metabolites. In this study we use human blood vessel organoids (BVOs) as a model of the microvasculature. BVOs fully recapitulate key features of the human microvasculature, including the reliance of mature endothelial cells on glycolytic metabolism, as concluded from metabolic flux assays and mass spectrometry-based metabolomics using stable tracing of 13C-glucose. Pharmacological targeting of PFKFB3, an activator of glycolysis, using two chemical inhibitors results in rapid BVO restructuring, vessel regression with reduced pericyte coverage. PFKFB3 mutant BVOs also display similar structural remodelling. Proteomic analysis of the BVO secretome reveal remodelling of the extracellular matrix and differential expression of paracrine mediators such as CTGF. Treatment with recombinant CTGF recovers microvessel structure. In this work we demonstrate that BVOs rapidly undergo restructuring in response to metabolic changes and identify CTGF as a critical paracrine regulator of microvascular integrity.
Collapse
Affiliation(s)
- Sara G Romeo
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Ilaria Secco
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Edoardo Schneider
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Christina M Reumiller
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Celio X C Santos
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Anna Zoccarato
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Vishal Musale
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Aman Pooni
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Xiaoke Yin
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Konstantinos Theofilatos
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Silvia Cellone Trevelin
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Lingfang Zeng
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Giovanni E Mann
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Varun Pathak
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Kevin Harkin
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Alan W Stitt
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Reinhold J Medina
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Andriana Margariti
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Manuel Mayr
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Ajay M Shah
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Mauro Giacca
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Anna Zampetaki
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK.
| |
Collapse
|
18
|
Balijepalli P, Meier KE. From outside to inside and back again: the lysophosphatidic acid-CCN axis in signal transduction. J Cell Commun Signal 2023; 17:845-849. [PMID: 36795277 PMCID: PMC10409932 DOI: 10.1007/s12079-023-00728-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 01/20/2023] [Indexed: 02/17/2023] Open
Abstract
CCN1 and CCN2 are matricellular proteins that are transcriptionally induced by various stimuli, including growth factors. CCN proteins act to facilitate signaling events involving extracellular matrix proteins. Lysophosphatidic acid (LPA) is a lipid that activates G protein-coupled receptors (GPCRs), enhancing proliferation, adhesion, and migration in many types of cancer cells. Our group previously reported that LPA induces production of CCN1 protein in human prostate cancer cell lines within 2-4 h. In these cells, the mitogenic activity of LPA is mediated by LPA Receptor 1 (LPAR1), a GPCR. There are multiple examples of the induction of CCN proteins by LPA, and by the related lipid mediator sphingosine-1-phosphate (S1P), in various cellular models. The signaling pathways responsible for LPA/S1P-induced CCN1/2 typically involve activation of the small GTP-binding protein Rho and the transcription factor YAP. Inducible CCNs can potentially play roles in downstream signal transduction events required for LPA and S1P-induced responses. Specifically, CCNs secreted into the extracellular space can facilitate the activation of additional receptors and signal transduction pathways, contributing to the biphasic delayed responses typically seen in response to growth factors acting via GPCRs. In some model systems, CCN1 and CCN2 play key roles in LPA/S1P-induced cell migration and proliferation. In this way, an extracellular signal (LPA or S1P) can activate GPCR-mediated intracellular signaling to induce the production of extracellular modulators (CCN1 and CCN2) that in turn initiate another round of intracellular signaling.
Collapse
Affiliation(s)
- Pravita Balijepalli
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA USA
| | - Kathryn E. Meier
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA USA
| |
Collapse
|
19
|
Zhao Y, Wang H, He T, Ma B, Chen G, Tzeng C. Knockdown of Yap attenuates TAA-induced hepatic fibrosis by interaction with hedgehog signals. J Cell Commun Signal 2023:10.1007/s12079-023-00775-6. [PMID: 37338798 DOI: 10.1007/s12079-023-00775-6] [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: 07/05/2022] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
Liver fibrosis is an aberrant wound healing response to tissue injury characterized by excessive extracellular matrix deposition and loss of normal liver architecture. Hepatic stellate cells (HSCs) activation is regards to be the major process in liver fibrogenesis which is dynamic and reversible. Both Hippo signaling core factor Yap and Hedgehog (Hh) signaling promote HSCs transdifferentiation thereby regulating the repair process of liver injury. However, the molecular function of YAP and the regulation between Yap and Hh during fibrogenesis remain uncertain. In this study, the essential roles of Yap in liver fibrosis were investigated. Yap was detected to be increased in liver fibrotic tissue by the thioacetamide (TAA)-induced zebrafish embryonic and adult models. Inhibition of Yap by both embryonic morpholino interference and adult's inhibitor treatment was proved to alleviate TAA-induced liver lesions by and histology and gene expression examination. Transcriptomic analysis and gene expression detection showed that Yap and Hh signaling pathway have a cross talking upon TAA-induced liver fibrosis. In addition, TAA induction promoted the nuclear colocalization of YAP and Hh signaling factor GLI2α. This study demonstrates that Yap and Hh play synergistic protective roles in liver fibrotic response and provides new theoretical insight concerning the mechanisms of fibrosis progression.
Collapse
Affiliation(s)
- Ye Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China.
| | - Huiling Wang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Tianhua He
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Bo Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Guoguang Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Chimeng Tzeng
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361005, China.
- Translational Medicine Research Center-Key Laboratory for Cancer T-Cell Theragnostic and Clinical Translation, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China.
- Xiamen Chang Gung Hospital Medical Research Center, Xiamen, Fujian, China.
| |
Collapse
|
20
|
Monsen VT, Attramadal H. Structural insights into regulation of CCN protein activities and functions. J Cell Commun Signal 2023:10.1007/s12079-023-00768-5. [PMID: 37245184 DOI: 10.1007/s12079-023-00768-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/07/2023] [Indexed: 05/29/2023] Open
Abstract
CCN proteins play important functions during development, in repair mechanisms following tissue injury, as well as in pathophysiologic mechanisms of metastasis of cancer. CCNs are secreted proteins that have a multimodular structure and are categorized as matricellular proteins. Although the prevailing view is that CCN proteins regulate biologic processes by interacting with a wide array of other proteins in the microenvironment of the extracellular matrix, the molecular mechanisms of action of CCN proteins are still poorly understood. Not dissuading the current view, however, the recent appreciation that these proteins are signaling proteins in their own right and may even be considered preproproteins controlled by endopeptidases to release a C-terminal bioactive peptide has opened new avenues of research. Also, the recent resolution of the crystal structure of two of the domains of CCN3 have provided new knowledge with implications for the entire CCN family. These resolved structures in combination with structural predictions based upon the AlphaFold artificial intelligence tool provide means to shed new light on CCN functions in context of the notable literature in the field. CCN proteins have emerged as important therapeutic targets in several disease conditions, and clinical trials are currently ongoing. Thus, a review that critically discusses structure - function relationship of CCN proteins, in particular as it relates to interactions with other proteins in the extracellular milieu and on the cell surface, as well as to cell signaling activities of these proteins, is very timely. Suggested mechanism for activation and inhibition of signaling by the CCN protein family (graphics generated with BioRender.com ).
Collapse
Affiliation(s)
- Vivi Talstad Monsen
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Håvard Attramadal
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
21
|
Qin Z, He T, Guo C, Kim JY, Quan T. CCN1 is predominantly elevated in human skin dermis by solar-simulated ultraviolet irradiation and accumulated in dermal extracellular matrix. J Cell Commun Signal 2023:10.1007/s12079-023-00767-6. [PMID: 37245186 DOI: 10.1007/s12079-023-00767-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/03/2023] [Indexed: 05/29/2023] Open
Abstract
Skin primarily comprises a collagen-rich extracellular matrix (ECM) that provides structural and functional support to the skin. Aging causes progressive loss and fragmentation of dermal collagen fibrils, leading to thin and weakened skin (Dermal aging). We previously reported that CCN1 is elevated in naturally aged human skin, photoaged human skin, and acute UV-irradiated human skin dermal fibroblasts in vivo. Elevated CCN1 alters the expression of numerous secreted proteins that have deleterious effects on the dermal microenvironment, impairing the structural integrity and function of the skin. Here we show that CCN1 is predominantly elevated in the human skin dermis by UV irradiation and accumulated in the dermal extracellular matrix. Laser capture microdissection indicated that CCN1 is predominantly induced in the dermis, not in the epidermis, by acute UV irradiation in human skin in vivo. Interestingly, while UV-induced CCN1 in the dermal fibroblasts and in the medium is transient, secreted CCN1 accumulates in the ECM. We explored the functionality of the matrix-bound CCN1 by culturing dermal fibroblasts on an acellular matrix plate that was enriched with a high concentration of CCN1. We observed that matrix-bound CCN1 activates integrin outside-in signaling resulting in the activation of FAK and its downstream target paxillin and ERK, as well as elevated MMP-1 and inhibition of collagen, in human dermal fibroblasts. These data suggest that accumulation of CCN1 in the dermal ECM is expected to progressively promote the aging of the dermis and thereby negatively impact the function of the dermis.
