1
|
Abdul-Azees PA, Rajesh R, Block TJ, Dean DD, Yeh CK, Capitano M, Kacena M, Chen XD, Marinković M. CCN Proteins as Matricellular Regulators of Bone in Aging and Disease. Curr Osteoporos Rep 2025; 23:23. [PMID: 40407982 PMCID: PMC12102002 DOI: 10.1007/s11914-025-00915-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/27/2025] [Indexed: 05/26/2025]
Abstract
PURPOSE OF REVIEW This review explores the role of cell communication network (CCN) proteins in regulating skeletal physiology, aging, and disease, particularly within the context of balanced bone remodeling. RECENT FINDINGS Recent conceptualization of paracrine and endocrine networks in bone marrow as a form of osteoimmunological crosstalk suggests a significant role for matricellular signaling in regulating bone homeostasis. As multifunctional adapters of cell-matrix interactions, CCNs are emerging as a focal point for parathyroid hormone (PTH) signaling and regulation of the RANKL/RANK/OPG axis in skeletal aging. Altered bone marrow CCN expression creates a permissive environment for accelerated postmenopausal bone loss and may contribute to the pathogenesis of osteoporosis and other diseases related to skeletal aging. CCNs modulate fundamental signaling mechanisms in bone development, homeostasis and repair. During aging, dysregulation of CCNs may negatively affect skeletal health and contribute to disease progression. As a result, CCNs may constitute promising therapeutic targets for improving and maintaining aging bone health.
Collapse
Affiliation(s)
- Parveez Ahamed Abdul-Azees
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229 - 3900, USA
| | - Rahul Rajesh
- Arkansas College of Osteopathic Medicine, Fort Smith, AR, 72916, USA
| | - Travis J Block
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229 - 3900, USA
| | - David D Dean
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229 - 3900, USA
| | - Chih-Ko Yeh
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229 - 3900, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
| | - Maegan Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Melissa Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Research Service, Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA
| | - Xiao-Dong Chen
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229 - 3900, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Miloš Marinković
- Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA.
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Research Service, Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
| |
Collapse
|
2
|
Fan H, Zhao H, Gao L, Dong Y, Zhang P, Yu P, Ji Y, Chen ZS, Liang X, Chen Y. CCN1 Enhances Tumor Immunosuppression through Collagen-Mediated Chemokine Secretion in Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500589. [PMID: 40287974 DOI: 10.1002/advs.202500589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/03/2025] [Indexed: 04/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense, immunosuppressive tumor microenvironment (TME) that limits therapeutic efficacy. This study investigates the role of cellular communication network factor 1 (CCN1, also known as Cyr61), an extracellular matrix-associated protein, in modulating the TME of PDAC. It is demonstrated that Ccn1 promotes PDAC progression by upregulating collagen and chemokine expression, thereby facilitating immune cell exclusion and enhancing tumor growth. Using a Ccn1-deficient PDAC model, decreased collagen and chemokine levels are observed, resulting in increased infiltration of cytotoxic immune cells and reduced myeloid-derived suppressor cells (MDSCs). Furthermore, Ccn1-deficient tumors exhibit heightened sensitivity to gemcitabine and show enhanced responsiveness to anti-programmed cell death 1 (anti-PD1) therapy. Mechanistically, Ccn1 regulates chemokine production through collagen expression, with chemokine levels remaining suppressed even upon interferon-gamma treatment in collagen-deficient cells. These findings highlight Ccn1 as a potential therapeutic target that reprograms the TME to enhance the efficacy of both chemotherapy and immunotherapy in PDAC, providing a novel approach for overcoming immune resistance in PDAC.
Collapse
Affiliation(s)
- Hongjie Fan
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Huzi Zhao
- Department of Pathology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, China
| | - Lili Gao
- Department of Pathology, Xinhua Hospital Affiliated to Medicine School of Shanghai Jiaotong University, Shanghai, 200082, China
| | - Yucheng Dong
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100006, China
| | - Pei Zhang
- Department of Mathematics, University of Maryland, College Park, Maryland, MD 20742, USA
| | - Pengfei Yu
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Yunfei Ji
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Xinmiao Liang
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Yang Chen
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| |
Collapse
|
3
|
Kong H, Qiao Y, Qi D, Zhao S, Cao Z, Feng J, Li Y, Liu Y, Liu T. circ_0002970 promotes fibroblast-like synoviocytes invasion and the inflammatory response through Hippo/YAP signaling to induce CTGF/CCN1 expression in rheumatoid arthritis. Arthritis Res Ther 2025; 27:97. [PMID: 40281642 PMCID: PMC12023364 DOI: 10.1186/s13075-025-03562-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by synovial inflammation, hyperplasia, and joint destruction. Fibroblast-like synoviocytes (FLSs) are key effector cells in RA, contributing to synovial invasion, extracellular matrix degradation, and inflammatory cytokine secretion. Recent studies suggest that circular RNAs (circRNAs) regulate cellular function and disease progression, but their role in RA remains unclear. The Hippo-YAP signaling pathway governs cell proliferation, apoptosis, and extracellular matrix remodeling, and its dysregulation is linked to RA synovial hyperplasia and inflammation. However, whether circRNAs regulate Hippo-YAP signaling in RA-FLSs has not been fully elucidated. This study investigates the role of circ_0002970 in RA progression and its regulation of the Hippo-YAP pathway. METHODS Synovial tissues from RA patients, osteoarthritis (OA) patients, and healthy controls were collected. Differentially expressed circRNAs were identified via RNA sequencing. The expression of circ_0002970 was validated via qRT‒PCR, FISH, and RNase R digestion assays. The functional experiments included transfection, migration/invasion assays, ELISA, and Western blotting to evaluate its role in RA-FLSs. RESULTS Circ_0002970 was significantly upregulated in RA-FLSs. Knockdown of circ_0002970 suppressed RA-FLS migration, invasion, and IL-6 secretion. Mechanistically, circ_0002970 knockdown downregulated the expression of Hippo-YAP pathway components (YAP, CTGF, and CCN1) and decreased the expression of MMP-9 and MMP-13, which are critical for cartilage degradation. Furthermore, verteporfin (VP)-mediated inhibition of Hippo-YAP reversed the effects of circ_0002970 overexpression. CONCLUSION These findings highlight circ_0002970 as a novel regulator of RA-FLS migration, invasion, and inflammation via the Hippo-YAP signaling pathway.
Collapse
Affiliation(s)
- Haoran Kong
- Zhengzhou University People's Hospital & Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, Henan, China
| | - Yushuang Qiao
- Zhengzhou University People's Hospital & Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, Henan, China
| | - Dahu Qi
- Zhengzhou University People's Hospital & Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, Henan, China
| | - Shixin Zhao
- Zhengzhou University People's Hospital & Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, Henan, China
| | - Zhiming Cao
- Zhengzhou University People's Hospital & Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, Henan, China
| | - Junhao Feng
- Zhengzhou University People's Hospital & Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, Henan, China
| | - Yitong Li
- Zhengzhou University People's Hospital & Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, Henan, China
| | - Yunke Liu
- Zhengzhou University People's Hospital & Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, Henan, China.
| | - Tao Liu
- Zhengzhou University People's Hospital & Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, Henan, China.
| |
Collapse
|
4
|
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
|
5
|
Lang A, Eastburn EA, Younesi M, Nijsure MP, Siciliano C, Pranatharthi Haran A, Panebianco CJ, Seidl E, Tang R, Alsberg E, Willett NJ, Gottardi R, Huh D, Boerckel JD. CYR61 delivery promotes angiogenesis during bone fracture repair. NPJ Regen Med 2025; 10:20. [PMID: 40263309 PMCID: PMC12015299 DOI: 10.1038/s41536-025-00398-y] [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: 05/09/2024] [Accepted: 01/31/2025] [Indexed: 04/24/2025] Open
Abstract
Compromised vascular supply and insufficient neovascularization impede bone repair, increasing risk of non-union. CYR61, Cysteine-rich angiogenic inducer of 61kD (also known as CCN1), is a matricellular growth factor that has been implicated in fracture repair. Here, we map the distribution of endogenous CYR61 during bone repair and evaluate the effects of recombinant CYR61 delivery on vascularized bone regeneration. In vitro, CYR61 treatment did not alter chondrogenesis or osteogenic gene expression, but significantly enhanced angiogenesis. In a mouse femoral fracture model, CYR61 delivery did not alter cartilage or bone formation, but accelerated neovascularization during fracture repair. Early initiation of ambulatory mechanical loading disrupted CYR61-induced neovascularization. Together, these data indicate that CYR61 delivery can enhance angiogenesis during bone repair, particularly for fractures with stable fixation, and may have therapeutic potential for fractures with limited blood vessel supply.
Collapse
Affiliation(s)
- Annemarie Lang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Emily A Eastburn
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Mousa Younesi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Madhura P Nijsure
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Carly Siciliano
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Annapurna Pranatharthi Haran
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Elizabeth Seidl
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rui Tang
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL, USA
| | - Nick J Willett
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
- The Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Riccardo Gottardi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Huang Y, Huang J, Zhan J, Chen M, Zheng J, He J, Fang W, Zhang L, Li J. CCN1 is a therapeutic target upregulated in EML4-ALK mutant lung adenocarcinoma reversibly resistant to alectinib. Cell Death Dis 2025; 16:303. [PMID: 40234387 PMCID: PMC12000322 DOI: 10.1038/s41419-025-07601-4] [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: 09/08/2024] [Revised: 03/11/2025] [Accepted: 03/27/2025] [Indexed: 04/17/2025]
Abstract
There is limited understanding of the phenomenon of reversible drug resistance, which is characterized by tumor cells regaining sensitivity when the drug is changed or withdrawn after a period of drug resistance. This phenomenon is usually not associated with genetic alterations of tumor cells. In this study, reversible resistant state was induced by alectinib in EML4-ALK mutant lung cancer cell. By performing RNA sequencing on reversible drug-resistant cell line to examine changes in transcriptional profile, significant change in CCN1 was detected after withdrawal and repeated administration of alectinib. Targeting CCN1 resulted in inhibition of tumor cell proliferation and angiogenesis, and restoration of sensitivity to alectinib in reversible drug-resistant cells. Further studies revealed that CCN1 could affect the expression of VEGFA by affecting AKT phosphorylation, and the change of NF-κB could impact the activation of CCN1-AKT-VEGFA pathway. Suppressing NF-κB or CCN1 receptor could improve the sensitivity to alectinib, further suggesting that NF-κB and CCN1 might play a key role in overcoming reversible drug resistance.
