1
|
Morishima N, Kamada Y, Ota H, Iwagami Y, Takahashi H, Shimosaka M, Sakon D, Kondo J, Yamada M, Kumada T, Eguchi H, Miyoshi E. Serum levels of the N-terminal fragment of connective tissue growth factor is a novel biomarker for chronic pancreatitis. Pract Lab Med 2024; 40:e00402. [PMID: 38828385 PMCID: PMC11143898 DOI: 10.1016/j.plabm.2024.e00402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024] Open
Abstract
Chronic inflammation of the pancreas is considered to be one of the causes of pancreatic cancer. However, the diagnosis of chronic pancreatitis (CP) is very difficult in the pancreas, where biopsies are difficult to perform. The prevalence of CP is estimated to be many times more common than in patients with actual symptomatic CP. In recent years, abnormal cleavage of certain proteins has attracted attention as a biomarker for CP other than pancreatic enzymes. Connective tissue growth factor (CTGF) is one of the growth factors involved in tissue repair and other processes and is increased by stimulation of transforming growth factor-β, suggesting a relationship of CTGF with fibrosis. In this study, we measured the total length of CTGF in blood and N-terminal fragment CTGF in 48 cases of chronic pancreatitis, 64 cases of pancreatic cancer and 45 healthy volunteers (HV). Interestingly, we found that blood N-terminal fragment CTGF level was significantly increased in CP and pancreatic cancer patients. Multiple logistic regression analysis showed serum levels of N-terminal fragment CTGF, CRP and amylase were significant and independent variables for the differential diagnosis of CP from HV. Receiver operating characteristic analysis showed that area under the curve (AUC) value of serum N-terminal fragment CTGF level was 0.933, which can differentiate between CP and HV. Several factors would be involved in the increase in serum N-terminal fragment CTGF level. In conclusion, serum N-terminal fragment CTGF level is a promising new biomarker for CP.
Collapse
Affiliation(s)
- Naoki Morishima
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hiyori Ota
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yoshifumi Iwagami
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Hidenori Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Munefumi Shimosaka
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Daisuke Sakon
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Jumpei Kondo
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | | | - Takashi Kumada
- Department of Nursing, Faculty of Nursing, Gifu Kyoritsu University, 5-50, Kitagata-cho, Ogaki, 503-8550, Gifu, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
2
|
Yoshida H, Yokota S, Satoh K, Ishisaki A, Chosa N. Connective tissue growth factor enhances TGF-β1-induced osteogenic differentiation via activation of p38 MAPK in mesenchymal stem cells. J Oral Biosci 2024; 66:68-75. [PMID: 38266705 DOI: 10.1016/j.job.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/26/2024]
Abstract
OBJECTIVES Cellular differentiation is based on the effects of various growth factors. Transforming growth factor (TGF)-β1 plays a pivotal role in inducing osteogenic differentiation of mesenchymal stem cells (MSCs). In this study, we investigated the influence of connective tissue growth factor (CTGF), known to function synergistically with TGF-β1, on osteogenic differentiation in MSCs. METHODS UE7T-13 cells were treated with TGF-β1 and/or CTGF. Subsequently, protein levels of intracellular signaling pathway molecules were determined through western blot analysis. The mRNA expression levels of osteogenic differentiation markers were investigated using reverse transcription-quantitative polymerase chain reaction. Bone matrix mineralization was evaluated through alizarin red staining. RESULTS Co-treatment with TGF-β1 and CTGF resulted in the suppression of TGF-β1-induced phosphorylation of extracellular signal-regulated kinase 1/2, an intracellular signaling pathway molecule in MSCs, while significantly enhancing the phosphorylation of p38 mitogen-activated protein kinase (MAPK). In MSCs, co-treatment with CTGF and TGF-β1 led to increased expression levels of alkaline phosphatase and type I collagen, markers of osteogenic differentiation induced by TGF-β1. Osteopontin expression was observed only after TGF-β1 and CTGF co-treatment. Notably, bone sialoprotein and osteocalcin were significantly upregulated by treatment with CTGF alone. Furthermore, CTGF enhanced the TGF-β1-induced mineralization in MSCs, with complete suppression observed after treatment with a p38 MAPK inhibitor. CONCLUSIONS CTGF enhances TGF-β1-induced osteogenic differentiation and subsequent mineralization in MSCs by predominantly activating the p38 MAPK-dependent pathway.
Collapse
Affiliation(s)
- Hironori Yoshida
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate, 028-3694, Japan; Division of Orthodontics, Department of Developmental Oral Health Science, Iwate Medical University, Morioka, Iwate, 020-8505, Japan
| | - Seiji Yokota
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate, 028-3694, Japan
| | - Kazuro Satoh
- Division of Orthodontics, Department of Developmental Oral Health Science, Iwate Medical University, Morioka, Iwate, 020-8505, Japan
| | - Akira Ishisaki
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate, 028-3694, Japan
| | - Naoyuki Chosa
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate, 028-3694, Japan.
| |
Collapse
|
3
|
Teisseire M, Giuliano S, Pagès G. Combination of Anti-Angiogenics and Immunotherapies in Renal Cell Carcinoma Show Their Limits: Targeting Fibrosis to Break through the Glass Ceiling? Biomedicines 2024; 12:385. [PMID: 38397987 PMCID: PMC10886484 DOI: 10.3390/biomedicines12020385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
This review explores treating metastatic clear cell renal cell carcinoma (ccRCC) through current therapeutic modalities-anti-angiogenic therapies and immunotherapies. While these approaches represent the forefront, their limitations and variable patient responses highlight the need to comprehend underlying resistance mechanisms. We specifically investigate the role of fibrosis, prevalent in chronic kidney disease, influencing tumour growth and treatment resistance. Our focus extends to unravelling the intricate interplay between fibrosis, immunotherapy resistance, and the tumour microenvironment for effective therapy development. The analysis centres on connective tissue growth factor (CTGF), revealing its multifaceted role in ccRCC-promoting fibrosis, angiogenesis, and cancer progression. We discuss the potential of targeting CTGF to address the problem of fibrosis in ccRCC. Emphasising the crucial relationship between fibrosis and the immune system in ccRCC, we propose that targeting CTGF holds promise for overcoming obstacles to cancer treatment. However, we recognise that an in-depth understanding of the mechanisms and potential limitations is imperative and, therefore, advocate for further research. This is an essential prerequisite for the successful integration of CTGF-targeted therapies into the clinical landscape.
Collapse
Affiliation(s)
| | - Sandy Giuliano
- University Cote d’Azur (UCA), Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, 06189 Nice, France;
| | - Gilles Pagès
- University Cote d’Azur (UCA), Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, 06189 Nice, France;
| |
Collapse
|
4
|
van Velthoven MJJ, Gudde AN, Struijs F, Oosterwijk E, Roovers JP, Guler Z, Hooijmans CR, Kouwer PHJ. The Effect of Growth Factors on Vaginal Wound Healing: A Systematic Review and Meta-analysis. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:429-440. [PMID: 37051705 DOI: 10.1089/ten.teb.2022.0225] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Surgical outcomes of pelvic organ prolapse (POP) surgery are poor, resulting in a 20% recurrence risk. Following the hypothesis that impaired wound healing is the main determinant of recurrent POP, growth factors have the potential to promote wound healing and may improve surgical outcomes. In this study, we systematically reviewed the effect of growth factors on vaginal wound healing in both in vitro and animal studies. For each independent comparison, the standardized mean difference and 95% CI were calculated using the Hedges' g correction. Of the 3858 retrieved studies, seven studies were included, of which six were included in meta-analysis (three in vitro studies and four in vivo studies). In vitro, basic fibroblast growth factor (bFGF) promotes proliferation, differentiation, and collagen types I and III production. Epidermal growth factor stimulates proliferation and connective tissue growth factor promotes Tenascin-C expression. These effects, however, are less pronounced in vivo; only bFGF slightly promotes collagen production. The review shows that growth factors, particularly bFGF, are able to promote vaginal wound healing in vitro. The uncertain in vivo findings suggest that preclinical models should be improved. The ultimate goal is to develop effective growth factor-supplemented therapies that improve surgical outcomes for POP.
Collapse
Affiliation(s)
- Melissa J J van Velthoven
- Institute of Molecules and Materials, Radboud University, Nijmegen, The Netherlands
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Aksel N Gudde
- Department of Obstetrics and Gynecology and Amsterdam University Medical Center, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Frederique Struijs
- Institute of Molecules and Materials, Radboud University, Nijmegen, The Netherlands
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Egbert Oosterwijk
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan-Paul Roovers
- Department of Obstetrics and Gynecology and Amsterdam University Medical Center, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Zeliha Guler
- Department of Obstetrics and Gynecology and Amsterdam University Medical Center, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Carlijn R Hooijmans
- Department of Anesthesiology, Pain and Palliative Care, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Paul H J Kouwer
- Institute of Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
5
|
Zhou L, McAdow AR, Yamada H, Burris B, Klatt Shaw D, Oonk K, Poss KD, Mokalled MH. Progenitor-derived glia are required for spinal cord regeneration in zebrafish. Development 2023; 150:dev201162. [PMID: 37213080 PMCID: PMC10233714 DOI: 10.1242/dev.201162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 04/26/2023] [Indexed: 05/10/2023]
Abstract
Unlike mammals, adult zebrafish undergo spontaneous recovery after major spinal cord injury. Whereas reactive gliosis presents a roadblock for mammalian spinal cord repair, glial cells in zebrafish elicit pro-regenerative bridging functions after injury. Here, we perform genetic lineage tracing, assessment of regulatory sequences and inducible cell ablation to define mechanisms that direct the molecular and cellular responses of glial cells after spinal cord injury in adult zebrafish. Using a newly generated CreERT2 transgenic line, we show that the cells directing expression of the bridging glial marker ctgfa give rise to regenerating glia after injury, with negligible contribution to either neuronal or oligodendrocyte lineages. A 1 kb sequence upstream of the ctgfa gene was sufficient to direct expression in early bridging glia after injury. Finally, ablation of ctgfa-expressing cells using a transgenic nitroreductase strategy impaired glial bridging and recovery of swim behavior after injury. This study identifies key regulatory features, cellular progeny, and requirements of glial cells during innate spinal cord regeneration.
Collapse
Affiliation(s)
- Lili Zhou
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anthony R. McAdow
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hunter Yamada
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brooke Burris
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dana Klatt Shaw
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kelsey Oonk
- Duke Regeneration Center, Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kenneth D. Poss
- Duke Regeneration Center, Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Mayssa H. Mokalled
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
6
|
Kubota S, Kawaki H, Perbal B, Takigawa M, Kawata K, Hattori T, Nishida T. Do not overwork: cellular communication network factor 3 for life in cartilage. J Cell Commun Signal 2023:10.1007/s12079-023-00723-4. [PMID: 36745317 DOI: 10.1007/s12079-023-00723-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 01/07/2023] [Indexed: 02/07/2023] Open
Abstract
Cellular communication network factor (CCN) 3, which is one of the founding members of the CCN family, displays diverse functions. However, this protein generally represses the proliferation of a variety of cells. Along with skeletal development, CCN3 is produced in cartilaginous anlagen, growth plate cartilage and epiphysial cartilage. Interestingly, CCN3 is drastically induced in the growth plates of mice lacking CCN2, which promotes endochondral ossification. Notably, chondrocytes in these mutant mice with elevated CCN3 production also suffer from impaired glycolysis and energy metabolism, suggesting a critical role of CCN3 in cartilage metabolism. Recently, CCN3 was found to be strongly induced by impaired glycolysis, and in our study, we located an enhancer that mediated CCN3 regulation via starvation. Subsequent investigations specified regulatory factor binding to the X-box 1 (RFX1) as a transcription factor mediating this CCN3 regulation. Impaired glycolysis is a serious problem, resulting in an energy shortage in cartilage without vasculature. CCN3 produced under such starved conditions restricts energy consumption by repressing cell proliferation, leading chondrocytes to quiescence and survival. This CCN3 regulatory system is indicated to play an important role in articular cartilage maintenance, as well as in skeletal development. Furthermore, CCN3 continues to regulate cartilage metabolism even during the aging process, probably utilizing this regulatory system. Altogether, CCN3 seems to prevent "overwork" by chondrocytes to ensure their sustainable life in cartilage by sensing energy metabolism. Similar roles are suspected to exist in relation to systemic metabolism, since CCN3 is found in the bloodstream.