Collapse
Affiliation(s)
- Zhaoping Qin
- Department of Dermatology, University of Michigan Medical School, 1301 Catherine, Medical Science I, Room 6447, Ann Arbor, MI, 48109-0609, USA
| | - Tianyuan He
- Department of Dermatology, University of Michigan Medical School, 1301 Catherine, Medical Science I, Room 6447, Ann Arbor, MI, 48109-0609, USA
| | - Chunfang Guo
- Department of Dermatology, University of Michigan Medical School, 1301 Catherine, Medical Science I, Room 6447, Ann Arbor, MI, 48109-0609, USA
| | - Jun Young Kim
- Department of Dermatology, University of Michigan Medical School, 1301 Catherine, Medical Science I, Room 6447, Ann Arbor, MI, 48109-0609, USA
- Department of Dermatology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Taihao Quan
- Department of Dermatology, University of Michigan Medical School, 1301 Catherine, Medical Science I, Room 6447, Ann Arbor, MI, 48109-0609, USA.
| |
Collapse
|
22
|
Wang YK, Weng HK, Mo FE. The regulation and functions of the matricellular CCN proteins induced by shear stress. J Cell Commun Signal 2023:10.1007/s12079-023-00760-z. [PMID: 37191841 DOI: 10.1007/s12079-023-00760-z] [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: 01/18/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023] Open
Abstract
Shear stress is a frictional drag generated by the flow of fluid, such as blood or interstitial fluid, and plays a critical role in regulating cellular gene expression and functional phenotype. The matricellular CCN family proteins are dynamically regulated by shear stress of different flow patterns, and their expression significantly alters the microenvironment of cells. Secreted CCN proteins mainly bind to several cell surface integrin receptors to mediate their diverse functions in regulating cell survival, function, and behavior. Gene-knockout studies indicate major functions of CCN proteins in the cardiovascular and skeletal systems, the two primary systems in which CCN expressions are regulated by shear stress. In the cardiovascular system, the endothelium is directly exposed to vascular shear stress. Unidirectional laminar blood flow generates laminar shear stress, which promotes a mature endothelial phenotype and upregulates anti-inflammatory CCN3 expression. In contrast, disturbed flow generates oscillatory shear stress, which induces endothelial dysfunction through the induction of CCN1 and CCN2. Shear-induced CCN1 binds to integrin α6β1 and promotes superoxide production, NF-κB activation, and inflammatory gene expression in endothelial cells. Although the interaction between shear stress and CCN4-6 is not clear, CCN 4 exhibits a proinflammatory property and CCN5 inhibits vascular cell growth and migration. The crucial roles of CCN proteins in cardiovascular development, homeostasis, and disease are evident but not fully understood. In the skeletal system, mechanical loading on bone generates shear stress from interstitial fluid in the lacuna-canalicular system and promotes osteoblast differentiation and bone formation. CCN1 and CCN2 are induced and potentially mediate fluid shear stress mechanosensing in osteocytes. However, the exact roles of interstitial shear stress-induced CCN1 and CCN2 in bone are still not clear. In contrast to other CCN family proteins, CCN3 inhibits osteoblast differentiation, although its regulation by interstitial shear stress in osteocytes has not been reported. The induction of CCN proteins by shear stress in bone and their functions remain largely unknown and merit further investigation. This review discusses the expression and functions of CCN proteins regulated by shear stress in physiological conditions, diseases, and cell culture models. The roles between CCN family proteins can be compensatory or counteractive in tissue remodeling and homeostasis.
Collapse
Affiliation(s)
- Yang-Kao Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 University Road, Tainan, 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Hung-Kai Weng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
- Department of Orthopedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Fan-E Mo
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 University Road, Tainan, 70101, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
23
|
Perbal B, Perbal M, Perbal A. Cooperation is the key: the CCN biological system as a gate to high complex protein superfamilies' signaling. J Cell Commun Signal 2023:10.1007/s12079-023-00749-8. [PMID: 37166690 DOI: 10.1007/s12079-023-00749-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
Cellular signaling is generally understood as the support of communication between contiguous cells belonging to the same tissue or cells being far apart of each other, at a molecular scale, when the message emitted by the transmitters is traveling in liquid or solid matter to reach recipient targets. Subcellular signaling is also important to ensure the proper cell constitution and functioning. However cell signaling is mostly used in the first understanding, to describe how the message sent from one point to another one, will reach a target where it will be interpreted. The Cellular Communication Network (CCN) factors (Perbal et al. 2018) constitute a family of biological regulators thought to be responsible for signaling pathways coordination (Perbal 2018). Indeed, these proteins interact with a diverse group of cell receptors, such as integrins, low density lipoprotein receptors, heparan sulfate proteoglycan receptors (HSPG), and the immunoglobulin superfamily expressed exclusively in the nervous system, or with soluble factors such as bone morphogenetic proteins (BMPS) and other growth factors such as vascular endothelial growth factor, fibroblastic growth factor, and transforming growth factor (TGFbeta). Starting from the recapitulation of basic concepts in enzymology and protein-ligands interactions, we consider, in this manuscript, interpretations of the mechanistic interactions that have been put forward to explain the diversity of CCN proteins biological activities. We suggest that the cross-talks between superfamilies of proteins under the control of CCNs might play a central role in the coordination of developmental signaling pathways.
Collapse
Affiliation(s)
| | - Matthieu Perbal
- M2 Probabilités et Modèles Aléatoires, Sorbonne Université, Paris, France
| | | |
Collapse
|
24
|
Garrett EC, Bielawski AM, Ruchti E, Sherer LM, Waghmare I, Hess-Homeier D, McCabe BD, Stowers RS, Certel SJ. The matricellular protein Drosophila Cellular Communication Network Factor is required for synaptic transmission and female fertility. Genetics 2023; 223:iyac190. [PMID: 36602539 PMCID: PMC9991515 DOI: 10.1093/genetics/iyac190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 01/06/2023] Open
Abstract
Within the extracellular matrix, matricellular proteins are dynamically expressed nonstructural proteins that interact with cell surface receptors, growth factors, and proteases, as well as with structural matrix proteins. The cellular communication network factors family of matricellular proteins serve regulatory roles to regulate cell function and are defined by their conserved multimodular organization. Here, we characterize the expression and neuronal requirement for the Drosophila cellular communication network factor family member. Drosophila cellular communication network factor is expressed in the nervous system throughout development including in subsets of monoamine-expressing neurons. Drosophila cellular communication network factor-expressing abdominal ganglion neurons innervate the ovaries and uterus and the loss of Drosophila cellular communication network factor results in reduced female fertility. In addition, Drosophila cellular communication network factor accumulates at the synaptic cleft and is required for neurotransmission at the larval neuromuscular junction. Analyzing the function of the single Drosophila cellular communication network factor family member will enhance our potential to understand how the microenvironment impacts neurotransmitter release in distinct cellular contexts and in response to activity.
Collapse
Affiliation(s)
| | - Ashley M Bielawski
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Evelyne Ruchti
- Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | - Lewis M Sherer
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Indrayani Waghmare
- Department of Cell and Developmental Biology, Program in Developmental Biology, Vanderbilt-Ingram Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - David Hess-Homeier
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Brian D McCabe
- Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | - R Steven Stowers
- Department of Cell Biology and Microbiology, Montana State University, Bozeman, MT 59717, USA
| | - Sarah J Certel
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
25
|
Kubota S, Kawaki H, Perbal B, Takigawa M, Kawata K, Hattori T, Nishida T. Do not overwork: cellular communication network factor 3 for life in cartilage. J Cell Commun Signal 2023:10.1007/s12079-023-00723-4. [PMID: 36745317 DOI: 10.1007/s12079-023-00723-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 01/07/2023] [Indexed: 02/07/2023] Open
Abstract
Cellular communication network factor (CCN) 3, which is one of the founding members of the CCN family, displays diverse functions. However, this protein generally represses the proliferation of a variety of cells. Along with skeletal development, CCN3 is produced in cartilaginous anlagen, growth plate cartilage and epiphysial cartilage. Interestingly, CCN3 is drastically induced in the growth plates of mice lacking CCN2, which promotes endochondral ossification. Notably, chondrocytes in these mutant mice with elevated CCN3 production also suffer from impaired glycolysis and energy metabolism, suggesting a critical role of CCN3 in cartilage metabolism. Recently, CCN3 was found to be strongly induced by impaired glycolysis, and in our study, we located an enhancer that mediated CCN3 regulation via starvation. Subsequent investigations specified regulatory factor binding to the X-box 1 (RFX1) as a transcription factor mediating this CCN3 regulation. Impaired glycolysis is a serious problem, resulting in an energy shortage in cartilage without vasculature. CCN3 produced under such starved conditions restricts energy consumption by repressing cell proliferation, leading chondrocytes to quiescence and survival. This CCN3 regulatory system is indicated to play an important role in articular cartilage maintenance, as well as in skeletal development. Furthermore, CCN3 continues to regulate cartilage metabolism even during the aging process, probably utilizing this regulatory system. Altogether, CCN3 seems to prevent "overwork" by chondrocytes to ensure their sustainable life in cartilage by sensing energy metabolism. Similar roles are suspected to exist in relation to systemic metabolism, since CCN3 is found in the bloodstream.
Collapse
Affiliation(s)
- Satoshi Kubota
- Department of Biochemistry and Molecular Dentistry, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan.
| | - Harumi Kawaki
- Department of Oral Biochemistry, Asahi University School of Dentistry, Mizuho, Japan
| | | | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences/Dental School, Okayama, Japan
| | - Kazumi Kawata
- Department of Biochemistry and Molecular Dentistry, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Takako Hattori
- Department of Biochemistry and Molecular Dentistry, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Takashi Nishida
- Department of Biochemistry and Molecular Dentistry, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences/Dental School, Okayama, Japan
| |
Collapse
|
26
|
Peidl A, Nguyen J, Chitturi P, Riser BL, Leask A. Using the Bleomycin-Induced Model of Fibrosis to Study the Contribution of CCN Proteins to Scleroderma Fibrosis. Methods Mol Biol 2023; 2582:309-321. [PMID: 36370359 DOI: 10.1007/978-1-0716-2744-0_21] [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] [Indexed: 06/16/2023]
Abstract
Approximately 45% of the deaths in the developed world result from conditions with a fibrotic component. Although no specific, focused anti-fibrotic therapies have been approved for clinical use, a long-standing concept is that targeting CCN proteins may be useful to treat fibrosis. Herein, we summarize current data supporting the concept that targeting CCN2 may be a viable anti-fibrotic approach to treat scleroderma. Testing this hypothesis has been made possible by using a mouse model of inflammation-driven skin and lung fibrosis.