Collapse
Affiliation(s)
- Yihua Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
- Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jie Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Jianhua Zhan
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Maojian Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiani Zheng
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Junyi He
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Wenfeng Fang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China.
| | - Li Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China.
| | - Jing Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
7
|
Di Patria L, Habel N, Olaso R, Fernandes R, Brenner C, Stefanovska B, Fromigue O. C-terminal binding protein-2 triggers CYR61-induced metastatic dissemination of osteosarcoma in a non-hypoxic microenvironment. J Exp Clin Cancer Res 2025; 44:83. [PMID: 40038783 PMCID: PMC11881356 DOI: 10.1186/s13046-025-03350-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/23/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Osteosarcoma is the most prevalent cancer-related bone disease diagnosed in the pediatric age group. The rapid development of metastatic lesions and resistance to chemotherapy remain major mechanisms responsible for the failure of treatments and poor outcome. We established that the expression level of Cysteine-rich protein 61 (CYR61/CCN1) correlates to tumor neo-vascularization and dissemination in preclinical and clinical osteosarcoma samples. The aim of this study was to investigate the CYR61-related mechanisms leading to the acquisition of metastatic capacity by osteosarcoma cells. METHODS Transcriptomic data issued from RNA-seq were subjected to pathways and gene set enrichment analyses. Murine and human cell lines with overexpressed or downregulated C-terminal Binding protein 2 (CtBP2) were established by lentiviral transduction. Cell metabolic activity was assessed by Seahorse XF Analyzer; cell replication rate by BrdU incorporation assay; stemness by clonogenicity assay and RT-qPCR detection of markers; cell migration by wound healing assay and Boyden chambers system; cell invasion using Matrigel coated Boyden chambers or fluorescence microscopy of Matrigel embedded 3D spheroids. FFPE samples derived from syngeneic tumor cells grafts into BALB/c mice were analyzed by IHC. The protein interactome was predicted in silico using the STRING database. RESULTS GSEA revealed that CYR61 modulate the transcription process. The in vitro expression level of CtBP2 and Cyr61 correlated positively in a panel of osteosarcoma cell lines. In silico analysis of protein-protein interaction network revealed a link with stemness markers. Variations in CtBP2 expression levels influenced stemness markers expression levels, cell clonogenicity, cell migration, Matrix Metalloproteinase activity and cell invasion. Surprisingly, while induction of CtBP2 expression under CYR61 correlated with the metastatic dissemination process in vivo, it occurred only at the invasive front of tumors. Hypoxic conditions in central tumor region interfered with CtBP2 induction of expression. CONCLUSIONS Our findings identify for the first time that CtBP2 acts as a required critical inducing factor in the CYR61-related metastatic progression of osteosarcoma, by favoring cell migration and invasiveness. Moreover, we demonstrate that while CtBP2 is a downstream transcriptional target of CYR61 signaling cascade, it occurs only under non-hypoxic conditions. The present study suggests that CtBP2 may represent a potential pivotal target for therapeutic management of metastases spreading in osteosarcoma.
Collapse
Affiliation(s)
- Laura Di Patria
- Inserm UMR981, Gustave Roussy Cancer Campus, Molecular Predictors and New Targets in Oncology, Université Paris Saclay, 39 Rue Camille Desmoulins, Villejuif, F-94805, France
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Nadia Habel
- Inserm UMR981, Gustave Roussy Cancer Campus, Molecular Predictors and New Targets in Oncology, Université Paris Saclay, 39 Rue Camille Desmoulins, Villejuif, F-94805, France
- Present Address : Centre de Traitement de L'Information Génétique (CTIG), INRAE, Jouy en Josas, France
| | - Robert Olaso
- Université Paris Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France
| | - Romain Fernandes
- CNRS UMR9018, Gustave Roussy, Metabolic and Systemic Aspects of Oncogenesis for New Therapeutic Approaches, Université Paris Saclay, Villejuif, France
| | - Catherine Brenner
- CNRS UMR9018, Gustave Roussy, Metabolic and Systemic Aspects of Oncogenesis for New Therapeutic Approaches, Université Paris Saclay, Villejuif, France
| | - Bojana Stefanovska
- Inserm UMR981, Gustave Roussy Cancer Campus, Molecular Predictors and New Targets in Oncology, Université Paris Saclay, 39 Rue Camille Desmoulins, Villejuif, F-94805, France
- Present Address: Department of Biochemistry and Structural Biology, Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Olivia Fromigue
- Inserm UMR981, Gustave Roussy Cancer Campus, Molecular Predictors and New Targets in Oncology, Université Paris Saclay, 39 Rue Camille Desmoulins, Villejuif, F-94805, France.
| |
Collapse
|
8
|
Hirabayashi S, Uyeda A, Manabe I, Yonezu Y, Saito T, Saido TC, Misawa H, Ogasawara Y, Kinoshita K, Muramatsu R. CCN1 derived from vascular endothelial cells impairs cognitive function in Alzheimer's disease model mice. J Pharmacol Sci 2025; 157:146-155. [PMID: 39929589 DOI: 10.1016/j.jphs.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 05/08/2025] Open
Abstract
Vascular endothelial cell-expressing molecules regulate neuronal function. Although cerebrovascular dysregulation is a hallmark of Alzheimer's disease (AD), the effect of changes in molecular expression on neuronal function in vascular endothelial cells during disease progression is not clear. In this study, we demonstrated that the cellular communication network factor 1 (CCN1), which is highly expressed in vascular endothelial cells during the chronic stage of AD in mice, is involved in the impairment of cognitive function. Vascular endothelial cells isolated from the brains of AppNL-G-F mice show differential expression of genes, including CCN1. CCN1 treatment decreased the synaptic number in cultured hippocampal cells, with changes in the expression of genes associated with morphological changes. In vivo, AppNL-G-F mice with CCN1 silencing in vascular endothelial cells demonstrated high spine density and improved spatial learning. No significant change was observed in the number of microglia/macrophages, astrocytes, and amyloid-beta (Aβ) accumulation in the hippocampus of the mice. These results suggest that CCN1 is a key factor modulating neurological dysfunction through neurovascular interactions.
Collapse
Affiliation(s)
- Shuntaro Hirabayashi
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Analytical Biochemistry, Graduate School of Pharmaceutical Sciences, Meiji Pharmaceutical University, Tokyo, Japan; Department of Pharmacognosy and Phytochemistry, Meiji Pharmaceutical University, Tokyo, Japan
| | - Akiko Uyeda
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ichiro Manabe
- Department of Systems Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoshino Yonezu
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Hidemi Misawa
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Yuki Ogasawara
- Department of Analytical Biochemistry, Graduate School of Pharmaceutical Sciences, Meiji Pharmaceutical University, Tokyo, Japan
| | - Kaoru Kinoshita
- Department of Pharmacognosy and Phytochemistry, Meiji Pharmaceutical University, Tokyo, Japan
| | - Rieko Muramatsu
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.
| |
Collapse
|
9
|
Zhao Y, Gu L, Chen Y, Lin Y, Xing J, Xu D, Su Z, Huang Z. Cysteine-Rich Protein 61 (CCN1) Deficiency Alleviated Cardiac Remodeling in 5/6 Nephrectomized Mice by Suppressing the MAPK Signaling Pathway. Cardiovasc Ther 2025; 2025:6813183. [PMID: 40225592 PMCID: PMC11986956 DOI: 10.1155/cdr/6813183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/28/2025] [Indexed: 04/15/2025] Open
Abstract
Background: With the progression of chronic kidney disease (CKD), we can often observe cardiac remodeling, fibrosis, and cardiac failure in patients. Cysteine-rich protein 61 (CCN1) is an extracellular matrix protein that plays a reuse role in cardiac remodeling. However, whether CCN1 participates in the crosslink between the heart and kidney in CKD and the potential mechanism remains unknown. Methods: We constructed a mouse model of CKD by 5/6 nephrectomy (5/6 Nx). Hematoxylin-eosin staining (H&E), Masson's trichrome staining, and Sirius red staining were used to observe cardiac morphology and fibrosis. H9c2 cells were treated with si-CCN1 or si-NC or mitogen-activated protein kinase (MAPK)-related inhibitors or agonist before being cultured with 5/6 Nx mouse serum. The relative protein level was detected by Western blotting. Results: We observed that CCN1 expression was markedly enhanced in the serum and heart tissues, accompanied by disordered myocardial arrangement, obvious cardiac fibrosis, hypertrophy, and decreased cardiac systolic function reflected by echocardiography. The relative markers collagen 1 (COL-1), transforming growth factor-β (TGF-β), heavy-chain cardiac myosin (MyHC), and atrial natriuretic peptide (ANP) presented an increase in expression. In vivo and in vitro, after the knockdown of CCN1, the above results in the CKD group or CKD serum group were reversed; in addition, the MAPK signaling pathway was obviously activated due to 5/6 Nx, which was abolished by CCN1 inhibition. CCN1 silencing or MAPK pathway inhibition also decreased the expression of myocardial fibrosis and hypertrophy markers in H9c2 cells, while MAPK-related agonist partly reversed the effect of CCN1 inhibition. Conclusion: Our in vivo and in vitro study showed that specific CCN1 deficiency markedly alleviated cardiac remodeling in 5/6 Nx mice through the inhibition of the MAPK pathway.
Collapse
MESH Headings
- Animals
- Nephrectomy
- Cysteine-Rich Protein 61/genetics
- Cysteine-Rich Protein 61/deficiency
- Cysteine-Rich Protein 61/metabolism
- Disease Models, Animal
- Ventricular Remodeling/drug effects
- Fibrosis
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/enzymology
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/physiopathology
- Male
- Mice, Inbred C57BL
- Cell Line
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/enzymology
- Ventricular Function, Left
- MAP Kinase Signaling System
- Rats
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/pathology
- Mice
- Mitogen-Activated Protein Kinase Kinases/metabolism
Collapse
Affiliation(s)
- Yihan Zhao
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Liang Gu
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of General Medicine, Mianyang Central Hospital, Mianyang, Sichuan, China
| | - Yunxuan Chen
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yibei Lin
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jincheng Xing
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Diyan Xu
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhen Su
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouqing Huang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
10
|
Chen Q, Li X, Li P, Liu H, Zhang Q, He L, Tang Z, Song H. Integrative bioinformatics analysis reveals novel insights into osteoarthritis pathogenesis and diagnostic biomarkers. BMC Musculoskelet Disord 2024; 25:999. [PMID: 39639239 PMCID: PMC11619307 DOI: 10.1186/s12891-024-08124-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent joint disorder characterized by degeneration and inflammation. Understanding its molecular mechanisms is crucial for diagnosis and treatment. METHODS We employed bioinformatics analyses to study OA using gene expression data. Differential expression analysis, weighted gene co-expression network analysis (WGCNA), and protein-protein interaction (PPI) network analysis were conducted. Enrichment analyses were performed to elucidate the biological significance of identified genes. Additionally, signature genes were identified using LASSO regression analysis, and a diagnostic nomogram was developed. qRT-PCR was conducted to confirm the expression levels of signature genes. RESULTS We identified 200 differentially expressed genes (DEGs) and a lightgreen module strongly correlated with OA. Within this module, 97 core genes were identified. Fifteen core lipopolysaccharide-related genes (LRGs) were found, enriched in immune and inflammatory pathways. Three hub genes (CCL3, ZFP36, and CCN1) emerged as potential biomarkers for OA diagnosis, with a nomogram showing high predictive accuracy, and validated by using clinical samples. Gene set enrichment analysis (GSEA) revealed distinct signaling pathways associated with the signature genes. Immunological analysis indicated altered immune profiles in OA, with the signature genes influencing immune cell infiltration and immune response pathways. CONCLUSION Our study provides insights into OA pathogenesis and identifies potential diagnostic biomarkers. The developed nomogram shows promise for accurate OA diagnosis. Furthermore, the signature genes play crucial roles in modulating the immune microenvironment in OA, suggesting their therapeutic potential. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Qipeng Chen
- Department of Orthopaedics and Traumatology III, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China
| | - Xiaodong Li
- Department of Orthopaedics, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China
| | - Pengfei Li
- Department of Orthopaedics and Traumatology I, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China
| | - Hongpeng Liu
- Department of Orthopaedics and Traumatology I, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China
| | - Qi Zhang
- Department of Orthopaedics and Traumatology III, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China
| | - Linqin He
- Department of Orthopaedics and Traumatology III, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China
| | - Zonghan Tang
- Department of Orthopaedics and Traumatology III, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China
| | - Hanbing Song
- Department of Orthopaedics and Traumatology III, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Zhou C, Sun C, Zhou W, Tian T, Schultz DC, Wu T, Yu M, Wu L, Pi L, Li C. Development of Novel Indole-Based Covalent Inhibitors of TEAD as Potential Antiliver Cancer Agents. J Med Chem 2024; 67:16270-16295. [PMID: 39270302 DOI: 10.1021/acs.jmedchem.4c00925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Abnormal activation of the YAP transcriptional signaling pathway drives proliferation in many hepatocellular carcinoma (HCC) and hepatoblastoma (HB) cases. Current treatment options often face resistance and toxicity, highlighting the need for alternative therapies. This article reports the discovery of a hit compound C-3 from docking-based virtual screening targeting TEAD lipid binding pocket, which inhibited TEAD-mediated transcription. Optimization led to the identification of a potent and covalent inhibitor CV-4-26 that exhibited great antitumor activity in HCC and HB cell lines in vitro, xenografted human HCC, and murine HB in vivo. These outcomes signify the potential of a highly promising therapeutic candidate for addressing a subset of HCC and HB cancers. In the cases of current treatment challenges due to high upregulation of YAP-TEAD activity, these findings offer a targeted alternative for more effective interventions against liver cancer.