Collapse
Affiliation(s)
- Satoshi Kubota
- Department of Biochemistry and Molecular Dentistry, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan.
| | - Harumi Kawaki
- Department of Oral Biochemistry, Asahi University School of Dentistry, Mizuho, Japan
| | | | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences/Dental School, Okayama, Japan
| | - Kazumi Kawata
- Department of Biochemistry and Molecular Dentistry, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Takako Hattori
- Department of Biochemistry and Molecular Dentistry, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Takashi Nishida
- Department of Biochemistry and Molecular Dentistry, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences/Dental School, Okayama, Japan
| |
Collapse
|
7
|
Rayrikar AY, Wagh GA, Santra MK, Patra C. Ccn2a-FGFR1-SHH signaling is necessary for intervertebral disc homeostasis and regeneration in adult zebrafish. Development 2023; 150:dev201036. [PMID: 36458546 PMCID: PMC10108606 DOI: 10.1242/dev.201036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022]
Abstract
Intervertebral disc (IVD) degeneration is the primary cause of back pain in humans. However, the cellular and molecular pathogenesis of IVD degeneration is poorly understood. This study shows that zebrafish IVDs possess distinct and non-overlapping zones of cell proliferation and cell death. We find that, in zebrafish, cellular communication network factor 2a (ccn2a) is expressed in notochord and IVDs. Although IVD development appears normal in ccn2a mutants, the adult mutant IVDs exhibit decreased cell proliferation and increased cell death leading to IVD degeneration. Moreover, Ccn2a overexpression promotes regeneration through accelerating cell proliferation and suppressing cell death in wild-type aged IVDs. Mechanistically, Ccn2a maintains IVD homeostasis and promotes IVD regeneration by enhancing outer annulus fibrosus cell proliferation and suppressing nucleus pulposus cell death through augmenting FGFR1-SHH signaling. These findings reveal that Ccn2a plays a central role in IVD homeostasis and regeneration, which could be exploited for therapeutic intervention in degenerated human discs.
Collapse
Affiliation(s)
- Amey Y. Rayrikar
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra 411004, India
- S P Pune University, Pune, Maharashtra 411007, India
| | - Ganesh A. Wagh
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra 411004, India
- S P Pune University, Pune, Maharashtra 411007, India
| | - Manas K. Santra
- National Centre for Cell Science, Pune, Maharashtra 411007, India
| | - Chinmoy Patra
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra 411004, India
- S P Pune University, Pune, Maharashtra 411007, India
| |
Collapse
|
8
|
Takigawa M. CCN Proteins (Cellular Communication Network Factors): Expanding Their Repertoire Toward a New Concept. Methods Mol Biol 2023; 2582:1-10. [PMID: 36370338 DOI: 10.1007/978-1-0716-2744-0_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
I herein report the general structures and functions of CCN proteins and possible molecular mechanisms involved in the unique biological actions of this family of intercellular signaling regulators, which are considered matricellular proteins and were once referred to as "signal conductors" but have recently been renamed "Cellular Communication Network Factors." Their repertoire of functions beyond their role as matricellular proteins is also described to aid in future studies. Advanced research concerning their relevance to pathology is briefly introduced as well. The information provided in this chapter is expected to be useful for readers of subsequent chapters.
Collapse
Affiliation(s)
- Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School/Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| |
Collapse
|
9
|
Negro S, Lauria F, Stazi M, Tebaldi T, D’Este G, Pirazzini M, Megighian A, Lessi F, Mazzanti CM, Sales G, Romualdi C, Fillo S, Lista F, Sleigh JN, Tosolini AP, Schiavo G, Viero G, Rigoni M. Hydrogen peroxide induced by nerve injury promotes axon regeneration via connective tissue growth factor. Acta Neuropathol Commun 2022; 10:189. [PMID: 36567321 PMCID: PMC9791753 DOI: 10.1186/s40478-022-01495-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/12/2022] [Indexed: 12/26/2022] Open
Abstract
Regeneration of the neuromuscular junction (NMJ) leverages on extensive exchange of factors released from motor axon terminals (MATs), muscle fibers and perisynaptic Schwann cells (PSCs), among which hydrogen peroxide (H2O2) is a major pro-regenerative signal. To identify critical determinants of NMJ remodeling in response to injury, we performed temporal transcriptional profiling of NMJs from 2 month-old mice during MAT degeneration/regeneration, and cross-referenced the differentially expressed genes with those elicited by H2O2 in SCs. We identified an enrichment in extracellular matrix (ECM) transcripts, including Connective Tissue Growth Factor (Ctgf), which is usually expressed during development. We discovered that Ctgf levels are increased in a Yes-associated protein (YAP)-dependent fashion in response to rapid, local H2O2 signaling generated by stressed mitochondria in the injured sciatic nerve, a finding highlighting the importance of signals triggered by mechanical force to motor nerve repair. Through sequestration of Ctgf or inactivation of H2O2, we delayed the recovery of neuromuscular function by impairing SC migration and, in turn, axon-oriented re-growth. These data indicate that H2O2 and its downstream effector Ctgf are pro-regenerative factors that enable axonal growth, and reveal a striking ECM remodeling process during nerve regeneration upon local H2O2 signaling. Our study identifies key transcriptomic changes at the regenerating NMJ, providing a rich source of pro-regenerative factors with potential for alleviating the consequences of peripheral nerve injuries.
Collapse
Affiliation(s)
- Samuele Negro
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy ,grid.5608.b0000 0004 1757 3470U.O.C. Clinica Neurologica, Azienda Ospedale, University of Padua, 35128 Padua, Italy
| | - Fabio Lauria
- grid.419463.d0000 0004 1756 3731Institute of Biophysics, CNR Unit at Trento, 38123 Povo, Italy
| | - Marco Stazi
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Toma Tebaldi
- grid.11696.390000 0004 1937 0351Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Povo, Italy ,grid.47100.320000000419368710Section of Hematology, Department of Internal Medicine, Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Giorgia D’Este
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Marco Pirazzini
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy ,grid.5608.b0000 0004 1757 3470Myology Center (CIR-Myo), University of Padua, 35129 Padua, Italy
| | - Aram Megighian
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy ,grid.5608.b0000 0004 1757 3470Padua Neuroscience Center, University of Padua, 35131 Padua, Italy
| | - Francesca Lessi
- Laboratory of Genomics, Pisa Science Foundation, 56017 San Giuliano Terme, Italy
| | - Chiara M. Mazzanti
- Laboratory of Genomics, Pisa Science Foundation, 56017 San Giuliano Terme, Italy
| | - Gabriele Sales
- grid.5608.b0000 0004 1757 3470Department of Biology, University of Padua, 35131 Padua, Italy
| | - Chiara Romualdi
- grid.5608.b0000 0004 1757 3470Department of Biology, University of Padua, 35131 Padua, Italy
| | - Silvia Fillo
- grid.470599.60000 0004 1760 920XCenter of Medical and Veterinary Research of the Ministry of Defence, 00184 Rome, Italy
| | - Florigio Lista
- grid.470599.60000 0004 1760 920XCenter of Medical and Veterinary Research of the Ministry of Defence, 00184 Rome, Italy
| | - James N. Sleigh
- grid.83440.3b0000000121901201Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UK Dementia Research Institute, University College London, London, WC1E 6BT UK
| | - Andrew P. Tosolini
- grid.83440.3b0000000121901201Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG UK
| | - Giampietro Schiavo
- grid.83440.3b0000000121901201Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UK Dementia Research Institute, University College London, London, WC1E 6BT UK
| | - Gabriella Viero
- grid.419463.d0000 0004 1756 3731Institute of Biophysics, CNR Unit at Trento, 38123 Povo, Italy
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padua, 35131, Padua, Italy. .,Myology Center (CIR-Myo), University of Padua, 35129, Padua, Italy.
| |
Collapse
|
10
|
Recent Advancements in Molecular Therapeutics for Corneal Scar Treatment. Cells 2022; 11:cells11203310. [PMID: 36291182 PMCID: PMC9600986 DOI: 10.3390/cells11203310] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
The process of corneal wound healing is complex and induces scar formation. Corneal scarring is a leading cause of blindness worldwide. The fibrotic healing of a major ocular wound disrupts the highly organized fibrillar collagen arrangement of the corneal stroma, rendering it opaque. The process of regaining this organized extracellular matrix (ECM) arrangement of the stromal layer to restore corneal transparency is complicated. The surface retention capacity of ocular drugs is poor, and there is a large gap between suitable corneal donors and clinical requirements. Therefore, a more efficient way of treating corneal scarring is needed. The eight major classes of interventions targeted as therapeutic tools for healing scarred corneas include those based on exosomes, targeted gene therapy, microRNAs, recombinant viral vectors, histone deacetylase inhibitors, bioactive molecules, growth factors, and nanotechnology. This review highlights the recent advancements in molecular therapeutics to restore a cornea without scarring. It also provides a scope to overcome the limitations of present studies and perform robust clinical research using these strategies.
Collapse
|
11
|
Mao Y, Zhao K, Li P, Sheng Y. The emerging role of leptin in obesity-associated cardiac fibrosis: evidence and mechanism. Mol Cell Biochem 2022; 478:991-1011. [PMID: 36214893 DOI: 10.1007/s11010-022-04562-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 09/15/2022] [Indexed: 11/24/2022]
Abstract
Cardiac fibrosis is a hallmark of various cardiovascular diseases, which is quite commonly found in obesity, and may contribute to the increased incidence of heart failure arrhythmias, and sudden cardiac death in obese populations. As an endogenous regulator of adiposity metabolism, body mass, and energy balance, obesity, characterized by increased circulating levels of the adipocyte-derived hormone leptin, is a critical contributor to the pathogenesis of cardiac fibrosis. Although there are some gaps in our knowledge linking leptin and cardiac fibrosis, this review will focus on the interplay between leptin and major effectors involved in the pathogenesis underlying cardiac fibrosis at both cellular and molecular levels based on the current reports. The profibrotic effect of leptin is predominantly mediated by activated cardiac fibroblasts but may also involve cardiomyocytes, endothelial cells, and immune cells. Moreover, a series of molecular signals with a known profibrotic property is closely involved in leptin-induced fibrotic events. A more comprehensive understanding of the underlying mechanisms through which leptin contributes to the pathogenesis of cardiac fibrosis may open up a new avenue for the rapid emergence of a novel therapy for preventing or even reversing obesity-associated cardiac fibrosis.