Collapse
Affiliation(s)
- Alexander Peidl
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - John Nguyen
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | | | - Bruce L Riser
- BLR Bio LLC, Kenosha, WI, USA
- Center for Cancer Cell Biology, Immunology and Infection, Department of Physiology and Biophysics, and Department of Medicine Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
27
|
Perbal B. Inception and establishment of the International CCN Society (ICCNS) and of the Journal of Cell Communication and Signaling (JCCS): A response to A. Leask's Editorial entitled "Modeling the microenvironment special issue". J Cell Commun Signal 2022; 16:627-629. [PMID: 36098895 PMCID: PMC9733759 DOI: 10.1007/s12079-022-00694-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A little over a year ago, on January 25, 2021, the new Editor-in-Chief (EiC) of JCCS stated in his Editorial: "ICCNS and JCCS were the brainchildren of Bernard Perbal, and without his energy and drive, neither would exist, to the detriment of us who are driven to solve difficult problems in science, and not picking low-hanging fruit. All one has to do is examine all the editorials written in JCCS (and CCS!) to see evidence of this. It will be tough to fill those shoes."I disagree with the assertion in the Editorial published on March 29, 2022 that G. Martin contributed "to the initial growth of the International CCN Society, and, ultimately, to the establishment of this journal." My opinion is based on the evidence that the International CCN Society (ICCNS) and its official organ journal, the Journal of Cell Communication and Signaling (JCCS), were created by myself. Over a span of 21 years until the present, and in spite of his contribution to the early history of CTGF, we never heard from G. Martin being involved or interested in any aspect of the ICCNS and its biannual meetings, nor in any aspect in the growth of JCCS.In order to further clarify the confusion stemming from the Editorial in question and to give credit where it is due, I provide below detailed evidence that undoubtedly ascribes the true inception of both ICCNS and JCCS, and merit to the efforts of all those who trusted and supported us during the initial difficult creative moments.I am of the opinion that the Editorial, and the implications that it carries do not justice to the efforts of those who were really involved in the creation of both the ICCNS and JCCS.In the name of respectful scientific integrity, I will provide the evidence that correctly attributes the inception of ICCNS and JCCS.
Collapse
|
28
|
Reprogramming of cancer-associated fibroblasts by apoptotic cancer cells inhibits lung metastasis via Notch1-WISP-1 signaling. Cell Mol Immunol 2022; 19:1373-1391. [PMID: 36241874 PMCID: PMC9708692 DOI: 10.1038/s41423-022-00930-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/14/2022] [Indexed: 12/05/2022] Open
Abstract
The interplay between apoptotic cancer cells and the tumor microenvironment modulates cancer progression and metastasis. Cancer-associated fibroblasts (CAFs) play a crucial role in promoting these events through paracrine communication. Here, we demonstrate that conditioned medium (CM) from lung CAFs exposed to apoptotic cancer cells suppresses TGF-β1-induced migration and invasion of cancer cells and CAFs. Direct exposure of CAFs to apoptotic 344SQ cells (ApoSQ) inhibited CAF migration and invasion and the expression of CAF activation markers. Enhanced secretion of Wnt-induced signaling protein 1 (WISP-1) by CAFs exposed to ApoSQ was required for these antimigratory and anti-invasive effects. Pharmacological inhibition of Notch1 activation or siRNA-mediated Notch1 silencing prevented WISP-1 production by CAFs and reversed the antimigratory and anti-invasive effects. Enhanced expression of the Notch ligand delta-like protein 1 on the surface of ultraviolet-irradiated apoptotic lung cancer cells triggered Notch1-WISP-1 signaling. Phosphatidylserine receptor brain-specific angiogenesis inhibitor 1 (BAI1)-Rac1 signaling, which facilitated efferocytosis by CAFs, participated in crosstalk with Notch1 signaling for optimal production of WISP-1. In addition, a single injection of ApoSQ enhanced WISP-1 production, suppressed the expression of CAF activation markers in isolated Thy1+ CAFs, and inhibited lung metastasis in syngeneic immunocompetent mice via Notch1 signaling. Treatment with CM from CAFs exposed to ApoSQ suppressed tumor growth and lung metastasis, whereas treatment with WISP-1-immunodepleted CM from CAFs exposed to ApoSQ reversed the antitumorigenic and antimetastatic effects. Therefore, treatment with CM from CAFs exposed to apoptotic lung cancer cells could be therapeutically applied to suppress CAF activation, thereby preventing cancer progression and metastasis.
Collapse
|
29
|
Cárdenas-León CG, Mäemets-Allas K, Klaas M, Lagus H, Kankuri E, Jaks V. Matricellular proteins in cutaneous wound healing. Front Cell Dev Biol 2022; 10:1073320. [PMID: 36506087 PMCID: PMC9730256 DOI: 10.3389/fcell.2022.1073320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Cutaneous wound healing is a complex process that encompasses alterations in all aspects of the skin including the extracellular matrix (ECM). ECM consist of large structural proteins such as collagens and elastin as well as smaller proteins with mainly regulative properties called matricellular proteins. Matricellular proteins bind to structural proteins and their functions include but are not limited to interaction with cell surface receptors, cytokines, or protease and evoking a cellular response. The signaling initiated by matricellular proteins modulates differentiation and proliferation of cells having an impact on the tissue regeneration. In this review we give an overview of the matricellular proteins that have been found to be involved in cutaneous wound healing and summarize the information known to date about their functions in this process.
Collapse
Affiliation(s)
| | - Kristina Mäemets-Allas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Mariliis Klaas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Heli Lagus
- Department of Plastic Surgery and Wound Healing Centre, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Viljar Jaks
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia,Dermatology Clinic, Tartu University Clinics, Tartu, Estonia,*Correspondence: Viljar Jaks,
| |
Collapse
|
30
|
Fuchs L, Mausner-Fainberg K, Luban A, Asseyer SE, Golan M, Benhamou M, Volovitz I, Regev K, Vigiser I, Piura Y, Kolb H, Paul F, Karni A. CTGF/CCN2 has a possible detrimental role in the inflammation and the remyelination failure in the early stages of multiple sclerosis. J Neuroimmunol 2022; 371:577936. [DOI: 10.1016/j.jneuroim.2022.577936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/19/2022] [Accepted: 07/24/2022] [Indexed: 11/15/2022]
|
31
|
Chang KS, Chen ST, Sung HC, Hsu SY, Lin WY, Hou CP, Lin YH, Feng TH, Tsui KH, Juang HH. WNT1 Inducible Signaling Pathway Protein 1 Is a Stroma-Specific Secreting Protein Inducing a Fibroblast Contraction and Carcinoma Cell Growth in the Human Prostate. Int J Mol Sci 2022; 23:ijms231911437. [PMID: 36232736 PMCID: PMC9570503 DOI: 10.3390/ijms231911437] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/18/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
The WNT1 inducible signaling pathway protein 1 (WISP1), a member of the connective tissue growth factor family, plays a crucial role in several important cellular functions in a highly tissue-specific manner. Results of a RT-qPCR indicated that WISP1 expressed only in cells of the human prostate fibroblasts, HPrF and WPMY-1, but not the prostate carcinoma cells in vitro. Two major isoforms (WISP1v1 and WISP1v2) were identified in the HPrF cells determined by RT-PCR and immunoblot assays. The knock-down of a WISP1 blocked cell proliferation and contraction, while treating respectively with the conditioned medium from the ectopic WISP1v1- and WISPv2-overexpressed 293T cells enhanced the migration of HPrF cells. The TNFα induced WISP1 secretion and cell contraction while the knock-down of WISP1 attenuated these effects, although TNFα did not affect the proliferation of the HPrF cells. The ectopic overexpression of WISP1v1 but not WISP1v2 downregulated the N-myc downstream regulated 1 (NDRG1) while upregulating N-cadherin, slug, snail, and vimentin gene expressions which induced not only the cell proliferation and invasion in vitro but also tumor growth of prostate carcinoma cells in vivo. The results confirmed that WISP1 is a stroma-specific secreting protein, enhancing the cell migration and contraction of prostate fibroblasts, as well as the proliferation, invasion, and tumor growth of prostate carcinoma cells.
