1
|
Huang J, Wan Z, Li J, Xiong X, Jiang R, Yang B, Xiong C, Chen H, Wan X, Luo Q, Zhao Y, Lin J, Ai X. Downregulation of NNMT affects trophoblast function via inhibiting COMP/CD36/ERK1/2 axis in recurrent spontaneous abortion. Cell Signal 2025; 132:111831. [PMID: 40274084 DOI: 10.1016/j.cellsig.2025.111831] [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: 12/08/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Recurrent spontaneous abortion (RSA) is closely associated with trophoblast dysfunction, yet the underlying regulatory mechanisms remain poorly understood. Herein, we found a significantly decreased level of nicotinamide N-methyltransferase (NNMT) in RSA villous tissues compared to normal pregnancies. NNMT knockdown suppressed trophoblast proliferation, migration and invasion in vitro, and increased embryo absorption rate in vivo. Upstream of NNMT, FOXA1 was identified as its transcriptional regulator, which was also downregulated in RSA villous tissues. Mechanistically, reduced NNMT led to the accumulation of methyl donor S-adenosyl methionine, thus promoting the methylation of histone H3 at lysine 27. This epigenetic modification further inhibited the expression of cartilage oligomeric matrix protein (COMP), along with its binding to CD36 receptor and subsequent activation of ERK1/2 pathway in trophoblast. Together, our study demonstrates the crucial role of NNMT at the maternal-fetal interface, provides mechanistic insights into the pathogenesis of RSA, and lays a basis for developing targeted therapies.
Collapse
Affiliation(s)
- Jialyu Huang
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Reproductive Health, Jiangxi Maternal and Child Health Hospital, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Nanchang Medical College, Nanchang, China
| | - Zhengwei Wan
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiaqi Li
- Department of Gynecologic Oncology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, China
| | - Xiangpeng Xiong
- Department of Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, China
| | - Ruiyin Jiang
- Department of Clinical Medicine, School of Queen Mary, Nanchang University, Nanchang, China
| | - Bin Yang
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Reproductive Health, Jiangxi Maternal and Child Health Hospital, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Nanchang Medical College, Nanchang, China
| | - Chaoyi Xiong
- Department of Pathology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, China
| | - Hong Chen
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Reproductive Health, Jiangxi Maternal and Child Health Hospital, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Nanchang Medical College, Nanchang, China
| | - Xinxia Wan
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Reproductive Health, Jiangxi Maternal and Child Health Hospital, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Nanchang Medical College, Nanchang, China
| | - Qimei Luo
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Reproductive Health, Jiangxi Maternal and Child Health Hospital, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Nanchang Medical College, Nanchang, China
| | - Yan Zhao
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Reproductive Health, Jiangxi Maternal and Child Health Hospital, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Nanchang Medical College, Nanchang, China.
| | - Jiaying Lin
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiaoyan Ai
- Department of Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, China.
| |
Collapse
|
2
|
Xiong Y, Li X, Sun B, Zhang J, Wu X, Guo F. Abnormal collagen deposition mediated by cartilage oligomeric matrix protein in the pathogenesis of oral submucous fibrosis. Int J Oral Sci 2025; 17:25. [PMID: 40148275 PMCID: PMC11950347 DOI: 10.1038/s41368-025-00355-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 02/05/2025] [Accepted: 02/14/2025] [Indexed: 03/29/2025] Open
Abstract
Abnormal accumulation of collagen fibrils is a hallmark feature of oral submucous fibrosis (OSF). However, the precise characteristics and underlying mechanisms remain unclear, impeding the advancement of potential therapeutic approaches. Here, we observed that collagen I, the main component of the extracellular matrix, first accumulated in the lamina propria and subsequently in the submucosa of OSF specimens as the disease progressed. Using RNA-seq and Immunofluorescence in OSF specimens, we screened the cartilage oligomeric matrix protein (COMP) responsible for the abnormal collagen accumulation. Genetic COMP deficiency reduced arecoline-stimulated collagen I deposition significantly in vivo. In comparison, both COMP and collagen I were upregulated under arecoline stimulation in wild-type mice. Human oral buccal mucosal fibroblasts (hBMFs) also exhibited increased secretion of COMP and collagen I after stimulation in vitro. COMP knockdown in hBMFs downregulates arecoline-stimulated collagen I secretion. We further demonstrated that hBMFs present heterogeneous responses to arecoline stimulation, of which COMP-positive fibroblasts secrete more collagen I. Since COMP is a molecular bridge with Fibril-associated collagens with Interrupted Triple helices (FACIT) in the collagen network, we further screened and identified collagen XIV, a FACIT member, co-localizing with both COMP and collagen I. Collagen XIV expression increased under arecoline stimulation in wild-type mice, whereas it was hardly expressed in the Comp-/- mice, even with under stimulation. In summary, we found that COMP may mediates abnormal collagen I deposition by functions with collagen XIV during the progression of OSF, suggesting its potential to be targeted in treating OSF.
Collapse
Affiliation(s)
- Yafei Xiong
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, China
- Academician Workstation for Oral-Maxillofacial Regenerative Medicine, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Xuechun Li
- Academician Workstation for Oral-Maxillofacial Regenerative Medicine, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Bincan Sun
- Academician Workstation for Oral-Maxillofacial Regenerative Medicine, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Zhang
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, China
- Academician Workstation for Oral-Maxillofacial Regenerative Medicine, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoshan Wu
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, China.
- Academician Workstation for Oral-Maxillofacial Regenerative Medicine, Central South University, Changsha, China.
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China.
| | - Feng Guo
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, China.
- Academician Workstation for Oral-Maxillofacial Regenerative Medicine, Central South University, Changsha, China.
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
3
|
Bhamidipati K, McIntyre ABR, Kazerounian S, Ce G, Tran M, Prell SA, Lau R, Khedgikar V, Altmann C, Small A, Wong V, Madhu R, Presti S, Anufrieva KS, Blazar PE, Lange JK, Seifert J, Moreland LW, Croft AP, Lewis MJ, Thomas R, Jonsson AH, Pitzalis C, Gravallese EM, Brenner MB, Korsunsky I, Wechalekar MD, Wei K. Spatial patterning of fibroblast TGFβ signaling underlies treatment resistance in rheumatoid arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.642821. [PMID: 40166167 PMCID: PMC11956964 DOI: 10.1101/2025.03.14.642821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Treatment-refractory rheumatoid arthritis (RA) is a major unmet need, and the mechanisms driving treatment resistance are poorly understood. To identify molecular determinants of RA non-remission, we performed spatial transcriptomic profiling on pre- and post-treatment synovial tissue biopsies from treatment naïve patients who received conventional DMARDs or adalimumab for 6 months. In the baseline biopsies of non-remission patients, we identified significant expansion of fibrogenic fibroblasts marked by high expression of COMP, a fibrosis-associated extracellular matrix protein. COMPhi fibroblasts localized to perivascular niches that, unexpectedly, served as transcriptional hubs for TGFβ activity. We identified endothelial-derived Notch signaling as an upstream regulator of fibroblast TGFβ signaling via its dual role in driving TGFβ isoform expression and suppressing TGFβ receptors, generating a proximal-distal gradient of TGFβ activity. Further, disruption of steady-state Notch signaling in vitro enabled fibrogenic fibroblast activation. Analysis of post-treatment biopsies revealed marked expansion of COMPhi fibroblasts in non-remission RA patients, despite evidence of successful immune cell depletion, suggesting a spatiotemporal process of fibrogenic remodeling linked to treatment resistance. Collectively, our data implicates targeting of TGFβ signaling to prevent exuberant synovial tissue fibrosis as a potential therapeutic strategy for refractory RA.
Collapse
Affiliation(s)
- Kartik Bhamidipati
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Alexa B R McIntyre
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Shideh Kazerounian
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Gao Ce
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Miles Tran
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Sean A Prell
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Rachel Lau
- Centre for Experimental Medicine and Rheumatology, EULAR Centre of Excellence, William Harvey Research Institute, Queen Mary University of London, London, UK
- Barts Health NHS Trust, Barts Biomedical Research Centre (BRC), National Institute for Health and Care Research (NIHR), London, UK
| | - Vikram Khedgikar
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Christopher Altmann
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Rheumatology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Annabelle Small
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Rheumatology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Vincent Wong
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Rheumatology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Roopa Madhu
- Division of Genetics, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Sonia Presti
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Ksenia S Anufrieva
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Philip E Blazar
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Jeffrey K Lange
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Jennifer Seifert
- Division of Rheumatology, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Larry W Moreland
- Division of Rheumatology, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Adam P Croft
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, EULAR Centre of Excellence, William Harvey Research Institute, Queen Mary University of London, London, UK
- Barts Health NHS Trust, Barts Biomedical Research Centre (BRC), National Institute for Health and Care Research (NIHR), London, UK
| | - Ranjeny Thomas
- Frazer Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Anna H Jonsson
- Division of Rheumatology, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, EULAR Centre of Excellence, William Harvey Research Institute, Queen Mary University of London, London, UK
- Barts Health NHS Trust, Barts Biomedical Research Centre (BRC), National Institute for Health and Care Research (NIHR), London, UK
| | - Ellen M Gravallese
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Michael B Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Ilya Korsunsky
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Mihir D Wechalekar
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Rheumatology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Zhao R, Fu M, Shu S, Chen X, Wang X, Zhang N, Yang K, Hua X, Wang X, Song J. Single-Cell Transcriptomics Identified Fibrosis-Activated Valve Interstitial Cells Involved in Functional Tricuspid Regurgitation. JACC. ASIA 2025; 5:478-495. [PMID: 40148022 PMCID: PMC12042980 DOI: 10.1016/j.jacasi.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 01/02/2025] [Accepted: 01/18/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND The treatment of functional tricuspid regurgitation (TR) is still controversial. Characterizing the cellular composition of the tricuspid valve and identifying the molecular alterations of each cell type in valves with TR will advance our understanding of the mechanisms of TR and guide improvements in treatment. OBJECTIVES The authors aimed to investigate the changes in cellular composition and gene expression patterns of cells in regurgitant tricuspid valves and shed light on the mechanisms of functional TR. METHODS To improve our understanding of the pathogenesis of functional TR, we performed single-cell RNA sequencing of tricuspid valve from 10 patients, including 5 patients with moderate-to-severe functional TR and 5 nondiseased control subjects. Multiplexed fluorescence was used to detect the spatial distributions of valvular cell states and validated the cell-cell interaction. RESULTS We assessed the transcriptional profiles of 84,102 cells and identified 6 major cell clusters, along with 25 cell subtypes, in the specimens. Valve interstitial cells (VICs) were the largest population. VICs and lymphoid cells exhibited more heterogeneity in TR patients. VICs exhibited higher transcriptional activity toward matrifibrocyte-like cells and myofibroblast-like cell differentiation, myeloid cells activated immune response, and lymphoid cells promoted fibrosis. In TR, the alternation of COMP-CD47 and FGF2-FGFR1 interaction may occur in TR specimens, which may serve as promising therapeutic targets for TR. CONCLUSIONS Our single-cell atlas highlights the transcriptomic heterogeneity underlying the cell functions and interactions in human tricuspid valves and defines molecular and cellular perturbations in functional TR. We identified VIC clusters with fibrosis activation accumulated in TR valves.
Collapse
Affiliation(s)
- Ruojin Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengxia Fu
- Galactophore Department, Galactophore Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Songren Shu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; The Cardiomyopathy Research Group, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; The Cardiomyopathy Research Group, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohu Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ningning Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; The Cardiomyopathy Research Group, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Keming Yang
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiumeng Hua
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; The Cardiomyopathy Research Group, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China; Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardiac Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
5
|
Liu Y, Yin W. CD36 in liver diseases. Hepatol Commun 2025; 9:e0623. [PMID: 39774047 PMCID: PMC11717518 DOI: 10.1097/hc9.0000000000000623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Cluster of differentiation 36 (CD36) is a transmembrane glycoprotein with the ability to bind to multiple ligands and perform diverse functions. Through the recognition of long-chain fatty acids, proteins containing thrombospondin structural homology repeat domains such as thrombospondin-1, and molecules with molecular structures consistent with danger- or pathogen-associated molecular patterns, CD36 participates in various physiological and pathological processes of the body. CD36 is widely expressed in various cell types, including hepatocytes and KCs in the liver, where it plays a pivotal role in lipid metabolism, inflammation, and oxidative stress. Accumulating evidence suggests that CD36 plays a complex role in the development of nonalcoholic simple fatty liver disease and NASH and contributes to the pathogenesis of inflammatory liver injury, hepatitis B/hepatitis C, liver fibrosis, and liver cancer. This review summarizes the current understanding of the structural properties, expression patterns, and functional mechanisms of CD36 in the context of liver pathophysiology. Furthermore, the potential of CD36 as a therapeutic target for the prevention and treatment of liver diseases is highlighted.
Collapse
|
6
|
Xu Y, Zhang Y, Tian H, Zhong Q, Yi K, Li F, Xue T, Wang H, Lao Y, Xu Y, Li Y, Long L, Li K, Tao Y, Li M. Smart Microneedle Arrays Integrating Cell-Free Therapy and Nanocatalysis to Treat Liver Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309940. [PMID: 38874114 PMCID: PMC11336984 DOI: 10.1002/advs.202309940] [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: 12/27/2023] [Revised: 06/02/2024] [Indexed: 06/15/2024]
Abstract
Liver fibrosis is a chronic pathological condition lacking specific clinical treatments. Stem cells, with notable potential in regenerative medicine, offer promise in treating liver fibrosis. However, stem cell therapy is hindered by potential immunological rejection, carcinogenesis risk, efficacy variation, and high cost. Stem cell secretome-based cell-free therapy offers potential solutions to address these challenges, but it is limited by low delivery efficiency and rapid clearance. Herein, an innovative approach for in situ implantation of smart microneedle (MN) arrays enabling precisely controlled delivery of multiple therapeutic agents directly into fibrotic liver tissues is developed. By integrating cell-free and platinum-based nanocatalytic combination therapy, the MN arrays can deactivate hepatic stellate cells. Moreover, they promote excessive extracellular matrix degradation by more than 75%, approaching normal levels. Additionally, the smart MN arrays can provide hepatocyte protection while reducing inflammation levels by ≈70-90%. They can also exhibit remarkable capability in scavenging almost 100% of reactive oxygen species and alleviating hypoxia. Ultimately, this treatment strategy can effectively restrain fibrosis progression. The comprehensive in vitro and in vivo experiments, supplemented by proteome and transcriptome analyses, substantiate the effectiveness of the approach in treating liver fibrosis, holding immense promise for clinical applications.