Collapse
Affiliation(s)
- Yukang Mao
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, People's Republic of China.,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Kun Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China.
| | - Yanhui Sheng
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, People's Republic of China. .,Department of Cardiology, Jiangsu Province Hospital, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
12
|
Fuchs L, Mausner-Fainberg K, Luban A, Asseyer SE, Golan M, Benhamou M, Volovitz I, Regev K, Vigiser I, Piura Y, Kolb H, Paul F, Karni A. CTGF/CCN2 has a possible detrimental role in the inflammation and the remyelination failure in the early stages of multiple sclerosis. J Neuroimmunol 2022; 371:577936. [DOI: 10.1016/j.jneuroim.2022.577936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/19/2022] [Accepted: 07/24/2022] [Indexed: 11/15/2022]
|
13
|
Kubota S, Kawata K, Hattori T, Nishida T. Molecular and Genetic Interactions between CCN2 and CCN3 behind Their Yin-Yang Collaboration. Int J Mol Sci 2022; 23:ijms23115887. [PMID: 35682564 PMCID: PMC9180607 DOI: 10.3390/ijms23115887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 12/15/2022] Open
Abstract
Cellular communication network factor (CCN) 2 and 3 are the members of the CCN family that conduct the harmonized development of a variety of tissues and organs under interaction with multiple biomolecules in the microenvironment. Despite their striking structural similarities, these two members show contrastive molecular functions as well as temporospatial emergence in living tissues. Typically, CCN2 promotes cell growth, whereas CCN3 restrains it. Where CCN2 is produced, CCN3 disappears. Nevertheless, these two proteins collaborate together to execute their mission in a yin–yang fashion. The apparent functional counteractions of CCN2 and CCN3 can be ascribed to their direct molecular interaction and interference over the cofactors that are shared by the two. Recent studies have revealed the mutual negative regulation systems between CCN2 and CCN3. Moreover, the simultaneous and bidirectional regulatory system of CCN2 and CCN3 is also being clarified. It is of particular note that these regulations were found to be closely associated with glycolysis, a fundamental procedure of energy metabolism. Here, the molecular interplay and metabolic gene regulation that enable the yin–yang collaboration of CCN2 and CCN3 typically found in cartilage development/regeneration and fibrosis are described.
Collapse
|
14
|
Kartamihardja AAP, Amalia SN, Sekiguchi A, Bhattarai A, Taketomi-Takahashi A, Motegi SI, Koyama H, Tsushima Y. Neutrophil elastase in the development of nephrogenic systemic fibrosis (NSF)-like skin lesion in renal failure mouse model. PLoS One 2021; 16:e0259211. [PMID: 34705860 PMCID: PMC8550606 DOI: 10.1371/journal.pone.0259211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/14/2021] [Indexed: 11/18/2022] Open
Abstract
Although neutrophil elastase (NE) may play a role in lung fibrosis and liver fibrosis, NE involvement in the development of nephrogenic systemic fibrosis has been unclear. We investigated the involvement of NE in the development of nephrogenic systemic fibrosis-like skin lesions post-injections of linear gadolinium-based contrast agents in renal failure mouse models. Renal failure mouse models were randomly divided into three groups: control group (saline), gadodiamide group, and gadopentetate group. Each solution was intravenously administered three times per week for three weeks. The mice were observed daily for skin lesions. Quantification of skin lesions, infiltrating inflammatory cells, and profibrotic cytokines in the affected skin was performed by immunostaining and reverse-transcription polymerase chain reaction (RT-PCR). Blood samples were collected from the facial vein to quantify NE enzymatic activity. The 158Gd concentrations in each sample were quantified using inductively coupled plasma mass spectrometry (ICP-MS). In the gadodiamide group, the mRNA expression of fibrotic markers was increased in the skin lesions compared to the control group. In the gadopentetate group, only collagen 1α and TGF-β mRNA expression were higher than in the control group. The expression of CD3+, CD68+, NE cells and the NE activity in the blood serum were significantly higher in the gadodiamide and gadopentetate groups compared to the control group. Gadolinium concentration in the skin of the gadodiamide group was significantly higher than the gadopentetate group, while almost no traces of gadolinium were found in the control group. Although gadopentetate and gadodiamide affected the fibrotic markers in the skin differently, NE may be involved in the development of fibrosis linked to the GBCAs injections in renal failure mouse models.
Collapse
Affiliation(s)
- A. Adhipatria P. Kartamihardja
- Department of Diagnostic Radiology and Nuclear Medicine Department, Gunma University Graduate School of Medicine, Maebashi, Japan
- Department of Nuclear Medicine and Molecular Imaging, Universitas Padjajaran, Sumedang, Indonesia
| | - Syahla Nisaa Amalia
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akiko Sekiguchi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Anu Bhattarai
- National Academy of Medical Sciences (NAMS), Bir Hospital, Kathmandu, Nepal
| | - Ayako Taketomi-Takahashi
- Department of Diagnostic Radiology and Nuclear Medicine Department, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Sei-ichiro Motegi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hiroshi Koyama
- Department of Public Health, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine Department, Gunma University Graduate School of Medicine, Maebashi, Japan
- Division of Integrated Oncology Research, Gunma Initiative for Advanced Research, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
15
|
Zaykov V, Chaqour B. The CCN2/CTGF interactome: an approach to understanding the versatility of CCN2/CTGF molecular activities. J Cell Commun Signal 2021; 15:567-580. [PMID: 34613590 DOI: 10.1007/s12079-021-00650-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/23/2021] [Indexed: 01/16/2023] Open
Abstract
Cellular communication network 2 (CCN2), also known as connective tissue growth factor (CTGF) regulates diverse cellular processes, some at odds with others, including adhesion, proliferation, apoptosis, and extracellular matrix (ECM) protein synthesis. Although a cause-and-effect relationship between CCN2/CTGF expression and local fibrotic reactions has initially been established, CCN2/CTGF manifests cell-, tissue-, and context-specific functions and differentially affects developmental and pathological processes ranging from progenitor cell fate decisions and angiogenesis to inflammation and tumorigenesis. CCN2/CTGF multimodular structure, binding to and activation or inhibition of multiple cell surface receptors, growth factors and ECM proteins, and susceptibility for proteolytic cleavage highlight the complexity to CCN2/CTGF biochemical attributes. CCN2/CTGF expression and dosage in the local environment affects a defined community of its interacting partners, and this results in sequestration of growth factors, interference with or potentiation of ligand-receptor binding, cellular internalization of CCN2/CTGF, inhibition or activation of proteases, and generation of CCN2/CTGF degradome products that add molecular diversity and expand the repertoire of functional modules in the cells and their microenvironment. Through these interactions, different intracellular signals and cellular responses are elicited culminating into physiological or pathological reactions. Thus, the CCN2/CTGF interactome is a defining factor of its tissue- and context-specific effects. Mapping of new CCN2/CTGF binding partners might shed light on yet unknown roles of CCN2/CTGF and provide a solid basis for tissue-specific targeting this molecule or its interacting partners in a therapeutic context.
Collapse
Affiliation(s)
- Viktor Zaykov
- Department of Cell Biology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA
| | - Brahim Chaqour
- Department of Cell Biology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA. .,Department of Ophthalmology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA.
| |
Collapse
|
16
|
Sun K, Xie Q, Hao CM. Mechanisms of Scarring in Focal Segmental Glomerulosclerosis. KIDNEY DISEASES (BASEL, SWITZERLAND) 2021; 7:350-358. [PMID: 34604342 PMCID: PMC8443927 DOI: 10.1159/000517108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/27/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Focal segmental glomerulosclerosis (FSGS) is a histologic pattern characterized by focal glomerular scarring, which often progresses to systemic and diffuse glomerulosclerosis. Previous studies have emphasized that the initiation of classic FSGS occurs in podocytes. The dysfunction and loss of podocytes have been associated with the development of proteinuria and the progression of various diseases. In addition, primary, secondary, and genetic FSGS are caused by different mechanisms of podocyte injury. SUMMARY The potential sources and mechanism of podocyte supplementation are the focus of our current research. Increasing attention has been paid to the role played by parietal epithelial cells (PECs) during the progression of FSGS. PECs are not only the primary influencing factors in glomerulosclerosis lesions but also have repair abilities, which remain a focus of debate. Notably, other resident glomerular cells also play significant roles in the progression of this disease. KEY MESSAGE In this review, we focus on the mechanism of scarring in FSGS and discuss current and potential therapeutic strategies.
Collapse
Affiliation(s)
- Ke Sun
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qionghong Xie
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuan-Ming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- Nephrology Division, Vanderbilt University Medical Center School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
17
|
Kikuchi R, Maeda Y, Tsuji T, Yamaguchi K, Abe S, Nakamura H, Aoshiba K. Fenofibrate inhibits TGF-β-induced myofibroblast differentiation and activation in human lung fibroblasts in vitro. FEBS Open Bio 2021; 11:2340-2349. [PMID: 34228906 PMCID: PMC8329776 DOI: 10.1002/2211-5463.13247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 06/11/2021] [Accepted: 07/05/2021] [Indexed: 02/05/2023] Open
Abstract
Fenofibrate (FF), a peroxisome proliferator-activated receptor-alpha (PPAR-α) agonist and a lipid-lowering agent, can decrease experimental pulmonary fibrosis. However, the mechanisms underlying the antifibrotic effect of FF remain unknown. Hence, this study was conducted to evaluate the effects of FF on transforming growth factor-beta (TGF-β)-induced myofibroblast differentiation and activation in lung fibroblasts. The results showed that FF inhibited alpha-smooth muscle actin (α-SMA) and connective tissue growth factor expression, collagen production, cell motility, SMAD3 phosphorylation and nuclear translocation, and metabolic reprogramming in TGF-β-exposed cells. The inhibitory effect of FF did not decrease with the addition of a PPAR-α antagonist. Moreover, the inhibitory effect given by FF could not be reproduced with the addition of an alternative PPAR-α agonist. FF inhibited mitochondrial respiration. However, rotenone, a complex I inhibitor, did not suppress TGF-β-induced myofibroblast differentiation. Furthermore, the TGF-β-induced nuclear reduction of protein phosphatase, Mg2+ /Mn2+ -dependent 1A (PPM1A), a SMAD phosphatase, was inhibited by FF. These results showed that FF suppressed TGF-β-induced myofibroblast differentiation and activation independent of PPAR-α activation and impaired mitochondrial respiration. In conclusion, this study provides information on the effects of FF on anti-TGF-β mechanisms.
Collapse
Affiliation(s)
- Ryota Kikuchi
- Department of Respiratory MedicineTokyo Medical University Ibaraki Medical CenterInashikiJapan
- Department of Respiratory MedicineTokyo Medical UniversityShinjuku‐kuJapan
| | - Yuki Maeda
- Department of Respiratory MedicineTokyo Medical University Ibaraki Medical CenterInashikiJapan
| | - Takao Tsuji
- Department of MedicineOtsuki Municipal HospitalJapan
| | - Kazuhiro Yamaguchi
- Department of Respiratory MedicineTokyo Medical University Ibaraki Medical CenterInashikiJapan
| | - Shinji Abe
- Department of Respiratory MedicineTokyo Medical UniversityShinjuku‐kuJapan
| | - Hiroyuki Nakamura
- Department of Respiratory MedicineTokyo Medical University Ibaraki Medical CenterInashikiJapan
| | - Kazutetsu Aoshiba
- Department of Respiratory MedicineTokyo Medical University Ibaraki Medical CenterInashikiJapan
| |
Collapse
|
18
|
Horlock D, Kaye DM, Winbanks CE, Gao XM, Kiriazis H, Donner DG, Gregorevic P, McMullen JR, Bernardo BC. Old Drug, New Trick: Tilorone, a Broad-Spectrum Antiviral Drug as a Potential Anti-Fibrotic Therapeutic for the Diseased Heart. Pharmaceuticals (Basel) 2021; 14:ph14030263. [PMID: 33804032 PMCID: PMC7998193 DOI: 10.3390/ph14030263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 11/16/2022] Open
Abstract
Cardiac fibrosis is associated with most forms of cardiovascular disease. No reliable therapies targeting cardiac fibrosis are available, thus identifying novel drugs that can resolve or prevent fibrosis is needed. Tilorone, an antiviral agent, can prevent fibrosis in a mouse model of lung disease. We investigated the anti-fibrotic effects of tilorone in human cardiac fibroblasts in vitro by performing a radioisotopic assay for [3H]-proline incorporation as a proxy for collagen synthesis. Exploratory studies in human cardiac fibroblasts treated with tilorone (10 µM) showed a significant reduction in transforming growth factor-β induced collagen synthesis compared to untreated fibroblasts. To determine if this finding could be recapitulated in vivo, mice with established pathological remodelling due to four weeks of transverse aortic constriction (TAC) were administered tilorone (50 mg/kg, i.p) or saline every third day for eight weeks. Treatment with tilorone was associated with attenuation of fibrosis (assessed by Masson's trichrome stain), a favourable cardiac gene expression profile and no further deterioration of cardiac systolic function determined by echocardiography compared to saline treated TAC mice. These data demonstrate that tilorone has anti-fibrotic actions in human cardiac fibroblasts and the adult mouse heart, and represents a potential novel therapy to treat fibrosis associated with heart failure.