Collapse
Affiliation(s)
- Chen Zhou
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Chunbao Sun
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, Louisiana 70112, United States
| | - Wei Zhou
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Tian Tian
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, Louisiana 70112, United States
| | - Daniel C Schultz
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, Louisiana 70112, United States
| | - Mu Yu
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida 32610, United States
- UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
| | - Lizi Wu
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida 32610, United States
- UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
- UF Institute of Genetics, University of Florida, Gainesville, Florida 32610, United States
| | - Liya Pi
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, Louisiana 70112, United States
| | - Chenglong Li
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
- Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
14
|
Zhou C, Sun C, Huang M, Tang X, Pi L, Li C. Exploring Degradation of Intrinsically Disordered Protein Yes-Associated Protein Induced by Proteolysis TArgeting Chimeras. J Med Chem 2024; 67:15168-15198. [PMID: 39189384 DOI: 10.1021/acs.jmedchem.4c00815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Yes-associated protein (YAP) is a key oncogene in the Hippo tumor suppression pathway, historically challenging to target due to its intrinsically disordered nature. Leveraging recent advances in high-throughput screening that identified several YAP binders, we employed proteolysis-targeting chimera technology to develop a series of YAP degraders. Utilizing NSC682769, a known YAP binder, linked with VHL ligand 2 or pomalidomide via diverse linkers, we synthesized degraders including YZ-6. This degrader not only recruits the E3 ligase VHL for the rapid and sustained degradation of YAP but also effectively inhibits its nuclear localization, curtailing YAP/TEAD-mediated transcription in cancer cell lines such as NCI-H226 and Huh7. This dual action significantly diminishes YAP's oncogenic activity, contributing to the potent antiproliferative effects observed both in vitro and in a Huh7 xenograft mouse model. These results underscore the potential of PROTAC-mediated YAP degradation as a strategy for treating YAP-driven cancers.
Collapse
Affiliation(s)
- Chen Zhou
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Chunbao Sun
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, Louisiana 70112, United States
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32610, United States
- UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32610, United States
- UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32610, United States
| | - Liya Pi
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, Louisiana 70112, United States
| | - Chenglong Li
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
15
|
Rana P, Ujjainiya R, Bharti V, Maiti S, Ekka MK. IGF2BP1-Mediated Regulation of CCN1 Expression by Specific Binding to G-Quadruplex Structure in Its 3'UTR. Biochemistry 2024; 63:2166-2182. [PMID: 39133064 DOI: 10.1021/acs.biochem.4c00172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The intricate regulation of gene expression is fundamental to the biological complexity of higher organisms, and is primarily governed by transcriptional and post-transcriptional mechanisms. The 3'-untranslated region (3'UTR) of mRNA is rich in cis-regulatory elements like G-quadruplexes (G4s), and plays a crucial role in post-transcriptional regulation. G4s have emerged as significant gene regulators, impacting mRNA stability, translation, and localization. In this study, we investigate the role of a robust parallel G4 structure situated within the 3'UTR of CCN1 mRNA in post-transcriptional regulation. This G4 structure is proximal to the stop codon of human CCN1, and evolutionarily conserved. We elucidated its interaction with the insulin-like growth factor 2 binding protein 1 (IGF2BP1), a noncanonical RNA N6-methyladenosine (m6A) modification reader, revealing a novel interplay between RNA modifications and G-quadruplex structures. Knockdown experiments and mutagenesis studies demonstrate that IGF2BP1 binds specifically to the G4 structure, modulating CCN1 mRNA stability. Additionally, we unveil the role of IGF2BP1's RNA recognition motifs in G4 recognition, highlighting this enthalpically driven interaction. Our findings offer fresh perspectives on the complex mechanisms of post-transcriptional gene regulation mediated by G4 RNA secondary structures.
Collapse
Affiliation(s)
- Priya Rana
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rajat Ujjainiya
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vishal Bharti
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
| | - Souvik Maiti
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mary K Ekka
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
16
|
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
|
17
|
Xu R, Jiang Z, Meng X, Xing L, Aladan W, Chi B, Dang T, Chai J. Cellular communication network 1 promotes CASP2 mRNA expression but suppresses its protein translation in esophageal adenocarcinoma. J Cell Commun Signal 2024; 18:e12046. [PMID: 39524140 PMCID: PMC11544643 DOI: 10.1002/ccs3.12046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/08/2024] [Accepted: 07/02/2024] [Indexed: 11/16/2024] Open
Abstract
Induction of apoptosis in tumor cells is one of the best ways to cure cancer. While most apoptosis requires a chain of caspase activation, CASP2 can do this all by itself. The matricellular protein cellular communication network 1 (CCN1) is known for supporting some cancer growth but suppressing others. Esophageal adenocarcinoma (EAC) belongs to the latter. CCN1 is capable of inducing TRAIL-mediated apoptosis in EAC cells. This study found that CCN1 upregulated CASP2 transcription but not its translation in EAC cells because, on one hand, CCN1 downregulated p16 and p21, which increased RB1 phosphorylation allowing E2F1 to transcribe more CASP2 mRNA, on the other hand, CCN1 also upregulated HuR, which is bound to CASP2 mRNA species and blocked its protein translation. As a result, CASP2 contributed nothing to CCN1-induced EAC cell apoptosis. On the contrary, CCN1 promoted CASP3, not only in its transcription but also in its translation and activation, which established the basis for CCN1-induced EAC cell apoptosis.
Collapse
Affiliation(s)
- Ruize Xu
- Inner Mongolia Institute of Digestive DiseasesInner Mongolia Engineering Research Center for Prevention and Treatment of Digestive DiseasesThe Second Affiliated Hospital of Baotou Medical CollegeInner Mongolia University of Science and TechnologyBaotouChina
| | - Zhenyu Jiang
- Inner Mongolia Institute of Digestive DiseasesInner Mongolia Engineering Research Center for Prevention and Treatment of Digestive DiseasesThe Second Affiliated Hospital of Baotou Medical CollegeInner Mongolia University of Science and TechnologyBaotouChina
| | - Xianmei Meng
- Inner Mongolia Institute of Digestive DiseasesInner Mongolia Engineering Research Center for Prevention and Treatment of Digestive DiseasesThe Second Affiliated Hospital of Baotou Medical CollegeInner Mongolia University of Science and TechnologyBaotouChina
| | - Lingling Xing
- Inner Mongolia Institute of Digestive DiseasesInner Mongolia Engineering Research Center for Prevention and Treatment of Digestive DiseasesThe Second Affiliated Hospital of Baotou Medical CollegeInner Mongolia University of Science and TechnologyBaotouChina
| | - Wula Aladan
- Inner Mongolia Institute of Digestive DiseasesInner Mongolia Engineering Research Center for Prevention and Treatment of Digestive DiseasesThe Second Affiliated Hospital of Baotou Medical CollegeInner Mongolia University of Science and TechnologyBaotouChina
| | - Baoxing Chi
- Inner Mongolia Institute of Digestive DiseasesInner Mongolia Engineering Research Center for Prevention and Treatment of Digestive DiseasesThe Second Affiliated Hospital of Baotou Medical CollegeInner Mongolia University of Science and TechnologyBaotouChina
| | - Tong Dang
- Inner Mongolia Institute of Digestive DiseasesInner Mongolia Engineering Research Center for Prevention and Treatment of Digestive DiseasesThe Second Affiliated Hospital of Baotou Medical CollegeInner Mongolia University of Science and TechnologyBaotouChina
| | - Jianyuan Chai
- Inner Mongolia Institute of Digestive DiseasesInner Mongolia Engineering Research Center for Prevention and Treatment of Digestive DiseasesThe Second Affiliated Hospital of Baotou Medical CollegeInner Mongolia University of Science and TechnologyBaotouChina
| |
Collapse
|
18
|
Gündel B, Liu X, Pfützenreuter A, Engelsberger V, Weiskirchen R, Löhr JM, Heuchel R. The Crosstalk Analysis between mPSCs and Panc1 Cells Identifies CCN1 as a Positive Regulator of Gemcitabine Sensitivity in Pancreatic Cancer Cells. Int J Mol Sci 2024; 25:9369. [PMID: 39273316 PMCID: PMC11394772 DOI: 10.3390/ijms25179369] [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: 06/26/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease that is almost entirely resistant to conventional chemotherapy and radiation therapy. A significant factor in this resistance appears to be the dense desmoplastic stroma, which contains various cancer-associated fibroblast (CAF) populations. However, our understanding of the communication between tumor cells and CAFs that contributes to this aggressive malignancy is still developing. Recently, we used an advanced three-dimensional heterospecies, heterospheroid co-culture model to investigate the signaling between human pancreatic tumor Panc1 cells and mouse pancreatic stellate cells (mPSCs) through global expression profiling. Upon discovering that CCN1 was significantly upregulated in Panc1 cells during co-culture, we decided to explore the role of CCN1 using CRISPR-Cas9 knockout technology. Panc1 cells lacking CCN1 showed reduced differentiation and decreased sensitivity to gemcitabine, primarily due to lower expression of genes involved in gemcitabine transport and metabolism. Additionally, we observed that stimulation with TGF-β1 and lysophosphatidic acid increased CCN1 expression in Panc1 cells and induced a shift in mPSCs towards a more myofibroblastic CAF-like phenotype.
Collapse
Affiliation(s)
- Beate Gündel
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE 141 86 Huddinge, Sweden; (B.G.); (J.-M.L.)
| | - Xinyuan Liu
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE 141 86 Huddinge, Sweden; (B.G.); (J.-M.L.)
| | - Anna Pfützenreuter
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE 141 86 Huddinge, Sweden; (B.G.); (J.-M.L.)
| | - Veronika Engelsberger
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE 141 86 Huddinge, Sweden; (B.G.); (J.-M.L.)