Collapse
Affiliation(s)
- Kang-Shuo Chang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Syue-Ting Chen
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Hsin-Ching Sung
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Shu-Yuan Hsu
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Wei-Yin Lin
- Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Chen-Pang Hou
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Tsui-Hsia Feng
- School of Nursing, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Ke-Hung Tsui
- Department of Urology, Shuang Ho Hospital, New Taipei City 235041, Taiwan
- TMU Research Center of Urology and Kidney, Department of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (K.-H.T.); (H.-H.J.); Tel.: +886-3-2118800 (ext. 5071) (H.-H.J.); Fax: +886-3-2118112 (H.-H.J.)
| | - Horng-Heng Juang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
- Correspondence: (K.-H.T.); (H.-H.J.); Tel.: +886-3-2118800 (ext. 5071) (H.-H.J.); Fax: +886-3-2118112 (H.-H.J.)
| |
Collapse
|
32
|
CCN1/Integrin α 5β 1 Instigates Free Fatty Acid-Induced Hepatocyte Lipid Accumulation and Pyroptosis through NLRP3 Inflammasome Activation. Nutrients 2022; 14:nu14183871. [PMID: 36145246 PMCID: PMC9505842 DOI: 10.3390/nu14183871] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 12/20/2022] Open
Abstract
Hyperlipidemia with high blood levels of free fatty acids (FFA) is the leading cause of non-alcoholic steatohepatitis. CCN1 is a secreted matricellular protein that drives various cellular functions, including proliferation, migration, and differentiation. However, its role in mediating FFA-induced pro-inflammatory cell death and its underlying molecular mechanisms have not been characterized. In this study, we demonstrated that CCN1 was upregulated in the livers of obese mice. The increase in FFA-induced CCN1 was evaluated in vitro by treating hepatocytes with a combination of oleic acid and palmitic acid (2:1). Gene silencing using specific small interfering RNAs (siRNA) revealed that CCN1 participated in FFA-induced intracellular lipid accumulation, caspase-1 activation, and hepatocyte pyroptosis. Next, we identified integrin α5β1 as a potential receptor of CCN1. Co-immunoprecipitation demonstrated that the binding between CCN1 and integrin α5β1 increased in hepatocytes upon FFA stimulation in the livers of obese mice. Similarly, the protein levels of integrin α5 and β1 were increased in vitro and in vivo. Experiments with specific siRNAs confirmed that integrin α5β1 played a part in FFA-induced intracellular lipid accumulation, NLRP3 inflammasome activation, and pyroptosis in hepatocytes. In conclusion, these results provide novel evidence that the CCN1/integrin α5β1 is a novel mediator that drives hepatic lipotoxicity via NLRP3-dependent pyroptosis.
Collapse
|
33
|
Xi Y, LaCanna R, Ma HY, N'Diaye EN, Gierke S, Caplazi P, Sagolla M, Huang Z, Lucio L, Arlantico A, Jeet S, Brightbill H, Emson C, Wong A, Morshead KB, DePianto DJ, Roose-Girma M, Yu C, Tam L, Jia G, Ramalingam TR, Marsters S, Ashkenazi A, Kim SH, Kelly R, Wu S, Wolters PJ, Feldstein AE, Vander Heiden JA, Ding N. A WISP1 antibody inhibits MRTF signaling to prevent the progression of established liver fibrosis. Cell Metab 2022; 34:1377-1393.e8. [PMID: 35987202 DOI: 10.1016/j.cmet.2022.07.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 06/06/2022] [Accepted: 07/19/2022] [Indexed: 01/18/2023]
Abstract
Fibrosis is the major risk factor associated with morbidity and mortality in patients with non-alcoholic steatohepatitis (NASH)-driven chronic liver disease. Although numerous efforts have been made to identify the mediators of the initiation of liver fibrosis, the molecular underpinnings of fibrosis progression remain poorly understood, and therapies to arrest liver fibrosis progression are elusive. Here, we identify a pathway involving WNT1-inducible signaling pathway protein 1 (WISP1) and myocardin-related transcription factor (MRTF) as a central mechanism driving liver fibrosis progression through the integrin-dependent transcriptional reprogramming of myofibroblast cytoskeleton and motility. In mice, WISP1 deficiency protects against fibrosis progression, but not fibrosis onset. Moreover, the therapeutic administration of a novel antibody blocking WISP1 halted the progression of existing liver fibrosis in NASH models. These findings implicate the WISP1-MRTF axis as a crucial determinant of liver fibrosis progression and support targeting this pathway by antibody-based therapy for the treatment of NASH fibrosis.
Collapse
Affiliation(s)
- Ying Xi
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Ryan LaCanna
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Hsiao-Yen Ma
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Elsa-Noah N'Diaye
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Sarah Gierke
- Department of Pathology, Genentech, South San Francisco, CA, USA
| | - Patrick Caplazi
- Department of Pathology, Genentech, South San Francisco, CA, USA
| | - Meredith Sagolla
- Department of Pathology, Genentech, South San Francisco, CA, USA
| | - Zhiyu Huang
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Laura Lucio
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Alexander Arlantico
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Surinder Jeet
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Hans Brightbill
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Claire Emson
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Aaron Wong
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Katrina B Morshead
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Daryle J DePianto
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Merone Roose-Girma
- Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Charles Yu
- Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Lucinda Tam
- Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Guiquan Jia
- Department of Biomarker Discovery, Genentech, South San Francisco, CA, USA
| | | | - Scot Marsters
- Department of Cancer Immunology, Genentech, South San Francisco, CA, USA
| | - Avi Ashkenazi
- Department of Cancer Immunology, Genentech, South San Francisco, CA, USA
| | - Si Hyun Kim
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Ryan Kelly
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Shuang Wu
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Paul J Wolters
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Ariel E Feldstein
- Department of Pediatrics, University of California, San Diego, San Diego, CA, USA
| | | | - Ning Ding
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
34
|
Fibroblast Growth Factors and Cellular Communication Network Factors: Intimate Interplay by the Founding Members in Cartilage. Int J Mol Sci 2022; 23:ijms23158592. [PMID: 35955724 PMCID: PMC9369280 DOI: 10.3390/ijms23158592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023] Open
Abstract
Fibroblast growth factors (FGFs) constitute a large family of signaling molecules that act in an autocrine/paracrine, endocrine, or intracrine manner, whereas the cellular communication network factors (CCN) family is composed of six members that manipulate extracellular signaling networks. FGFs and CCNs are structurally and functionally distinct, except for the common characteristics as matricellular proteins. Both play significant roles in the development of a variety of tissues and organs, including the skeletal system. In vertebrates, most of the skeletal parts are formed and grow through a process designated endochondral ossification, in which chondrocytes play the central role. The growth plate cartilage is the place where endochondral ossification occurs, and articular cartilage is left to support the locomotive function of joints. Several FGFs, including FGF-2, one of the founding members of this family, and all of the CCNs represented by CCN2, which is required for proper skeletal development, can be found therein. Research over a decade has revealed direct binding of CCN2 to FGFs and FGF receptors (FGFRs), which occasionally affect the biological outcome via FGF signaling. Moreover, a recent study uncovered an integrated regulation of FGF and CCN genes by FGF signaling. In this review, after a brief introduction of these two families, molecular and genetic interactions between CCN and FGF family members in cartilage, and their biological effects, are summarized. The molecular interplay represents the mutual involvement of the other in their molecular functions, leading to collaboration between CCN2 and FGFs during skeletal development.
Collapse
|
35
|
Li Z, Cong X, Kong W. Matricellular proteins: Potential biomarkers and mechanistic factors in aortic aneurysms. J Mol Cell Cardiol 2022; 169:41-56. [DOI: 10.1016/j.yjmcc.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/30/2022] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
|
36
|
Marinkovic M, Dai Q, Gonzalez AO, Tran ON, Block TJ, Harris SE, Salmon AB, Yeh CK, Dean DD, Chen XD. Matrix-bound Cyr61/CCN1 is required to retain the properties of the bone marrow mesenchymal stem cell niche but is depleted with aging. Matrix Biol 2022; 111:108-132. [PMID: 35752272 PMCID: PMC10069241 DOI: 10.1016/j.matbio.2022.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 05/30/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022]
Abstract
Previously, we showed that extracellular matrices (ECMs), produced ex vivo by various types of stromal cells, direct bone marrow mesenchymal stem cells (BM-MSCs) in a tissue-specific manner and recapitulate physiologic changes characteristic of the aging microenvironment. In particular, BM-MSCs obtained from elderly donors and cultured on ECM produced by young BM stromal cells showed improved quantity, quality and osteogenic differentiation. In the present study, we searched for matrix components that are required for a functional BM-MSC niche by comparing ECMs produced by BM stromal cells from "young" (≤25 y/o) versus "elderly" (≥60 y/o) donors. With increasing donor age, ECM fibrillar organization and mechanical integrity deteriorated, along with the ability to promote BM-MSC proliferation and responsiveness to growth factors. Proteomic analyses revealed that the matricellular protein, Cyr61/CCN1, was present in young, but undetectable in elderly, BM-ECM. To assess the role of Cyr61 in the BM-MSC niche, we used genetic methods to down-regulate the incorporation of Cyr61 during production of young ECM and up-regulate its incorporation in elderly ECM. The results showed that Cyr61-depleted young ECM lost the ability to promote BM-MSC proliferation and growth factor responsiveness. However, up-regulating the incorporation of Cyr61 during synthesis of elderly ECM restored its ability to support BM-MSC responsiveness to osteogenic factors such as BMP-2 and IGF-1. We next examined aging bone and compared bone mineral density and Cyr61 content of L4-L5 vertebral bodies in "young" (9-11 m/o) and "elderly" (21-33 m/o) mice. Our analyses showed that low bone mineral density was associated with decreased amounts of Cyr61 in osseous tissue of elderly versus young mice. Our results strongly demonstrate a novel role for ECM-bound Cyr61 in the BM-MSC niche, where it is responsible for retention of BM-MSC proliferation and growth factor responsiveness, while depletion of Cyr61 from the BM niche contributes to an aging-related dysregulation of BM-MSCs. Our results also suggest new potential therapeutic targets for treating age-related bone loss by restoring specific ECM components to the stem cell niche.