Collapse
Affiliation(s)
- Duncan Horlock
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
| | - David M. Kaye
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- Department of Cardiology, Alfred Hospital, Melbourne, VIC 3004, Australia
- Department of Medicine, Monash University, Clayton, VIC 3800, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Catherine E. Winbanks
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
| | - Xiao-Ming Gao
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Disease in Central Asia, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi 830054, China
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Daniel G. Donner
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Paul Gregorevic
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- Centre for Muscle Research, Department of Physiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Julie R. McMullen
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- Department of Medicine, Monash University, Clayton, VIC 3800, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia
- Department of Physiology, Monash University, Clayton, VIC 3800, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia
| | - Bianca C. Bernardo
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia
- Department of Paediatrics, University of Melbourne, VIC 3010, Australia
- Correspondence: ; Tel.: +61-(0)3-8532-1167; Fax: +61-(0)3-8532-1100
| |
Collapse
|
19
|
JNK and p38 Inhibitors Prevent Transforming Growth Factor-β1-Induced Myofibroblast Transdifferentiation in Human Graves' Orbital Fibroblasts. Int J Mol Sci 2021; 22:ijms22062952. [PMID: 33799469 PMCID: PMC7998969 DOI: 10.3390/ijms22062952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1)-induced myofibroblast transdifferentiation from orbital fibroblasts is known to dominate tissue remodeling and fibrosis in Graves’ ophthalmopathy (GO). However, the signaling pathways through which TGF-β1 activates Graves’ orbital fibroblasts remain unclear. This study investigated the role of the mitogen-activated protein kinase (MAPK) pathway in TGF-β1-induced myofibroblast transdifferentiation in human Graves’ orbital fibroblasts. The MAPK pathway was assessed by measuring the phosphorylation of p38, c-Jun N-terminal kinase (JNK), and extracellular-signal-regulated kinase (ERK) by Western blots. The expression of connective tissue growth factor (CTGF), α-smooth muscle actin (α-SMA), and fibronectin representing fibrogenesis was estimated. The activities of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) responsible for extracellular matrix (ECM) metabolism were analyzed. Specific pharmacologic kinase inhibitors were used to confirm the involvement of the MAPK pathway. After treatment with TGF-β1, the phosphorylation levels of p38 and JNK, but not ERK, were increased. CTGF, α-SMA, and fibronectin, as well as TIMP-1 and TIMP-3, were upregulated, whereas the activities of MMP-2/-9 were inhibited. The effects of TGF-β1 on the expression of these factors were eliminated by p38 and JNK inhibitors. The results suggested that TGF-β1 could induce myofibroblast transdifferentiation in human Graves’ orbital fibroblasts through the p38 and JNK pathways.
Collapse
|
20
|
Leguit RJ, Raymakers RAP, Hebeda KM, Goldschmeding R. CCN2 (Cellular Communication Network factor 2) in the bone marrow microenvironment, normal and malignant hematopoiesis. J Cell Commun Signal 2021; 15:25-56. [PMID: 33428075 PMCID: PMC7798015 DOI: 10.1007/s12079-020-00602-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 12/20/2020] [Indexed: 02/06/2023] Open
Abstract
CCN2, formerly termed Connective Tissue Growth Factor, is a protein belonging to the Cellular Communication Network (CCN)-family of secreted extracellular matrix-associated proteins. As a matricellular protein it is mainly considered to be active as a modifier of signaling activity of several different signaling pathways and as an orchestrator of their cross-talk. Furthermore, CCN2 and its fragments have been implicated in the regulation of a multitude of biological processes, including cell proliferation, differentiation, adhesion, migration, cell survival, apoptosis and the production of extracellular matrix products, as well as in more complex processes such as embryonic development, angiogenesis, chondrogenesis, osteogenesis, fibrosis, mechanotransduction and inflammation. Its function is complex and context dependent, depending on cell type, state of differentiation and microenvironmental context. CCN2 plays a role in many diseases, especially those associated with fibrosis, but has also been implicated in many different forms of cancer. In the bone marrow (BM), CCN2 is highly expressed in mesenchymal stem/stromal cells (MSCs). CCN2 is important for MSC function, supporting its proliferation, migration and differentiation. In addition, stromal CCN2 supports the maintenance and longtime survival of hematopoietic stem cells, and in the presence of interleukin 7, stimulates the differentiation of pro-B lymphocytes into pre-B lymphocytes. Overexpression of CCN2 is seen in the majority of B-acute lymphoblastic leukemias, especially in certain cytogenetic subgroups associated with poor outcome. In acute myeloid leukemia, CCN2 expression is increased in MSCs, which has been associated with leukemic engraftment in vivo. In this review, the complex function of CCN2 in the BM microenvironment and in normal as well as malignant hematopoiesis is discussed. In addition, an overview is given of data on the remaining CCN family members regarding normal and malignant hematopoiesis, having many similarities and some differences in their function.
Collapse
Affiliation(s)
- Roos J. Leguit
- Department of Pathology, University Medical Center Utrecht, H04-312, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| | - Reinier A. P. Raymakers
- Department of Hematology, UMCU Cancer Center, Heidelberglaan 100 B02.226, 3584 CX Utrecht, The Netherlands
| | - Konnie M. Hebeda
- Department of Pathology, Radboud University Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Centre Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
21
|
Molecular Changes Underlying Hypertrophic Scarring Following Burns Involve Specific Deregulations at All Wound Healing Stages (Inflammation, Proliferation and Maturation). Int J Mol Sci 2021; 22:ijms22020897. [PMID: 33477421 PMCID: PMC7831008 DOI: 10.3390/ijms22020897] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/14/2022] Open
Abstract
Excessive connective tissue accumulation, a hallmark of hypertrophic scaring, results in progressive deterioration of the structure and function of organs. It can also be seen during tumor growth and other fibroproliferative disorders. These processes result from a wide spectrum of cross-talks between mesenchymal, epithelial and inflammatory/immune cells that have not yet been fully understood. In the present review, we aimed to describe the molecular features of fibroblasts and their interactions with immune and epithelial cells and extracellular matrix. We also compared different types of fibroblasts and their roles in skin repair and regeneration following burn injury. In summary, here we briefly review molecular changes underlying hypertrophic scarring following burns throughout all basic wound healing stages, i.e. during inflammation, proliferation and maturation.
Collapse
|
22
|
Grossi S, Grimaldi A, Congiu T, Parnigoni A, Campanelli G, Campomenosi P. Human Primary Dermal Fibroblasts Interacting with 3-Dimensional Matrices for Surgical Application Show Specific Growth and Gene Expression Programs. Int J Mol Sci 2021; 22:ijms22020526. [PMID: 33430241 PMCID: PMC7825678 DOI: 10.3390/ijms22020526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/28/2020] [Accepted: 01/05/2021] [Indexed: 01/06/2023] Open
Abstract
Several types of 3-dimensional (3D) biological matrices are employed for clinical and surgical applications, but few indications are available to guide surgeons in the choice among these materials. Here we compare the in vitro growth of human primary fibroblasts on different biological matrices commonly used for clinical and surgical applications and the activation of specific molecular pathways over 30 days of growth. Morphological analyses by Scanning Electron Microscopy and proliferation curves showed that fibroblasts have different ability to attach and proliferate on the different biological matrices. They activated similar gene expression programs, reducing the expression of collagen genes and myofibroblast differentiation markers compared to fibroblasts grown in 2D. However, differences among 3D matrices were observed in the expression of specific metalloproteinases and interleukin-6. Indeed, cell proliferation and expression of matrix degrading enzymes occur in the initial steps of interaction between fibroblast and the investigated meshes, whereas collagen and interleukin-6 expression appear to start later. The data reported here highlight features of fibroblasts grown on different 3D biological matrices and warrant further studies to understand how these findings may be used to help the clinicians choose the correct material for specific applications.
Collapse
Affiliation(s)
- Sarah Grossi
- Department of Biotechnology and Life Sciences, University of Insubria, DBSV, Via J.H. Dunant 3, 21100 Varese, Italy; (S.G.); (A.G.); (A.P.)
| | - Annalisa Grimaldi
- Department of Biotechnology and Life Sciences, University of Insubria, DBSV, Via J.H. Dunant 3, 21100 Varese, Italy; (S.G.); (A.G.); (A.P.)
| | - Terenzio Congiu
- Department of Surgical Sciences, University of Cagliari, 09100 Cagliari, Italy;
| | - Arianna Parnigoni
- Department of Biotechnology and Life Sciences, University of Insubria, DBSV, Via J.H. Dunant 3, 21100 Varese, Italy; (S.G.); (A.G.); (A.P.)
| | - Giampiero Campanelli
- Milano Hernia Center, Department of Surgical Science, Istituto Clinico Sant’Ambrogio, Via Luigi Giuseppe Faravelli 16, 20149 Milan, Italy;
- Department of Medicine and Surgery, University of Insubria, DMC, Via Guicciardini 9, 21100 Varese, Italy
| | - Paola Campomenosi
- Department of Biotechnology and Life Sciences, University of Insubria, DBSV, Via J.H. Dunant 3, 21100 Varese, Italy; (S.G.); (A.G.); (A.P.)
- Correspondence: ; Tel.: +39-0332-421322
| |
Collapse
|
23
|
Chen Z, Zhang N, Chu HY, Yu Y, Zhang ZK, Zhang G, Zhang BT. Connective Tissue Growth Factor: From Molecular Understandings to Drug Discovery. Front Cell Dev Biol 2020; 8:593269. [PMID: 33195264 PMCID: PMC7658337 DOI: 10.3389/fcell.2020.593269] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/09/2020] [Indexed: 01/18/2023] Open
Abstract
Connective tissue growth factor (CTGF) is a key signaling and regulatory molecule involved in different biological processes, such as cell proliferation, angiogenesis, and wound healing, as well as multiple pathologies, such as tumor development and tissue fibrosis. Although the underlying mechanisms of CTGF remain incompletely understood, a commonly accepted theory is that the interactions between different protein domains in CTGF and other various regulatory proteins and ligands contribute to its variety of functions. Here, we highlight the structure of each domain of CTGF and its biology functions in physiological conditions. We further summarized main diseases that are deeply influenced by CTGF domains and the potential targets of these diseases. Finally, we address the advantages and disadvantages of current drugs targeting CTGF and provide the perspective for the drug discovery of the next generation of CTGF inhibitors based on aptamers.
Collapse
Affiliation(s)
- Zihao Chen
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ning Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hang Yin Chu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Zong-Kang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Bao-Ting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
24
|
Pemmari T, Ivanova L, May U, Lingasamy P, Tobi A, Pasternack A, Prince S, Ritvos O, Makkapati S, Teesalu T, Cairo MS, Järvinen TAH, Liao Y. Exposed CendR Domain in Homing Peptide Yields Skin-Targeted Therapeutic in Epidermolysis Bullosa. Mol Ther 2020; 28:1833-1845. [PMID: 32497513 PMCID: PMC7403337 DOI: 10.1016/j.ymthe.2020.05.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/05/2020] [Accepted: 05/14/2020] [Indexed: 01/12/2023] Open
Abstract
Systemic skin-selective therapeutics would be a major advancement in the treatment of diseases affecting the entire skin, such as recessive dystrophic epidermolysis bullosa (RDEB), which is caused by mutations in the COL7A1 gene and manifests in transforming growth factor-β (TGF-β)-driven fibrosis and malignant transformation. Homing peptides containing a C-terminal R/KXXR/K motif (C-end rule [CendR] sequence) activate an extravasation and tissue penetration pathway for tumor-specific drug delivery. We have previously described a homing peptide CRKDKC (CRK) that contains a cryptic CendR motif and homes to angiogenic blood vessels in wounds and tumors, but it cannot penetrate cells or tissues. In this study, we demonstrate that removal of the cysteine from CRK to expose the CendR sequence confers the peptide novel ability to home to normal skin. Fusion of the truncated CRK (tCRK) peptide to the C terminus of an extracellular matrix protein decorin (DCN), a natural TGF-β inhibitor, resulted in a skin-homing therapeutic molecule (DCN-tCRK). Systemic DCN-tCRK administration in RDEB mice led to inhibition of TGF-β signaling in the skin and significant improvement in the survival of RDEB mice. These results suggest that DCN-tCRK has the potential to be utilized as a novel therapeutic compound for the treatment of dermatological diseases such as RDEB.