Collapse
Affiliation(s)
- Yanteng Xu
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Yixin Zhang
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Hao Tian
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
- Department of NeurologyThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Qingguo Zhong
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Ke Yi
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Fenfang Li
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Tiantian Xue
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Yeh‐Hsing Lao
- Department of Pharmaceutical SciencesUniversity at BuffaloThe State University of New YorkBuffaloNY14214USA
| | - Yingying Xu
- Center for Health ResearchGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou510530China
- University of China Academy of SciencesBeijing100049China
| | - Yinxiong Li
- Center for Health ResearchGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou510530China
- University of China Academy of SciencesBeijing100049China
| | - Ling Long
- Department of NeurologyThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Kai Li
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Yu Tao
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
- Guangdong Provincial Key Laboratory of Liver DiseaseGuangzhou510630China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
- Guangdong Provincial Key Laboratory of Liver DiseaseGuangzhou510630China
| |
Collapse
|
7
|
Ye T, Huang H, Chen K, Yu Y, Yue D, Jiang L, Wu H, Zhang N. Development and validation of prognostic signatures of NAD+ metabolism and immune-related genes in colorectal cancer. Heliyon 2024; 10:e34403. [PMID: 39130406 PMCID: PMC11315184 DOI: 10.1016/j.heliyon.2024.e34403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 07/01/2024] [Accepted: 07/09/2024] [Indexed: 08/13/2024] Open
Abstract
Background Colorectal cancer (CRC) is a prevalent cause of death from malignant tumors. This study aimed to develop a nicotinamide adenine dinucleotide (NAD+) metabolism and immune-related prognostic signature, providing a theoretical foundation for prognosis and therapy in CRC patients. Methods NAD + metabolism-related and immune-related subtypes of CRC patients were identified by consistent clustering. Differentially expressed genes (DEGs) between the two subtypes of CRC were identified by overlapping. A risk signature was constructed using univariate Cox and least absolute shrinkage and selection operator (LASSO) regression analyses. Independent prognostic predictors were authenticated by Cox analysis. Gene set variation analysis (GSVA) and single-sample gene set enrichment analysis (ssGSEA) were applied to investigate the connection between the prognostic signature and the immune microenvironment. Chemotherapy drug sensitivity and immunotherapy responsiveness were projected using the 'pRRophetic' package and Tumor Immune Dysfunction and Exclusion (TIDE) website. The Human Protein Atlas (HPA) database was used to assess the protein expression of prognostic genes in CRC and normal tissues. Results Using bioinformatics methods, three prognostic genes related to immune-related NAD + metabolism were identified, and the results were used to establish and verify a prognostic signature related to immune-related NAD + metabolism in CRC patients. Cox regression analysis confirmed that the risk score was a reliable independent prognostic predictor. GSVA and ssGSEA indicated that the prognostic signature was associated with the immune microenvironment. TIDE analysis suggested that the signature might act as an immunotherapy predictor. Chemotherapy sensitivity analysis revealed that COMP was correlated with chemotherapy sensitivity in CRC patients and might be a potential therapeutic target. Conclusion This study identified NAD + metabolism-immune-related prognostic genes (MOGAT2, COMP, and DNASE1L3) and developed a prognostic signature for CRC prognosis, which is significant for clinical prognosis prediction and treatment strategy decisions for CRC patients.
Collapse
Affiliation(s)
- Tao Ye
- Department of Rehabilitation, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Hong Huang
- The First Clinical College of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Kangli Chen
- The First Clinical College of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Yuanao Yu
- The First Clinical College of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Dongqin Yue
- The First Clinical College of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Li Jiang
- The First Clinical College of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Huixian Wu
- The First Clinical College of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Ning Zhang
- Department of Pharmacy, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| |
Collapse
|
8
|
Wang Y, Li HT, Liu G, Jiang CS, Ni YH, Zeng JH, Lin X, Wang QY, Li DZ, Wang W, Zeng XP. COMP promotes pancreatic fibrosis by activating pancreatic stellate cells through CD36-ERK/AKT signaling pathways. Cell Signal 2024; 118:111135. [PMID: 38479555 DOI: 10.1016/j.cellsig.2024.111135] [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: 11/25/2023] [Revised: 02/22/2024] [Accepted: 03/08/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Pancreatic fibrosis is one of the most important pathological features of chronic pancreatitis (CP) and pancreatic stellate cells (PSCs) are the key cells of fibrosis. As an extracellular matrix (ECM) glycoprotein, cartilage oligomeric matrix protein (COMP) is critical for collagen assembly and ECM stability and recent studies showed that COMP exert promoting fibrosis effect in the skin, lungs and liver. However, the role of COMP in activation of PSCs and pancreatic fibrosis remain unclear. We aimed to investigate the role and specific mechanisms of COMP in regulating the profibrotic phenotype of PSCs and pancreatic fibrosis. METHODS ELISA method was used to determine serum COMP in patients with CP. Mice model of CP was established by repeated intraperitoneal injection of cerulein and pancreatic fibrosis was evaluated by Hematoxylin-Eosin staining (H&E) and Sirius red staining. Immunohistochemical staining was used to detect the expression changes of COMP and fibrosis marker such as α-SMA and Fibronectin in pancreatic tissue of mice. Cell Counting Kit-8, Wound Healing and Transwell assessed the proliferation and migration of human pancreatic stellate cells (HPSCs). Western blotting, qRT-PCR and immunofluorescence staining were performed to detect the expression of fibrosis marker, AKT and MAPK family proteins in HPSCs. RNA-seq omics analysis as well as small interfering RNA of COMP, recombinant human COMP (rCOMP), MEK inhibitors and PI3K inhibitors were used to study the effect and mechanism of COMP on activation of HPSCs. RESULTS ELISA showed that the expression of COMP significantly increased in the serum of CP patients. H&E and Sirius red staining analysis showed that there was a large amount of collagen deposition in the mice in the CP model group and high expression of COMP, α-SMA, Fibronectin and Vimentin were observed in fibrotic tissues. TGF-β1 stimulates the activation of HPSCs and increases the expression of COMP. Knockdown of COMP inhibited proliferation and migration of HPSCs. Further, RNA-seq omics analysis and validation experiments in vitro showed that rCOMP could significantly promote the proliferation and activation of HPSCs, which may be due to promoting the phosphorylation of ERK and AKT through membrane protein receptor CD36. rCOMP simultaneously increased the expression of α-SMA, Fibronectin and Collagen I in HPSCs. CONCLUSION In conclusion, this study showed that COMP was up-regulated in CP fibrotic tissues and COMP induced the activation, proliferation and migration of PSCs through the CD36-ERK/AKT signaling pathway. COMP may be a potential therapeutic candidate for the treatment of CP. Interfering with the expression of COMP or the communication between COMP and CD36 on PSCs may be the next direction for therapeutic research.
Collapse
Affiliation(s)
- Yi Wang
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hai-Tao Li
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China; Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, China; Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Gang Liu
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China; Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, China; Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Chuan-Shen Jiang
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China; Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, China; Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Yan-Hong Ni
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jing-Hui Zeng
- Department of Presbyatrics, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xia Lin
- Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Qing-Yun Wang
- Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Da-Zhou Li
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China; Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, China; Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China.
| | - Wen Wang
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China; Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, China; Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China.
| | - Xiang-Peng Zeng
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China; Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, China; Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China.
| |
Collapse
|
9
|
Graf SD, Keber CU, Hattesohl A, Teply-Szymanski J, Hattesohl S, Guder M, Gercke N, Di Fazio P, Slater EP, Jesinghaus M, Denkert C, Bartsch DK, Lehman B. Mesenteric fibrosis in patients with small intestinal neuroendocrine tumors is associated with enrichment of alpha-smooth muscle actin-positive fibrosis and COMP-expressing stromal cells. J Neuroendocrinol 2024; 36:e13364. [PMID: 38246597 DOI: 10.1111/jne.13364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/09/2023] [Accepted: 12/08/2023] [Indexed: 01/23/2024]
Abstract
Neuroendocrine tumors of the small intestine (SI-NETs) often develop lymph node metastasis (LNM)-induced mesenteric fibrosis (MF). MF can cause intestinal obstruction as well as ischemia and render surgical resection technically challenging. The underlying pathomechanisms of MF are still not well understood. We examined mesenteric LNM and the surrounding stroma compartment from 24 SI-NET patients, including 11 with in situ presentation of strong MF (MF+) and 13 without MF (MF-). Differential gene expression was assessed with the HTG EdgeSeq Oncology Biomarker Panel comparing MF+ with MF- within LNM and paired stromal samples, respectively. Most interesting differentially expressed genes were validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) in combination with validation of associated protein levels utilizing immunohistochemistry (IHC) staining of MF+ and MF- formalin-fixed, paraffin-embedded (FFPE) patient samples. Overall, 14 genes measured with a 2549-gene expression panel were differentially expressed in MF+ patients compared to MF-. Of those, nine were differentially expressed genes in LNM and five genes in the stromal tissue (>2-fold change, p < .05). The top hits included increased COMP and COL11A1 expression in the stroma of MF+ patients compared to MF-, as well as decreased HMGA2, COL6A6, and SLC22A3 expression in LNM of MF+ patients compared to LNM of MF- patients. RT-qPCR confirmed high levels of COMP and COL11A1 in stroma samples of MF+ compared to MF- patients. IHC staining confirmed the enrichment of α-smooth muscle actin-positive fibrosis in MF+ compared to MF- patients with corresponding increase of COMP-expressing stromal cells in MF+. Since COMP is associated with the known driver for fibrosis development transforming growth factor beta and with a cancer-associated fibroblasts enriched environment, it seems to be a promising new target for MF research.
Collapse
Affiliation(s)
- Sebastian D Graf
- Department of Visceral, Thoracic and Vascular Surgery, Philipps-University Marburg, Marburg, Germany
| | - Corinna U Keber
- Institute of Pathology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Akira Hattesohl
- Institute of Pathology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Julia Teply-Szymanski
- Institute of Pathology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Sophia Hattesohl
- Institute of Pathology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Marc Guder
- Institute of Pathology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Norman Gercke
- Department of Visceral, Thoracic and Vascular Surgery, Philipps-University Marburg, Marburg, Germany
| | - Pietro Di Fazio
- Department of Visceral, Thoracic and Vascular Surgery, Philipps-University Marburg, Marburg, Germany
| | - Emily P Slater
- Department of Visceral, Thoracic and Vascular Surgery, Philipps-University Marburg, Marburg, Germany
| | - Moritz Jesinghaus
- Institute of Pathology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Carsten Denkert
- Institute of Pathology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Detlef K Bartsch
- Department of Visceral, Thoracic and Vascular Surgery, Philipps-University Marburg, Marburg, Germany
| | - Bettina Lehman
- Department of Visceral, Thoracic and Vascular Surgery, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
10
|
Xia L, Zhou Z, Chen X, Luo W, Ding L, Xie H, Zhuang W, Ni K, Li G. Ligand-dependent CD36 functions in cancer progression, metastasis, immune response, and drug resistance. Biomed Pharmacother 2023; 168:115834. [PMID: 37931517 DOI: 10.1016/j.biopha.2023.115834] [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: 07/12/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023] Open
Abstract
CD36, a multifunctional glycoprotein, has been shown to play critical roles in tumor initiation, progression, metastasis, immune response, and drug resistance. CD36 serves as a receptor for a wide range of ligands, including lipid-related ligands (e.g., long-chain fatty acid (LCFA), oxidized low-density lipoprotein (oxLDL), and oxidized phospholipids), as well as protein-related ligands (e.g., thrombospondins, amyloid proteins, collagens I and IV). CD36 is overexpressed in various cancers and may act as an independent prognostic marker. While it was initially identified as a mediator of anti-angiogenesis through its interaction with thrombospondin-1 (TSP1), recent research has highlighted its role in promoting tumor growth, metastasis, drug resistance, and immune suppression. The varied impact of CD36 on cancer is likely ligand-dependent. Therefore, we focus specifically on the ligand-dependent role of CD36 in cancer to provide a critical review of recent advances, perspectives, and challenges.
Collapse
Affiliation(s)
- Liqun Xia
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhenwei Zhou
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianjiong Chen
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenqin Luo
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lifeng Ding
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiyun Xie
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zhuang
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Kangxin Ni
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Gonghui Li
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
11
|
Dong Y, Wang X, Xu L, Li X, Dai H, Mao X, Chu Y, Yuan X, Liu H. Development of a Chimeric Protein BiPPB-mIFNγ-tTβRII for Improving the Anti-Fibrotic Activity in Vivo by Targeting Fibrotic Liver and Dual Inhibiting the TGF-β1/Smad Signaling Pathway. Protein J 2023; 42:753-765. [PMID: 37690089 DOI: 10.1007/s10930-023-10147-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2023] [Indexed: 09/12/2023]
Abstract
Excessive production of transforming growth factor β1 (TGF-β1) in activated hepatic stellate cells (aHSCs) promotes liver fibrosis by activating the TGF-β1/Smad signaling pathway. Thus, specifically inhibiting the pro-fibrotic activity of TGF-β1 in aHSCs is an ideal strategy for treating liver fibrosis. Overexpression of platelet-derived growth factor β receptor (PDGFβR) has been demonstrated on the surface of aHSCs relative to normal cells in liver fibrosis. Interferon-gamma peptidomimetic (mIFNγ) and truncated TGF-β receptor type II (tTβRII) inhibit the TGF-β1/Smad signaling pathway by different mechanisms. In this study, we designed a chimeric protein by the conjugation of (1) mIFNγ and tTβRII coupled via plasma protease-cleavable linker sequences (FNPKTP) to (2) PDGFβR-recognizing peptide (BiPPB), namely BiPPB-mIFNγ-tTβRII. This novel protein BiPPB-mIFNγ-tTβRII was effectively prepared using Escherichia coli expression system. The active components BiPPB-mIFNγ and tTβRII were slowly released from BiPPB-mIFNγ-tTβRII by hydrolysis using the plasma protease thrombin in vitro. Moreover, BiPPB-mIFNγ-tTβRII highly targeted to fibrotic liver tissues, markedly ameliorated liver morphology and fibrotic responses in chronic liver fibrosis mice by both inhibiting the phosphorylation of Smad2/3 and inducing the expression of Smad7. Meanwhile, BiPPB-mIFNγ-tTβRII markedly reduced the deposition of collagen fibrils and expression of fibrosis-related proteins in acute liver fibrosis mice. Furthermore, BiPPB-mIFNγ-tTβRII showed a good safety performance in both liver fibrosis mice. Taken together, BiPPB-mIFNγ-tTβRII improved the in vivo anti-liver fibrotic activity due to its high fibrotic liver-targeting potential and the dual inhibition of the TGF-β1/Smad signaling pathway, which may be a potential candidate for targeting therapy on liver fibrosis.