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| | - J.-Matthias Löhr
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE 141 86 Huddinge, Sweden; (B.G.); (J.-M.L.)
| | - Rainer Heuchel
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE 141 86 Huddinge, Sweden; (B.G.); (J.-M.L.)
| |
Collapse
|
19
|
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
|
20
|
Fujita M, Sasada M, Iyoda T, Fukai F. Involvement of Matricellular Proteins in Cellular Senescence: Potential Therapeutic Targets for Age-Related Diseases. Int J Mol Sci 2024; 25:6591. [PMID: 38928297 PMCID: PMC11204155 DOI: 10.3390/ijms25126591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Senescence is a physiological and pathological cellular program triggered by various types of cellular stress. Senescent cells exhibit multiple characteristic changes. Among them, the characteristic flattened and enlarged morphology exhibited in senescent cells is observed regardless of the stimuli causing the senescence. Several studies have provided important insights into pro-adhesive properties of cellular senescence, suggesting that cell adhesion to the extracellular matrix (ECM), which is involved in characteristic morphological changes, may play pivotal roles in cellular senescence. Matricellular proteins, a group of structurally unrelated ECM molecules that are secreted into the extracellular environment, have the unique ability to control cell adhesion to the ECM by binding to cell adhesion receptors, including integrins. Recent reports have certified that matricellular proteins are closely involved in cellular senescence. Through this biological function, matricellular proteins are thought to play important roles in the pathogenesis of age-related diseases, including fibrosis, osteoarthritis, intervertebral disc degeneration, atherosclerosis, and cancer. This review outlines recent studies on the role of matricellular proteins in inducing cellular senescence. We highlight the role of integrin-mediated signaling in inducing cellular senescence and provide new therapeutic options for age-related diseases targeting matricellular proteins and integrins.
Collapse
Affiliation(s)
- Motomichi Fujita
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Chiba, Japan
| | - Manabu Sasada
- Clinical Research Center in Hiroshima, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8551, Japan
| | - Takuya Iyoda
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-Doori, Sanyo-Onoda 756-0884, Yamaguchi, Japan
| | - Fumio Fukai
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Chiba, Japan
| |
Collapse
|
21
|
Hu K, Xie L, Wu W, Zhang J, Li Y, He J, Zhang Y, Lu D, Koeffler HP, Lin L, Yin D. CYR61 Acts as an Intracellular Microtubule-Associated Protein and Coordinates Mitotic Progression via PLK1-FBW7 Pathway. Int J Biol Sci 2024; 20:3140-3155. [PMID: 38904029 PMCID: PMC11186368 DOI: 10.7150/ijbs.93335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Cysteine-rich angiogenic inducer 61 (CYR61), also called CCN1, has long been characterized as a secretory protein. Nevertheless, the intracellular function of CYR61 remains unclear. Here, we found that CYR61 is important for proper cell cycle progression. Specifically, CYR61 interacts with microtubules and promotes microtubule polymerization to ensure mitotic entry. Moreover, CYR61 interacts with PLK1 and accumulates during the mitotic process, followed by degradation as mitosis concludes. The proteolysis of CYR61 requires the PLK1 kinase activity, which directly phosphorylates two conserved motifs on CYR61, enhancing its interaction with the SCF E3 complex subunit FBW7 and mediating its degradation by the proteasome. Mutations of phosphorylation sites of Ser167 and Ser188 greatly increase CYR61's stability, while deletion of CYR61 extends prophase and metaphase and delays anaphase onset. In summary, our findings highlight the precise control of the intracellular CYR61 by the PLK1-FBW7 pathway, accentuating its significance as a microtubule-associated protein during mitotic progression.
Collapse
Affiliation(s)
- Kaishun Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Limin Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Wenjing Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
- Department of Breast Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Jingyuan Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Yu Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen 518000, P.R. China
| | - Jiehua He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Yin Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Daning Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - H. Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Lehang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| |
Collapse
|
22
|
Kim JY, Quan T. Emerging Perspectives of YAP/TAZ in Human Skin Epidermal and Dermal Aging. Ann Dermatol 2024; 36:135-144. [PMID: 38816974 PMCID: PMC11148314 DOI: 10.5021/ad.23.156] [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: 12/11/2023] [Revised: 01/25/2024] [Accepted: 02/18/2024] [Indexed: 06/01/2024] Open
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are key downstream effectors of the Hippo signaling pathway, which plays a central role in tissue homeostasis, organ development, and regeneration. While the dysregulation of YAP/TAZ has been linked to various human diseases, their involvement in the aging of human skin has only recently begun to manifest. In the skin, the YAP/TAZ effectors emerge as central regulators in maintaining homeostasis of epidermal stem cells and dermal extracellular matrix, and thus intimately linked to skin aging processes. This review underscores recent molecular breakthroughs highlighting how age-related decline of YAP/TAZ activity impacts human epidermal and dermal aging. Gaining insight into the evolving roles of YAP/TAZ in human skin aging presents a promising avenue for the development of innovative therapeutic approaches aimed at enhancing skin health and addressing age-related skin conditions.
Collapse
Affiliation(s)
- Jun Young Kim
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Dermatology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
23
|
Lang A, Eastburn EA, Younesi M, Nijsure M, Siciliano C, Haran AP, Panebianco CJ, Seidl E, Tang R, Alsberg E, Willett NJ, Gottardi R, Huh D, Boerckel JD. Cyr61 delivery promotes angiogenesis during bone fracture repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588239. [PMID: 38617208 PMCID: PMC11014620 DOI: 10.1101/2024.04.05.588239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Compromised vascular supply and insufficient neovascularization impede bone repair, increasing risk of non-union. Cyr61, Cysteine-rich angiogenic inducer of 61kD (also known as CCN1), is a matricellular growth factor that is regulated by mechanical cues during fracture repair. Here, we map the distribution of endogenous Cyr61 during bone repair and evaluate the effects of recombinant Cyr61 delivery on vascularized bone regeneration. In vitro, Cyr61 treatment did not alter chondrogenesis or osteogenic gene expression, but significantly enhanced angiogenesis. In a mouse femoral fracture model, Cyr61 delivery did not alter cartilage or bone formation, but accelerated neovascularization during fracture repair. Early initiation of ambulatory mechanical loading disrupted Cyr61-induced neovascularization. Together, these data indicate that Cyr61 delivery can enhance angiogenesis during bone repair, particularly for fractures with stable fixation, and may have therapeutic potential for fractures with limited blood vessel supply.
Collapse
Affiliation(s)
- Annemarie Lang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Emily A. Eastburn
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Mousa Younesi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Madhura Nijsure
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Carly Siciliano
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Annapurna Pranatharthi Haran
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Elizabeth Seidl
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Rui Tang
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Eben Alsberg
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Nick J. Willett
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States
- The Veterans Affairs Portland Health Care System, Portland, OR, United States
| | - Riccardo Gottardi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Joel D. Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
24
|
McCallum S, Suresh KB, Islam T, Saustad AW, Shelest O, Patil A, Lee D, Kwon B, Yenokian I, Kawaguchi R, Beveridge CH, Manchandra P, Randolph CE, Meares GP, Dutta R, Plummer J, Knott SRV, Chopra G, Burda JE. Lesion-remote astrocytes govern microglia-mediated white matter repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585251. [PMID: 38558977 PMCID: PMC10979953 DOI: 10.1101/2024.03.15.585251] [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
Spared regions of the damaged central nervous system undergo dynamic remodeling and exhibit a remarkable potential for therapeutic exploitation. Here, lesion-remote astrocytes (LRAs), which interact with viable neurons, glia and neural circuitry, undergo reactive transformations whose molecular and functional properties are poorly understood. Using multiple transcriptional profiling methods, we interrogated LRAs from spared regions of mouse spinal cord following traumatic spinal cord injury (SCI). We show that LRAs acquire a spectrum of molecularly distinct, neuroanatomically restricted reactivity states that evolve after SCI. We identify transcriptionally unique reactive LRAs in degenerating white matter that direct the specification and function of local microglia that clear lipid-rich myelin debris to promote tissue repair. Fueling this LRA functional adaptation is Ccn1 , which encodes for a secreted matricellular protein. Loss of astrocyte CCN1 leads to excessive, aberrant activation of local microglia with (i) abnormal molecular specification, (ii) dysfunctional myelin debris processing, and (iii) impaired lipid metabolism, culminating in blunted debris clearance and attenuated neurological recovery from SCI. Ccn1 -expressing white matter astrocytes are specifically induced by local myelin damage and generated in diverse demyelinating disorders in mouse and human, pointing to their fundamental, evolutionarily conserved role in white matter repair. Our findings show that LRAs assume regionally divergent reactivity states with functional adaptations that are induced by local context-specific triggers and influence disorder outcome. Astrocytes tile the central nervous system (CNS) where they serve vital roles that uphold healthy nervous system function, including regulation of synapse development, buffering of neurotransmitters and ions, and provision of metabolic substrates 1 . In response to diverse CNS insults, astrocytes exhibit disorder-context specific transformations that are collectively referred to as reactivity 2-5 . The characteristics of regionally and molecularly distinct reactivity states are incompletely understood. The mechanisms through which distinct reactivity states arise, how they evolve or resolve over time, and their consequences for local cell function and CNS disorder progression remain enigmatic. Immediately adjacent to CNS lesions, border-forming astrocytes (BFAs) undergo transcriptional reprogramming and proliferation to form a neuroprotective barrier that restricts inflammation and supports axon regeneration 6-9 . Beyond the lesion, spared but dynamic regions of the injured CNS exhibit varying degrees of synaptic circuit remodeling and progressive cellular responses to secondary damage that have profound consequences for neural repair and recovery 10,11 . Throughout these cytoarchitecturally intact, but injury-reactive regions, lesion-remote astrocytes (LRAs) intermingle with neurons and glia, undergo little to no proliferation, and exhibit varying degrees of cellular hypertrophy 7,12,13 . The molecular and functional properties of LRAs remain grossly undefined. Therapeutically harnessing spared regions of the injured CNS will require a clearer understanding of the accompanying cellular and molecular landscape. Here, we leveraged integrative transcriptional profiling methodologies to identify multiple spatiotemporally resolved, molecularly distinct states of LRA reactivity within the injured spinal cord. Computational modeling of LRA-mediated heterotypic cell interactions, astrocyte-specific conditional gene deletion, and multiple mouse models of acute and chronic CNS white matter degeneration were used to interrogate a newly identified white matter degeneration-reactive astrocyte subtype. We define how this reactivity state is induced and its role in governing the molecular and functional specification of local microglia that clear myelin debris from the degenerating white matter to promote repair.
Collapse
|
25
|
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
|
26
|
Hutchenreuther J, Nguyen J, Quesnel K, Vincent KM, Petitjean L, Bourgeois S, Boyd M, Bou-Gharios G, Postovit LM, Leask A. Cancer-associated Fibroblast-specific Expression of the Matricellular Protein CCN1 Coordinates Neovascularization and Stroma Deposition in Melanoma Metastasis. CANCER RESEARCH COMMUNICATIONS 2024; 4:556-570. [PMID: 38363129 PMCID: PMC10898341 DOI: 10.1158/2767-9764.crc-23-0571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/19/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Melanoma is the leading cause of skin cancer-related death. As prognosis of patients with melanoma remains problematic, identification of new therapeutic targets remains essential. Matricellular proteins are nonstructural extracellular matrix proteins. They are secreted into the tumor microenvironment to coordinate behavior among different cell types, yet their contribution to melanoma is underinvestigated. Examples of matricellular proteins include those comprising the CCN family. The CCN family member, CCN1, is highly proangiogenic. Herein, we show that, in human patients with melanoma, although found in several tumor cell types, CCN1 is highly expressed by a subset of cancer-associated fibroblasts (CAF) in patients with melanoma and this expression correlates positively with expression of proangiogenic genes and progressive disease/resistance to anti-PD1 checkpoint inhibitors. Consistent with these observations, in a syngeneic C57BL6 mouse model of melanoma, loss of CCN1 expression from Col1A2-Cre-, herein identified as "universal," fibroblasts, impaired metastasis of subcutaneously injected B16F10 tumor cells to lung, concomitant with disrupted neovascularization and collagen organization. Disruption of the extracellular matrix in the loss of CCN1 was validated using a novel artificial intelligence-based image analysis platform that revealed significantly decreased phenotypic fibrosis and composite morphometric collagen scores. As drug resistance is linked to matrix deposition and neoangiogenesis, these data suggest that CCN1, due to its multifaceted role, may represent a novel therapeutic target for drug-resistant melanoma. Our data further emphasize the essential role that cancer-associated, (universal) Col1A2-Cre-fibroblasts and extracellular matrix remodeling play in coordinating behavior among different cell types within the tumor microenvironment. SIGNIFICANCE In human patients, the expression of proangiogenic matricellular protein CCN1 in CAFs correlates positively with expression of stroma and angiogenic markers and progressive disease/resistance to checkpoint inhibitor therapy. In an animal model, loss of CCN1 from CAFs impaired metastasis of melanoma cells, neovascularization, and collagen deposition, emphasizing that CAFs coordinate cellular behavior in a tumor microenvironment and that CCN1 may be a novel target.