Collapse
Affiliation(s)
- Toini Pemmari
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, 33520 Tampere, Finland
| | - Larisa Ivanova
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Ulrike May
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, 33520 Tampere, Finland
| | - Prakash Lingasamy
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Allan Tobi
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Anja Pasternack
- Department of Physiology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Stuart Prince
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, 33520 Tampere, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Shreya Makkapati
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Tambet Teesalu
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; Center for Nanomedicine, University of California, Santa Barbara, CA 93106, USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA; Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA; Department of Pathology, New York Medical College, Valhalla, NY 10595, USA; Department of Medicine, New York Medical College, Valhalla, NY 10595, USA; Deparmtent of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA
| | - Tero A H Järvinen
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, 33520 Tampere, Finland.
| | - Yanling Liao
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA.
| |
Collapse
|
25
|
Antibiotics Used in Regenerative Endodontics Modify Immune Response of Macrophages to Bacterial Infection. J Endod 2019; 45:1349-1356. [DOI: 10.1016/j.joen.2019.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/08/2019] [Accepted: 08/06/2019] [Indexed: 12/24/2022]
|
26
|
Connective Tissue Growth Factor Is Related to All-cause Mortality in Hemodialysis Patients and Is Lowered by On-line Hemodiafiltration: Results from the Convective Transport Study. Toxins (Basel) 2019; 11:toxins11050268. [PMID: 31086050 PMCID: PMC6563290 DOI: 10.3390/toxins11050268] [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] [Received: 04/05/2019] [Revised: 04/26/2019] [Accepted: 05/08/2019] [Indexed: 11/17/2022] Open
Abstract
Connective tissue growth factor (CTGF) plays a key role in the pathogenesis of tissue fibrosis. The aminoterminal fragment of CTGF is a middle molecule that accumulates in chronic kidney disease. The aims of this study are to explore determinants of plasma CTGF in hemodialysis (HD) patients, investigate whether CTGF relates to all-cause mortality in HD patients, and investigate whether online-hemodiafiltration (HDF) lowers CTGF. Data from 404 patients participating in the CONvective TRAnsport STudy (CONTRAST) were analyzed. Patients were randomized to low-flux HD or HDF. Pre-dialysis CTGF was measured by sandwich ELISA at baseline, after six and 12 months. CTGF was inversely related in multivariable analysis to glomerular filtration rate (GFR) (p < 0.001) and positively to cardiovascular disease (CVD) (p = 0.006), dialysis vintage (p < 0.001), interleukin-6 (p < 0.001), beta-2-microglobulin (p = 0.045), polycystic kidney disease (p < 0.001), tubulointerstitial nephritis (p = 0.002), and renal vascular disease (p = 0.041). Patients in the highest quartile had a higher mortality risk compared to those in the lowest quartile (HR 1.7, 95% CI: 1.02-2.88, p = 0.043). HDF lowered CTGF with 4.8% between baseline and six months, whereas during HD, CTGF increased with 4.9% (p < 0.001). In conclusion, in HD patients, CTGF is related to GFR, CVD and underlying renal disease and increased the risk of all-cause mortality. HDF reduces CTGF.
Collapse
|
27
|
Ramani K, Biswas PS. Interleukin-17: Friend or foe in organ fibrosis. Cytokine 2019; 120:282-288. [PMID: 30772195 DOI: 10.1016/j.cyto.2018.11.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 02/07/2023]
Abstract
Fibrosis affects all vital organs accounting for a staggering 45% of deaths worldwide and no effective therapies are currently available. Unresolved inflammation triggers downstream signaling events that lead to organ fibrosis. In recent years, proinflammatory cytokine Interleukin-17 (IL-17) has been implicated in several chronic inflammatory diseases that often culminate in organ damage followed by impaired wound healing and fibrosis. In this review, we outline the contribution of the IL-17 in mediating fibrotic diseases in various organs. A comprehensive understanding of the inflammatory events, and particularly the details of IL-17 signaling in vivo, could be beneficial in designing new therapeutic or preventive approaches to treat fibrosis. Additionally, understanding organ-specific differences in IL-17 activity could lead to targeted therapies and help spare other organs from unwanted side effects.
Collapse
Affiliation(s)
- Kritika Ramani
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Partha S Biswas
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
28
|
Chang YM, Tamilselvi S, Lin HJ, Tsai CC, Lin YM, Day CH, Viswanadha VP, Chang HN, Kuo WW, Huang CY. Alpinia oxyphylla Miq extract ameliorates cardiac fibrosis associated with D-galactose induced aging in rats. ENVIRONMENTAL TOXICOLOGY 2019; 34:172-178. [PMID: 30367734 DOI: 10.1002/tox.22671] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/14/2018] [Accepted: 09/16/2018] [Indexed: 06/08/2023]
Abstract
Cardiac fibrosis is a common pathophysiological process observed during chronic and stress-induced acceleration of cardiac aging. Fibrosis is a necessary process during wound healing and tissue repair. However, its deposition in organs would proceed to scarring and organ damage. Here Alpinate Oxyphyllae Fructus (AOF), a Chinese medicine extract was used to protect aging heart from collagen accumulation. About 8 weeks old, male SD rats were randomly divided into (i) Control, (ii) D-galactose induced aging (IA), (iii) IA + AOF 50 (AOF low, AL), (iv) IA + AOF 100 (AOF medium, AM), (v) IA + AOF 150 (AOF high, AH) mg/kg/day, AOF was administered orally. After 8 weeks rats were sacrificed and hearts were collected. Results showed collagen deposition and up-regulation of matrix metalloproteinases-MMP-2 and -9 in D-galactose-induced aging rats. Furthermore, western blotting and immunostaining were also confirmed the upregulation of TGF-β1 mediated fibrosis in aging induced rats. However, collagen deposition and fibrosis were significantly decreased by AOF treatments (AM and AH). AOF treatments salvaged the cardiac fibrosis. Hence, AOF might be a potential therapeutic agent in the prevention of cardiac fibrosis associated with aging. The protective effects of AOF might have promising results in anti-aging treatments.
Collapse
Affiliation(s)
- Yung-Ming Chang
- The School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung, Taiwan
- Chinese Medicine Department, E-DA Hospital, Kaohsiung, Taiwan
- 1PT Biotechnology Co., Ltd., Taichung, Taiwan
| | - Shanmugam Tamilselvi
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Hung-Jen Lin
- Departments of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chin-Chuan Tsai
- The School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung, Taiwan
- Chinese Medicine Department, E-DA Hospital, Kaohsiung, Taiwan
| | - Yueh-Min Lin
- Department of pathology, Changhua Christian Hospital, Changhua, Taiwan
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | | | | | - Hsin-Nung Chang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Departments of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
- Department of Biological Science and Technology, Asia University, Taichung, Taiwan
| |
Collapse
|
29
|
Xiong S, Gao H, Qin L, Jia Y, Gao M, Ren L. Microgrooved collagen-based corneal scaffold for promoting collective cell migration and antifibrosis. RSC Adv 2019; 9:29463-29473. [PMID: 35528407 PMCID: PMC9071845 DOI: 10.1039/c9ra04009a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 07/19/2019] [Indexed: 02/04/2023] Open
Abstract
Microgrooved collagen membrane can effectively promote the epithelialization of corneal epithelial cells and inhibit the fibrosis of corneal stromal cells.
Collapse
Affiliation(s)
- Sijia Xiong
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510641
- China
- National Engineering Research Centre for Tissue Restoration and Reconstruction
| | - Huichang Gao
- School of Medicine
- South China University of Technology
- Guangzhou 510006
- China
| | - Lanfeng Qin
- National Engineering Research Centre for Tissue Restoration and Reconstruction
- Guangzhou 510006
- China
- Guangdong Province Key Laboratory of Biomedical Engineering
- South China University of Technology
| | - Yongguang Jia
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510641
- China
- National Engineering Research Centre for Tissue Restoration and Reconstruction
| | - Meng Gao
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510641
- China
- National Engineering Research Centre for Tissue Restoration and Reconstruction
| | - Li Ren
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510641
- China
- National Engineering Research Centre for Tissue Restoration and Reconstruction
| |
Collapse
|
30
|
Kaasbøll OJ, Gadicherla AK, Wang JH, Monsen VT, Hagelin EMV, Dong MQ, Attramadal H. Connective tissue growth factor (CCN2) is a matricellular preproprotein controlled by proteolytic activation. J Biol Chem 2018; 293:17953-17970. [PMID: 30262666 DOI: 10.1074/jbc.ra118.004559] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/23/2018] [Indexed: 11/06/2022] Open
Abstract
Connective tissue growth factor (CTGF; now often referred to as CCN2) is a secreted protein predominantly expressed during development, in various pathological conditions that involve enhanced fibrogenesis and tissue fibrosis, and in several cancers and is currently an emerging target in several early-phase clinical trials. Tissues containing high CCN2 activities often display smaller degradation products of full-length CCN2 (FL-CCN2). Interpretation of these observations is complicated by the fact that a uniform protein structure that defines biologically active CCN2 has not yet been resolved. Here, using DG44 CHO cells engineered to produce and secrete FL-CCN2 and cell signaling and cell physiological activity assays, we demonstrate that FL-CCN2 is itself an inactive precursor and that a proteolytic fragment comprising domains III (thrombospondin type 1 repeat) and IV (cystine knot) appears to convey all biologically relevant activities of CCN2. In congruence with these findings, purified FL-CCN2 could be cleaved and activated following incubation with matrix metalloproteinase activities. Furthermore, the C-terminal fragment of CCN2 (domains III and IV) also formed homodimers that were ∼20-fold more potent than the monomeric form in activating intracellular phosphokinase cascades. The homodimer elicited activation of fibroblast migration, stimulated assembly of focal adhesion complexes, enhanced RANKL-induced osteoclast differentiation of RAW264.7 cells, and promoted mammosphere formation of MCF-7 mammary cancer cells. In conclusion, CCN2 is synthesized and secreted as a preproprotein that is autoinhibited by its two N-terminal domains and requires proteolytic processing and homodimerization to become fully biologically active.