Collapse
Affiliation(s)
- Yixin Dong
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Xiaohua Wang
- Laboratory of Pathogenic Microbiology and Immunology, Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Liming Xu
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Xin Li
- Department of Pediatrics, Hongqi Hospital Affiliated to Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Haibing Dai
- Department of Biology, Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Xu Mao
- Department of Pharmacology, Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Yanhui Chu
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Xiaohuan Yuan
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, 157011, Mudanjiang, PR China.
| | - Haifeng Liu
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, 157011, Mudanjiang, PR China.
- Laboratory of Pathogenic Microbiology and Immunology, Mudanjiang Medical University, 157011, Mudanjiang, PR China.
| |
Collapse
|
12
|
Wu T, Jin Y, Chen F, Xuan X, Cao J, Liang Y, Wang Y, Zhan J, Zhao M, Huang C. Identification and characterization of bone/cartilage-associated signatures in common fibrotic skin diseases. Front Genet 2023; 14:1121728. [PMID: 37082197 PMCID: PMC10111020 DOI: 10.3389/fgene.2023.1121728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
Background: Fibrotic skin diseases are characterized by excessive accumulation of the extracellular matrix (ECM) and activation of fibroblasts, leading to a global healthcare burden. However, effective treatments of fibrotic skin diseases remain limited, and their pathological mechanisms require further investigation. This study aims to investigate the common biomarkers and therapeutic targets in two major fibrotic skin diseases, namely, keloid and systemic sclerosis (SSc), by bioinformatics analysis.Methods: The keloid (GSE92566) and SSc (GSE95065) datasets were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified, followed by functional enrichment analysis using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). We then constructed a protein–protein interaction (PPI) network for the identification of hub genes. We explored the possibility of further functional enrichment analysis of hub genes on the Metascape, GeneMANIA, and TissueNexus platforms. Transcription factor (TF)–hub gene and miRNA–hub gene networks were established using NetworkAnalyst. We fixed GSE90051 and GSE76855 as the external validation datasets. Student’s t-test and receiver operating characteristic (ROC) curve were used for candidate hub gene validation. Hub gene expression was assessed in vitro by quantitative real-time PCR.Results: A total of 157 overlapping DEGs (ODEGs) were retrieved from the GSE92566 and GSE95065 datasets, and five hub genes (COL11A1, COL5A2, ASPN, COL10A1, and COMP) were identified and validated. Functional studies revealed that hub genes were predominantly enriched in bone/cartilage-related and collagen-related processes. FOXC1 and miR-335-5p were predicted to be master regulators at both transcriptional and post‐transcriptional levels.Conclusion: COL11A1, COL5A2, ASPN, COL10A1, and COMP may help understand the pathological mechanism of the major fibrotic skin diseases; moreover, FOXC1 and miR-355-5p could build a regulatory network in keloid and SSc.
Collapse
Affiliation(s)
- Ting Wu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Jin
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangqi Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiuyun Xuan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juanmei Cao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Liang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqing Wang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinshan Zhan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengjie Zhao
- Department of Dermatology, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Mengjie Zhao, ; Changzheng Huang,
| | - Changzheng Huang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Mengjie Zhao, ; Changzheng Huang,
| |
Collapse
|
13
|
Wang J, Yang Y, Sun F, Luo Y, Yang Y, Li J, Hu W, Tao H, Lu C, Yang JJ. ALKBH5 attenuates mitochondrial fission and ameliorates liver fibrosis by reducing Drp1 methylation. Pharmacol Res 2023; 187:106608. [PMID: 36566000 DOI: 10.1016/j.phrs.2022.106608] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Mitochondrial metabolism plays a pivotal role in various cellular processes and fibrosis. However, the mechanism underlying mitochondrial metabolic function and liver fibrosis remains poorly understood. In this study, we determined whether mitochondrial metabolism mediates liver fibrosis using cells, animal models, and clinical samples to elucidate the potential effects and underlying mechanism of mitochondrial metabolism in liver fibrosis. We report that AlkB Homolog 5 (ALKBH5) decreases mitochondrial membrane potential (MMP) and oxygen consumption rate (OCR), suppresses mitochondrial fission and hepatic stellate cell (HSC) proliferation and migration and ameliorates liver fibrosis. Enhancement of mitochondrial fission, an essential event during HSC proliferation and migration, is dependent on decreased ALKBH5 expression. Furthermore, we reveal that low ALKBH5 expression is associated with elevated N6-methyladenosine (m6A) mRNA levels. Mechanistically, ALKBH5 mediates m6A demethylation in the 3'UTR of Drp1 mRNA and induces its translation in a YTH domain family proteins 1 (YTHDF1)-independent manner. Subsequently, in transforming growth factor-β1 (TGF-β1) induced HSC, Dynamin-related protein 1 (Drp1) mediates mitochondrial fission and increases cell proliferation and migration. Decreased Drp1 expression inhibits mitochondrial fission and suppresses HSC proliferation and migration. Notably, human fibrotic liver and heart tissue exhibited enhanced mitochondrial fission; increased YTHDF1, Drp1, alpha-smooth muscle actin (α-SMA) and collagen I expression; decreased ALKBH5 expression and increased liver fibrosis. Our results highlight a novel mechanism by which ALKBH5 suppresses mitochondrial fission and HSC proliferation and migration by reducing Drp1 methylation in an m6A-YTHDF1-dependent manner, which may indicate a demethylation-based approach for liver fibrosis diagnosis and therapy.
Collapse
Affiliation(s)
- Juan Wang
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yang Yang
- Department of Surgical Oncology, Suzhou Science & Technology Town Hospital, Suzhou 215153, China
| | - Feng Sun
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yong Luo
- Department of Scientific research and experimental center, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Yan Yang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Hui Tao
- Department of Anesthesiology, The Second Hospital of Anhui Medical University, Hefei 230601, China.
| | - Chao Lu
- First Affiliated Hospital, Anhui University of Science & Technology, Huainan 232001, China; School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China.
| |
Collapse
|
14
|
Guo B, Wang Y, Liu W, Zhang S. Cartilage oligomeric matrix protein acts as a molecular biomarker in multiple cancer types. Clin Transl Oncol 2022; 25:535-554. [DOI: 10.1007/s12094-022-02968-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/26/2022] [Indexed: 04/07/2023]
|
15
|
Cartilage Oligomeric Matrix Protein, Diseases, and Therapeutic Opportunities. Int J Mol Sci 2022; 23:ijms23169253. [PMID: 36012514 PMCID: PMC9408827 DOI: 10.3390/ijms23169253] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/05/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Cartilage oligomeric matrix protein (COMP) is an extracellular matrix (ECM) glycoprotein that is critical for collagen assembly and ECM stability. Mutations of COMP cause endoplasmic reticulum stress and chondrocyte apoptosis, resulting in rare skeleton diseases. The bouquet-like structure of COMP allows it to act as a bridging molecule that regulates cellular phenotype and function. COMP is able to interact with many other ECM components and binds directly to a variety of cellular receptors and growth factors. The roles of COMP in other skeleton diseases, such as osteoarthritis, have been implied. As a well-established biochemical marker, COMP indicates cartilage turnover associated with destruction. Recent exciting achievements indicate its involvement in other diseases, such as malignancy, cardiovascular diseases, and tissue fibrosis. Here, we review the basic concepts of COMP and summarize its novel functions in the regulation of signaling events. These findings renew our understanding that COMP has a notable function in cell behavior and disease progression as a signaling regulator. Interestingly, COMP shows distinct functions in different diseases. Targeting COMP in malignancy may withdraw its beneficial effects on the vascular system and induce or aggravate cardiovascular diseases. COMP supplementation is a promising treatment for OA and aortic aneurysms while it may induce tissue fibrosis or cancer metastasis.
Collapse
|
16
|
Tan J, Yu Z, Ling X, Qiu G, Yang X, Tang Y, Yang D, Yang M, Gao F. Main Pathological Changes of Benign Ureteral Strictures. Front Med (Lausanne) 2022; 9:916145. [PMID: 35872768 PMCID: PMC9300898 DOI: 10.3389/fmed.2022.916145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo identify the pathological classification of benign ureteral strictures according to the histological features and explore the relationship between various pathological types and inflammatory cells, fibroblasts, and collagen.Patients and MethodsThirty one specimens from patients diagnosed with ureteral strictures between 2013 and 2021 were included and classified according to the histopathological characteristics. The number of fibroblasts and inflammatory cells was counted, and the proportion of type I and type III collagen in ureteral stricture tissues was detected by picrosirius red staining.ResultsWe identified three types of benign ureteral strictures in 31 specimens: inflammatory cell infiltration (n = 10, 32%), fibroplasia (n = 14, 45%), and hyalinization (n = 7, 23%), with significant differences in obstruction history and hydronephrosis grades among the three types. The number of inflammatory cells (lymphocytes, neutrophils and eosinophils) was significantly lower in hyalinization ureteral strictures than in the other two types (p < 0.05). The number of foreign-body giant cells associated with foreign-body reactions increased significantly in suture-induced ureteral strictures (p < 0.05). Fibroplasia type had the largest number of fibroblasts, whereas the other two types had smaller numbers. The results of type I and III collagen analysis showed that type I and III collagen were the most abundant in hyalinization among all ureteral stricture types (p < 0.05). Compared to ureteral strictures, the content of type I and III collagen in atresia increased significantly (p < 0.05).ConclusionCommon pathological types of benign ureteral strictures include inflammatory cell infiltration, fibroplasia, and hyalinization. Changes in type I and III collagen, inflammatory cells, and fibroblasts in different pathological types may be related to the progression of ureteral strictures.
Collapse
Affiliation(s)
- Jiang Tan
- Department of Anatomy, Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Zhuoyuan Yu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyi Ling
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guoping Qiu
- Department of Anatomy, Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Xin Yang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Tang
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China
| | - Dong Yang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mei Yang
- Department of Anatomy, Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- *Correspondence: Mei Yang,
| | - Fei Gao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Fei Gao,
| |
Collapse
|
17
|
Yu S, Ericson M, Fanjul A, Erion DM, Paraskevopoulou M, Smith EN, Cole B, Feaver R, Holub C, Gavva N, Horman SR, Huang J. Genome-wide CRISPR Screening to Identify Drivers of TGF-β-Induced Liver Fibrosis in Human Hepatic Stellate Cells. ACS Chem Biol 2022; 17:918-929. [PMID: 35274923 PMCID: PMC9016707 DOI: 10.1021/acschembio.2c00006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Liver fibrosis progression in chronic liver disease leads to cirrhosis, liver failure, or hepatocellular carcinoma and often ends in liver transplantation. Even with an increased understanding of liver fibrogenesis and many attempts to generate therapeutics specifically targeting fibrosis, there is no approved treatment for liver fibrosis. To further understand and characterize the driving mechanisms of liver fibrosis, we developed a high-throughput genome-wide CRISPR/Cas9 screening platform to identify hepatic stellate cell (HSC)-derived mediators of transforming growth factor (TGF)-β-induced liver fibrosis. The functional genomics phenotypic screening platform described here revealed the novel biology of TGF-β-induced fibrogenesis and potential drug targets for liver fibrosis.
Collapse
Affiliation(s)
- Shan Yu
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Matthew Ericson
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Andrea Fanjul
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Derek M. Erion
- Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts 02139, United States
| | - Maria Paraskevopoulou
- Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts 02139, United States
| | - Erin N. Smith
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Banumathi Cole
- HemoShear Therapeutics, Inc., Charlottesville, Virginia 22902, United States
| | - Ryan Feaver
- HemoShear Therapeutics, Inc., Charlottesville, Virginia 22902, United States
| | - Corine Holub
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Narender Gavva
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Shane R. Horman
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Jie Huang
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| |
Collapse
|
18
|
Chen L, Zhu D, Huang J, Zhang H, Zhou G, Zhong X. Identification of Hub Genes Associated with COPD Through Integrated Bioinformatics Analysis. Int J Chron Obstruct Pulmon Dis 2022; 17:439-456. [PMID: 35273447 PMCID: PMC8901430 DOI: 10.2147/copd.s353765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/20/2022] [Indexed: 12/30/2022] Open
Affiliation(s)
- Lin Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
- Department of Respiratory and Critical Care Medicine, Liuzhou People’s Hospital, LiuZhou, Guangxi, People’s Republic of China
| | - Donglan Zhu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Jinfu Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Hui Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Guang Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Xiaoning Zhong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
- Correspondence: Xiaoning Zhong, Tel +86 13607881203, Fax +86 771-5356702, Email
| |
Collapse
|
19
|
Abdel-Azeez HA, Elhady HA, Fikry AA. Cartilage oligomeric matrix protein as a non-invasive biomarker for diagnosis of hepatocellular carcinoma in patients with liver cirrhosis. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2022; 15:139-145. [PMID: 35845304 PMCID: PMC9275745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/12/2022] [Indexed: 11/08/2022]
Abstract
Aim The current study purposed to evaluate serum COMP (Cartilage oligomeric matrix protein) as a diagnostic marker for HCC in patients with cirrhosis and to correlate it with other parameters of disease progression. Background COMP is known to promote fibrosis in various tissues. Emerging evidence shows that COMP plays critical roles in tumor development. It can serve as a fibrosis and cancer biomarkers. Methods The study included 24 subjects who serve as the healthy control, 24 cirrhotic patients without HCC, and 24 HCC patients with cirrhosis. All participants were subjected to liver function tests, AFP, calculation of fibrotic indices (APRI and FIB-4), and serum COMP by ELISA. Results COMP was significantly increased in cirrhotic patients when compared to healthy controls and in HCC patients when compared to cirrhotic patients and healthy controls. A significant positive correlation was observed between COMP and APRI and FIB-4 in cirrhotic and HCC patients. Based on receiver operating characteristic (ROC) curve analysis, COMP had an area under curve (AUC) of 0.943 with 87.5% sensitivity and 79.2% specificity for diagnosis of HCC in cirrhotic patients. In combination with AFP, the sensitivity was increased to 100%. Conclusion COMP might act as a promising non-invasive biomarker for HCC either alone or in combination with AFP. It was correlated with the degree of fibrosis and associated with advanced cancer staging.