Collapse
Affiliation(s)
- James Hutchenreuther
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - John Nguyen
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Katherine Quesnel
- Department of Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Krista M. Vincent
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
- Department of Medical Genetics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | | | - Sophia Bourgeois
- Department of Biology, University of Western Ontario, London, Ontario, Canada
| | - Mark Boyd
- Office of the Vice President of Research, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - George Bou-Gharios
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Lynne-Marie Postovit
- Department of Biomedical and Molecular Sciences, Queens University, Kingston, Ontario, Canada
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
27
|
Liu S, Jia X, Liu B, Liu Y, Yin H. Suppression of cerebral ischemia injury induced blood brain barrier breakdown by dexmedetomidine via promoting CCN1. Aging (Albany NY) 2024; 16:3750-3762. [PMID: 38364236 PMCID: PMC10929797 DOI: 10.18632/aging.205557] [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: 10/12/2023] [Accepted: 01/03/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND Blood-brain barrier (BBB) could aggravate cerebral ischemia injury. Dexmedetomidine (Dex) has been believed to play a protective role in cerebral ischemia injury-induced BBB injury. METHODS Middle cerebral artery occlusion (MCAO) and oxygen-glucose deprivation (OGD) models were established to simulate cerebral ischemia injury. Animal experiments included 4 groups, Sham, MCAO, MCAO+Dex, MCAO+Dex+sh-CCN1. Generally applicable gene set enrichment analysis was performed to analyze gene expression difference. Total collagen content and Evans blue staining were performed to measure infarct ratio and BBB breakdown, respectively. The cell apoptosis, mRNA and protein expression were measured through flow cytometry, PCR, and western blotting, respectively. The levels of IL-1β, TNF-α, and IL-6 in serum were measured with commercial ELISA kits. RESULTS Dex greatly promoted the expression level of CCN1. Dex suppressed cerebral ischemia injury, increased tight junction protein expression, improved the memory ability and neurological function of MCAO rats through targeting CCN1. The significant increase of inflammatory factors in the serum of MCAO rats were suppressed by Dex. Dex suppressed OGD induced increase of HRP permeability and promoting tight junction protein expression in vitro through regulating CCN1. The neurological function evaluation was performed with Neurological Severity Score (NSS) and Longa Score Scale. CONCLUSIONS Dex could remarkably alleviate cerebral ischemia injury by inhibiting BBB breakdown, inflammatory response, and promoting neurological function and tight junction protein expression via up-regulating CCN1. This study might provide a novel therapeutic target for the prevention and treatment of cerebral ischemia injury-induced BBB.
Collapse
Affiliation(s)
- Shuangmei Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Xuepeng Jia
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Bo Liu
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
- Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Shenyang 110004, Liaoning, China
| | - Yue Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Hong Yin
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| |
Collapse
|
28
|
Li X, Xie Z, Zhou Q, Tan X, Meng W, Pang Y, Huang L, Ding Z, Hu Y, Li R, Huang G, Li H. TGN-020 Alleviate Inflammation and Apoptosis After Cerebral Ischemia-Reperfusion Injury in Mice Through Glymphatic and ERK1/2 Signaling Pathway. Mol Neurobiol 2024; 61:1175-1186. [PMID: 37695472 PMCID: PMC10861636 DOI: 10.1007/s12035-023-03636-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/30/2023] [Indexed: 09/12/2023]
Abstract
Post-stroke acute inhibition of aquaporin 4 (AQP4) is known to exacerbate inflammation and apoptosis, yet the underlying mechanisms are not fully understood. The objective of this study was to investigate the specific mechanism of inflammation and apoptosis following cerebral ischemia-reperfusion (I/R) injury using the AQP4-specific inhibitor, N-(1,3,4-thiadiazol-2-yl) pyridine-3-carboxamide dihydrochloride (TGN-020). Ischemic stroke was induced in mice using the middle cerebral artery occlusion (MCAO) model. The C57/BL6 mice were randomly divided into three groups as follows: sham operation, I/R 48 h, and TGN-020 + I/R 48 h treatment. All mice were subjected to a series of procedures. These procedures encompassed 2,3,5-triphenyltetrazolium chloride (TTC) staining, neurological scoring, fluorescence tracing, western blotting, immunofluorescence staining, and RNA sequencing (RNA-seq). The glymphatic function in the cortex surrounding cerebral infarction was determined using tracer, glial fibrillary acid protein (GFAP), AQP4 co-staining, and beta-amyloid precursor protein (APP) staining; differential genes were detected using RNA-seq. The influence of TGN-020 on the extracellular signal-regulated kinase 1/2 (ERK) 1/2 pathway was confirmed using the ERK1/2 pathway agonists Ro 67-7467. Additionally, we examined the expression of inflammation associated with microglia and astrocytes after TGN-020 and Ro 67-7467 treatment. Compared with I/R group, TGN-020 alleviated glymphatic dysfunction by inhibiting astrocyte proliferation and reducing tracer accumulation in the peri-infarct area. RNA-seq showed that the differentially expressed genes were mainly involved in the activation of astrocytes and microglia and in the ERK1/2 pathway. Western blot and immunofluorescence further verified the expression of associated inflammation. The inflammation and cell apoptosis induced by I/R are mitigated by TGN-020. This mitigation occurs through the improvement of glymphatic function and the inhibition of the ERK1/2 pathway.
Collapse
Affiliation(s)
- Xiaohong Li
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Zhuoxi Xie
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Qian Zhou
- Department of Neurology, the Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
| | - Xiaoli Tan
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Weiting Meng
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Yeyu Pang
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Lizhen Huang
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Zhihao Ding
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Yuanhong Hu
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Ruhua Li
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Guilan Huang
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Hao Li
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| |
Collapse
|
29
|
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
|
30
|
Quan T. Human Skin Aging and the Anti-Aging Properties of Retinol. Biomolecules 2023; 13:1614. [PMID: 38002296 PMCID: PMC10669284 DOI: 10.3390/biom13111614] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
The skin is the most-extensive and -abundant tissue in the human body. Like many organs, as we age, human skin experiences gradual atrophy in both the epidermis and dermis. This can be primarily attributed to the diminishing population of epidermal stem cells and the reduction in collagen, which is the primary structural protein in the human body. The alterations occurring in the epidermis and dermis due to the aging process result in disruptions to the structure and functionality of the skin. This creates a microenvironment conducive to age-related skin conditions such as a compromised skin barrier, slowed wound healing, and the onset of skin cancer. This review emphasizes the recent molecular discoveries related to skin aging and evaluates preventive approaches, such as the use of topical retinoids. Topical retinoids have demonstrated promise in enhancing skin texture, diminishing fine lines, and augmenting the thickness of both the epidermal and dermal layers.
Collapse
Affiliation(s)
- Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
31
|
Bauerschmitz G, Hüchel S, Gallwas J, Gründker C. Inhibition of Increased Invasiveness of Breast Cancer Cells With Acquired Tamoxifen Resistance by Suppression of CYR61. Cancer Genomics Proteomics 2023; 20:531-538. [PMID: 37889058 PMCID: PMC10614060 DOI: 10.21873/cgp.20403] [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: 07/20/2023] [Revised: 08/23/2023] [Accepted: 09/01/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND/AIM Hormone sensitivity-targeted therapy with selective estrogen receptor modulators (SERMs), such as 4-hydroxytamoxifen (4-OHT), is the mainstay of treatment for breast cancers (BCs) that express estrogen receptor α (ERα). However, development of resistance limits this therapy approach. The question arises whether changes associated with 4-OHT resistance could be exploited therapeutically. MATERIALS AND METHODS First, 4-OHT-resistant sublines of ERα-positive breast carcinoma cell lines MCF-7 and T47D were generated. Viability was assessed by the Alamar Blue assay. Cell invasion was quantified in modified Boyden chambers with Matrigel. Changes in expression of CYR61, S100A4, and ERα were examined by RT-qPCR. Expression of CYR61 was suppressed by transient gene silencing using siRNA. Successful suppression was verified by western blot. Efficacy of 4-OHT treatment was analyzed by quantification of viability using Alamar Blue assay. Correlation of CYR61 levels in patients with luminal A BC to distant metastases-free survival was determined by Kaplan-Meier analysis. RESULTS ERα-positive MCF-7 and T47D BC cells exhibit an extremely weak invasion rate. Acquired tamoxifen resistance significantly increased the invasive behavior of both tamoxifen-resistant MCF-7-TR and T47D-TR sublines. In addition, expression of CYR61 and S100A4 showed significantly increased levels, whereas expression of ERα was decreased. Suppression of CYR61 expression resulted in a significant decreased invasion rate. In addition, expression of S100A4 was reduced, whereas expression of ERα was increased. Furthermore, suppression of CYR61 resulted in re-sensitization to 4-OHT. High CYR61 levels in patients with luminal A BC resulted in reduced distant metastases-free survival. CONCLUSION The prometastatic factor CYR61 appears to play an important role in the increased invasiveness of tamoxifen-resistant ERα-positive BC cells. Its suppression leads to a lower invasion rate. Given the few therapeutic options available for tamoxifen-resistant BC, therapy that reduces CYR61 may improve its treatability in future.
Collapse
Affiliation(s)
- Gerd Bauerschmitz
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Silke Hüchel
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Julia Gallwas
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Carsten Gründker
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
32
|
Israel LL, Braubach O, Shatalova ES, Chepurna O, Sharma S, Klymyshyn D, Galstyan A, Chiechi A, Cox A, Herman D, Bliss B, Hasen I, Ting A, Arechavala R, Kleinman MT, Patil R, Holler E, Ljubimova JY, Koronyo-Hamaoui M, Sun T, Black KL. Exposure to environmental airborne particulate matter caused wide-ranged transcriptional changes and accelerated Alzheimer's-related pathology: A mouse study. Neurobiol Dis 2023; 187:106307. [PMID: 37739136 DOI: 10.1016/j.nbd.2023.106307] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/04/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023] Open
Abstract
Air pollution poses a significant threat to human health, though a clear understanding of its mechanism remains elusive. In this study, we sought to better understand the effects of various sized particulate matter from polluted air on Alzheimer's disease (AD) development using an AD mouse model. We exposed transgenic Alzheimer's mice in their prodromic stage to different sized particulate matter (PM), with filtered clean air as control. After 3 or 6 months of exposure, mouse brains were harvested and analyzed. RNA-seq analysis showed that various PM have differential effects on the brain transcriptome, and these effects seemed to correlate with PM size. Many genes and pathways were affected after PM exposure. Among them, we found a strong activation in mRNA Nonsense Mediated Decay pathway, an inhibition in pathways related to transcription, neurogenesis and survival signaling as well as angiogenesis, and a dramatic downregulation of collagens. Although we did not detect any extracellular Aβ plaques, immunostaining revealed that both intracellular Aβ1-42 and phospho-Tau levels were increased in various PM exposure conditions compared to the clean air control. NanoString GeoMx analysis demonstrated a remarkable activation of immune responses in the PM exposed mouse brain. Surprisingly, our data also indicated a strong activation of various tumor suppressors including RB1, CDKN1A/p21 and CDKN2A/p16. Collectively, our data demonstrated that exposure to airborne PM caused a profound transcriptional dysregulation and accelerated Alzheimer's-related pathology.