Collapse
Affiliation(s)
- Ole Jørgen Kaasbøll
- From the Institute for Surgical Research, Oslo University Hospital and University of Oslo, NO-0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, NO-0316 Oslo, Norway
| | - Ashish K Gadicherla
- From the Institute for Surgical Research, Oslo University Hospital and University of Oslo, NO-0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, NO-0316 Oslo, Norway
| | - Jian-Hua Wang
- National Institute of Biological Sciences, 102206 Beijing, China
| | - Vivi Talstad Monsen
- From the Institute for Surgical Research, Oslo University Hospital and University of Oslo, NO-0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, NO-0316 Oslo, Norway
| | - Else Marie Valbjørn Hagelin
- From the Institute for Surgical Research, Oslo University Hospital and University of Oslo, NO-0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, NO-0316 Oslo, Norway
| | - Meng-Qiu Dong
- National Institute of Biological Sciences, 102206 Beijing, China
| | - Håvard Attramadal
- From the Institute for Surgical Research, Oslo University Hospital and University of Oslo, NO-0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, NO-0316 Oslo, Norway.
| |
Collapse
|
31
|
Tsai CC, Wu SB, Kau HC, Wei YH. Essential role of connective tissue growth factor (CTGF) in transforming growth factor-β1 (TGF-β1)-induced myofibroblast transdifferentiation from Graves' orbital fibroblasts. Sci Rep 2018; 8:7276. [PMID: 29739987 PMCID: PMC5940888 DOI: 10.1038/s41598-018-25370-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 04/11/2018] [Indexed: 12/12/2022] Open
Abstract
Connective tissue growth factor (CTGF) associated with transforming growth factor-β (TGF-β) play a pivotal role in the pathophysiology of many fibrotic disorders. However, it is not clear whether this interaction also takes place in GO. In this study, we investigated the role of CTGF in TGF-β-induced extracellular matrix production and myofibroblast transdifferentiation in Graves’ orbital fibroblasts. By Western blot analysis, we demonstrated that TGF-β1 induced the expression of CTGF, fibronectin, and alpha-smooth muscle actin (α-SMA) in Graves’ orbital fibroblasts. In addition, the protein levels of fibronectin and α-SMA in Graves’ orbital fibroblasts were also increased after treatment with a recombinant human protein CTGF (rhCTGF). Moreover, we transfected the orbital fibroblasts with a small hairpin RNA of CTGF gene (shCTGF) to knockdown the expression levels of CTGF, which showed that knockdown of CTGF significantly diminished TGF-β1-induced expression of CTGF, fibronectin and α-SMA proteins in Graves’ orbital fibroblasts. Furthermore, the addition of rhCTGF to the shCTGF-transfected orbital fibroblasts could restore TGF-β1-induced expression of fibronectin and α-SMA proteins. Our findings demonstrate that CTGF is an essential downstream mediator for TGF-β1-induced extracellular matrix production and myofibroblast transdifferentiation in Graves’ orbital fibroblasts and thus may provide with a potential therapeutic target for treatment of GO.
Collapse
Affiliation(s)
- Chieh-Chih Tsai
- Department of Ophthalmology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan.
| | - Shi-Bei Wu
- Biomedical Commercialization Center, Taipei Medical University, Taipei, Taiwan
| | - Hui-Chuan Kau
- Department of Ophthalmology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan.,Department of Ophthalmology, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Yau-Huei Wei
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City, Taiwan
| |
Collapse
|
32
|
Zhou T, Che D, Lan Y, Fang Z, Xie J, Gong H, Li C, Feng J, Hong H, Qi W, Ma C, Yang Z, Cai W, Zhong J, Ma J, Yang X, Gao G. Mesenchymal marker expression is elevated in Müller cells exposed to high glucose and in animal models of diabetic retinopathy. Oncotarget 2018; 8:4582-4594. [PMID: 27999189 PMCID: PMC5354856 DOI: 10.18632/oncotarget.13945] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 12/01/2016] [Indexed: 12/21/2022] Open
Abstract
Müller cells are retinal glial cells and exhibit a fibroblast-like phenotype and ability to migrate in diabetic retinopathy (DR). However, expression of mesenchymal markers, which promote fibrosis in various organs, has not been characterized in the diabetic retina. We examined changes in the expression of these markers in Müller cells exposed to high glucose and in animal models of diabetic retinopathy. High glucose conditions increased mesenchymal maker expression and migration in Müller cells. Snail, N-cadherin, Vimentin, β-catenin, and α-smooth muscle actin (α-SMA) levels were all dramatically increased in retinas from humans with diabetic retinopathy (DR) and from DR mouse models. In addition, Snail overexpression increased the expression of connective tissue growth factor (CTGF) and fibronectin, while Snail knockdown attenuated high glucose-induced increases in fibronectin and CTGF expression. These results demonstrate for the first time that mesenchymal markers are upregulated in retinas from a diabetic mouse model, and that Snail and N-cadherin levels are also increased in Müller cells exposed to high glucose. This suggests mesenchymal proteins may play a crucial role in the development of DR.
Collapse
Affiliation(s)
- Ti Zhou
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Di Che
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yuqing Lan
- Department of Ophthalmology, Second Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhenzhen Fang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinye Xie
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - HaiJun Gong
- Department of Ophthalmology, Second Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - ChaoYang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Juan Feng
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Honghai Hong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weiwei Qi
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Caiqi Ma
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhonghan Yang
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - WeiBin Cai
- Guangdong Engineering and Technology Research Center for Disease-Model Animals, Sun Yat-Sen University, Guangzhou, China
| | - Jun Zhong
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jianxing Ma
- Department of Physiology, University of Oklahoma, Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Xia Yang
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Functional Molecules from Marine Microorganisms (Sun Yat-sen University), Department of Education of Guangdong Province, Guangzhou, China
| | - Guoquan Gao
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,China Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| |
Collapse
|
33
|
Matricellular CCN6 (WISP3) protein: a tumor suppressor for mammary metaplastic carcinomas. J Cell Commun Signal 2018; 12:13-19. [PMID: 29357008 DOI: 10.1007/s12079-018-0451-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 02/07/2023] Open
Abstract
Located at 6q22-23, Ccn6 (WISP3) encodes for a matrix-associated protein of the CCN family, characterized by regulatory, rather than structural, roles in development and cancer. CCN6, the least studied member of the CCN family, shares the conserved multimodular structure of CCN proteins, as well as their tissue and cell-type specific functions. In the breast, CCN6 is a critical regulator of epithelial-to-mesenchymal transitions (EMT) and tumor initiating cells. Studies using human breast cancer tissue samples demonstrated that CCN6 messenger RNA and protein are expressed in normal breast epithelia but reduced or lost in aggressive breast cancer phenotypes, especially inflammatory breast cancer and metaplastic carcinomas. Metaplastic carcinomas are mesenchymal-like triple negative breast carcinomas, enriched for markers of EMT and stemness. RNAseq analyses of the TCGA Breast Cancer cohort show reduced CCN6 expression in approximately 50% of metaplastic carcinomas compared to normal breast. Our group identified frameshift mutations of Ccn6 in a subset of human metaplastic breast carcinoma. Importantly, conditional, mammary epithelial-cell specific ccn6 (wisp3) knockout mice develop invasive high-grade mammary carcinomas that recapitulate human spindle cell metaplastic carcinomas, demonstrating a tumor suppressor function for ccn6. Our studies on CCN6 functions in metaplastic carcinoma highlight the potential of CCN6 as a novel therapeutic approach for this specific type of breast cancer.
Collapse
|
34
|
Rayego-Mateos S, Morgado-Pascual JL, Rodrigues-Diez RR, Rodrigues-Diez R, Falke LL, Mezzano S, Ortiz A, Egido J, Goldschmeding R, Ruiz-Ortega M. Connective tissue growth factor induces renal fibrosis via epidermal growth factor receptor activation. J Pathol 2018; 244:227-241. [PMID: 29160908 DOI: 10.1002/path.5007] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/20/2017] [Accepted: 11/14/2017] [Indexed: 01/04/2023]
Abstract
Connective tissue growth factor (CCN2/CTGF) is a matricellular protein that is overexpressed in progressive human renal diseases, mainly in fibrotic areas. In vitro studies have demonstrated that CCN2 regulates the production of extracellular matrix (ECM) proteins and epithelial-mesenchymal transition (EMT), and could therefore contribute to renal fibrosis. CCN2 blockade ameliorates experimental renal damage, including diminution of ECM accumulation. We have reported that CCN2 and its C-terminal degradation product CCN2(IV) bind to epidermal growth factor receptor (EGFR) to modulate renal inflammation. However, the receptor involved in CCN2 profibrotic actions has not been described so far. Using a murine model of systemic administration of CCN2(IV), we have unveiled a fibrotic response in the kidney that was diminished by EGFR blockade. Additionally, in conditional CCN2 knockout mice, renal fibrosis elicited by folic acid-induced renal damage was prevented, and this was linked to inhibition of EGFR pathway activation. Our in vitro studies demonstrated a direct effect of CCN2 via the EGFR pathway on ECM production by fibroblasts and the induction of EMT in tubular epithelial cells. Our studies clearly show that the EGFR regulates CCN2 fibrotic signalling in the kidney, and suggest that EGFR pathway blockade could be a potential therapeutic option to block CCN2-mediated profibrotic effects in renal diseases. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory. School of Medicine, Universidad Autónoma Madrid, Madrid, Spain
| | - José Luis Morgado-Pascual
- Cellular Biology in Renal Diseases Laboratory. School of Medicine, Universidad Autónoma Madrid, Madrid, Spain
| | | | - Raquel Rodrigues-Diez
- Cellular Biology in Renal Diseases Laboratory. School of Medicine, Universidad Autónoma Madrid, Madrid, Spain
| | - Lucas L Falke
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sergio Mezzano
- Division of Nephrology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Alberto Ortiz
- IIS-Fundación Jiménez Díaz-UAM, School of Medicine, UAM, Madrid, Spain
| | - Jesús Egido
- IIS-Fundación Jiménez Díaz-UAM, School of Medicine, UAM, Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory. School of Medicine, Universidad Autónoma Madrid, Madrid, Spain
| |
Collapse
|
35
|
Ebnerasuly F, Hajebrahimi Z, Tabaie SM, Darbouy M. Effect of Simulated Microgravity Conditions on Differentiation of Adipose Derived Stem Cells towards Fibroblasts Using Connective Tissue Growth Factor. IRANIAN JOURNAL OF BIOTECHNOLOGY 2017; 15:241-251. [PMID: 29845076 DOI: 10.15171/ijb.1747] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 06/14/2017] [Accepted: 07/08/2017] [Indexed: 12/12/2022]
Abstract
Background: Mesenchymal stem cells (MSCs) are multipotent cells able to differentiating into a variety of mesenchymal tissues including osteoblasts, adipocytes and several other tissues. Objectives: Differentiation of MSCs into fibroblast cells in vitro is an attractive strategy to achieve fibroblast cell and use them for purposes such as regeneration medicine. The goal of this study was investigate the simulated microgravity effect on differentiation of Adipose Derived Stem Cells (ADSCs) to fibroblasts. Materials and Methods: To fibroblast differentiation 100 ng.mL-1 of connective tissue growth factor (CTGF), and for simulation microgravity, 2D clinostat was used. After isolation the human ADSCs from adipose, cells were passaged, and at passages 3 they were used for characterization and subsequent steps. After 7 days of CTGF and simulated microgravity treatment, proliferation, and differentiation were analyzed collectively by MTT assay, quantitative PCR analyses, and Immunocytochemistry staining. Results: MTT assay revealed that CTGF stimulate the proliferation but simulated microgravity didn't have statistically significant effect on cell proliferation. In RNA level the expression of these genes are investigated: collagen type I (COLI), elastin (ELA), collagen type III (ColIII), Matrix Metalloproteinases I(MMP1), Fibronectin 1 (FN1), CD44, Fibroblast Specific protein (FSP-1), Integrin Subunit Beta 1 (ITGB1), Vimentin (VIM) and Fibrillin (FBN). We found that expression of ELN, FN1, FSP1, COL1A1, ITGB1, MMP1 and COL3A1 in both condition, and VIM and FBN1 just in differentiation medium in normal gravity increased. In protein level the expression of COL III and ELN in simulated microgravity increased. Conclusions: These findings collectively demonstrate that the simulated microgravity condition alters the marker fibroblast gene expression in fibroblast differentiation process.