Collapse
Affiliation(s)
- Hala A Abdel-Azeez
- Clinical PathologyDepartment, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hoda A Elhady
- Internal Medicine Department, Faculty of Medicine, Zagazig University,Zagazig, Egypt
| | - Abeer A Fikry
- Clinical PathologyDepartment, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
20
|
Janša V, Klančič T, Pušić M, Klein M, Vrtačnik Bokal E, Ban Frangež H, Rižner TL. Proteomic analysis of peritoneal fluid identified COMP and TGFBI as new candidate biomarkers for endometriosis. Sci Rep 2021; 11:20870. [PMID: 34686725 PMCID: PMC8536751 DOI: 10.1038/s41598-021-00299-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/30/2021] [Indexed: 11/09/2022] Open
Abstract
Endometriosis is a common non-malignant gynecological disease that significantly compromises fertility and quality of life of the majority of patients. The gold standard for diagnosis is visual inspection of the pelvic organs by surgical laparoscopy and there are no biomarkers that would allow non-invasive diagnosis. The pathogenesis of endometriosis is not completely understood, thus analysis of peritoneal fluid might contribute in this respect. Our prospective case-control study included 58 patients undergoing laparoscopy due to infertility, 32 patients with peritoneal endometriosis (cases) and 26 patients with unexplained primary infertility (controls). Discovery proteomics using antibody microarrays that covered 1360 proteins identified 16 proteins with different levels in cases versus the control patients. The validation using an ELISA approach confirmed significant differences in the levels of cartilage oligomeric matrix protein (COMP) and transforming growth factor-β-induced protein ig-h3 (TGFBI) and nonsignificant differences in angiotensinogen (AGT). A classification model based on a linear support vector machine revealed AUC of > 0.83, sensitivity of 0.81 and specificity of 1.00. Differentially expressed proteins represent candidates for diagnostic and prognostic biomarkers or drug targets. Our findings have brought new knowledge that will be helpful in the understanding of the pathophysiology of endometriosis and warrant further studies in blood samples.
Collapse
Affiliation(s)
- V Janša
- Department of Obstetrics and Gynecology, University Medical Centre Ljubljana, Šlajmerjeva 3, 1000, Ljubljana, Slovenia
| | - T Klančič
- Faculty of Medicine, Institute of Biochemistry, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - M Pušić
- Faculty of Medicine, Institute of Biochemistry, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - M Klein
- Sciomics GmbH, Karl-Landsteiner-Straße 6, 69151, Neckargemünd, Germany
| | - E Vrtačnik Bokal
- Department of Obstetrics and Gynecology, University Medical Centre Ljubljana, Šlajmerjeva 3, 1000, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - H Ban Frangež
- Department of Obstetrics and Gynecology, University Medical Centre Ljubljana, Šlajmerjeva 3, 1000, Ljubljana, Slovenia.
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia.
| | - T Lanišnik Rižner
- Faculty of Medicine, Institute of Biochemistry, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia.
| |
Collapse
|
21
|
Tumor Nonimmune-Microenvironment-Related Gene Expression Signature Predicts Brain Metastasis in Lung Adenocarcinoma Patients after Surgery: A Machine Learning Approach Using Gene Expression Profiling. Cancers (Basel) 2021; 13:cancers13174468. [PMID: 34503278 PMCID: PMC8430997 DOI: 10.3390/cancers13174468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/30/2021] [Accepted: 09/02/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary It is important to be able to predict brain metastasis in lung adenocarcinoma patients; however, research in this area is still lacking. Much of the previous work on tumor microenvironments in lung adenocarcinoma with brain metastasis concerns the tumor immune microenvironment. The importance of the tumor nonimmune microenvironment (extracellular matrix (ECM), epithelial–mesenchymal transition (EMT) feature, and angiogenesis) has been overlooked with regard to brain metastasis. We evaluated tumor nonimmune-microenvironment-related gene expression signatures that could predict brain metastasis after the surgical resection of lung adenocarcinoma using a machine learning approach. We identified a tumor nonimmune-microenvironment-related 17-gene expression signature, and this signature showed high brain metastasis predictive power in four machine learning classifiers. The immunohistochemical expression of the top three genes of the 17-gene expression signature yielded similar results to NanoString tests. Our tumor nonimmune-microenvironment-related gene expression signatures are important biological markers that can predict brain metastasis and provide patient-specific treatment options. Abstract Using a machine learning approach with a gene expression profile, we discovered a tumor nonimmune-microenvironment-related gene expression signature, including extracellular matrix (ECM) remodeling, epithelial–mesenchymal transition (EMT), and angiogenesis, that could predict brain metastasis (BM) after the surgical resection of 64 lung adenocarcinomas (LUAD). Gene expression profiling identified a tumor nonimmune-microenvironment-related 17-gene expression signature that significantly correlated with BM. Of the 17 genes, 11 were ECM-remodeling-related genes. The 17-gene expression signature showed high BM predictive power in four machine learning classifiers (areas under the receiver operating characteristic curve = 0.845 for naïve Bayes, 0.849 for support vector machine, 0.858 for random forest, and 0.839 for neural network). Subgroup analysis revealed that the BM predictive power of the 17-gene signature was higher in the early-stage LUAD than in the late-stage LUAD. Pathway enrichment analysis showed that the upregulated differentially expressed genes were mainly enriched in the ECM–receptor interaction pathway. The immunohistochemical expression of the top three genes of the 17-gene expression signature yielded similar results to NanoString tests. The tumor nonimmune-microenvironment-related gene expression signatures found in this study are important biological markers that can predict BM and provide patient-specific treatment options.
Collapse
|
22
|
Udomsinprasert W, Angkathunyakul N, Jittikoon J, Chaikledkaew U, Vejchapipat P, Poovorawan Y, Honsawek S. Cartilage oligomeric matrix protein as a marker of progressive liver fibrosis in biliary atresia. Sci Rep 2021; 11:16695. [PMID: 34404836 PMCID: PMC8371124 DOI: 10.1038/s41598-021-95805-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/29/2021] [Indexed: 12/15/2022] Open
Abstract
This study aimed to determine whether mRNA and protein levels of cartilage oligomeric matrix protein (COMP), a glycoprotein responsible for modulating homeostasis of extracellular matrix, in the systemic and local liver environments were associated with clinical parameters of biliary atresia (BA) patients and might serve as a biomarker for BA severity. COMP protein levels in the circulation of 96 BA patients and 56 healthy controls and its mRNA and protein expressions in the liver of 20 BA patients and 5 non-BA patients were evaluated using enzyme-linked immunosorbent assay, real-time polymerase chain reaction, and immunohistochemistry, respectively. In the circulation of BA patients, COMP levels were significantly higher than those in healthy controls. Compared with early-stage BA patients, those with advanced-stage including jaundice, fibrosis, and hepatic dysfunction had significantly increased circulating COMP levels. Raised circulating COMP levels were found to be independently correlated with degree of liver fibrosis. Survival analysis showed that elevated circulating COMP levels were significantly associated with decreased survival of BA patients. Receiver-operating characteristic curve analysis unveiled a diagnostic value of circulating COMP as a non-invasive biomarker of BA (AUC = 0.99), with a sensitivity of 100.0% and a specificity of 98.2%. In the liver, both COMP mRNA and protein expressions of BA patients with fibrosis were significantly greater than those of BA patients without fibrosis and non-BA patients. Collectively, increased circulating COMP might reflect unfavorable outcome of BA patients and have potential as a novel biomarker for the disease severity following Kasai-operation.
Collapse
Affiliation(s)
- Wanvisa Udomsinprasert
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayudthaya Road, Rajathevi, Bangkok, 10400, Thailand.
| | - Napat Angkathunyakul
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Jiraphun Jittikoon
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayudthaya Road, Rajathevi, Bangkok, 10400, Thailand
| | - Usa Chaikledkaew
- Social and Administrative Pharmacy Division, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
- Mahidol University Health Technology Assessment (MUHTA) Graduate Program, Mahidol University, Bangkok, 10400, Thailand
| | - Paisarn Vejchapipat
- Department of Surgery, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Yong Poovorawan
- Department of Pediatrics, Faculty of Medicine, Center of Excellence in Clinical Virology, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sittisak Honsawek
- Osteoarthritis and Musculoskeleton Research Unit, Department of Biochemistry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
23
|
Wei D, Wu S, Liu J, Zhang X, Guan X, Gao L, Xu Z. Theobromine ameliorates nonalcoholic fatty liver disease by regulating hepatic lipid metabolism via mTOR signaling pathway in vivo and in vitro. Can J Physiol Pharmacol 2021; 99:775-785. [PMID: 33290156 DOI: 10.1139/cjpp-2020-0259] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Theobromine, a methylxanthine present in cocoa, has been shown to possess many beneficial pharmacological properties such as anti-oxidative stress, anti-inflammatory property, and anti-microbial activity. In this study, we investigated the effects of theobromine on nonalcoholic fatty liver disease (NAFLD) and the possible underlying mechanisms in vivo and in vitro. The results showed that theobromine reduced body weight and fat mass and improved dyslipidemia. Theobromine mitigated liver injury and significantly reduced hepatic triglyceride level in mice with obesity. Histological examinations also showed hepatic steatosis was alleviated after theobromine treatment. Furthermore, theobromine reversed the elevated mRNA and protein expression of SREBP-1c, FASN, CD36, FABP4, and the suppressed expression of PPARα and CPT1a in the liver of mice with obesity, which were responsible for lipogenesis, fatty acid uptake, and fatty acid oxidation respectively. In vitro, theobromine also downregulated SREBP-1c, FASN, CD36, FABP4 and upregulated PPARα and CPT1a mRNA and protein levels in hepatocytes in a dose-dependent manner, while these changes were reversed by L-leucine, a mammalian target of rapamycin (mTOR) agonist. The present study demonstrated that theobromine improved NAFLD by inhibiting lipogenesis and fatty acid uptake and promoting fatty acid oxidation in the liver and hepatocytes, which might be associated with its suppression of mTOR signaling pathway. Novelty: Theobromine protects against high-fat diet - induced NAFLD. Theobromine inhibits lipogenesis and fatty acid uptake and promotes fatty acid oxidation in the liver and hepatocytes via inhibiting mTOR signaling pathway.
Collapse
Affiliation(s)
- Dan Wei
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Shaofei Wu
- Department of Hepatology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Jie Liu
- Department of Public Health, Tengzhou Central People's Hospital, Zaozhuang, Shandong, China
| | - Xiaoqian Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoling Guan
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Li Gao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Zhipeng Xu
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
24
|
Serum Cartilage Oligomeric Matrix Protein and Golgi Protein-73: New Diagnostic and Predictive Tools for Liver Fibrosis and Hepatocellular Cancer? Cancers (Basel) 2021; 13:cancers13143510. [PMID: 34298722 PMCID: PMC8304371 DOI: 10.3390/cancers13143510] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/07/2021] [Accepted: 07/10/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) ranks as the sixth most common malignancy and represents the fourth leading cause of cancer-related deaths. However, most HCC cases are insidious in the early stages leading to a delay in diagnosis with limited treatment options. In patients with chronic liver diseases, advanced liver fibrosis and cirrhosis are the leading risk factors for the development of HCC. Cartilage oligomeric matrix protein (COMP) and Golgi protein-73 (GP73) are two biomarkers that have been associated with the progression of chronic liver disease, including inflammation, fibrosis, and HCC. The aim of our study was to assess the performance of the combination of these biomarkers. We confirmed, in a large cohort of 288 patients with chronic liver diseases, that the combination of GP73 and COMP had a high discriminative ability to detect severe fibrosis/cirrhosis and is efficient in predicting the development of HCC. Abstract The cartilage oligomeric matrix protein (COMP) and Golgi-protein-73 (GP73) have been proposed as markers of liver fibrosis and hepatocellular carcinoma (HCC). Our aim was to assess the performance of the combination of these markers in diagnosing cirrhosis and predicting HCC development. Sera from 288 consecutive patients with chronic liver diseases were investigated by using COMP and GP73-ELISAs. Dual positivity for COMP (>15 U/L) and GP73 (>20 units) was observed in 24 (8.3%) patients, while 30 (10.4%) were GP73(+)/COMP(−), 37/288 (12.8%) GP73(−)/COMP(+), and 197 (68.5%) GP73(−)/COMP(−). Positivity for both markers was associated with cirrhosis [23/24 (95.8%) for GP73(+)/COMP(+) vs. 22/30 (73.3%) for GP73(+)/COMP(−) vs. 25/37 (67.6%) for GP73(−)/COMP(+) vs. 46/197 (23.4%) for GP73(−)/COMP(−); P < 0.001]. The combination of GP73, COMP, the aspartate aminotransferase/platelets ratio index, and the Fibrosis-4 score had even higher diagnostic accuracy to detect the presence of cirrhosis [AUC (95% CI): 0.916 (0.878–0.946)] or significant liver fibrosis (METAVIR ≥ F2) [AUC (95% CI): 0.832 (0.768–0.883)] than each marker alone. Kaplan-Meier analysis showed that positivity for both GP73 and COMP was associated with higher rates of HCC development (P < 0.001) and liver-related deaths (P < 0.001) during follow-up. In conclusion, the combination of GP73 and COMP seems efficient to detect cirrhosis and predict worse outcomes and the development of HCC in patients with chronic liver diseases.