Collapse
Affiliation(s)
- Liron L Israel
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Oliver Braubach
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Ekaterina S Shatalova
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Oksana Chepurna
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Sachin Sharma
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Dmytro Klymyshyn
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Anna Galstyan
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Antonella Chiechi
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Alysia Cox
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - David Herman
- Department of Environmental and Occupational Health, University of California, Irvine 92697, United States of America
| | - Bishop Bliss
- Department of Environmental and Occupational Health, University of California, Irvine 92697, United States of America
| | - Irene Hasen
- Department of Environmental and Occupational Health, University of California, Irvine 92697, United States of America
| | - Amanda Ting
- Department of Environmental and Occupational Health, University of California, Irvine 92697, United States of America
| | - Rebecca Arechavala
- Department of Environmental and Occupational Health, University of California, Irvine 92697, United States of America
| | - Michael T Kleinman
- Department of Environmental and Occupational Health, University of California, Irvine 92697, United States of America
| | - Rameshwar Patil
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Eggehard Holler
- Terasaki Institute, Los Angeles, CA 90024, United States of America
| | | | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Tao Sun
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America.
| | - Keith L Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America.
| |
Collapse
|
33
|
Fisher GJ, Wang B, Cui Y, Shi M, Zhao Y, Quan T, Voorhees JJ. Skin aging from the perspective of dermal fibroblasts: the interplay between the adaptation to the extracellular matrix microenvironment and cell autonomous processes. J Cell Commun Signal 2023; 17:523-529. [PMID: 37067763 PMCID: PMC10409944 DOI: 10.1007/s12079-023-00743-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 03/10/2023] [Indexed: 04/18/2023] Open
Abstract
This article summarizes important molecular mechanisms that drive aging in human skin from the perspective of dermal fibroblasts. The dermis comprises the bulk of the skin and is largely composed of a collagen-rich extracellular matrix (ECM). The dermal ECM provides mechanical strength, resiliency, and an environment that supports the functions of ibroblasts and other types of dermal cells. Fibroblasts produce the dermal ECM and maintain its homeostasis. Fibroblasts attach to the ECM and this attachment controls their morphology and function. During aging, the ECM undergoes gradual degradation that is nitiated by matrix metalloproteinases (MMPs). This degradation alters mechanical forces within the dermal ECM and disrupts he interactions between fibroblasts and the ECM thereby generating an aged fibroblast phenotype. This aged fibroblast phenotype is characterized by collapsed morphology, altered mechanosignaling, induction of CCN1, and activation of transcription factor AP-1, with consequent upregulation of target genes including MMPs and pro-inflammatory mediators. The TGF-beta pathway coordinately regulates ECM production and turnover. Altered mechanical forces, due to ECM fragmentation, down-regulate the type II TGF-beta receptor, thereby reducing ECM production and further increasing ECM breakdown. Thus, dermal aging involves a feed-forward process that reinforces the aged dermal fibroblast phenotype and promotes age-related dermal ECM deterioration. As discussed in the article, the expression of the aged dermal fibroblast phenotype involves both adaptive and cell-autonomous mechanisms.
Collapse
Affiliation(s)
- Gary J Fisher
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA.
- Department of Dermatology, Tsinghua Changgung Hospital, Tsinghua University, Beijing, China.
| | - Bo Wang
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Yilei Cui
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Mai Shi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Yi Zhao
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Taihao Quan
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - John J Voorhees
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| |
Collapse
|
34
|
Alkon N, Assen FP, Arnoldner T, Bauer WM, Medjimorec MA, Shaw LE, Rindler K, Holzer G, Weber P, Weninger W, Freystätter C, Chennareddy S, Kinaciyan T, Farlik M, Jonak C, Griss J, Bangert C, Brunner PM. Single-cell RNA sequencing defines disease-specific differences between chronic nodular prurigo and atopic dermatitis. J Allergy Clin Immunol 2023; 152:420-435. [PMID: 37210042 DOI: 10.1016/j.jaci.2023.04.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/12/2023] [Accepted: 04/19/2023] [Indexed: 05/22/2023]
Abstract
BACKGROUND Chronic nodular prurigo (CNPG) is an inflammatory skin disease that is maintained by a chronic itch-scratch cycle likely rooted in neuroimmunological dysregulation. This condition may be associated with atopy in some patients, and there are now promising therapeutic results from blocking type 2 cytokines such as IL-4, IL-13, and IL-31. OBJECTIVES This study aimed to improve the understanding of pathomechanisms underlying CNPG as well as molecular relationships between CNPG and atopic dermatitis (AD). METHODS We profiled skin lesions from patients with CNPG in comparison with AD and healthy control individuals using single-cell RNA sequencing combined with T-cell receptor sequencing. RESULTS We found type 2 immune skewing in both CNPG and AD, as evidenced by CD4+ helper T cells expressing IL13. However, only AD harbored an additional, oligoclonally expanded CD8A+IL9R+IL13+ cytotoxic T-cell population, and immune activation pathways were highly upregulated in AD, but less so in CNPG. Conversely, CNPG showed signatures of extracellular matrix organization, collagen synthesis, and fibrosis, including a unique population of CXCL14-IL24+ secretory papillary fibroblasts. Besides known itch mediators such as IL31 and oncostatin M, we also detected increased levels of neuromedin B in fibroblasts of CNPG lesions compared with AD and HC, with neuromedin B receptors detectable on some nerve endings. CONCLUSIONS These data show that CNPG does not harbor the strong disease-specific immune activation pathways that are typically found in AD but is rather characterized by upregulated stromal remodeling mechanisms that might have a direct impact on itch fibers.
Collapse
Affiliation(s)
- Natalia Alkon
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Frank P Assen
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Tamara Arnoldner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang M Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Marco A Medjimorec
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Lisa E Shaw
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Katharina Rindler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Gregor Holzer
- Department of Dermatology, Klinik Donaustadt, Vienna, Austria
| | - Philipp Weber
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Christian Freystätter
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Sumanth Chennareddy
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Tamar Kinaciyan
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Constanze Jonak
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Johannes Griss
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Christine Bangert
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Patrick M Brunner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
35
|
Kozlova NI, Morozevich GE, Gevorkian NM, Kurbatov LK, Berman AE. Implication of integrin α5β1 in senescence of SK-Mel-147 human melanoma cells. BIOMEDITSINSKAIA KHIMIIA 2023; 69:156-164. [PMID: 37384907 DOI: 10.18097/pbmc20236903156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Downregulation of α5β1 integrin in the SK-Mel-147 human melanoma culture model sharply inhibits the phenotypic manifestations of tumor progression: cell proliferation and clonal activity. This was accompanied by a 2-3-fold increase in the content of SA-β-Gal positive cells thus indicating an increase in the cellular senescence phenotype. These changes were accompanied by a significant increase in the activity of p53 and p21 tumor suppressors and components of the PI3K/Akt/mTOR/p70 signaling pathway. Pharmacological inhibition of mTORC1 reduced the content of SA-β-Gal positive cells in the population of α5β1-deficient SK-Mel-147 cells. A similar effect was observed with pharmacological and genetic inhibition of the activity of Akt1, one of the three Akt protein kinase isoenzymes; suppression of other Akt isozymes did not affect melanoma cell senescence. The results presented in this work and previously obtained indicate that α5β1 shares with other integrins of the β1 family the function of cell protection from senescence. This function is realized via regulation of the PI3K/Akt1/mTOR signaling pathway, in which Akt1 exhibits a non-canonical activity.
Collapse
Affiliation(s)
- N I Kozlova
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | - L K Kurbatov
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A E Berman
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
36
|
Pervaiz N, Kathuria I, Aithabathula RV, Singla B. Matricellular proteins in atherosclerosis development. Matrix Biol 2023; 120:1-23. [PMID: 37086928 PMCID: PMC10225360 DOI: 10.1016/j.matbio.2023.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/24/2023]
Abstract
The extracellular matrix (ECM) is an intricate network composed of various multi-domain macromolecules like collagen, proteoglycans, and fibronectin, etc., that form a structurally stable composite, contributing to the mechanical properties of tissue. However, matricellular proteins are non-structural, secretory extracellular matrix proteins, which modulate various cellular functions via interacting with cell surface receptors, proteases, hormones, and cell-matrix. They play essential roles in maintaining tissue homeostasis by regulating cell differentiation, proliferation, adhesion, migration, and several signal transduction pathways. Matricellular proteins display a broad functionality regulated by their multiple structural domains and their ability to interact with different extracellular substrates and/or cell surface receptors. The expression of these proteins is low in adults, however, gets upregulated following injuries, inflammation, and during tumor growth. The marked elevation in the expression of these proteins during atherosclerosis suggests a positive association between their expression and atherosclerotic lesion formation. The role of matricellular proteins in atherosclerosis development has remained an area of research interest in the last two decades and studies revealed these proteins as important players in governing vascular function, remodeling, and plaque formation. Despite extensive research, many aspects of the matrix protein biology in atherosclerosis are still unknown and future studies are required to investigate whether targeting pathways stimulated by these proteins represent viable therapeutic approaches for patients with atherosclerotic vascular diseases. This review summarizes the characteristics of distinct matricellular proteins, discusses the available literature on the involvement of matrix proteins in the pathogenesis of atherosclerosis and suggests new avenues for future research.
Collapse
Affiliation(s)
- Naveed Pervaiz
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, USA
| | - Ishita Kathuria
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, USA
| | - Ravi Varma Aithabathula
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, USA
| | - Bhupesh Singla
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, USA.
| |
Collapse
|
37
|
Bresgen N, Kovacs M, Lahnsteiner A, Felder TK, Rinnerthaler M. The Janus-Faced Role of Lipid Droplets in Aging: Insights from the Cellular Perspective. Biomolecules 2023; 13:912. [PMID: 37371492 PMCID: PMC10301655 DOI: 10.3390/biom13060912] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
It is widely accepted that nine hallmarks-including mitochondrial dysfunction, epigenetic alterations, and loss of proteostasis-exist that describe the cellular aging process. Adding to this, a well-described cell organelle in the metabolic context, namely, lipid droplets, also accumulates with increasing age, which can be regarded as a further aging-associated process. Independently of their essential role as fat stores, lipid droplets are also able to control cell integrity by mitigating lipotoxic and proteotoxic insults. As we will show in this review, numerous longevity interventions (such as mTOR inhibition) also lead to strong accumulation of lipid droplets in Saccharomyces cerevisiae, Caenorhabditis elegans, Drosophila melanogaster, and mammalian cells, just to name a few examples. In mammals, due to the variety of different cell types and tissues, the role of lipid droplets during the aging process is much more complex. Using selected diseases associated with aging, such as Alzheimer's disease, Parkinson's disease, type II diabetes, and cardiovascular disease, we show that lipid droplets are "Janus"-faced. In an early phase of the disease, lipid droplets mitigate the toxicity of lipid peroxidation and protein aggregates, but in a later phase of the disease, a strong accumulation of lipid droplets can cause problems for cells and tissues.