Collapse
Affiliation(s)
- Milos Marinkovic
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States; Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX 78249, United States; Research Service, South Texas Veterans Health Care System, Audie Murphy VA Medical Center, San Antonio, TX 78229(,) United States
| | - Qiuxia Dai
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Aaron O Gonzalez
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States; Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX 78249, United States
| | - Olivia N Tran
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States; Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX 78249, United States
| | - Travis J Block
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States; Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX 78249, United States
| | - Stephen E Harris
- Department of Periodontics, University of Texas Health Science Center at San Antonio, TX 78229, United States
| | - Adam B Salmon
- Department of Molecular Medicine, Barshop Institute for Longevity and Aging Studies at The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, United States; Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, Audie Murphy VA Medical Center, San Antonio, TX 78229, United States
| | - Chih-Ko Yeh
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States; Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, Audie Murphy VA Medical Center, San Antonio, TX 78229, United States
| | - David D Dean
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States; Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX 78249, United States
| | - Xiao-Dong Chen
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States; Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX 78249, United States; Research Service, South Texas Veterans Health Care System, Audie Murphy VA Medical Center, San Antonio, TX 78229(,) United States.
| |
Collapse
|
37
|
Targeting fibrosis, mechanisms and cilinical trials. Signal Transduct Target Ther 2022; 7:206. [PMID: 35773269 PMCID: PMC9247101 DOI: 10.1038/s41392-022-01070-3] [Citation(s) in RCA: 217] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 02/05/2023] Open
Abstract
Fibrosis is characterized by the excessive extracellular matrix deposition due to dysregulated wound and connective tissue repair response. Multiple organs can develop fibrosis, including the liver, kidney, heart, and lung. Fibrosis such as liver cirrhosis, idiopathic pulmonary fibrosis, and cystic fibrosis caused substantial disease burden. Persistent abnormal activation of myofibroblasts mediated by various signals, such as transforming growth factor, platelet-derived growth factor, and fibroblast growh factor, has been recongized as a major event in the occurrence and progression of fibrosis. Although the mechanisms driving organ-specific fibrosis have not been fully elucidated, drugs targeting these identified aberrant signals have achieved potent anti-fibrotic efficacy in clinical trials. In this review, we briefly introduce the aetiology and epidemiology of several fibrosis diseases, including liver fibrosis, kidney fibrosis, cardiac fibrosis, and pulmonary fibrosis. Then, we summarise the abnormal cells (epithelial cells, endothelial cells, immune cells, and fibroblasts) and their interactions in fibrosis. In addition, we also focus on the aberrant signaling pathways and therapeutic targets that regulate myofibroblast activation, extracellular matrix cross-linking, metabolism, and inflammation in fibrosis. Finally, we discuss the anti-fibrotic drugs based on their targets and clinical trials. This review provides reference for further research on fibrosis mechanism, drug development, and clinical trials.
Collapse
|
38
|
Cheng N, Kim KH, Lau LF. Senescent hepatic stellate cells promote liver regeneration through IL-6 and ligands of CXCR2. JCI Insight 2022; 7:158207. [PMID: 35708907 PMCID: PMC9431681 DOI: 10.1172/jci.insight.158207] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/15/2022] [Indexed: 11/29/2022] Open
Abstract
Senescent cells have long been associated with deleterious effects in aging-related pathologies, although recent studies have uncovered their beneficial roles in certain contexts, such as wound healing. We have found that hepatic stellate cells (HSCs) underwent senescence within 2 days after 2/3 partial hepatectomy (PHx) in young (2–3 months old) mice, and the elimination of these senescent cells by using the senolytic drug ABT263 or by using a genetic mouse model impaired liver regeneration. Senescent HSCs secrete IL-6 and CXCR2 ligands as part of the senescence-associated secretory phenotype, which induces multiple signaling pathways to stimulate liver regeneration. IL-6 activates STAT3, induces Yes-associated protein (YAP) activation through SRC family kinases, and synergizes with CXCL2 to activate ERK1/2 to stimulate hepatocyte proliferation. The administration of either IL-6 or CXCL2 partially restored liver regeneration in mice with senescent cell elimination, and the combination of both fully restored liver weight recovery. Furthermore, the matricellular protein central communication network factor 1 (CCN1, previously called CYR61) was rapidly elevated in response to PHx and induced HSC senescence. Knockin mice expressing a mutant CCN1 unable to bind integrin α6β1 were deficient in senescent cells and liver regeneration after PHx. Thus, HSC senescence, largely induced by CCN1, is a programmed response to PHx and plays a critical role in liver regeneration through signaling pathways activated by IL-6 and ligands of CXCR2.
Collapse
Affiliation(s)
- Naiyuan Cheng
- Biochemistry and Molecular Genetics, University of Illinois at Chicago College of Medicine, Chicago, United States of America
| | - Ki-Hyun Kim
- Biochemistry and Molecular Genetics, University of Illinois at Chicago College of Medicine, Chicago, United States of America
| | - Lester F Lau
- Biochemistry and Molecular Genetics, University of Illinois at Chicago College of Medicine, Chicago, United States of America
| |
Collapse
|
39
|
Won JH, Choi JS, Jun JI. CCN1 interacts with integrins to regulate intestinal stem cell proliferation and differentiation. Nat Commun 2022; 13:3117. [PMID: 35660741 PMCID: PMC9166801 DOI: 10.1038/s41467-022-30851-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/20/2022] [Indexed: 12/14/2022] Open
Abstract
Intestinal stem cells (ISCs) at the crypt base contribute to intestinal homeostasis through a balance between self-renewal and differentiation. However, the molecular mechanisms regulating this homeostatic balance remain elusive. Here we show that the matricellular protein CCN1/CYR61 coordinately regulates ISC proliferation and differentiation through distinct pathways emanating from CCN1 interaction with integrins αvβ3/αvβ5. Mice that delete Ccn1 in Lgr5 + ISCs or express mutant CCN1 unable to bind integrins αvβ3/αvβ5 exhibited exuberant ISC expansion and enhanced differentiation into secretory cells at the expense of absorptive enterocytes in the small intestine, leading to nutrient malabsorption. Analysis of crypt organoids revealed that through integrins αvβ3/αvβ5, CCN1 induces NF-κB-dependent Jag1 expression to regulate Notch activation for differentiation and promotes Src-mediated YAP activation and Dkk1 expression to control Wnt signaling for proliferation. Moreover, CCN1 and YAP amplify the activities of each other in a regulatory loop. These findings establish CCN1 as a niche factor in the intestinal crypts, providing insights into how matrix signaling exerts overarching control of ISC homeostasis. Intestinal stem cells contribute to homeostasis through a balance between self-renewal and differentiation. Here the authors show that CCN1 is an intestinal stem cell niche factor that activates integrin αvβ3/αvβ5 signaling to regulate proliferation and differentiation through distinct downstream pathways.
Collapse
Affiliation(s)
- Jong Hoon Won
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, 900 South Ashland Avenue, Chicago, IL, 60607, USA
| | - Jacob S Choi
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, 900 South Ashland Avenue, Chicago, IL, 60607, USA.,Department of Medicine, Northwestern University, 676 North St. Clair street Arkes Suite 2330, Chicago, IL, 60611, USA
| | - Joon-Il Jun
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, 900 South Ashland Avenue, Chicago, IL, 60607, USA.
| |
Collapse
|
40
|
Song MH, Yoo J, Oh JG, Kook H, Park WJ, Jeong D. Matricellular Protein CCN5 Gene Transfer Ameliorates Cardiac and Skeletal Dysfunction in mdx/utrn (±) Haploinsufficient Mice by Reducing Fibrosis and Upregulating Utrophin Expression. Front Cardiovasc Med 2022; 9:763544. [PMID: 35557546 PMCID: PMC9088811 DOI: 10.3389/fcvm.2022.763544] [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: 08/24/2021] [Accepted: 03/31/2022] [Indexed: 12/21/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disorder characterized by progressive muscle degeneration due to dystrophin gene mutations. Patients with DMD initially experience muscle weakness in their limbs during adolescence. With age, patients develop fatal respiratory and cardiac dysfunctions. During the later stages of the disease, severe cardiac fibrosis occurs, compromising cardiac function. Previously, our research showed that the matricellular protein CCN5 has antifibrotic properties. Therefore, we hypothesized that CCN5 gene transfer would ameliorate cardiac fibrosis and thus improve cardiac function in DMD-induced cardiomyopathy. We utilized mdx/utrn (±) haploinsufficient mice that recapitulated the DMD-disease phenotypes and used an adeno-associated virus serotype-9 viral vector for CCN5 gene transfer. We evaluated the onset of cardiac dysfunction using echocardiography and determined the experimental starting point in 13-month-old mice. Two months after CCN5 gene transfer, cardiac function was significantly enhanced, and cardiac fibrosis was ameliorated. Additionally, running performance was improved in CCN5 gene-transfected mice. Furthermore, in silico gene profiling analysis identified utrophin as a novel transcriptional target of CCN5. This was supplemented by a utrophin promoter assay and RNA-seq analysis, which confirmed that CCN5 was directly associated with utrophin expression. Our results showed that CCN5 may be a promising therapeutic molecule for DMD-induced cardiac and skeletal dysfunction.