Collapse
|
25
|
Liver fibrosis promotes immune escape in hepatocellular carcinoma via GOLM1-mediated PD-L1 upregulation. Cancer Lett 2021; 513:14-25. [PMID: 33992711 DOI: 10.1016/j.canlet.2021.05.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/25/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023]
Abstract
Immune checkpoint blockade is considered a breakthrough in cancer treatment. However, with the low response rates and therapeutic resistance of patients with hepatocellular carcinoma (HCC), the challenges facing the application of this treatment are tremendous. Liver fibrosis is a key driver of tumor immune escape, the underlying mechanism has never been clarified. This study sought to explore the role of liver fibrosis in regulating tumor-infiltrating lymphocytes (TILs) and inducing tumor immunosuppression. Ninety-nine fixed HCC tissue samples were used to analyze the association between liver fibrosis and immune escape using immunohistochemistry. In HCC patients, low FIB-4 values and high CD8+ T cell infiltration were correlated with prolonged survival. Elevated expression of immune checkpoints and attenuated antitumor immunity were observed in CCl4-induced mice liver fibrosis models and human fibrotic livers compared to control group. GOLM1 levels were increased in livers of patients with fibrosis and mice in response to CCl4-induced liver fibrosis. CD8+ T cell infiltrations were significantly decreased and PD-L1 expression was significantly increased in tumor tissues from hepatocyte-specific GOLM1 transgenic mice (Alb/GOLM1 mice) inducing chemical carcinogenesis compared to their corresponding control WT mice. GOLM1 induced PD-L1 expression via EGFR pathway activation. EGFR inhibitors, especially together with anti-PD-L1 therapy, improved the efficacy of immunotherapy in HCC. These findings illustrate the importance of liver fibrosis-induced immunosuppression as a tumor-promoting mechanism. GOLM1, which is highly upregulated in the fibrotic liver, regulates tumor microenvironmental immune escape via the EGFR/PD-L1 signaling pathway. EGFR blockade may bolster the efficacy of immune checkpoint inhibitors for HCC treatment.
Collapse
|
26
|
Gonzalez E, Azkargorta M, Garcia-Vallicrosa C, Prieto-Elordui J, Elortza F, Blanco-Sampascual S, Falcon-Perez JM. Could protein content of Urinary Extracellular Vesicles be useful to detect Cirrhosis in Alcoholic Liver Disease? Int J Biol Sci 2021; 17:1864-1877. [PMID: 34131392 PMCID: PMC8193259 DOI: 10.7150/ijbs.59725] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/10/2021] [Indexed: 12/13/2022] Open
Abstract
Alcohol abuse has a high impact on the mortality and morbidity related to a great number of diseases and is responsible for the development of alcoholic liver disease (ALD). It remains challenging to detect and evaluate its severity, which is crucial for prognosis. In this work, we studied if urinary EVs (uEVs) could serve in diagnose and evaluate cirrhosis in ALD. To this purpose, uEVs characterization by cryo-electron microscopy (Cryo-EM), Nanoparticle Tracking Analysis (NTA) and Western blotting (WB) was performed in a cohort of 21 controls and 21 cirrhotic patients. Then, proteomics of uEVs was carried out in a second cohort of 6 controls and 8 patients in order to identify new putative biomarkers for cirrhosis in ALD. Interestingly, uEVs concentration, size and protein composition were altered in cirrhotic patients. From a total of 1304 proteins identified in uEVs, 90 of them were found to be altered in cirrhotic patients. The results suggest that uEVs could be considered as a tool and a supplier of new biomarkers for cirrhosis in ALD, whose application would be especially relevant in chronic patients. Yet, further research is necessary to obtain more relevant result in clinical terms.
Collapse
Affiliation(s)
- Esperanza Gonzalez
- Exosomes Laboratory. Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Mikel Azkargorta
- Proteomics Platform. Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Clara Garcia-Vallicrosa
- Exosomes Laboratory. Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | | | - Felix Elortza
- Proteomics Platform. Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | | | - Juan Manuel Falcon-Perez
- Exosomes Laboratory. Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd), Madrid, Spain
- IKERBASQUE Basque Foundation for Science Bilbao Spain
| |
Collapse
|
27
|
COMP and TSP-4: Functional Roles in Articular Cartilage and Relevance in Osteoarthritis. Int J Mol Sci 2021; 22:ijms22052242. [PMID: 33668140 PMCID: PMC7956748 DOI: 10.3390/ijms22052242] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/16/2021] [Accepted: 02/20/2021] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) is a slow-progressing joint disease, leading to the degradation and remodeling of the cartilage extracellular matrix (ECM). The usually quiescent chondrocytes become reactivated and accumulate in cell clusters, become hypertrophic, and intensively produce not only degrading enzymes, but also ECM proteins, like the cartilage oligomeric matrix protein (COMP) and thrombospondin-4 (TSP-4). To date, the functional roles of these newly synthesized proteins in articular cartilage are still elusive. Therefore, we analyzed the involvement of both proteins in OA specific processes in in vitro studies, using porcine chondrocytes, isolated from femoral condyles. The effect of COMP and TSP-4 on chondrocyte migration was investigated in transwell assays and their potential to modulate the chondrocyte phenotype, protein synthesis and matrix formation by immunofluorescence staining and immunoblot. Our results demonstrate that COMP could attract chondrocytes and may contribute to a repopulation of damaged cartilage areas, while TSP-4 did not affect this process. In contrast, both proteins similarly promoted the synthesis and matrix formation of collagen II, IX, XII and proteoglycans, but inhibited that of collagen I and X, resulting in a stabilized chondrocyte phenotype. These data suggest that COMP and TSP-4 activate mechanisms to protect and repair the ECM in articular cartilage.
Collapse
|
28
|
Shi Y, Sun L, Zhang R, Hu Y, Wu Y, Dong X, Dong D, Chen C, Geng Z, Li E, Fan Y. Thrombospondin 4/integrin α2/HSF1 axis promotes proliferation and cancer stem-like traits of gallbladder cancer by enhancing reciprocal crosstalk between cancer-associated fibroblasts and tumor cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:14. [PMID: 33407730 PMCID: PMC7789630 DOI: 10.1186/s13046-020-01812-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/11/2020] [Indexed: 12/11/2022]
Abstract
Background Cancer-associated fibroblasts (CAFs), the primary component of tumor stroma in tumor microenvironments, are well-known contributors to the malignant progression of gallbladder cancer (GBC). Thrombospondins (THBSs or TSPs) comprise a family of five adhesive glycoproteins that are overexpressed in many types of cancers. However, the expression and potential roles of TSPs in the crosstalk between CAFs and GBC cells has remained unclear. Methods Peritumoral fibroblasts (PTFs) and CAFs were extracted from GBC tissues. Thrombospondin expression in GBC was screened by RT-qPCR. MTT viability assay, colony formation, EdU incorporation assay, flow cytometry analysis, Transwell assay, tumorsphere formation and western blot assays were performed to investigate the effects of CAF-derived TSP-4 on GBC cell proliferation, EMT and cancer stem-like features. Subcutaneous tumor formation models were established by co-implanting CAFs and GBC cells or GBC cells overexpressing heat shock factor 1 (HSF1) to evaluate the roles of TSP-4 and HSF1 in vivo. To characterize the mechanism by which TSP-4 is involved in the crosstalk between CAFs and GBC cells, the levels of a variety of signaling molecules were detected by coimmunoprecipitation, immunofluorescence staining, and ELISA assays. Results In the present study, we showed that TSP-4, as the stromal glycoprotein, is highly expressed in CAFs from GBC and that CAF-derived TSP-4 induces the proliferation, EMT and cancer stem-like features of GBC cells. Mechanistically, CAF-secreted TSP-4 binds to the transmembrane receptor integrin α2 on GBC cells to induce the phosphorylation of HSF1 at S326 and maintain the malignant phenotypes of GBC cells. Moreover, the TSP-4/integrin α2 axis-induced phosphorylation of HSF1 at S326 is mediated by Akt activation (p-Akt at S473) in GBC cells. In addition, activated HSF1 signaling increased the expression and paracrine signaling of TGF-β1 to induce the transdifferentiation of PTFs into CAFs, leading to their recruitment into GBC and increased TSP-4 expression in CAFs, thereby forming a positive feedback loop to drive the malignant progression of GBC. Conclusions Our data indicate that a complex TSP-4/integrin α2/HSF1/TGF-β cascade mediates reciprocal interactions between GBC cells and CAFs, providing a promising therapeutic target for gallbladder cancer patients.
Collapse
Affiliation(s)
- Yu Shi
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Liankang Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Rui Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Yuan Hu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Yinying Wu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Xuyuan Dong
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Danfeng Dong
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Chen Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Zhimin Geng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Enxiao Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China.
| | - Yangwei Fan
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China.
| |
Collapse
|
29
|
Tian T, Xie R, Ding K, Han B, Yang Q, Yang X. IOX1 protects from TGF-β induced fibrosis in LX-2 cells via the regulation of extracellular matrix protein expression. Exp Ther Med 2021; 21:180. [PMID: 33488789 PMCID: PMC7812578 DOI: 10.3892/etm.2021.9611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/17/2020] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the effect of the histone H3K9 demethylase inhibitor, IOX1, on the mechanism of hepatic fibrosis in TGF-β-induced human hepatic stellate LX-2 cells. Cellular proliferation, apoptosis, histone H3K9 dimethylation (H3K9me2), protein expression of extracellular matrix (ECM)-related proteins α-smooth muscle actin (SMA), type I collagen (Col I), MMP-1 and TIMP-1 were measured. H3K9me2 levels in the promoter region of ECM-related genes were detected by real-time cell analysis (RTCA), flow cytometry, western blotting and chromatin immunoprecipitation (ChIP) in LX-2 cells. IOX1 significantly inhibited cell proliferation and the IC50 of IOX1 was 100 µM in cells treated with IOX1 for 48 h. IOX1 significantly induced apoptosis in LX-2 cells in a concentration-dependent manner. In addition, different concentration of IOX1 increased the level of H3K9me2 and downregulated the expression of α-SMA, Col I, MMP-1 and TIMP-1 in TGF-β-induced LX-2 cells. ChIP measurements indicated that H3K9me2 levels in the promotor region of the corresponding genes were increased in TGF-β-induced LX-2 cells. IOX1 may elevate H3K9me2 in the promotor region of Col I, MMP-1, and TIMP-1 genes to regulate α-SMA, Col I, MMP-1 and TIMP-1 protein expression to induce cell apoptosis, inhibit LX-2 cell proliferation and oppose hepatic fibrotic activity.
Collapse
Affiliation(s)
- Tian Tian
- Department of Eugenic Genetics, Guiyang Maternal and Child Health Care Hospital, Guiyang, Guizhou 550003, P.R. China
| | - Rujia Xie
- Department of Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Kaize Ding
- Department of Assisted Reproduction, Guiyang Maternal and Child Health Care Hospital, Guiyang, Guizhou 550003, P.R. China
| | - Bing Han
- Department of Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Qin Yang
- Department of Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Xue Yang
- Department of Eugenic Genetics, Guiyang Maternal and Child Health Care Hospital, Guiyang, Guizhou 550003, P.R. China
| |
Collapse
|
30
|
Wang L, Sun Y, Luo X, Han H, Yin H, Zhao B, Chen X, Yu Q, Qiu H, Yuan X. Prophylactical Low Dose Whole-Liver Irradiation Inhibited Colorectal Liver Metastasis by Regulating Hepatic Niche in Mice. Onco Targets Ther 2020; 13:8451-8462. [PMID: 32922035 PMCID: PMC7455754 DOI: 10.2147/ott.s263858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/03/2020] [Indexed: 12/23/2022] Open
Abstract
Background The liver is the most common target for metastatic colorectal cancer. Changes of the local hepatic niche due to hepatic diseases such as cirrhosis decrease the incidence of colorectal cancer liver metastasis. Hepatic niche heterogeneity could influence the risk of hepatic metastasis. Materials and Methods We simulated changes of the hepatic niche via prophylactical liver irradiation with a safe dose of 6 Gy. GEO dataset and GO analysis revealed a difference in the expression of matrix metalloproteinase 1 (MMP1) in primary colorectal cancer versus liver metastasis, as well as synchronous versus metachronous liver metastasis. Western blotting, Immunofluorescence and qRT-PCR were conducted to measure protein expressions, location and RNA expressions. Colony formation, wound-healing, transwell assays experiments were performed to determine the malignant biological properties of colorectal cancer cells. shRNA transfection was used to conduct stable transfected cell lines. Results Tissue inhibitor of metalloproteinases 1 (TIMP1) expression was significantly higher in metastases lesions than primary tumors. In vivo, TIMP1 expression in the hepatic niche increased after a safe dose of 6 Gy irradiation, along with MMP1 decreased, leading to collagen fiber deposition and impairment of hepatic microcirculation. In vitro, irradiated hepatic stellate cells-conditioned media reduced the migration and clone formation ability of colon cancer cells SW480 and HCT116. Low TIMP1 expression in hepatic stellate cells reduced tumor cell invasion and migration. Conclusion Prophylactical 6 Gy whole-liver irradiation could inhibit colorectal cancer liver metastasis by regulating TIMP1/MMP1 balance in the hepatic niche before liver metastatic lesion formed.