Collapse
Affiliation(s)
- Farid Ebnerasuly
- Department of Biology, Fars Science and Research Branch , Islamic Azad University, Marvdasht, Iran.,Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Zahra Hajebrahimi
- Aerospace Research Institute, Ministry of Science Research and Technology, Tehran, Iran
| | - Seyed Mehdi Tabaie
- Medical Laser Research Center, Iranian Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Mojtaba Darbouy
- Department of Biology, Fars Science and Research Branch , Islamic Azad University, Marvdasht, Iran.,Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| |
Collapse
|
36
|
Brummer G, Acevedo DS, Hu Q, Portsche M, Fang WB, Yao M, Zinda B, Myers M, Alvarez N, Fields P, Hong Y, Behbod F, Cheng N. Chemokine Signaling Facilitates Early-Stage Breast Cancer Survival and Invasion through Fibroblast-Dependent Mechanisms. Mol Cancer Res 2017; 16:296-308. [PMID: 29133591 DOI: 10.1158/1541-7786.mcr-17-0308] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/20/2017] [Accepted: 11/02/2017] [Indexed: 12/13/2022]
Abstract
Ductal carcinoma in situ (DCIS) is the most common form of breast cancer, with 50,000 cases diagnosed every year in the United States. Overtreatment and undertreatment remain significant clinical challenges in patient care. Identifying key mechanisms associated with DCIS progression could uncover new biomarkers to better predict patient prognosis and improve guided treatment. Chemokines are small soluble molecules that regulate cellular homing through molecular gradients. CCL2-mediated recruitment of CCR2+ macrophages are a well-established mechanism for metastatic progression. Although the CCL2/CCR2 pathway is a therapeutic target of interest, little is known about the role of CCR2 expression in breast cancer. Here, using a mammary intraductal injection (MIND) model to mimic DCIS formation, the role of CCR2 was explored in minimally invasive SUM225 and highly invasive DCIS.com breast cancer cells. CCR2 overexpression increased SUM225 breast cancer survival and invasion associated with accumulation of CCL2 expressing fibroblasts. CCR2-deficient DCIS.com breast cancer cells formed fewer invasive lesions with fewer CCL2+ fibroblasts. Cografting CCL2-deficient fibroblasts with DCIS.com breast cancer cells in the subrenal capsule model inhibited tumor invasion and survival associated with decreased expression of aldehyde dehydrogenase (ALDH1), a proinvasive factor, and decreased expression of HTRA2, a proapoptotic serine protease. Through data mining analysis, high expression of CCR2 and ALDH1 and low HTRA2 expression were correlated with poor prognosis of breast cancer patients.Implications: This study demonstrates that CCR2 overexpression in breast cancer drives early-stage breast cancer progression through stromal-dependent expression of CCL2 with important insight into prognosis and treatment of DCIS. Mol Cancer Res; 16(2); 296-308. ©2017 AACR.
Collapse
Affiliation(s)
- Gage Brummer
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Diana S Acevedo
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Qingting Hu
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Mike Portsche
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Wei Bin Fang
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Min Yao
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Brandon Zinda
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Megan Myers
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Nehemiah Alvarez
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Patrick Fields
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Yan Hong
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Fariba Behbod
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Nikki Cheng
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
37
|
Duan X, Ji M, Deng F, Sun Z, Lin Z. Effects of connective tissue growth factor on human periodontal ligament fibroblasts. Arch Oral Biol 2017; 84:37-44. [PMID: 28941713 DOI: 10.1016/j.archoralbio.2017.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 08/28/2017] [Accepted: 09/16/2017] [Indexed: 12/01/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate the effects of different concentrations of connective tissue growth factor (CTGF) on human periodontal ligament fibroblasts(HPLFs). DESIGN HPLFs were cultured and identified. Then, different concentrations of CTGF (1, 5, 10, 50, 100ng/ml) were added to the HPLF culture. Next, CCK-8 assays, alkaline phosphatase (ALP) assays, hydroxyproline determination, alizarin red staining methods, Transwell chambers and real-time PCR methods were applied to observe the effects of CTGF on the proliferation, ALP activity, synthesis of collagen, formation of mineralized nodules and migration. We also studied expression of ALP, fiber link protein (FN), integrin-binding sialoprotein (IBSP), osteocalcin (OC), and integrin beta 1 (ITGB1) mRNA by HPLFs. Statistical significance was assumed if P<0.05 or P<0.01. RESULTS The addition of CTGF (1, 5, 10ng/ml) remarkably promoted the proliferation and collagen synthesis of HPLFs compared with controls. CTGF (1, 5, 10, 50ng/ml) improved ALP activity of HPLFs, and at all concentrations, CTGF (1, 5, 10, 50, 100ng/ml) improved the expression of ALP, FN, IBSP and ITGB1 mRNA. In addition, CTGF (1, 5, 10, 50, 100ng/ml) promoted the migration of HPLFs, which was dose-dependent, with maximal promotion in the 10ng/ml group (P<0.05 or P<0.01). CONCLUSIONS Thus, in a certain range of concentrations, CTGF can promote the biological effects, including proliferation, migration and collagen synthesis of HPLFs, to promote the differentiation of HPLFs in the process of osteogenesis.
Collapse
Affiliation(s)
- Xuejing Duan
- School of Stomatology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Mei Ji
- School of Stomatology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Fengying Deng
- School of Stomatology, Shandong University, Jinan, Shandong Province, China
| | - Zhe Sun
- School of Stomatology, Shandong University, Jinan, Shandong Province, China
| | - Zhiyong Lin
- School of Stomatology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
38
|
George J, Tsutsumi M, Tsuchishima M. MMP-13 deletion decreases profibrogenic molecules and attenuates N-nitrosodimethylamine-induced liver injury and fibrosis in mice. J Cell Mol Med 2017; 21:3821-3835. [PMID: 28782260 PMCID: PMC5706575 DOI: 10.1111/jcmm.13304] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/05/2017] [Indexed: 12/16/2022] Open
Abstract
Connective tissue growth factor (CTGF) is involved in inflammation, pathogenesis and progression of liver fibrosis. Matrix metalloproteinase‐13 (MMP‐13) cleaves CTGF and releases several fragments, which are more potent than the parent molecule to induce fibrosis. The current study was aimed to elucidate the significance of MMP‐13 and CTGF and their downstream effects in liver injury and fibrosis. Hepatic fibrosis was induced using intraperitoneal injections of N‐nitrosodimethylamine (NDMA) in doses of 10 μg/g body weight on three consecutive days of each week over a period of 4 weeks in both wild‐type (WT) and MMP‐13 knockout mice. Administration of NDMA resulted in marked elevation of AST, ALT, TGF‐β1 and hyaluronic acid in the serum and activation of stellate cells, massive necrosis, deposition of collagen fibres and increase in total collagen in the liver of WT mice with a significant decrease in MMP‐13 knockout mice. Protein and mRNA levels of CTGF, TGF‐β1, α‐SMA and type I collagen and the levels of MMP‐2, MMP‐9 and cleaved products of CTGF were markedly increased in NDMA‐treated WT mice compared to the MMP‐13 knockout mice. Blocking of MMP‐13 with CL‐82198 in hepatic stellate cell cultures resulted in marked decrease of the staining intensity of CTGF as well as protein levels of full‐length CTGF and its C‐terminal fragments and active TGF‐β1. The data demonstrate that MMP‐13 and CTGF play a crucial role in modulation of fibrogenic mediators and promote hepatic fibrogenesis. Furthermore, the study suggests that blocking of MMP‐13 and CTGF has potential therapeutic implications to arrest liver fibrosis.
Collapse
Affiliation(s)
- Joseph George
- Department of Medicine, Division of Molecular Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Mikihiro Tsutsumi
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Mutsumi Tsuchishima
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| |
Collapse
|
39
|
Mokalled MH, Patra C, Dickson AL, Endo T, Stainier DYR, Poss KD. Injury-induced ctgfa directs glial bridging and spinal cord regeneration in zebrafish. Science 2017; 354:630-634. [PMID: 27811277 DOI: 10.1126/science.aaf2679] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022]
Abstract
Unlike mammals, zebrafish efficiently regenerate functional nervous system tissue after major spinal cord injury. Whereas glial scarring presents a roadblock for mammalian spinal cord repair, glial cells in zebrafish form a bridge across severed spinal cord tissue and facilitate regeneration. We performed a genome-wide profiling screen for secreted factors that are up-regulated during zebrafish spinal cord regeneration. We found that connective tissue growth factor a (ctgfa) is induced in and around glial cells that participate in initial bridging events. Mutations in ctgfa disrupted spinal cord repair, and transgenic ctgfa overexpression or local delivery of human CTGF recombinant protein accelerated bridging and functional regeneration. Our study reveals that CTGF is necessary and sufficient to stimulate glial bridging and natural spinal cord regeneration.
Collapse
Affiliation(s)
- Mayssa H Mokalled
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Chinmoy Patra
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Amy L Dickson
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Toyokazu Endo
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Kenneth D Poss
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
40
|
Feng X, Pi L, Sriram S, Schultz GS, Gibson DJ. Connective tissue growth factor is not necessary for haze formation in excimer laser wounded mouse corneas. PLoS One 2017; 12:e0172304. [PMID: 28207886 PMCID: PMC5313228 DOI: 10.1371/journal.pone.0172304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/02/2017] [Indexed: 11/19/2022] Open
Abstract
We sought to determine if connective tissue growth factor (CTGF) is necessary for the formation of corneal haze after corneal injury. Mice with post-natal, tamoxifen-induced, knockout of CTGF were subjected to excimer laser phototherapeutic keratectomy (PTK) and the corneas were allowed to heal. The extent of scaring was observed in non-induced mice, heterozygotes, and full homozygous knockout mice and quantified by macrophotography. The eyes from these mice were collected after euthanization for re-genotyping to control for possible Cre-mosaicism. Primary corneal fibroblasts from CTGF knockout corneas were established in a gel plug assay. The plug was removed, simulating an injury, and the rate of hole closure and the capacity for these cells to form light reflecting cells in response to CTGF and platelet-derived growth factor B (PDGF-B) were tested and compared to wild-type cells. We found that independent of genotype, each group of mice was still capable of forming light reflecting haze in the cornea after laser ablation (p = 0.40). Results from the gel plug closure rate in primary cell cultures of knockout cells were not statistically different from serum starved wild-type cells, independent of treatment. Compared to the serum starved wild-type cells, stimulation with PDGF-BB significantly increased the KO cell culture's light reflection (p = 0.03). Most interestingly, both reflective cultures were positive for α-SMA, but the cellular morphology and levels of α-SMA were distinct and not in proportion to the light reflection seen. This new work demonstrates that corneas without CTGF can still form sub-epithelial haze, and that the light reflecting phenotype can be reproduced in culture. These data support the possibilities of growth factor redundancy and that multiple pro-haze pathways exist.
Collapse
Affiliation(s)
- Xiaodi Feng
- Institute of Wound Research, University of Florida, Gainesville, Florida, United States of America
| | - Liya Pi
- Department of Pediatrics, University of Florida, Gainesville, Florida, United States of America
| | - Sriniwas Sriram
- Department of Biomedical Engineering, University of Florida, Gainesville, Florida, United States of America
| | - Gregory S. Schultz
- Institute of Wound Research, University of Florida, Gainesville, Florida, United States of America
| | - Daniel J. Gibson
- Institute of Wound Research, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
41
|
Abstract
I introduce the general structures and functions of CCN proteins and possible molecular mechanisms regarding the unique biological actions of this new family of signaling regulators, which may be referred to as "signal conductors." Relevance to pathology is also briefly introduced. The information provided in this overview should be useful for readers of the following chapters.