Collapse
Affiliation(s)
- Min Ho Song
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Jimeen Yoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jae Gyun Oh
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Hyun Kook
- Basic Research Laboratory, Chonnam National University Medical School, Gwangju, South Korea
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Dongtak Jeong
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Molecular and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea
| |
Collapse
|
41
|
Song MH, Jo Y, Kim YK, Kook H, Jeong D, Park WJ. The TSP-1 domain of the matricellular protein CCN5 is essential for its nuclear localization and anti-fibrotic function. PLoS One 2022; 17:e0267629. [PMID: 35476850 PMCID: PMC9045603 DOI: 10.1371/journal.pone.0267629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/12/2022] [Indexed: 02/07/2023] Open
Abstract
The matricellular protein CCN5 exerts anti-fibrotic activity in hearts partly by inducing reverse trans-differentiation of myofibroblasts (MyoFBs) to fibroblasts (FBs). CCN5 consists of three structural domains: an insulin-like growth factor binding protein (IGFBP), a von Willebrand factor type C (VWC), and a thrombospondin type 1 (TSP-1). In this study, we set out to elucidate the roles of these domains in the context of the reverse trans-differentiation of MyoFBs to FBs. First, human cardiac FBs were trans-differentiated to MyoFBs by treatment with TGF-β; this was then reversed by treatment with recombinant human CCN5 protein or various recombinant proteins comprising individual or paired CCN5 domains. Subcellular localization of these recombinant proteins was analyzed by immunocytochemistry, cellular fractionation, and western blotting. Anti-fibrotic activity was also evaluated by examining expression of MyoFB-specific markers, α-SMA and fibronectin. Our data show that CCN5 is taken up by FBs and MyoFBs mainly via clathrin-mediated endocytosis, which is essential for the function of CCN5 during the reverse trans-differentiation of MyoFBs. Furthermore, we showed that the TSP-1 domain is essential and sufficient for endocytosis and nuclear localization of CCN5. However, the TSP-1 domain alone is not sufficient for the anti-fibrotic function of CCN5; either the IGFBP or VWC domain is needed in addition to the TSP-1 domain.
Collapse
Affiliation(s)
- Min Ho Song
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Yongjoon Jo
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Jeollanam-do, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanam-do, Republic of Korea
| | - Dongtak Jeong
- Department of Molecular & Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, Gyeonggi-do, Republic of Korea
- * E-mail: (WJP); (DJ)
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
- * E-mail: (WJP); (DJ)
| |
Collapse
|
42
|
Zhou YT, Yu YQ, Yang H, Yang H, Huo YF, Huang Y, Tian XX, Fang WG. Extracellular ATP promotes angiogenesis and adhesion of TNBC cells to endothelial cells via up-regulation of CTGF. Cancer Sci 2022; 113:2457-2471. [PMID: 35441763 PMCID: PMC9277410 DOI: 10.1111/cas.15375] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 12/01/2022] Open
Abstract
Our previous works have indicated that extracellular ATP is an important prometastasis factor. However, the molecular mechanism involved needs to be further studied. We demonstrated that extracellular ATP treatment could upregulate the expression of connective tissue growth factor (CTGF) in both triple‐negative breast cancer (TNBC) cells and endothelial cells (ECs). Extracellular ATP stimulated the migration of TNBC cells and ECs, and angiogenesis of ECs via the P2Y2––YAP‐CTGF axis. Furthermore, we demonstrated that adenosine triphosphate (ATP) stimulated TNBC cell adhesion to ECs and transmigration through the EC layer via CTGF by upregulation of integrin β1 on TNBC cells and VCAM‐1 on ECs. Both apyrase (ATP‐diphosphohydrolase) and CTGF shRNA treatments could inhibit the metastasis of inoculated tumors to lung and liver in a mouse model, and these treated tumors had fewer blood vessels. Collectively, our data indicated that extracellular ATP promotes tumor angiogenesis and the interactions between TNBC cells and ECs through upregulation of CTGF, thereby stimulating TNBC metastasis. The pleiotropic effects of ATP in angiogenesis and cell adhesion suggest that extracellular ATP or CTGF could be an effective target for TNBC therapy.
Collapse
Affiliation(s)
- Yan-Ting Zhou
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Yu-Qing Yu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Hui Yang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Han Yang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Yan-Fei Huo
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Yang Huang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Xin-Xia Tian
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Wei-Gang Fang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| |
Collapse
|
43
|
Zhou H, Zhang Y, Wang J, Yan Y, Liu Y, Shi X, Zhang Q, Xu X. The CREB and AP-1-Dependent Cell Communication Network Factor 1 Regulates Porcine Epidemic Diarrhea Virus-Induced Cell Apoptosis Inhibiting Virus Replication Through the p53 Pathway. Front Microbiol 2022; 13:831852. [PMID: 35418961 PMCID: PMC8996185 DOI: 10.3389/fmicb.2022.831852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/14/2022] [Indexed: 12/15/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) infection causes severe diarrhea, dehydration, and high mortality in sick pigs, causing huge economic losses to the pig industry. However, the relationship between cell communication network factor 1 (CCN1) and PEDV infection has not been reported. In this study, we showed that the expression of CCN1 was enhanced by PEDV infection, and we observed that PEDV promotes the CREB and AP-1 activation to promote CCN1 expression. The PKA and p38 inhibitors significantly suppress CCN1 expression, indicating that PEDV-induced CCN1 expression may be through PKA and p38 pathway. Further tests confirmed that CREB and AP-1 are regulated by PKA and p38, respectively. Overexpression of CCN1 decreased the replication of PEDV, whereas knockdown of CCN1 increased the replication of PEDV. We proved that the overexpression of CCN1 increased the phosphorylation level of p53, promoted the expresion of Bax and the cleavage of caspase 9 and caspase 3, and inhibited the production of Bcl-2. CCN1 knockdown decreased the phosphorylation level of p53, inhibited the production of Bax and the cleavage of caspase 9 and caspase 3, and promoted the expression of Bcl-2. The treatment of PFT-α (p53 inhibitor) significantly suppressed the expression of cleaved caspase 9 and caspase 3, leading to the decrease of apoptosis. Together, these studies showed that PEDV promotes the activation of CREB and AP-1 to increase the expression of CCN1. Overexpression of CCN1 promotes apoptosis by elevating p53 protein phosphorylation and inhibits PEDV replication, and knockdown of CCN1 inhibits apoptosis by decreasing p53 protein phosphorylation and promotes PEDV replication. Our study could provide some reference for the molecular mechanisms of PEDV-induced CCN1 induction and supply a new therapeutic target for PEDV.
Collapse
Affiliation(s)
- Hongchao Zhou
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Yuting Zhang
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Jingjing Wang
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Yuchao Yan
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Yi Liu
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Xiaojie Shi
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Qi Zhang
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Xingang Xu
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| |
Collapse
|
44
|
Hellewell AL, Heesom KJ, Jepson MA, Adams JC. PDIA3/ERp57 promotes a matrix-rich secretome that stimulates fibroblast adhesion through CCN2. Am J Physiol Cell Physiol 2022; 322:C624-C644. [PMID: 35196163 PMCID: PMC8977143 DOI: 10.1152/ajpcell.00258.2021] [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] [Indexed: 11/22/2022]
Abstract
The matricellular glycoprotein thrombospondin1 (TSP1) has complex roles in the extracellular matrix and at cell surfaces, but relatively little is known about its intracellular associations prior to secretion. To search for novel intracellular interactions of TSP1 in situ, we carried out a biotin ligase-based TSP1 interactome screen and identified protein disulphide isomerase A3 (PDIA3/ERp57) as a novel candidate binding protein. In validation, TSP1 and PDIA3 were established to bind in vitro and to colocalise in the endoplasmic reticulum of human dermal fibroblasts (HDF). Loss of PDIA3 function, either by pharmacological inhibition in HDF or in Pdia3-/- mouse embryo fibroblasts (Pdia3-/-MEF), led to alterations in the composition of cell-derived ECM, involving changed abundance of fibronectin and TSP1, and was correlated with reduced cell spreading, altered organisation of F-actin and reduced focal adhesions. These cellular phenotypes of Pdia3-/-MEF were normalised by exposure to conditioned medium (WTCM) or extracellular matrix (WTECM) from wild-type (WT)-MEF. Rescue depended on PDIA3 activity in WT-MEF, and was not prevented by immunodepletion of fibronectin. Heparin-binding proteins in WTCM were found to be necessary for rescue. Comparative quantitative tandem-mass-tag proteomics and functional assays on the heparin-binding secretomes of WT-MEF and Pdia3-/- MEF identified multiple ECM and growth factor proteins to be down-regulated in the CM of Pdia3-/- MEF. Of these, CCN2 was identified to be necessary for the adhesion-promoting activity of WTCM on Pdia3-/- MEF and to bind TSP1. Thus, PDIA3 coordinates fibroblast production of an ECM-rich, pro-adhesive microenvironment, with implications for PDIA3 as a translational target.
Collapse
Affiliation(s)
| | - Kate J Heesom
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Mark A Jepson
- Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
45
|
Espinoza I, Kurapaty C, Park CH, Vander Steen T, Kleer CG, Wiley E, Rademaker A, Cuyàs E, Verdura S, Buxó M, Reynolds C, Menendez JA, Lupu R. Depletion of CCN1/CYR61 reduces triple-negative/basal-like breast cancer aggressiveness. Am J Cancer Res 2022; 12:839-851. [PMID: 35261806 PMCID: PMC8899977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 02/03/2022] [Indexed: 06/14/2023] Open
Abstract
Triple-negative/basal-like breast cancer (BC) is characterized by aggressive biological features, which allow relapse and metastatic spread to occur more frequently than in hormone receptor-positive (luminal) subtypes. The molecular complexity of triple-negative/basal-like BC poses major challenges for the implementation of targeted therapies, and chemotherapy remains the standard approach at all stages. The matricellular protein cysteine-rich angiogenic inducer 61 (CCN1/CYR61) is associated with aggressive metastatic phenotypes and poor prognosis in BC, but it is unclear whether anti-CCN1 approaches can be successfully applied in triple-negative/basal-like BC. Herein, we first characterized the prevalence of CNN1 expression in matched samples of primary tumors and metastatic relapse in a series of patients with BC. We then investigated the biological effect of CCN1 depletion on tumorigenic traits in vitro and in vivo using archetypal TNBC cell lines. Immunohistochemical analyses of tissue microarrays revealed a significant increase of the highest CCN1 score in recurrent tissues of triple-negative/basal-like BC tumors. Stable silencing of CCN1 in triple-negative/basal-like BC cells promoted a marked reduction in the expression of the CCN1 integrin receptor αvβ3, inhibited anchorage-dependent cell growth, reduced clonogenicity, and impaired migration capacity. In an orthotopic model of triple-negative/basal-like BC, silencing of CCN1 notably reduced tumor burden, which was accompanied by decreased microvessel density and concurrent induction of the luminal epithelial marker E-cadherin. Thus, CNN1/CYR61-targeting strategies might have therapeutic value in suppressing the biological aggressiveness of triple-negative/basal-like BC.