Collapse
Affiliation(s)
- Lu Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yinan Sun
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Hu Han
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Han Yin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ben Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Xinyi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Qianqian Yu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Hong Qiu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
31
|
Yu Y, Jiang L, Wang H, Shen Z, Cheng Q, Zhang P, Wang J, Wu Q, Fang X, Duan L, Wang S, Wang K, An P, Shao T, Chung RT, Zheng S, Min J, Wang F. Hepatic transferrin plays a role in systemic iron homeostasis and liver ferroptosis. Blood 2020; 136:726-739. [PMID: 32374849 PMCID: PMC7414596 DOI: 10.1182/blood.2019002907] [Citation(s) in RCA: 399] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 04/19/2020] [Indexed: 02/08/2023] Open
Abstract
Although the serum-abundant metal-binding protein transferrin (encoded by the Trf gene) is synthesized primarily in the liver, its function in the liver is largely unknown. Here, we generated hepatocyte-specific Trf knockout mice (Trf-LKO), which are viable and fertile but have impaired erythropoiesis and altered iron metabolism. Moreover, feeding Trf-LKO mice a high-iron diet increased their susceptibility to developing ferroptosis-induced liver fibrosis. Importantly, we found that treating Trf-LKO mice with the ferroptosis inhibitor ferrostatin-1 potently rescued liver fibrosis induced by either high dietary iron or carbon tetrachloride (CCl4) injections. In addition, deleting hepatic Slc39a14 expression in Trf-LKO mice significantly reduced hepatic iron accumulation, thereby reducing ferroptosis-mediated liver fibrosis induced by either a high-iron diet or CCl4 injections. Finally, we found that patients with liver cirrhosis have significantly lower levels of serum transferrin and hepatic transferrin, as well as higher levels of hepatic iron and lipid peroxidation, compared with healthy control subjects. Taken together, these data indicate that hepatic transferrin plays a protective role in maintaining liver function, providing a possible therapeutic target for preventing ferroptosis-induced liver fibrosis.
Collapse
Affiliation(s)
- Yingying Yu
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- Precision Nutrition Innovation Center, Department of Nutrition, School of Public Health, Zhengzhou University, Zhengzhou, China; and
| | - Li Jiang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Wang
- Precision Nutrition Innovation Center, Department of Nutrition, School of Public Health, Zhengzhou University, Zhengzhou, China; and
| | - Zhe Shen
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Cheng
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Pan Zhang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaming Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Wu
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuexian Fang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingyan Duan
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Shufen Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng An
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Tuo Shao
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Raymond T Chung
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Shusen Zheng
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Junxia Min
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- Precision Nutrition Innovation Center, Department of Nutrition, School of Public Health, Zhengzhou University, Zhengzhou, China; and
| |
Collapse
|
32
|
Su S, Tian H, Jia X, Zhu X, Wu J, Zhang Y, Chen Y, Li Z, Zhou Y. Mechanistic insights into the effects of SREBP1c on hepatic stellate cell and liver fibrosis. J Cell Mol Med 2020; 24:10063-10074. [PMID: 32678475 PMCID: PMC7520338 DOI: 10.1111/jcmm.15614] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 01/19/2023] Open
Abstract
Sterol regulatory element-binding protein 1c (SREBP1c) plays key roles in maintenance of hepatic stellate cell (HSC) quiescence. The present researches investigated the mechanisms underlying the effects of SREBP1c on HSCs and liver fibrogenesis by HSC-targeted overexpression of the active SREBP1c using adenovirus in vitro and in vivo. Results demonstrated that SREBP1c exerted inhibitory effects on TAA-induced liver fibrosis. SREBP1c down-regulated TGFβ1 level in liver, reduced the receptors for TGFβ1 and PDGFβ, and interrupted the signalling pathways of Smad3 and Akt1/2/3 but not ERK1/2 in HSCs. SREBP1c also led to the decreases in the protein levels of the bromodomain-containing chromatin-modifying factor bromodomain protein 4, methionine adenosyltransferase 2B (MAT2B) and TIMP1 in HSCs. In vivo activated HSCs did not express cyclin D1 and cyclin E1 but SREBP1c down-regulated both cyclins in vitro. SREBP1c elevated PPARγ and MMP1 protein levels in the model of liver fibrosis. The effect of SREBP1c on MAT2B expression was associated with its binding to MAT2B1 promoter. Taken together, the mechanisms underlying the effects of SREBP1c on HSC activation and liver fibrosis were involved in its influences on TGFβ1 level, the receptors for TGFβ1 and PDGFβ and their downstream signalling, and the molecules for epigenetic regulation of genes.
Collapse
Affiliation(s)
- Shengyan Su
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Nantong, China
| | - Haimeng Tian
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Nantong, China
| | - Xin Jia
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Nantong, China
| | - Xiaofei Zhu
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Nantong, China
| | - Juanjuan Wu
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Nantong, China
| | - Yali Zhang
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Nantong, China
| | - Yuanyuan Chen
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Nantong, China
| | - Ziqiang Li
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Nantong, China
| | - Yajun Zhou
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Nantong, China.,Key Laboratory of Microenvironment and Translational Cancer Research, Medical College, Nantong University, Nantong, China
| |
Collapse
|
33
|
Metabolism of N-nitrosodimethylamine, methylation of macromolecules, and development of hepatic fibrosis in rodent models. J Mol Med (Berl) 2020; 98:1203-1213. [PMID: 32666246 DOI: 10.1007/s00109-020-01950-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/04/2020] [Accepted: 07/09/2020] [Indexed: 12/15/2022]
Abstract
Hepatic fibrosis and cirrhosis are chronic diseases affecting liver and a major health problem throughout the world. The hallmark of fibrosis and cirrhosis is inordinate synthesis and deposition of fibril forming collagens in the extracellular matrix of the liver leading to nodule formation and loss of normal architecture. Hepatic stellate cells play a crucial role in the pathogenesis and progression of liver fibrosis through secretion of several potent fibrogenic factors that trigger hepatocytes, portal fibrocytes, and bone marrow-derived fibroblasts to synthesize and deposit several connective tissue proteins, especially collagens between hepatocytes and space of Disse. Regulation of various events involved in the activation and transformation of hepatic stellate cells seems to be an appropriate strategy for the arrest of hepatic fibrosis and liver cirrhosis. In order to unravel the molecular mechanisms involved in the pathogenesis and progression of hepatic fibrosis, to determine proper and potent targets to arrest fibrosis, and to discover powerful therapeutic agents, a quick and reproducible animal model of hepatic fibrosis and liver cirrhosis that display all decompensating features of human condition is required. This review thoroughly evaluates the biochemical, histological, and pathological features of N-nitrosodimethylamine-induced model of liver injury, hepatic fibrosis, and early cirrhosis in rodents.
Collapse
|
34
|
Park S, Ranjbarvaziri S, Zhao P, Ardehali R. Cardiac Fibrosis Is Associated With Decreased Circulating Levels of Full-Length CILP in Heart Failure. ACTA ACUST UNITED AC 2020; 5:432-443. [PMID: 32478206 PMCID: PMC7251193 DOI: 10.1016/j.jacbts.2020.01.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 01/09/2023]
Abstract
After in vitro stimulation or in vivo pressure overload injury, activated cardiac fibroblasts express Ltbp2, Comp, and Cilp. In ischemic heart disease, LTBP2, COMP, and CILP localize to the fibrotic regions of the injured heart. Circulating levels of full-length CILP are decreased in patients with heart failure, suggestive of the potential to use this protein as a biomarker for the presence of cardiac fibrosis.
Cardiac fibrosis is a pathological process associated with various forms of heart failure. This study identified latent transforming growth factor-β binding protein 2, cartilage oligomeric matrix protein, and cartilage intermediate layer protein 1 as potential biomarkers for cardiac fibrosis. All 3 encoded proteins showed increased expression in fibroblasts after transforming growth factor-β stimulation in vitro and localized specifically to fibrotic regions in vivo. Of the 3, only the full-length cartilage intermediate layer protein 1 showed a significant decrease in circulating levels in patients with heart failure compared with healthy volunteers. Further studies on these 3 proteins will lead to a better understanding of their biomarker potential for cardiac fibrosis.
Collapse
Key Words
- CFB, cardiac fibroblast
- CILP, cartilage intermediate layer protein 1
- COMP, cartilage oligomeric matrix protein
- ECM, extracellular matrix
- ELISA, enzyme-linked immunosorbent assay
- Ltbp2, latent transforming growth factor-β binding protein 2
- PCR, polymerase chain reaction
- RNA, ribonucleic acid
- TAC, transverse aortic constriction
- TGF, transforming growth factor
- biomarker
- cardiac fibrosis
- extracellular matrix protein
- heart failure
Collapse
Affiliation(s)
- Shuin Park
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, California.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles (UCLA), Los Angeles, California.,Molecular, Cellular, and Integrative Physiology Graduate Program, University of California, Los Angeles (UCLA), Los Angeles, California
| | - Sara Ranjbarvaziri
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, California.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles (UCLA), Los Angeles, California.,Molecular, Cellular, and Integrative Physiology Graduate Program, University of California, Los Angeles (UCLA), Los Angeles, California
| | - Peng Zhao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, California
| | - Reza Ardehali
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, California.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles (UCLA), Los Angeles, California.,Molecular, Cellular, and Integrative Physiology Graduate Program, University of California, Los Angeles (UCLA), Los Angeles, California.,Molecular Biology Institute, UCLA, Los Angeles, California
| |
Collapse
|
35
|
Papadakos KS, Darlix A, Jacot W, Blom AM. High Levels of Cartilage Oligomeric Matrix Protein in the Serum of Breast Cancer Patients Can Serve as an Independent Prognostic Marker. Front Oncol 2019; 9:1141. [PMID: 31737569 PMCID: PMC6831625 DOI: 10.3389/fonc.2019.01141] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022] Open
Abstract
Background: Cartilage oligomeric matrix protein (COMP) is a pentameric cartilage protein also expressed in breast cancer tumors. A high expression of COMP evaluated by immunohistochemical staining is as an independent prognostic marker associated with poor patients' prognosis. Methods: Herein, levels of COMP were analyzed using an IVD approved ELISA in serum samples from 233 well-characterized breast cancer patients; 176 with metastatic breast cancer; and 57 in an early stage of the disease. Results: The metastatic patients had double the concentration of serum COMP compared with those with early breast cancer. High levels of COMP in sera of metastatic patients were associated with the histological subtype (p = 0.025) and estrogen receptor positivity (p = 0.019) at the time of breast cancer diagnosis. Further, correlation was observed between the serum levels of COMP and the presence of liver (p = 0.010) or bone (p = 0.010) metastases in this population. Most importantly, elevated serum levels of COMP appear to serve as an independent prognostic marker of survival as assessed by Cox proportional hazard regression analysis (p = 0.001) for the metastatic patients. Among metastatic patients treated with taxanes (Docetaxel-Paclitaxel) as part of their first metastatic line (n = 25), those with high levels of serum COMP detected in the metastatic stage of the disease had a shorter median survival (0.2 years) compared with those with low levels of serum COMP (1.1 years) (p = 0.001). Conclusions: Taken together, the serum levels of COMP are elevated in the metastatic patients and may be a potential novel biomarker for the evaluation of the prognosis in this population.
Collapse
Affiliation(s)
- Konstantinos S Papadakos
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Amélie Darlix
- Department of Medical Oncology, Institut Régional du Cancer Montpellier ICM, University of Montpellier, Montpellier, France
| | - William Jacot
- Department of Medical Oncology, Institut Régional du Cancer Montpellier ICM, University of Montpellier, Montpellier, France
| | - Anna M Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
36
|
Locostatin Alleviates Liver Fibrosis Induced by Carbon Tetrachloride in Mice. Dig Dis Sci 2019; 64:2570-2580. [PMID: 30874989 DOI: 10.1007/s10620-019-05588-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 03/08/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Liver fibrosis is featured with excessive deposition of extracellular matrix and fibrous connective tissue hyperplasia. The specific inhibitor of Raf-1 kinase inhibitor protein, locostatin, inhibits the migration of hepatic stellate cells. In this study, we investigated the effect of locostatin on liver fibrosis and its underlying mechanism. METHODS Carbon tetrachloride (CCl4) was used to induce liver fibrosis in mice, and locostatin was injected intraperitoneally. Liver fibrosis was assessed by Masson and Sirius red staining, hydroxyproline (HYP) assay, and collagen percentage area. Collagen I, collagen III, and α-SMA were detected by RT-PCR and western blot. The levels of MMP-13, MMP-2, TIMP-1, and TIMP-2 were estimated by ELISA. Liver inflammation was evaluated by HE staining and immunohistochemistry; liver myeloperoxidase (MPO), superoxide dismutase, and malondialdehyde were measured by ELISA; and cytokines were by Mouse Cytokine Array Q4000. RESULTS Compared to the CCl4 group, HYP (208.56 ± 6.12) µg/g, percentage of total collagen at overall region (1.91 ± 0.13), MMP-13/TIMP-1 (0.19 ± 0.01), MPO (1.45 ± 0.04) U/g, TGF-β (2652 ± 91.20), PDGF-AA (3897 ± 290.69), and E-selectin (1569 ± 66.48) in the liver tissues were decreased significantly in the locostatin-treated group. CONCLUSIONS Locostatin mitigated liver fibrosis and inflammation induced by CCl4. The mechanism is via inhibition inflammatory cytokines, TGF-β, PDGF-AA, and E-selectin.
Collapse
|
37
|
Hepatoprotection of yangonin against hepatic fibrosis in mice via farnesoid X receptor activation. Int Immunopharmacol 2019; 75:105833. [PMID: 31450152 DOI: 10.1016/j.intimp.2019.105833] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/06/2019] [Accepted: 08/16/2019] [Indexed: 01/05/2023]
Abstract
Hepatic fibrosis is a reversible would-healing response following chronic liver injury of different aetiologies and represents a major worldwide health problem. Up to date, there is no satisfactory drugs treated for liver fibrosis. The present study was to investigate hepatoprotection of yangonin against liver fibrosis induced by thioacetamide (TAA) in mice and further to clarify the involvement of farnesoid X receptor (FXR) in vivo and in vitro. Yangonin treatment remarkably ameliorated TAA-induced liver injury by reducing relative liver weight, as well as serum ALT and AST activities. Moreover, yangonin alleviated TAA-induced accumulation of bile acids through increasing the expression of bile acid efflux transporters such as Bsep and Mrp2, and reducing hepatic uptake transporter Ntcp expression, all of these are FXR-target genes. The liver sections stained by H&E indicated that the histopathological change induced by TAA was improved by yangonin. Masson and Sirius red staining indicated the obvious anti-fibrotic effect of yangonin. The mechanism of anti-fibrotic effect of yangonin was that yangonin reduced collagen content by regulating the genes involved in hepatic fibrosis including COL1-α1 and TIMP-1. Besides, yangonin inhibited hepatic stellate cell activation by reducing TGF-β1 and α-SMA expression. In addition, yangonin protected against TAA-induced hepatic inflammation via its inhibition of NF-κB and TNF-α. These hepatoprotective effects of yangonin were abrogated by guggulsterone which is a FXR antagonist. In vitro experiment further demonstrated dose-dependent activation of FXR by yangonin using dual-luciferase reporter assay. In summary, yangonin produces hepatoprotection against TAA-induced liver fibrosis via FXR activation.