Collapse
Affiliation(s)
- Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School/Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8525, Japan.
| |
Collapse
|
42
|
Sakai N, Chun J, Duffield JS, Lagares D, Wada T, Luster AD, Tager AM. Lysophosphatidic acid signaling through its receptor initiates profibrotic epithelial cell fibroblast communication mediated by epithelial cell derived connective tissue growth factor. Kidney Int 2016; 91:628-641. [PMID: 27927603 DOI: 10.1016/j.kint.2016.09.030] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 08/22/2016] [Accepted: 09/15/2016] [Indexed: 02/06/2023]
Abstract
The expansion of the fibroblast pool is a critical step in organ fibrosis, but the mechanisms driving expansion remain to be fully clarified. We previously showed that lysophosphatidic acid (LPA) signaling through its receptor LPA1 expressed on fibroblasts directly induces the recruitment of these cells. Here we tested whether LPA-LPA1 signaling drives fibroblast proliferation and activation during the development of renal fibrosis. LPA1-deficient (LPA1-/-) or -sufficient (LPA1+/+) mice were crossed to mice with green fluorescent protein expression (GFP) driven by the type I procollagen promoter (Col-GFP) to identify fibroblasts. Unilateral ureteral obstruction-induced increases in renal collagen were significantly, though not completely, attenuated in LPA1-/-Col-GFP mice, as were the accumulations of both fibroblasts and myofibroblasts. Connective tissue growth factor was detected mainly in tubular epithelial cells, and its levels were suppressed in LPA1-/-Col-GFP mice. LPA-LPA1 signaling directly induced connective tissue growth factor expression in primary proximal tubular epithelial cells, through a myocardin-related transcription factor-serum response factor pathway. Proximal tubular epithelial cell-derived connective tissue growth factor mediated renal fibroblast proliferation and myofibroblast differentiation. Administration of an inhibitor of myocardin-related transcription factor/serum response factor suppressed obstruction-induced renal fibrosis. Thus, targeting LPA-LPA1 signaling and/or myocardin-related transcription factor/serum response factor-induced transcription could be promising therapeutic strategies for renal fibrosis.
Collapse
Affiliation(s)
- Norihiko Sakai
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Division of Nephrology, Kanazawa University Hospital, Kanazawa, Japan; Division of Blood Purification, Kanazawa University Hospital, Kanazawa, Japan.
| | - Jerold Chun
- Department of Molecular Biology, Helen L. Dorris Institute for Neurological and Psychiatric Disorders, The Scripps Research Institute, La Jolla, California, USA
| | - Jeremy S Duffield
- Division of Nephrology, Department of Medicine, Center for Lung Biology and Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA; Biogen, Cambridge, Massachusetts, USA
| | - David Lagares
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Takashi Wada
- Division of Nephrology, Kanazawa University Hospital, Kanazawa, Japan; Department of Laboratory Medicine and Nephrology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew M Tager
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Fibrosis Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
43
|
Jiang J, Leong NL, Khalique U, Phan TM, Lyons KM, Luck JV. Connective tissue growth factor (CTGF/CCN2) in haemophilic arthropathy and arthrofibrosis: a histological analysis. Haemophilia 2016; 22:e527-e536. [PMID: 27704689 DOI: 10.1111/hae.13049] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2016] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Joint haemorrhage is the principal clinical manifestation of haemophilia frequently leading to advanced arthropathy and arthrofibrosis, resulting in severe disability. The degree and prevalence of arthrofibrosis in hemophilic arthropathy is more severe than in other forms of arthropathy. Expression of connective tissue growth factor (CTGF) has been linked to many fibrotic diseases, but has not been studied in the context of haemophilic arthropathy. AIM We aim to compare synovial tissues histologically from haemophilia and osteoarthritis patients with advanced arthropathy in order to compare expression of proteins that are possibly aetiologic in the development of arthrofibrosis. METHODS Human synovial tissues were obtained from 10 haemophilia and 10 osteoarthritis patients undergoing joint surgery and processed for histology and immunohistochemistry. RESULTS All samples from haemophilia patients had synovitis with hypertrophy and hyperplasia of synovial villi. Histologically, synovial tissues contained hyperplastic villi with increased cellularity and abundant haemosiderin- and ferritin-pigmented macrophage-like cells (HMCs), with a perivascular localization in the sub-surface layer. CTGF staining was observed in the surface layer and sub-surface layer in all haemophilia patients, exclusively co-localizing with HMCs. Quantification showed that the extent of CTGF-positive areas was correlated with the degree of detection of HMCs. CTGF was not observed in any of the samples from osteoarthritis patients. CONCLUSION Using histological analysis, we showed that CTGF expression is elevated in haemophilia patients with arthrofibrosis and absent in patients with osteoarthritis. Additionally, we found that CTGF is always associated with haemosiderin-pigmented macrophage-like cells, which suggests that CTGF is produced by synovial A cells following the uptake of blood breakdown products.
Collapse
Affiliation(s)
- J Jiang
- Hemophilia Treatment Center at Orthopaedic Institute for Children, Los Angeles, CA, USA.,Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - N L Leong
- Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - U Khalique
- Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - T M Phan
- Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - K M Lyons
- Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - J V Luck
- Hemophilia Treatment Center at Orthopaedic Institute for Children, Los Angeles, CA, USA.,Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| |
Collapse
|
44
|
Yang Y, Li Y, Ma Z, Jiang S, Fan C, Hu W, Wang D, Di S, Sun Y, Yi W. A brief glimpse at CTRP3 and CTRP9 in lipid metabolism and cardiovascular protection. Prog Lipid Res 2016; 64:170-177. [DOI: 10.1016/j.plipres.2016.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/14/2016] [Accepted: 10/11/2016] [Indexed: 01/19/2023]
|
45
|
A virus-like particle-based connective tissue growth factor vaccine suppresses carbon tetrachloride-induced hepatic fibrosis in mice. Sci Rep 2016; 6:32155. [PMID: 27562139 PMCID: PMC4999884 DOI: 10.1038/srep32155] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/03/2016] [Indexed: 12/30/2022] Open
Abstract
Connective tissue growth factor (CTGF) has been recognized as a central mediator and promising therapeutic target in hepatic fibrosis. In this study, we generated a novel virus-like particle (VLP) CTGF vaccine by inserting the 138–159 amino acid (aa) fragment of CTGF into the central c/e1 epitope of C-terminus truncated hepatitis B virus core antigen (HBc, aa 1–149) using a prokaryotic expression system. Immunization of BALB/c mice with the VLP vaccine efficiently elicited the production of anti-CTGF neutralizing antibodies. Vaccination with this CTGF vaccine significantly protected BALB/c mice from carbon tetrachloride (CCl4)-induced hepatic fibrosis, as indicated by decreased hepatic hydroxyproline content and lower fibrotic score. CCl4 intoxication-induced hepatic stellate cell activation was inhibited by the vaccination, as indicated by decreased α-smooth muscle actin expression and Smad2 phosphorylation. Vaccination against CTGF also attenuated the over-expression of some profibrogenic factors, such as CTGF, transforming growth factor-β1, platelet-derived growth factor-B and tissue inhibitor of metalloproteinase-1 in the fibrotic mouse livers, decreased hepatocyte apoptosis and accelerated hepatocyte proliferation in the fibrotic mouse livers. Our results clearly indicate that vaccination against CTGF inhibits fibrogenesis, alleviates hepatocyte apoptosis and facilitate hepatic regeneration. We suggest that the vaccine should be developed into an effective therapeutic measure for hepatic fibrosis.
Collapse
|
46
|
Dual roles of CCN proteins in breast cancer progression. J Cell Commun Signal 2016; 10:217-222. [PMID: 27520547 DOI: 10.1007/s12079-016-0345-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 07/30/2016] [Indexed: 01/10/2023] Open
Abstract
The tumor microenvironment has a powerful effect on the development and progression of human breast cancer, which may be used therapeutically. Despite efforts to understand the complex role of the tumor microenvironment in breast cancer development, the specific players and their contributions to tumorigenesis need further investigation. The CCN family of matricellular proteins comprises six members (CCN1-6; CYR61, CTGF, NOV, WISP1-3) with central roles in development, inflammation, and tissue repair. CCN proteins also exert functions during pathological processes including fibrosis and cancer by regulating extracellular signals in the cellular environment. Studies have demonstrated that all six CCN proteins exert functions in breast tumorigenesis. Although CCN proteins share a multimodular structure in which most cysteine residues are conserved within structural motifs, they may have opposing functions in breast cancer progression. A better understanding of the functions of each CCN member will assist in the development of specific therapeutic approaches for breast cancer.
Collapse
|
47
|
Zhao L, Mi Y, Guan H, Xu Y, Mei Y. Velvet antler peptide prevents pressure overload-induced cardiac fibrosis via transforming growth factor (TGF)-β1 pathway inhibition. Eur J Pharmacol 2016; 783:33-46. [DOI: 10.1016/j.ejphar.2016.04.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 04/14/2016] [Accepted: 04/20/2016] [Indexed: 12/21/2022]
|
48
|
Novel therapeutic strategies targeting fibroblasts and fibrosis in heart disease. Nat Rev Drug Discov 2016; 15:620-638. [PMID: 27339799 DOI: 10.1038/nrd.2016.89] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Our understanding of the functions of cardiac fibroblasts has moved beyond their roles in heart structure and extracellular matrix generation and now includes their contributions to paracrine, mechanical and electrical signalling during ontogenesis and normal cardiac activity. Fibroblasts also have central roles in pathogenic remodelling during myocardial ischaemia, hypertension and heart failure. As key contributors to scar formation, they are crucial for tissue repair after interventions including surgery and ablation. Novel experimental approaches targeting cardiac fibroblasts are promising potential therapies for heart disease. Indeed, several existing drugs act, at least partially, through effects on cardiac connective tissue. This Review outlines the origins and roles of fibroblasts in cardiac development, homeostasis and disease; illustrates the involvement of fibroblasts in current and emerging clinical interventions; and identifies future targets for research and development.
Collapse
|
49
|
Xu H, Li P, Liu M, Liu C, Sun Z, Guo X, Zhang Y. CCN2 and CCN5 exerts opposing effect on fibroblast proliferation and transdifferentiation induced by TGF-β. Clin Exp Pharmacol Physiol 2016. [PMID: 26218313 DOI: 10.1111/1440-1681.12470] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epidural fibrosis might occur after lumbar discectomy and contributes to failed back syndrome. Transforming growth factor (TGF)-β has been reported to influence multiple organ fibrosis, in which connective tissue growth factor/cysteine-rich 61/nephroblastoma overexpressed 2 (CCN2) and CCN5 are involved. However, the effect of CCN2 and CCN5 on TGF-β induced fibrosis has not yet been elucidated. This study reports that CCN2 and CCN5 play opposing roles in cell proliferation and transdifferentiation of human skin fibroblasts or rabbit epidural scar-derived fibroblasts exposed to TGF-β. We observed that TGF-β1 induced fibroblasts proliferation and differentiation in a dose-dependent manner (from 0 μg/L to 20 μg/L). Meanwhile, CCN2 expression is up-regulated while CCN5 expression is inhibited by TGF-β1 exposure. Furthermore, it is demonstrated that CCN2 overexpression leads to promoted proliferation and elevated collagen and α-smooth muscle actin (α-SMA) expression, which are inhibited by CCN5 overexpression. Moreover, it is shown that the cysteine knot (CT) domain, present in CCN2 but absent in CCN5, plays an essential part in fibroblast proliferation and differentiation. Additionally, enhanced TGF-β and CCN2 expression but decreased CCN5 expression is found in rabbit epidural scar-derived fibroblasts. Overall, the results show the opposing effects of CCN2 and CCN5 on fibroblast proliferation and transdifferentiation induced by TGF-β.
Collapse
Affiliation(s)
- Honghai Xu
- Department of Orthopaedics, Third Affiliated Hospital (Shaanxi Provincial People's Hospital), Xi'an, Shaanxi, China
| | - Peng Li
- Xi 'an Medical College, Xi'an, Shaanxi, China
| | | | - Cong Liu
- Xi 'an Medical College, Xi'an, Shaanxi, China
| | - Zhengming Sun
- Department of Orthopaedics, Third Affiliated Hospital (Shaanxi Provincial People's Hospital), Xi'an, Shaanxi, China
| | - Xiong Guo
- Department of the Faculty of Public Health, Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuelin Zhang
- Department of Neurosurgery, Third Affiliated Hospital (Shaanxi Provincial People's Hospital), Xi'an, Shaanxi, China
| |
Collapse
|
50
|
Luo W, Hu L, Li W, Xu G, Xu L, Zhang C, Wang F. Epo inhibits the fibrosis and migration of Müller glial cells induced by TGF-β and high glucose. Graefes Arch Clin Exp Ophthalmol 2016; 254:881-90. [DOI: 10.1007/s00417-016-3290-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 01/06/2016] [Accepted: 02/08/2016] [Indexed: 12/11/2022] Open
|