Collapse
Affiliation(s)
- Ingrid Espinoza
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo ClinicRochester, MN 55905, USA
- Department of Preventive Medicine, John D. Bower School of Population Health, University of Mississippi Medical CenterJackson, MS 39216, USA
- Cancer Institute, School of Medicine, University of Mississippi Medical CenterJackson, MS 39216, USA
| | - Chandra Kurapaty
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo ClinicRochester, MN 55905, USA
| | - Cheol-Hong Park
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo ClinicRochester, MN 55905, USA
| | - Travis Vander Steen
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo ClinicRochester, MN 55905, USA
| | - Celina G Kleer
- Department of Pathology, University of MichiganAnn Arbor, MI 48109, USA
| | - Elizabeth Wiley
- Department of Pathology, University of Illinois at ChicagoChicago, IL 60607, USA
| | - Alfred Rademaker
- Department of Preventive Medicine, Northwestern University Feinberg School of MedicineChicago, IL 60611, USA
| | - Elisabet Cuyàs
- Girona Biomedical Research Institute17190 Salt, Girona, Spain
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism & Cancer Group, Catalan Institute of Oncology17007 Girona, Spain
| | - Sara Verdura
- Girona Biomedical Research Institute17190 Salt, Girona, Spain
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism & Cancer Group, Catalan Institute of Oncology17007 Girona, Spain
| | - Maria Buxó
- Statistical and Methodological Advice Unit, Girona Biomedical Research Institute17190 Salt, Girona, Spain
| | - Carol Reynolds
- Department of Pathology, Division of Anatomic Pathology, Mayo ClinicRochester, MN 55905, USA
| | - Javier A Menendez
- Girona Biomedical Research Institute17190 Salt, Girona, Spain
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism & Cancer Group, Catalan Institute of Oncology17007 Girona, Spain
| | - Ruth Lupu
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo ClinicRochester, MN 55905, USA
- Mayo Clinic Cancer CenterRochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology Laboratory, Mayo Clinic MinnesotaRochester, MN 55905, USA
| |
Collapse
|
46
|
Espinoza I, Yang L, Steen TV, Vellon L, Cuyàs E, Verdura S, Lau L, Menendez JA, Lupu R. Binding of the angiogenic/senescence inducer CCN1/CYR61 to integrin α 6β 1 drives endocrine resistance in breast cancer cells. Aging (Albany NY) 2022; 14:1200-1213. [PMID: 35148282 PMCID: PMC8876916 DOI: 10.18632/aging.203882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 01/29/2022] [Indexed: 11/25/2022]
Abstract
CCN1/CYR61 promotes angiogenesis, tumor growth and chemoresistance by binding to its integrin receptor αvβ3 in endothelial and breast cancer (BC) cells. CCN1 controls also tissue regeneration by engaging its integrin receptor α6β1 to induce fibroblast senescence. Here, we explored if the ability of CCN1 to drive an endocrine resistance phenotype in estrogen receptor-positive BC cells relies on interactions with either αvβ3 or α6β1. First, we took advantage of site-specific mutagenesis abolishing the CCN1 receptor-binding sites to αvβ3 and α6β1 to determine the integrin partner responsible for CCN1-driven endocrine resistance. Second, we explored a putative nuclear role of CCN1 in regulating ERα-driven transcriptional responses. Retroviral forced expression of a CCN1 derivative with a single amino acid change (D125A) that abrogates binding to αvβ3 partially phenocopied the endocrine resistance phenotype induced upon overexpression of wild-type (WT) CCN1. Forced expression of the CCN1 mutant TM, which abrogates all the T1, H1, and H2 binding sites to α6β1, failed to bypass the estrogen requirement for anchorage-independent growth or to promote resistance to tamoxifen. Wild-type CCN1 promoted estradiol-independent transcriptional activity of ERα and enhanced ERα agonist response to tamoxifen. The α6β1-binding-defective TM-CCN1 mutant lost the ERα co-activator-like behavior of WT-CCN1. Co-immunoprecipitation assays revealed a direct interaction between endogenous CCN1 and ERα, and in vitro approaches confirmed the ability of recombinant CCN1 to bind ERα. CCN1 signaling via α6β1, but not via αvβ3, drives an endocrine resistance phenotype that involves a direct binding of CCN1 to ERα to regulate its transcriptional activity in ER+ BC cells.
Collapse
Affiliation(s)
- Ingrid Espinoza
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Mayo Clinic, Rochester, 55905 MN, USA.,Current address: Department of Preventive Medicine, John D. Bower School of Population Health, University of Mississippi Medical Center, Jackson, MS 39216, USA.,Current address: Cancer Institute, School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Lin Yang
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Mayo Clinic, Rochester, 55905 MN, USA
| | - Travis Vander Steen
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Mayo Clinic, Rochester, 55905 MN, USA
| | - Luciano Vellon
- Stem Cells Laboratory, Institute of Biology and Experimental Medicine (IBYME-CONICET), Buenos Aires C1428ADN, Argentina
| | - Elisabet Cuyàs
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona 17005, Spain.,Girona Biomedical Research Institute, Salt, Girona 17190, Spain
| | - Sara Verdura
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona 17005, Spain.,Girona Biomedical Research Institute, Salt, Girona 17190, Spain
| | - Lester Lau
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Javier A Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona 17005, Spain.,Girona Biomedical Research Institute, Salt, Girona 17190, Spain
| | - Ruth Lupu
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Mayo Clinic, Rochester, 55905 MN, USA.,Department of Biochemistry and Molecular Biology Laboratory, Mayo Clinic Minnesota, Rochester, MN 55905, USA.,Mayo Clinic Cancer Center, Rochester, MN 55905, USA
| |
Collapse
|
47
|
Rayego-Mateos S, Morgado-Pascual JL, Lavoz C, Rodrigues-Díez RR, Márquez-Expósito L, Tejera-Muñoz A, Tejedor-Santamaría L, Rubio-Soto I, Marchant V, Ruiz-Ortega M. CCN2 Binds to Tubular Epithelial Cells in the Kidney. Biomolecules 2022; 12:biom12020252. [PMID: 35204752 PMCID: PMC8869303 DOI: 10.3390/biom12020252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 02/01/2023] Open
Abstract
Cellular communication network-2 (CCN2), also called connective tissue growth factor (CTGF), is considered a fibrotic biomarker and has been suggested as a potential therapeutic target for kidney pathologies. CCN2 is a matricellular protein with four distinct structural modules that can exert a dual function as a matricellular protein and as a growth factor. Previous experiments using surface plasmon resonance and cultured renal cells have demonstrated that the C-terminal module of CCN2 (CCN2(IV)) interacts with the epidermal growth factor receptor (EGFR). Moreover, CCN2(IV) activates proinflammatory and profibrotic responses in the mouse kidney. The aim of this paper was to locate the in vivo cellular CCN2/EGFR binding sites in the kidney. To this aim, the C-terminal module CCN2(IV) was labeled with a fluorophore (Cy5), and two different administration routes were employed. Both intraperitoneal and direct intra-renal injection of Cy5-CCN2(IV) in mice demonstrated that CCN2(IV) preferentially binds to the tubular epithelial cells, while no signal was detected in glomeruli. Moreover, co-localization of Cy5-CCN2(IV) binding and activated EGFR was found in tubules. In cultured tubular epithelial cells, live-cell confocal microscopy experiments showed that EGFR gene silencing blocked Cy5-CCN2(IV) binding to tubuloepithelial cells. These data clearly show the existence of CCN2/EGFR binding sites in the kidney, mainly in tubular epithelial cells. In conclusion, these studies show that circulating CCN2(IV) can directly bind and activate tubular cells, supporting the role of CCN2 as a growth factor involved in kidney damage progression.
Collapse
Affiliation(s)
- Sandra Rayego-Mateos
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (S.R.-M.); (L.M.-E.); (A.T.-M.); (L.T.-S.); (I.R.-S.); (V.M.)
- Red de Investigación Renal (REDinREN), Av. de Monforte de Lemos, 5, 28029 Madrid, Spain;
| | - José Luis Morgado-Pascual
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Cordoba, Spain;
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14071 Cordoba, Spain
| | - Carolina Lavoz
- Division of Nephrology, School of Medicine, Universidad Austral Chile, Valdivia 5090000, Chile;
| | - Raúl R. Rodrigues-Díez
- Red de Investigación Renal (REDinREN), Av. de Monforte de Lemos, 5, 28029 Madrid, Spain;
- Translational Immunology Laboratory, Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
| | - Laura Márquez-Expósito
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (S.R.-M.); (L.M.-E.); (A.T.-M.); (L.T.-S.); (I.R.-S.); (V.M.)
- Red de Investigación Renal (REDinREN), Av. de Monforte de Lemos, 5, 28029 Madrid, Spain;
| | - Antonio Tejera-Muñoz
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (S.R.-M.); (L.M.-E.); (A.T.-M.); (L.T.-S.); (I.R.-S.); (V.M.)