Collapse
|
38
|
Ballengee CR, Stidham RW, Liu C, Kim MO, Prince J, Mondal K, Baldassano R, Dubinsky M, Markowitz J, Leleiko N, Hyams J, Denson L, Kugathasan S. Association Between Plasma Level of Collagen Type III Alpha 1 Chain and Development of Strictures in Pediatric Patients With Crohn's Disease. Clin Gastroenterol Hepatol 2019; 17:1799-1806. [PMID: 30213581 PMCID: PMC6531351 DOI: 10.1016/j.cgh.2018.09.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/04/2018] [Accepted: 09/04/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS There are few serum biomarkers to identify patients with Crohn's disease (CD) who are at risk for stricture development. The extracellular matrix components, collagen type III alpha 1 chain (COL3A1) and cartilage oligomeric matrix protein (COMP), could contribute to intestinal fibrosis. We investigated whether children with inflammatory CD (B1) who later develop strictures (B2) have increased plasma levels of COL3A1 or COMP at diagnosis, compared with children who remain B1. We compared results with previously studied biomarkers, including autoantibodies against colony-stimulating factor 2 (CSF2). METHODS We selected 161 subjects (mean age, 12.2 y; 62% male) from the Risk Stratification and Identification of Immunogenic and Microbial Markers of Rapid Disease Progression in Children with Crohn's cohort, completed at 28 sites in the United States and Canada from 2008 through 2012. The children underwent colonoscopy and upper endoscopy at diagnosis and were followed up every 6 months for 36 months; plasma samples were collected at baseline. Based on CD phenotype, children were separated to group 1 (B1 phenotype at diagnosis and follow-up evaluation), group 2 (B2 phenotype at diagnosis), or group 3 (B1 phenotype at diagnosis who developed strictures during follow-up evaluation). Plasma samples were collected from patients and 40 children without inflammatory bowel disease (controls) at baseline and analyzed by enzyme-linked immunosorbent assay to measure COL3A1 and COMP. These results were compared with those from a previous biomarker study. The Kruskal-Wallis test and the pairwise Dunn test with Bonferroni correction were used to compare differences among groups. RESULTS The median baseline concentration of COL3A1 was significantly higher in plasma from group 3 vs group 1 (P < .01) and controls (P = .01). Median baseline plasma concentrations of COMP did not differ significantly among groups. A model comprising baseline concentrations of COL3A1 and anti-CSF2 identified patients with B2 vs B1 CD with an area under the curve of 0.80 (95% CI, 0.71-0.89); the combined concentration identified patients with strictures with a sensitivity value of 0.70 (95% CI, 0.55-0.83) and a specificity value of 0.83 (95% CI, 0.67-0.93). CONCLUSIONS We found median plasma concentrations of COL3A1, measured by enzyme-linked immunosorbent assay at diagnosis, to be significantly higher in patients with CD who later developed strictures than in patients without strictures. The combination of concentrations of COL3A1 and anti-CSF2 might be used to identify pediatric patients at CD diagnosis who are at risk for future strictures. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT00790543.
Collapse
Affiliation(s)
| | | | - Chunyan Liu
- Cincinnati Children’s Hospital Medical Center
| | - Mi-Ok Kim
- University of California San Francisco
| | | | | | | | | | | | | | | | - Lee Denson
- Cincinnati Children’s Hospital Medical Center
| | - Subra Kugathasan
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, Georgia.
| |
Collapse
|
39
|
Sun L, Wang Y, Wang L, Yao B, Chen T, Li Q, Liu Z, Liu R, Niu Y, Song T, Liu Q, Tu K. Resolvin D1 prevents epithelial-mesenchymal transition and reduces the stemness features of hepatocellular carcinoma by inhibiting paracrine of cancer-associated fibroblast-derived COMP. J Exp Clin Cancer Res 2019; 38:170. [PMID: 30999932 PMCID: PMC6472102 DOI: 10.1186/s13046-019-1163-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) require stromal signals for maintaining pluripotency and self-renewal capacities to confer tumor metastasis. Resolvin D1 (RvD1), an endogenous anti-inflammatory lipid mediator, has recently been identified to display anti-cancer effects by acting on stroma cells. Our previous study reveals that hepatic stellate cells (HSCs)-derived cartilage oligomeric matrix protein (COMP) contributes to hepatocellular carcinoma (HCC) progression. However, whether RvD1 inhibits paracrine of cancer-associated fibroblasts (CAFs)-derived COMP to prevent epithelial-mesenchymal transition (EMT) and cancer stemness in HCC remains to be elucidated. METHODS CAFs were isolated from HCC tissues. Direct and indirect co-culture models were established to analyze the interactions between HCC cells and CAFs in the presence of RvD1 in vitro. The transwell and tumor sphere formation assays were used to determine invasion and stemness of HCC cells. The subcutaneous tumor formation and orthotopic liver tumor models were established by co-implantation of CAFs and HCC cells to evaluate the role of RvD1 in vivo. To characterize the mechanism of RvD1 inhibited paracrine of COMP in CAFs, various signaling molecules were analyzed by ELISA, western blotting, reactive oxygen species (ROS) detection, immunofluorescence staining, dual luciferase reporter assay and chromatin immunoprecipitation assay. RESULTS Our data revealed that RvD1 treatment can impede the CAFs-induced cancer stem-like properties and the EMT of HCC cells under co-culture conditions. In vivo studies indicated that RvD1 intervention repressed the promoting effects of CAFs on tumor growth and metastasis of HCC. Furthermore, RvD1 inhibited CAF-induced EMT and stemness features of HCC cells by suppressing the secretion of COMP. Mechanistically, formyl peptide receptor 2 (FPR2) receptor mediated the suppressive effects of RvD1 on COMP and forkhead box M1 (FOXM1) expression in CAFs. Notably, RvD1 impaired CAF-derived COMP in a paracrine manner by targeting FPR2/ROS/FOXM1 signaling to ultimately abrogate FOXM1 recruitment to the COMP promoter. CONCLUSION Our results indicated that RvD1 impaired paracrine of CAFs-derived COMP by targeting FPR2/ROS/FOXM1 signaling to repress EMT and cancer stemness in HCC. Thus, RvD1 may be a potential agent to promote treatment outcomes in HCC.
Collapse
Affiliation(s)
- Liankang Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi Province 710061 China
| | - Yufeng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi Province 710061 China
| | - Liang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi Province 710061 China
| | - Bowen Yao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi Province 710061 China
| | - Tianxiang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi Province 710061 China
| | - Qing Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi Province 710061 China
| | - Zhikui Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi Province 710061 China
| | - Runkun Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi Province 710061 China
| | - Yongshen Niu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi Province 710061 China
| | - Tao Song
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi Province 710061 China
| | - Qingguang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi Province 710061 China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi Province 710061 China
| |
Collapse
|
40
|
Andréasson K, Jönsson G, Hesselstrand R, Norrgren H. Persistent elevation of fibrosis biomarker cartilage oligomeric matrix protein following hepatitis C virus eradication. World J Hepatol 2019; 11:330-334. [PMID: 30967910 PMCID: PMC6447423 DOI: 10.4254/wjh.v11.i3.330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/27/2019] [Accepted: 03/16/2019] [Indexed: 02/06/2023] Open
Abstract
Serum levels of cartilage oligomeric matrix protein (COMP) has been presented as a biomarker of liver fibrosis in several cross-sectional studies. COMP is also an essential mediator in carcinoma development and has also been associated with hepatocellular carcinoma. We present a prospective analysis of this biomarker in 38 patients with chronic hepatitis C who were subject to eradication therapy with direct acting antivirals. We confirm previous studies associating COMP elevation with liver cirrhosis. We also show how viral levels are correlated with COMP at baseline. In our prospective analysis, we report that successful eradication of hepatitis C results in improvement in liver stiffness and laboratory liver function tests at 1 year follow-up. In contrast, median COMP-levels remain unchanged during the study period. We conclude that the biomarker potential of COMP in the prospective evaluation of liver diseases, remains to be elucidated.
Collapse
Affiliation(s)
- Kristofer Andréasson
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund S-221 85, Sweden
| | - Göran Jönsson
- Section of Infectious Diseases, Department of Clinical Sciences Lund, Lund University, Lund S-221 85, Sweden
| | - Roger Hesselstrand
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund S-221 85, Sweden
| | - Hans Norrgren
- Section of Infectious Diseases, Department of Clinical Sciences Lund, Lund University, Lund S-221 85, Sweden
| |
Collapse
|
41
|
Metformin Inhibits the Expression of Biomarkers of Fibrosis of EPCs In Vitro. Stem Cells Int 2019; 2019:9019648. [PMID: 31011335 PMCID: PMC6442487 DOI: 10.1155/2019/9019648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 12/16/2018] [Accepted: 01/28/2019] [Indexed: 02/08/2023] Open
Abstract
Endothelial progenitor cells (EPCs) are a group of circulating cells with important functions in vascular repair and treatment of cardiovascular diseases. However, in patients with atrial fibrillation (AF), the number and function of EPCs reportedly are decreased. TGF-β is highly expressed in AF patients. In this study, we examined the effect of TGF-β1 on EPCs and the therapeutic outcome of metformin treatment on TGF-β1-induced EPCs. EPCs were induced with TGF-β1 at different concentrations (5 ng/ml, 10 ng/ml, and 20 ng/ml) for 48 h followed by western blot, qPCR, and immunofluorescence analyses to investigate changes in the levels of the fibrosis-related proteins, α-SMA, Col I, Col III, CTGF, and MMP-1. Live-dead cell staining was used to evaluate cell apoptosis. Compared with the control, TGF-β1 treatment significantly (p < 0.05) enhanced the levels of α-SMA, Col I, Col III, CTGF, and MMP-1 in a dose-dependent manner. The most effective concentration of TGF-β1 (20 ng/ml) was then used to induce fibrosis biomarker expression in EPCs, followed by treatment with metformin at different concentrations (0.5, 1, and 2 mmol/l). Metformin treatment suppressed TGF-β-induced expression of all above factors, with the effect at 2 mmol/l being significant (p < 0.05). Live-dead cell staining showed no difference among the control, TGF-β1-treated, and metformin-treated groups. In conclusion, our study showed that TGF-β1 induces the expression of fibrosis biomarkers in EPCs, which is attenuated by treatment with metformin. Thus, metformin may have therapeutic potential for improving EPC function in cardiovascular diseases.
Collapse
|
42
|
Samovski D, Abumrad NA. Regulation of lipophagy in NAFLD by cellular metabolism and CD36. J Lipid Res 2019; 60:755-757. [PMID: 30819696 DOI: 10.1194/jlr.c093674] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Dmitri Samovski
- Center for Human Nutrition, Washington University School of Medicine, Saint Louis, MO 63108
| | - Nada A Abumrad
- Center for Human Nutrition, Washington University School of Medicine, Saint Louis, MO 63108
| |
Collapse
|
43
|
Gebauer JM, Köhler A, Dietmar H, Gompert M, Neundorf I, Zaucke F, Koch M, Baumann U. COMP and TSP-4 interact specifically with the novel GXKGHR motif only found in fibrillar collagens. Sci Rep 2018; 8:17187. [PMID: 30464261 PMCID: PMC6249252 DOI: 10.1038/s41598-018-35447-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/05/2018] [Indexed: 12/19/2022] Open
Abstract
COMP (cartilage oligomeric matrix protein) is a member of the thrombospondin family and forms homopentamers as well as mixed heterooligomers with its closely related family member TSP-4. COMP is long known to bind to collagens and to influence collagen fibril formation. Recent work indicates that already intracellular interaction with collagen is important for collagen secretion. However, the exact binding site of COMP on the collagen triple helix has not been described up to now. In this study we have identified a GXKGHR motif on the collagen II helix to bind to COMP, using a recombinantly expressed collagen II peptide library. This binding sequence is conserved throughout evolution and we demonstrate that TSP-4 binds to the same sequence. The identified binding motif overlaps with the recognition sites of many other collagen-binding partners (e.g. PEDF, Heparin) and also spans the lysine residues, which form collagen cross-links. COMP might thereby protect collagen helices from premature modification and cross-linking. Interestingly, this motif is only found in classical fibrillar collagens, although COMP is known to also bind other types. This might indicate that COMP has a unique interface for fibrillar collagens, thus making it an interesting target for the development of antifibrotic drugs.
Collapse
Affiliation(s)
- Jan M Gebauer
- Institute of Biochemistry, University of Cologne, Cologne, Germany.
| | - Anna Köhler
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Helen Dietmar
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Monika Gompert
- Institute of Biochemistry, University of Cologne, Cologne, Germany
| | - Ines Neundorf
- Institute of Biochemistry, University of Cologne, Cologne, Germany
| | - Frank Zaucke
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Dr. Rolf Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim gGmbH, Frankfurt, Germany
| | - Manuel Koch
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute for Dental Research and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ulrich Baumann
- Institute of Biochemistry, University of Cologne, Cologne, Germany
| |
Collapse
|
44
|
Posey KL, Coustry F, Hecht JT. Cartilage oligomeric matrix protein: COMPopathies and beyond. Matrix Biol 2018; 71-72:161-173. [PMID: 29530484 PMCID: PMC6129439 DOI: 10.1016/j.matbio.2018.02.023] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 02/06/2023]
Abstract
Cartilage oligomeric matrix protein (COMP) is a large pentameric glycoprotein that interacts with multiple extracellular matrix proteins in cartilage and other tissues. While, COMP is known to play a role in collagen secretion and fibrillogenesis, chondrocyte proliferation and mechanical strength of tendons, the complete range of COMP functions remains to be defined. COMPopathies describe pseudoachondroplasia (PSACH) and multiple epiphyseal dysplasia (MED), two skeletal dysplasias caused by autosomal dominant COMP mutations. The majority of the mutations are in the calcium binding domains and compromise protein folding. COMPopathies are ER storage disorders in which the retention of COMP in the chondrocyte ER stimulates overwhelming cellular stress. The retention causes oxidative and inflammation processes leading to chondrocyte death and loss of long bone growth. In contrast, dysregulation of wild-type COMP expression is found in numerous diseases including: fibrosis, cardiomyopathy and breast and prostate cancers. The most exciting clinical application is the use of COMP as a biomarker for idiopathic pulmonary fibrosis and cartilage degeneration associated osteoarthritis and rheumatoid and, as a prognostic marker for joint injury. The ever expanding roles of COMP in single gene disorders and multifactorial diseases will lead to a better understanding of its functions in ECM and tissue homeostasis towards the goal of developing new therapeutic avenues.