Collapse
Affiliation(s)
- Nikolaus Bresgen
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Melanie Kovacs
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Angelika Lahnsteiner
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Thomas Klaus Felder
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Mark Rinnerthaler
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| |
Collapse
|
38
|
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
|
39
|
Zhao Z, Behm C, Tian Z, Rausch MA, Rausch-Fan X, Andrukhov O. Cyclic tensile strain-induced yes-associated protein activity modulates the response of human periodontal ligament mesenchymal stromal cells to tumor necrosis factor-α. Arch Oral Biol 2022; 143:105527. [PMID: 36055128 DOI: 10.1016/j.archoralbio.2022.105527] [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: 04/06/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/02/2022]
Abstract
OBJECTIVES This study aimed to evaluate the role of yes-associated protein (YAP) in the inflammatory processes induced in human periodontal ligament-derived mesenchymal stromal cells (hPDL-MSCs) by cyclic tensile strain (CTS). DESIGN hPDL-MSCs from five periodontally healthy individuals were stimulated with 12% CTS and/or TNF-α for 24 h. YAP activity was determined by analyzing the YAP nuclear localization and the target genes expression, using immunofluorescence and qPCR, respectively. Verteporfin was used to inhibit the activation of YAP. The gene expression of interleukin (IL)-6, IL-8, vascular cell adhesion molecule (VCAM)-1, and intercellular adhesion molecule (ICAM)-1 was analyzed by qPCR. RESULTS In the absence of TNF-α, application of CTS resulted in the nuclear YAP translocation and upregulation of YAP target genes. Verteporfin inhibited the activation of YAP pathway and upregulated the basal expression of IL-6 and IL-8. TNF-α induced the activation of YAP pathway, which was inhibited by verteporfin. However, application of CTS under these conditions diminished TNF-α-induced YAP activation. TNF-α-induced expression of IL-6, VCAM-1, and ICAM-1 was inhibited after the application of CTS. Inhibition of YAP activation by verteporfin diminished TNF-α-induced gene expression of IL-6, VCAM-1, and ICAM-1, and under these conditions no inhibitory effect of CTS on these parameters was observed. CONCLUSIONS YAP is at least partially involved in the CTS-activated mechanotransduction pathway. The effects of CTS and YAP on the inflammatory responses depend on the inflammatory environment. A better understanding of the inflammatory modulation by mechanical stress may help improve the orthodontic strategies, especially in the patient with periodontitis.
Collapse
Affiliation(s)
- Zhongqi Zhao
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Christian Behm
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; Division of Orthodontics, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Zhiwei Tian
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Marco Aoqi Rausch
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; Division of Orthodontics, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Xiaohui Rausch-Fan
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Oleh Andrukhov
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
40
|
Gao F, Li C, Smith SM, Peinado N, Kohbodi G, Tran E, Loh YHE, Li W, Borok Z, Minoo P. Decoding the IGF1 signaling gene regulatory network behind alveologenesis from a mouse model of bronchopulmonary dysplasia. eLife 2022; 11:e77522. [PMID: 36214448 PMCID: PMC9581530 DOI: 10.7554/elife.77522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 10/07/2022] [Indexed: 11/13/2022] Open
Abstract
Lung development is precisely controlled by underlying gene regulatory networks (GRN). Disruption of genes in the network can interrupt normal development and cause diseases such as bronchopulmonary dysplasia (BPD) - a chronic lung disease in preterm infants with morbid and sometimes lethal consequences characterized by lung immaturity and reduced alveolarization. Here, we generated a transgenic mouse exhibiting a moderate severity BPD phenotype by blocking IGF1 signaling in secondary crest myofibroblasts (SCMF) at the onset of alveologenesis. Using approaches mirroring the construction of the model GRN in sea urchin's development, we constructed the IGF1 signaling network underlying alveologenesis using this mouse model that phenocopies BPD. The constructed GRN, consisting of 43 genes, provides a bird's eye view of how the genes downstream of IGF1 are regulatorily connected. The GRN also reveals a mechanistic interpretation of how the effects of IGF1 signaling are transduced within SCMF from its specification genes to its effector genes and then from SCMF to its neighboring alveolar epithelial cells with WNT5A and FGF10 signaling as the bridge. Consistently, blocking WNT5A signaling in mice phenocopies BPD as inferred by the network. A comparative study on human samples suggests that a GRN of similar components and wiring underlies human BPD. Our network view of alveologenesis is transforming our perspective to understand and treat BPD. This new perspective calls for the construction of the full signaling GRN underlying alveologenesis, upon which targeted therapies for this neonatal chronic lung disease can be viably developed.
Collapse
Affiliation(s)
- Feng Gao
- Division of Neonatology, Department of Pediatrics, University of Southern CaliforniaLos AngelesUnited States
| | - Changgong Li
- Division of Neonatology, Department of Pediatrics, University of Southern CaliforniaLos AngelesUnited States
| | - Susan M Smith
- Division of Neonatology, Department of Pediatrics, University of Southern CaliforniaLos AngelesUnited States
| | - Neil Peinado
- Division of Neonatology, Department of Pediatrics, University of Southern CaliforniaLos AngelesUnited States
| | - Golenaz Kohbodi
- Division of Neonatology, Department of Pediatrics, University of Southern CaliforniaLos AngelesUnited States
| | - Evelyn Tran
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Yong-Hwee Eddie Loh
- Norris Medical Library, University of Southern CaliforniaLos AngelesUnited States
| | - Wei Li
- Department of Nephrology, Jiangsu Provincial Hospital of Traditional Chinese MedicineNanjingChina
| | - Zea Borok
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San DiegoSan DiegoUnited States
| | - Parviz Minoo
- Division of Neonatology, Department of Pediatrics, University of Southern CaliforniaLos AngelesUnited States
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
41
|
Li Z, Wu Z, Xi X, Zhao F, Liu H, Liu D. Cellular communication network factor 1 interlinks autophagy and ERK signaling to promote osteogenesis of periodontal ligament stem cells. J Periodontal Res 2022; 57:1169-1182. [PMID: 36199215 DOI: 10.1111/jre.13054] [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: 06/07/2022] [Revised: 08/23/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To investigate the effects of cellular communication network factor 1 (CCN1), a critical matricellular protein, on alveolar bone regeneration, and to elucidate the underlying molecular mechanism. BACKGROUND In the process of orthodontic tooth movement, bone deposition on the tension side of human periodontal ligament stem cells (hPDLSCs) ensured high efficiency and long-term stability of the treatment. The matricellular protein CCN1 is responsive to mechanical stimulation, exhibiting important tasks in bone homoeostasis. However, the role and mechanism of CCN1 on alveolar bone remodeling of hPDLSCs remains unclear. METHODS The expression and distribution of CCN1 in rat periodontal ligament were detected by immunofluorescence staining and immunohistochemical staining. ELISA verified the secretion of CCN1 triggered by stretch loading. To examine the mineralization ability of hPDLSCs induced by CCN1, Western blotting, qRT-PCR, ARS, and ALP staining were performed. CCK-8 and cell migration assay were performed to detect the cell proliferation rate and the wound healing. PI3K/Akt, MAPK, and autophagy activation were examined via Western blotting and immunofluorescence. RESULTS Mechanical stimuli induced the release of CCN1 into extracellular environment by hPDLSCs. Knockdown of CCN1 attenuated the osteogenesis of hPDLSCs while rhCCN1 enhanced the expression of Runx2, Col 1, ALPL, and promoted the mineralization nodule formation. CCN1 activated PI3K/Akt and ERK signaling, and blockage of PI3K/Akt signaling reversed the accelerated cell migration triggered by CCN1. The enhanced osteogenesis induced by CCN1 was abolished by ERK signaling inhibitor PD98059 or autophagy inhibitor 3-MA. Further investigation demonstrated PD98059 abrogated the activation of autophagy. CONCLUSION This study demonstrated that CCN1 promotes osteogenesis in hPDLSCs via autophagy and MAPK/ERK pathway.
Collapse
Affiliation(s)
- Zixuan Li
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Zuping Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Xun Xi
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Fang Zhao
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Hong Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Dongxu Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
42
|
Ascenção K, Lheimeur B, Szabo C. Regulation of CyR61 expression and release by 3-mercaptopyruvate sulfurtransferase in colon cancer cells. Redox Biol 2022; 56:102466. [PMID: 36113340 PMCID: PMC9482125 DOI: 10.1016/j.redox.2022.102466] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 10/28/2022] Open
Abstract
Cysteine-rich angiogenic inducer 61 (CYR61, also termed CCN family member 1 or CCN1), is a matricellular protein encoded by the CYR61 gene. This protein has been implicated in the regulation of various cancer-associated processes including tumor growth, angiogenesis, tumor cell adhesion, migration, and invasion as well as the regulation of anticancer drug resistance. Hydrogen sulfide (H2S) is a gaseous endogenous biological mediator, involved in the regulation of cellular bioenergetics, angiogenesis, invasion, and chemotherapeutic resistance in several types of cancer. H2S is produced by three enzymes: cystathionine-β-synthase (CBS), cystathionine-γ-lyase (CSE) and 3-mercaptopyruvate sulfurtransferase (3-MST). The current studies were set up to investigate if CBS or 3-MST regulates CyR61 in colon cancer cells in the context of the regulation of proliferation, migration, and survival. The study mainly utilized HCT116 cells, in which two of the principal H2S-producing enzymes, CBS and 3-MST, are highly expressed. The H2S donor GYY4137 and the polysulfide donor Na2S3 activated the CyR61 promoter in a concentration-dependent fashion. Aminooxyacetic acid (AOAA), a pharmacological inhibitor of CBS as well as HMPSNE: 2-[(4-hydroxy-6- methylpyrimidin-2-yl)sulfanyl]-1-(naphthalen-1-yl)ethan-1-one, a pharmacological inhibitor of 3-MST inhibited CyR61 mRNA expression. This effect was more pronounced in response to HMPSNE than to AOAA and occurred through the modulation of S1PR via ATF1 and CREB. CyR61 was found to play an active, but relatively minor role in maintaining colon cell proliferation. HMPSNE markedly suppressed the secretion/release of CyR61 from the colon cancer cells. Moreover, HMPSNE promoted colon cancer cell apoptosis; endogenously produced CyR61 was found to counteract this effect, at least in part via RhoA activation. Taken together, we conclude that the upregulation of 3-MST in cancer cells exerts cytoprotective effects and confers the cancer cells a more aggressive phenotype - at least in part via the modulation of CyR61 expression and release.
Collapse
Affiliation(s)
- Kelly Ascenção
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Bassma Lheimeur
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Csaba Szabo
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
43
|
Kim K, Cheng N, Lau LF. Cellular communication network factor 1-stimulated liver macrophage efferocytosis drives hepatic stellate cell activation and liver fibrosis. Hepatol Commun 2022; 6:2798-2811. [PMID: 35929736 PMCID: PMC9512468 DOI: 10.1002/hep4.2057] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 06/26/2022] [Accepted: 07/10/2022] [Indexed: 11/06/2022] Open
Abstract
Following inflammatory injury in the liver, neutrophils quickly infiltrate the injured tissue to defend against microbes and initiate the repair process; these neutrophils are short lived and rapidly undergo apoptosis. Hepatic stellate cells (HSCs) are the principal precursor cells that transdifferentiate into myofibroblast-like cells, which produce a large amount of extracellular matrix that promotes repair but can also lead to fibrosis if the injury becomes chronic. The matricellular protein cellular communication network factor 1 (CCN1) acts as a bridging molecule by binding phosphatidylserine in apoptotic cells and integrin αv β3 in phagocytes, thereby triggering efferocytosis or phagocytic clearance of the apoptotic cells. Here, we show that CCN1 induces liver macrophage efferocytosis of apoptotic neutrophils in carbon tetrachloride (CCl4 )-induced liver injury, leading to the production of activated transforming growth factor (TGF)-β1, which in turn induces HSC transdifferentiation into myofibroblast-like cells that promote fibrosis development. Consequently, knock-in mice expressing a single amino acid substitution in CCN1 rendering it unable to bind αv β3 or induce efferocytosis are impaired in neutrophil clearance, production of activated TGF-β1, and HSC transdifferentiation, resulting in greatly diminished liver fibrosis following exposure to CCl4 . Conclusion: These results reveal the crucial role of CCN1 in stimulating liver macrophage clearance of apoptotic neutrophils, a process that drives HSC transdifferentiation into myofibroblastic cells and underlies fibrogenesis in chronic liver injury.