- Red de Investigación Renal (REDinREN), Av. de Monforte de Lemos, 5, 28029 Madrid, Spain;
| | - Lucía Tejedor-Santamaría
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (S.R.-M.); (L.M.-E.); (A.T.-M.); (L.T.-S.); (I.R.-S.); (V.M.)
- Red de Investigación Renal (REDinREN), Av. de Monforte de Lemos, 5, 28029 Madrid, Spain;
| | - Irene Rubio-Soto
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (S.R.-M.); (L.M.-E.); (A.T.-M.); (L.T.-S.); (I.R.-S.); (V.M.)
- Red de Investigación Renal (REDinREN), Av. de Monforte de Lemos, 5, 28029 Madrid, Spain;
| | - Vanessa Marchant
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (S.R.-M.); (L.M.-E.); (A.T.-M.); (L.T.-S.); (I.R.-S.); (V.M.)
- Red de Investigación Renal (REDinREN), Av. de Monforte de Lemos, 5, 28029 Madrid, Spain;
| | - Marta Ruiz-Ortega
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (S.R.-M.); (L.M.-E.); (A.T.-M.); (L.T.-S.); (I.R.-S.); (V.M.)
- Red de Investigación Renal (REDinREN), Av. de Monforte de Lemos, 5, 28029 Madrid, Spain;
- Correspondence:
| |
Collapse
|
48
|
Zhong T, Jiang Z, Wang X, Wang H, Song M, Chen W, Yang S. Key genes associated with prognosis and metastasis of clear cell renal cell carcinoma. PeerJ 2022; 10:e12493. [PMID: 35036081 PMCID: PMC8740509 DOI: 10.7717/peerj.12493] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/25/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is a tumor that frequently shows the hematogenous pathway and tends to be resistant to radiotherapy and chemotherapy. However, the exact mechanism of ccRCC metastasis remains unknown. METHODS Differentially expressed genes (DEGs) of three gene expression profiles (GSE85258, GSE105288 and GSE22541) downloaded from the Gene Expression Omnibus (GEO) database were analyzed by GEO2R analysis, and co-expressed DEGs among the datasets were identified using a Venn drawing tool. The co-expressed DEGs were investigated using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, and hub genes were determined based on the protein-protein interaction network established by STRING. After survival analysis performed on UALCAN website, possible key genes were selected and verified in ccRCC cell lines and ccRCC tissues (n = 44). Statistical analysis was conducted using GraphPad Prism (Version 8.1.1). RESULTS A total of 104 co-expressed DEGs were identified in the three datasets. Pathway analysis revealed that these genes were enriched in the extracellular matrix (ECM)-receptor interaction, protein digestion and absorption and focal adhesion. Survival analysis on 17 hub genes revealed that four key genes with a significant impact on survival: procollagen C-endopeptidase enhancer (PCOLCE), prolyl 4-hydroxylase subunit beta (P4HB), collagen type VI alpha 2 (COL6A2) and collagen type VI alpha 3 (COL6A3). Patients with higher expression of these key genes had worse survival than those with lower expression. In vitro experiments revealed that the mRNA expression levels of PCOLCE, P4HB and COL6A2 were three times higher and that of COL6A3 mRNA was 16 times higher in the metastatic ccRCC cell line Caki-1 than the corresponding primary cell line Caki-2. Immunohistochemistry revealed higher expression of the proteins encoded by these four genes in metastatic ccRCC compared with tumors from the corresponding primary sites, with statistical significance. CONCLUSION PCOLCE, P4HB, COL6A2 and COL6A3 are upregulated in metastatic ccRCC and might be related to poor prognosis and distant metastases.
Collapse
Affiliation(s)
- Tingting Zhong
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zeying Jiang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiangdong Wang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Honglei Wang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Meiyi Song
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenfang Chen
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shicong Yang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
49
|
Tadijan A, Humphries JD, Samaržija I, Stojanović N, Zha J, Čuljak K, Tomić M, Paradžik M, Nestić D, Kang H, Humphries MJ, Ambriović-Ristov A. The Tongue Squamous Carcinoma Cell Line Cal27 Primarily Employs Integrin α6β4-Containing Type II Hemidesmosomes for Adhesion Which Contribute to Anticancer Drug Sensitivity. Front Cell Dev Biol 2021; 9:786758. [PMID: 34977030 PMCID: PMC8716755 DOI: 10.3389/fcell.2021.786758] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022] Open
Abstract
Integrins are heterodimeric cell surface glycoproteins used by cells to bind to the extracellular matrix (ECM) and regulate tumor cell proliferation, migration and survival. A causative relationship between integrin expression and resistance to anticancer drugs has been demonstrated in different tumors, including head and neck squamous cell carcinoma. Using a Cal27 tongue squamous cell carcinoma model, we have previously demonstrated that de novo expression of integrin αVβ3 confers resistance to several anticancer drugs (cisplatin, mitomycin C and doxorubicin) through a mechanism involving downregulation of active Src, increased cell migration and invasion. In the integrin αVβ3 expressing Cal27-derived cell clone 2B1, αVβ5 expression was also increased, but unrelated to drug resistance. To identify the integrin adhesion complex (IAC) components that contribute to the changes in Cal27 and 2B1 cell adhesion and anticancer drug resistance, we isolated IACs from both cell lines. Mass spectrometry (MS)-based proteomics analysis indicated that both cell lines preferentially, but not exclusively, use integrin α6β4, which is classically found in hemidesmosomes. The anticancer drug resistant cell clone 2B1 demonstrated an increased level of α6β4 accompanied with increased deposition of a laminin-332-containing ECM. Immunofluorescence and electron microscopy demonstrated the formation of type II hemidesmosomes by both cell types. Furthermore, suppression of α6β4 expression in both lines conferred resistance to anticancer drugs through a mechanism independent of αVβ3, which implies that the cell clone 2B1 would have been even more resistant had the upregulation of α6β4 not occurred. Taken together, our results identify a key role for α6β4-containing type II hemidesmosomes in regulating anticancer drug sensitivity.
Collapse
Affiliation(s)
- Ana Tadijan
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
- Laboratory for Protein Dynamics, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Jonathan D. Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Ivana Samaržija
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Nikolina Stojanović
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Junzhe Zha
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Kristina Čuljak
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Marija Tomić
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Mladen Paradžik
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Davor Nestić
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, South Korea
| | - Martin J. Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Andreja Ambriović-Ristov
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
50
|
Cialdai F, Bolognini D, Vignali L, Iannotti N, Cacchione S, Magi A, Balsamo M, Vukich M, Neri G, Donati A, Monici M, Capaccioli S, Lulli M. Effect of space flight on the behavior of human retinal pigment epithelial ARPE-19 cells and evaluation of coenzyme Q10 treatment. Cell Mol Life Sci 2021; 78:7795-7812. [PMID: 34714361 PMCID: PMC11073052 DOI: 10.1007/s00018-021-03989-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/20/2021] [Accepted: 10/13/2021] [Indexed: 10/20/2022]
Abstract
Astronauts on board the International Space Station (ISS) are exposed to the damaging effects of microgravity and cosmic radiation. One of the most critical and sensitive districts of an organism is the eye, particularly the retina, and > 50% of astronauts develop a complex of alterations designated as spaceflight-associated neuro-ocular syndrome. However, the pathogenesis of this condition is not clearly understood. In the current study, we aimed to explore the cellular and molecular effects induced in the human retinal pigment ARPE-19 cell line by their transfer to and 3-day stay on board the ISS in the context of an experiment funded by the Agenzia Spaziale Italiana. Treatment of cells on board the ISS with the well-known bioenergetic, antioxidant, and antiapoptotic coenzyme Q10 was also evaluated. In the ground control experiment, the cells were exposed to the same conditions as on the ISS, with the exception of microgravity and radiation. The transfer of ARPE-19 retinal cells to the ISS and their living on board for 3 days did not affect cell viability or apoptosis but induced cytoskeleton remodeling consisting of vimentin redistribution from the cellular boundaries to the perinuclear area, underlining the collapse of the network of intermediate vimentin filaments under unloading conditions. The morphological changes endured by ARPE-19 cells grown on board the ISS were associated with changes in the transcriptomic profile related to the cellular response to the space environment and were consistent with cell dysfunction adaptations. In addition, the results obtained from ARPE-19 cells treated with coenzyme Q10 indicated its potential to increase cell resistance to damage.
Collapse
Affiliation(s)
- Francesca Cialdai
- ASAcampus Joint Laboratory, ASA Res. Div., Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università Degli Studi Di Firenze, Firenze, Italy
| | - Davide Bolognini
- Department of Experimental and Clinical Medicine, Università Degli Studi Di Firenze, Firenze, Italy
| | - Leonardo Vignali
- ASAcampus Joint Laboratory, ASA Res. Div., Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università Degli Studi Di Firenze, Firenze, Italy
| | - Nicola Iannotti
- Department of Life Sciences, Università Degli Studi Di Siena, Siena, Italy
| | - Stefano Cacchione
- Department of Biology and Biotechnology "Charles Darwin", Università Di Roma "La Sapienza", Roma, Italy
| | - Alberto Magi
- Department of Information Engineering, Università Degli Studi Di Firenze, Firenze, Italy
| | | | | | | | | | - Monica Monici
- ASAcampus Joint Laboratory, ASA Res. Div., Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università Degli Studi Di Firenze, Firenze, Italy
| | - Sergio Capaccioli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università Degli Studi Di Firenze, viale Morgagni 50, 50134, Firenze, Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università Degli Studi Di Firenze, viale Morgagni 50, 50134, Firenze, Italy.
| |
Collapse
|