Collapse
Affiliation(s)
- Karen L Posey
- McGovern Medical School, UTHealth, Department of Pediatrics, United States.
| | - Francoise Coustry
- McGovern Medical School, UTHealth, Department of Pediatrics, United States
| | - Jacqueline T Hecht
- McGovern Medical School, UTHealth, Department of Pediatrics, United States; UTHealth, School of Dentistry, United States
| |
Collapse
|
45
|
Li Q, Wang C, Wang Y, Sun L, Liu Z, Wang L, Song T, Yao Y, Liu Q, Tu K. HSCs-derived COMP drives hepatocellular carcinoma progression by activating MEK/ERK and PI3K/AKT signaling pathways. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:231. [PMID: 30231922 PMCID: PMC6146743 DOI: 10.1186/s13046-018-0908-y] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022]
Abstract
Background Cartilage oligomeric matrix protein (COMP) is known to promote fibrosis in skin, lung and liver. Emerging evidence shows that COMP plays critical roles in tumor development, including breast cancer, colon cancer and hepatocellular carcinoma (HCC). Nevertheless, the role of COMP in HCC proliferation and metastasis and its underlying mechanisms remain fully unclear. Methods Serum COMP was determined by ELISA. Cell Counting Kit-8 and plate colony formation were performed to evaluate cell proliferation. Wound healing and transwell assays were used to determine migration and invasion of HCC cells. Western blotting and immunofluorescence were carried out for detection of epithelial-to-mesenchymal transition (EMT) markers and MMPs in HCC cells. The in vivo role of COMP was evaluated using mouse models. We also measured effects of hepatic stellate cells (HSCs)-conditioned medium (CM) on HCC progression using transwell coculture system. Results Here, we found that serum COMP levels in HCC patients were significantly higher than those in healthy controls. Accordingly, high serum COMP levels in HCC patients significantly correlated with malignant clinical characteristics and poor clinical outcomes. Next, we investigated that recombinant human COMP protein (rCOMP) treatment resulted in increased abilities of proliferation, invasion and migration of HCC cells. Furthermore, rCOMP treatment enhanced proliferative and metastatic colonization of HCC cells in vivo. Mechanistically, CD36 receptor played an essential role in COMP-mediated HCC cell proliferation and metastasis. Functionally, COMP/CD36 signaling caused phosphorylation of ERK and AKT, resulting in the upregulation of tumor-progressive genes such as EMT markers, MMP-2/9, Slug and Twist in HCC cells. Interestingly, we revealed that COMP was secreted by HSCs. CM of LX2 cells with COMP knockdown showed weaker effects on the activation of MEK/ERK and PI3K/AKT signaling pathways in HCC cells compared to control CM. Conclusions Our findings indicated that HSCs-derived COMP collaborated with CD36 and subsequently played an essential role in MEK/ERK and PI3K/AKT-mediated HCC progression. COMP might act as a promising target for the diagnosis and treatment of aggressive HCC. Electronic supplementary material The online version of this article (10.1186/s13046-018-0908-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qing Li
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Cong Wang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Yufeng Wang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Liankang Sun
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Zhikui Liu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Liang Wang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Tao Song
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Yingmin Yao
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China
| | - Qingguang Liu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China.
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi Province, China.
| |
Collapse
|
46
|
Denton N, Pinnick KE, Karpe F. Cartilage oligomeric matrix protein is differentially expressed in human subcutaneous adipose tissue and regulates adipogenesis. Mol Metab 2018; 16:172-179. [PMID: 30100245 PMCID: PMC6157646 DOI: 10.1016/j.molmet.2018.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 07/17/2018] [Accepted: 07/23/2018] [Indexed: 12/15/2022] Open
Abstract
Objective The composition of the extracellular matrix (ECM) impacts adipocyte function and might determine adipose tissue (AT) function and distribution. Cartilage oligomeric matrix protein (COMP), a matricellular protein usually studied in bone and cartilage, is highly differentially expressed between subcutaneous abdominal and gluteal AT. This study aimed to explore COMP's role in human subcutaneous abdominal and gluteal AT and preadipocyte biology. Methods COMP mRNA levels were measured in whole AT and immortalised preadipocytes via quantitative (q)-PCR. Tissue and cellular COMP protein were measured via Western blot and immunohistochemistry; plasma COMP was measured by ELISA. The effect of COMP on adipogenesis in immortalised preadipocytes was evaluated by qPCR of adipogenic markers and cellular triacylglycerol (TAG) accumulation. Results qPCR analysis of paired subcutaneous abdominal and gluteal AT biopsies (n = 190) across a range of BMI (20.7–45.5 kg/m2) indicated ∼3-fold higher COMP expression in gluteal AT (P = 1.7 × 10−31); protein levels mirrored this. Immunohistochemistry indicated COMP was abundant in gluteal AT ECM and co-localised with collagen-1. AT COMP mRNA levels and circulating COMP protein levels were positively associated with BMI/adiposity but unrelated to AT distribution. COMP expression changed dynamically during adipogenesis (time × depot, P = 0.01). Supplementation of adipogenic medium with exogenous COMP protein (500 ng/ml) increased PPARG2 expression ∼1.5-fold (P = 0.0003) and TAG accumulation ∼1.25-fold in abdominal and gluteal preadipocytes (P = 0.02). Conclusions We confirmed that COMP is an ECM protein which is differentially expressed between subcutaneous abdominal and gluteal AT. Despite its depot-specific expression pattern, however, AT COMP mRNA levels and plasma COMP concentration correlated positively with overall obesity but not body fat distribution. Exogenous COMP enhanced adipogenesis. These data identify COMP as a novel regulator of AT and highlight the importance of the ECM to AT biology. COMP is a matricellular protein which is abundant in bone and cartilage. COMP is differentially expressed between subcutaneous abdominal and gluteal AT. COMP expression was positively correlated with overall AT mass but not distribution. COMP promoted adipogenesis in subcutaneous abdominal and gluteal preadipocytes. COMP represents a novel regulator of AT biology that resides in the ECM.
Collapse
Affiliation(s)
- Nathan Denton
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Katherine E Pinnick
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospital NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
47
|
Wang J, Pan W, Wang Y, Lei W, Feng B, Du C, Wang XJ. Enhanced efficacy of curcumin with phosphatidylserine-decorated nanoparticles in the treatment of hepatic fibrosis. Drug Deliv 2018; 25:1-11. [PMID: 29214887 PMCID: PMC6058669 DOI: 10.1080/10717544.2017.1399301] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatic macrophages have been considered as a therapeutic target for liver fibrosis treatment, and phosphatidylserine (PS)-containing nanoparticles are commonly used to mimic apoptotic cells that can specifically regulate macrophage functions, resulting in anti-inflammatory effects. This study was designed to test the efficacy of PS-modified nanostructured lipid carriers (mNLCs) containing curcumin (Cur) (Cur-mNLCs) in the treatment of liver fibrosis in a rat model. Carbon tetrachloride-induced liver fibrosis in rats was used as an experimental model, and the severity of the disease was examined by both biochemical and histological methods. Here, we showed that mNLCs were spherical nanoparticles with decreased negative zeta potentials due to PS decoration, and significantly increased both mean residence time and area under the curve of Cur. In the rats with liver fibrosis, PS-modification of NLCs enhanced the nanoparticles targeting to the diseased liver, which was evidenced by their highest accumulation in the liver. As compared to all the controls, Cur-mNLCs were significantly more effective at reducing the liver damage and fibrosis, which were indicated by in Cur-mNLCs-treated rats the least increase in liver enzymes and pro-inflammatory cytokines in the circulation, along with the least increase in collagen fibers and alpha smooth muscle actin and the most increased hepatocyte growth factors (HGF) and matrix metalloprotease (MMP) two in the livers. In conclusion, PS-modified NLCs nanoparticles prolonged the retention time of Cur, and enhanced its bioavailability and delivery efficiency to the livers, resulting in reduced liver fibrosis and up-regulating hepatic expression of HGF and MMP-2.
Collapse
Affiliation(s)
- Ji Wang
- a State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy , School of Stomatology, The Fourth Military Medical University , Xi'an , PR China
| | - Wen Pan
- a State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy , School of Stomatology, The Fourth Military Medical University , Xi'an , PR China
| | - Ying Wang
- a State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy , School of Stomatology, The Fourth Military Medical University , Xi'an , PR China
| | - Wan Lei
- a State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy , School of Stomatology, The Fourth Military Medical University , Xi'an , PR China
| | - Bin Feng
- a State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy , School of Stomatology, The Fourth Military Medical University , Xi'an , PR China
| | - Caigan Du
- b Department of Urologic Sciences , University of British Columbia, Jack Bell Research Centre , Vancouver , BC , Canada
| | - Xiao-Juan Wang
- a State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy , School of Stomatology, The Fourth Military Medical University , Xi'an , PR China
| |
Collapse
|
48
|
Wu W, Li J, Yu C, Gao Y, Fan S, Ye X, Wang Y, Zheng J. Association of serum ADAMTS-7 levels with left ventricular reverse remodeling after ST-elevation myocardial infarction. Eur J Med Res 2018. [PMID: 29523183 PMCID: PMC5845142 DOI: 10.1186/s40001-018-0305-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Left ventricular reverse remodeling (LVRR) in patients with ST-elevation myocardial infarction (STEMI) is associated with a good prognosis. Serum levels of ADAMTS-7 might be used for the prognosis of STEMI. This study aimed to investigate the relationship between serum ADAMTS-7 levels and LVRR. METHODS This was a prospective study of 104 patients with STEMI who underwent revascularization and 63 controls. ADAMTS-7 serum levels were measured on days 1, 3, and 7 and in months 1 and 6 after STEMI. A decrease ≥ 15% of the left ventricular end-systolic volume at 6 months was defined as LVRR. RESULTS The serum levels of ADAMTS-7 in patients with LVRR were lower than those without LVRR (3.84 ± 2.26 vs. 5.02 ± 2.54, P = 0.032) 7 days after STEMI and the difference between day 7 and day 1 (ΔADAMTS-7) was even significantly lower (- 1.31 ± 0.94 vs. - 0.30 ± 0.22, P = 0.021). Multivariate analysis showed that ΔADAMTS-7(day 7 minus day 1) was independently associated with LVRR (OR = - 0.322, 95% CI = - 0.996 to - 0.074, P = 0.028). Receiver operating characteristic (ROC) curve analysis showed that LVRR could be predicted (sensitivity 89%, specificity 82%, and area under the curve 0.896) when ΔADAMTS-7(day 7 minus day 1) was < - 0.39. CONCLUSIONS ΔADAMTS-7(day 7 minus day 1) might be a potential predictive factor for LVRR.
Collapse
Affiliation(s)
- Wenjing Wu
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jiahui Li
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Changan Yu
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yanxiang Gao
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Shuying Fan
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiaojun Ye
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yong Wang
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jingang Zheng
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
49
|
Cardiovascular Disease: An Introduction. BIOMATHEMATICAL AND BIOMECHANICAL MODELING OF THE CIRCULATORY AND VENTILATORY SYSTEMS 2018. [PMCID: PMC7123129 DOI: 10.1007/978-3-319-89315-0_1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Cardiovascular disease (CVD) is a collective term designating all types of affliction affecting the blood circulatory system, including the heart and vasculature, which, respectively, displaces and conveys the blood.
Collapse
|
50
|
Lin Z, Changfu H, Fengling Z, Wei G, Lei B, Yiping L, Miao Z, Zhongzheng Y, Youliang Z, Shuyin D, Wu Y. Long non-coding RNA deep sequencing reveals the role of macrophage in liver disorders. Oncotarget 2017; 8:114966-114979. [PMID: 29383134 PMCID: PMC5777746 DOI: 10.18632/oncotarget.23154] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 11/15/2017] [Indexed: 12/31/2022] Open
Abstract
Liver disorders such as hepatitis, cirrhosis and hepatocellular carcinoma are a series of the most life threatening diseases along with extensive inflammatory cellular infiltrations. Macrophage has been proved to be key regulators and initiators of inflammation, and long non-coding RNAs (lncRNAs) are recommended to play critical roles in the occurrence and development of a variety of diseases. To uncover the role of macrophage in liver disorders via lncRNA sequencing method, we first applied a lncRNA classification pipeline to identify 1247 lncRNAs represented on the Affymetrix Mouse Genome 430/430A 2.0 array. We then analyzed the lncRNA expression patterns in a set of previously published gene expression profiles of silica particle exposed macrophages and liver respectively, and identified and validated sets of differentially expressed lncRNAs shared by macrophages and liver. The functional enrichment analysis of these lncRNAs was processed on the basis of their expression signatures, three aspects including cis, trans and co-acting proteins were proposed. This is the first time to correlate macrophage with liver disorders via co-expressed lncRNAs. Our findings indicated that roles of macrophage in liver disorders were double-edged, the differentially expressed lncRNAs and their corresponding regulatory genes or proteins may serve as potential diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Zhang Lin
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China.,Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250001, China.,Key Laboratory of Reproductive Endocrinology, Shandong University, Ministry of Education, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan 250001, China
| | - Hao Changfu
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Zhao Fengling
- Department of Occupational Disease, Henan Provincial Institute of Occupational Health, Zhengzhou 450052, China
| | - Guo Wei
- Department of Occupational Disease, Henan Provincial Institute of Occupational Health, Zhengzhou 450052, China
| | - Bao Lei
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Li Yiping
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Zhang Miao
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Yue Zhongzheng
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Zhao Youliang
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Duan Shuyin
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Yao Wu
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|