Collapse
Affiliation(s)
- Ki‐Hyun Kim
- Department of Biochemistry and Molecular GeneticsUniversity of Illinois at Chicago, College of MedicineChicagoIllinoisUSA
| | - Naiyuan Cheng
- Department of Biochemistry and Molecular GeneticsUniversity of Illinois at Chicago, College of MedicineChicagoIllinoisUSA
| | - Lester F. Lau
- Department of Biochemistry and Molecular GeneticsUniversity of Illinois at Chicago, College of MedicineChicagoIllinoisUSA
| |
Collapse
|
44
|
ZeXie decoction alleviates non-alcoholic fatty liver disease in rats: The study of genes, lipids, and gut microbiotas. Biochem Biophys Res Commun 2022; 632:129-138. [DOI: 10.1016/j.bbrc.2022.09.097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 09/17/2022] [Accepted: 09/24/2022] [Indexed: 11/23/2022]
|
45
|
Ren J, Wang X, Parry SN, Yee C, Gorrell MD, McLennan SV, Twigg SM. Targeting CCN2 protects against progressive non-alcoholic steatohepatitis in a preclinical model induced by high-fat feeding and type 2 diabetes. J Cell Commun Signal 2022; 16:447-460. [PMID: 35038159 PMCID: PMC9411483 DOI: 10.1007/s12079-022-00667-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes is an independent risk factor for non-alcoholic steatohepatitis (NASH) progression and its mediators have not been resolved. In this study, a pathogenic role of cellular communication network factor 2 (CCN2) protein in NASH pathology, was investigated in an established preclinical NASH model. Male wild type C57BL/6 mice received either Chow or high fat diet (HFD) for 26 weeks, with some mice in each group randomly selected to receive low dose streptozotocin (STZ: 3 i.p. injections, 65 mg/kg) at 15 weeks to induce type 2 diabetes. In the final 10 of the 26 weeks mice from each group were administered i.p. either rabbit anti-CCN2 neutralizing antibody (CCN2Ab) or as control normal rabbit IgG, at a dose of 150 µg per mouse twice/week. NASH developed in the HFD plus diabetes (HFD+DM) group. Administration of CCN2Ab significantly downregulated collagen I and collagen III mRNA induction and prevented pro-inflammatory MCP-1 mRNA induction in HFD+DM mice. At the protein level, CCN2Ab significantly attenuated collagen accumulation by PSR stain and collagen I protein induction in HFD+DM. Phosphorylation of the pro-fibrotic ERK signalling pathway in liver in HFD+DM was attenuated by CCN2Ab treatment. Intrahepatic CCN1 mRNA was induced, whereas CCN3 was downregulated at both the mRNA and protein levels in HFD+DM. CCN3 down-regulation was prevented by CCN2Ab treatment. This in vivo study indicates that CCN2 is a molecular target in NASH with high fat diet and diabetes, and that regulation of ERK signalling is implicated in this process.
Collapse
Affiliation(s)
- Jing Ren
- Greg Brown Diabetes and Endocrinology Research Laboratories, Sydney Medical School (Central), Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Xiaoyu Wang
- Greg Brown Diabetes and Endocrinology Research Laboratories, Sydney Medical School (Central), Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Sarah N Parry
- Greg Brown Diabetes and Endocrinology Research Laboratories, Sydney Medical School (Central), Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Christine Yee
- Greg Brown Diabetes and Endocrinology Research Laboratories, Sydney Medical School (Central), Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Mark D Gorrell
- Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Susan V McLennan
- Greg Brown Diabetes and Endocrinology Research Laboratories, Sydney Medical School (Central), Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
- New South Wales Health Pathology, Camperdown, NSW, 2050, Australia
| | - Stephen M Twigg
- Greg Brown Diabetes and Endocrinology Research Laboratories, Sydney Medical School (Central), Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.
- Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
46
|
Mechanical Stretch Induced Skin Regeneration: Molecular and Cellular Mechanism in Skin Soft Tissue Expansion. Int J Mol Sci 2022; 23:ijms23179622. [PMID: 36077018 PMCID: PMC9455829 DOI: 10.3390/ijms23179622] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Skin soft tissue expansion is one of the most basic and commonly used techniques in plastic surgery to obtain excess skin for a variety of medical uses. However, skin soft tissue expansion is faced with many problems, such as long treatment process, poor skin quality, high retraction rate, and complications. Therefore, a deeper understanding of the mechanisms of skin soft tissue expansion is needed. The key to skin soft tissue expansion lies in the mechanical stretch applied to the skin by an inflatable expander. Mechanical stimulation activates multiple signaling pathways through cellular adhesion molecules and regulates gene expression profiles in cells. Meanwhile, various types of cells contribute to skin expansion, including keratinocytes, dermal fibroblasts, and mesenchymal stem cells, which are also regulated by mechanical stretch. This article reviews the molecular and cellular mechanisms of skin regeneration induced by mechanical stretch during skin soft tissue expansion.
Collapse
|
47
|
Yao MX, Cheng JY, Liu Y, Sun J, Hua DX, He QY, Liu HY, Fu L, Zhao H. Cross-sectional and longitudinal associations of serum Cysteine-rich 61 with severity and prognosis among community-acquired pneumonia patients in China. Front Med (Lausanne) 2022; 9:939002. [PMID: 36035395 PMCID: PMC9403795 DOI: 10.3389/fmed.2022.939002] [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: 05/15/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundCysteine-rich 61 (CYR61) is implicated in many pulmonary diseases. However, the relationship between CYR61 and community-acquired pneumonia (CAP) patients was unknown. This research aimed to estimate the correlations of serum CYR61 with severity and prognosis in CAP patients through a prospective cohort study.MethodsAll 541 CAP patients were enrolled in this study. Fasting venous blood was collected. Clinical characteristics and demographic information were obtained. CYR61 and inflammatory cytokines were detected in serum using ELISA.ResultsSerum CYR61 was gradually increased in parallel with severity scores in CAP patients. Correlative analysis indicated that serum CYR61 was strongly associated with many clinical parameters in CAP patients. Moreover, mixed logistic and linear regression models found that there were positive correlations between serum CYR61 and CAP severity scores after adjusted for age, BMI, and respiratory rate. Stratified analyses suggested that age affected the associations between serum CYR61 and severity scores. On admission, higher serum CYR61 levels elevated the risks of mechanical ventilation, vasoactive agent, ICU admission, death, and longer hospital stays during hospitalization. Moreover, serum CYR61 in combination with severity scores upregulated the predictive capacities for severity and death than single serum CYR61 or severity scores in CAP patients.ConclusionThere are significantly positive dose-response associations of serum CYR61 on admission with the severity and adverse prognostic outcomes, demonstrating that CYR61 is involved in the pathophysiology of CAP. Serum CYR61 may be used as a potential biomarker for the diagnosis and prognosis in CAP patients.
Collapse
Affiliation(s)
- Meng-Xing Yao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jia-Yi Cheng
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ying Liu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Sun
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dong-Xu Hua
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qi-Yuan He
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hong-Yan Liu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lin Fu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Toxicology, Anhui Medical University, Hefei, China
- *Correspondence: Lin Fu, ;
| | - Hui Zhao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Hui Zhao,
| |
Collapse
|
48
|
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
|
49
|
Machcinska S, Walendzik K, Kopcewicz M, Wisniewska J, Rokka A, Pääkkönen M, Slowinska M, Gawronska-Kozak B. Hypoxia reveals a new function of Foxn1 in the keratinocyte antioxidant defense system. FASEB J 2022; 36:e22436. [PMID: 35792861 DOI: 10.1096/fj.202200249rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 01/12/2023]
Abstract
Skin exposed to environmental threats, including injuries and oxidative stress, develops an efficient but not fully recognized system of repair and antioxidant protection. Here, using mass spectrometry analysis (LC-MS/MS), followed by in vitro and in vivo experiments, we provided evidence that Foxn1 in keratinocytes regulates elements of the electron transport chain and participates in the thioredoxin system (Txn2, Txnrd3, and Srxn1) induction, particularly in a hypoxic environment. We first showed that Foxn1 in keratinocytes upregulates glutathione thioredoxin reductase 3 (Txnrd3) protein expression, and high levels of Txnrd3 mRNA were detected in injured skin of Foxn1+/+ mice. We also showed that Foxn1 strongly downregulated the Ccn2 protein expression, participating in epidermal reconstruction after injury. An in vitro assay revealed that Foxn1 controls keratinocyte migration, stimulating it under normoxia and suppressing it under hypoxia. Keratinocytes overexpressing Foxn1 and exposed to hypoxia displayed a reduced ability to promote angiogenesis by downregulating Vegfa expression. In conclusion, this study showed a new mechanism in which Foxn1, along with hypoxia, participates in the activation of antioxidant defense and controls the functional properties of keratinocytes.
Collapse
Affiliation(s)
- Sylwia Machcinska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Katarzyna Walendzik
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Marta Kopcewicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Wisniewska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Anne Rokka
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Mirva Pääkkönen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Mariola Slowinska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Barbara Gawronska-Kozak
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
50
|
HDAC5 inactivates CYR61-regulated CD31/mTOR axis to prevent the occurrence of preeclampsia. Cell Tissue Res 2022; 390:281-292. [PMID: 35900603 DOI: 10.1007/s00441-022-03652-7] [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: 07/26/2021] [Accepted: 06/09/2022] [Indexed: 11/02/2022]
Abstract
Our study was to pinpoint the significance of histone deacetylase 5 (HDAC5) affecting the pathogenesis of preeclampsia (PE) via CD31/mammalian target of rapamycin (mTOR) axis by regulating cysteine-rich angiogenic inducer 61 (CYR61). Expression of HDAC5, CYR61, and CD31/mTOR in placental tissues of patients with PE and trophoblast cells HTR-8/SVneo cells was determined first followed by their interaction analysis. Following different transfection, the significance of HDAC5 in cell functions was assayed in relation to CYR61 and CD31/mTOR. An in vivo PE mouse model was constructed for further validation. The clinical tissue and in vitro cell experimentations discovered that HDAC5 was downregulated in placental tissues of PE patients and trophoblast cells, while CYR61, CD31, mTOR, and p-mTOR displayed upregulation. After overexpression of HDAC5, trophoblast cell functions were enhanced. HDAC5 reduced the acetylation enrichment of H3K27 to inhibit the expression of CYR61. Furthermore, CYR61 promoted the activation of CD31/mTOR axis, thereby inhibiting HTR-8/SVneo cell functions. The in vivo rat model confirmed the above alterations. Taken together, HDAC5 contributes to downregulation of CYR61 through histone deacetylation, inactivating CD31/mTOR axis, which prevents the occurrence and development of PE.
Collapse
|