1
|
Ooka A, Yamaguchi M, Suzuki K, Saito SY, Kaneko YK, Kimura T, Ishikawa T. Differentiation-inducing factor-1 ameliorates liver fibrosis through the reversion of activated hepatic stellate cells. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167802. [PMID: 40101372 DOI: 10.1016/j.bbadis.2025.167802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/25/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Affiliation(s)
- Akira Ooka
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Momoka Yamaguchi
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka City, Shizuoka 422-8526, Japan.
| | - Kensuke Suzuki
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Shin-Ya Saito
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka City, Shizuoka 422-8526, Japan; Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoinooka, Imabari City, Ehime 794-8555, Japan
| | - Yukiko K Kaneko
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Toshihide Kimura
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Tomohisa Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka City, Shizuoka 422-8526, Japan
| |
Collapse
|
2
|
Tanaka S, Elgaabari A, Seki M, Kuwakado S, Zushi K, Miyamoto J, Sawano S, Mizunoya W, Ehara K, Watanabe N, Ogawa Y, Imakyure H, Fujimaru R, Osaki R, Shitamitsu K, Mizoguchi K, Ushijima T, Maeno T, Nakashima T, Suzuki T, Nakamura M, Anderson JE, Tatsumi R. In vitro immuno-prevention of nitration/dysfunction of myogenic stem cell activator HGF, towards developing a strategy for age-related muscle atrophy. Aging Cell 2024; 23:e14337. [PMID: 39297318 PMCID: PMC11464115 DOI: 10.1111/acel.14337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/24/2024] [Accepted: 09/02/2024] [Indexed: 10/11/2024] Open
Abstract
In response to peroxynitrite (ONOO-) generation, myogenic stem satellite cell activator HGF (hepatocyte growth factor) undergoes nitration of tyrosine residues (Y198 and Y250) predominantly on fast IIa and IIx myofibers to lose its binding to the signaling receptor c-met, thereby disturbing muscle homeostasis during aging. Here we show that rat anti-HGF monoclonal antibody (mAb) 1H41C10, which was raised in-house against a synthetic peptide FTSNPEVRnitroY198EV, a site well-conserved in mammals, functions to confer resistance to nitration dysfunction on HGF. 1H41C10 was characterized by recognizing both nitrated and non-nitrated HGF with different affinities as revealed by Western blotting, indicating that the paratope of 1H41C10 may bind to the immediate vicinity of Y198. Subsequent experiments showed that 1H41C10-bound HGF resists peroxynitrite-induced nitration of Y198. A companion mAb-1H42F4 presented similar immuno-reactivity, but did not protect Y198 nitration, and thus served as the control. Importantly, 1H41C10-HGF also withstood Y250 nitration to retain c-met binding and satellite cell activation functions in culture. The Fab region of 1H41C10 exerts resistivity to Y250 nitration possibly due to its localization in the immediate vicinity to Y250, as supported by an additional set of experiments showing that the 1H41C10-Fab confers Y250-nitration resistance which the Fc segment does not. Findings highlight the in vitro preventive impact of 1H41C10 on HGF nitration-dysfunction that strongly impairs myogenic stem cell dynamics, potentially pioneering cogent strategies for counteracting or treating age-related muscle atrophy with fibrosis (including sarcopenia and frailty) and the therapeutic application of investigational HGF drugs.
Collapse
Affiliation(s)
- Sakiho Tanaka
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Alaa Elgaabari
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
- Department of Physiology, Faculty of Veterinary MedicineKafrelsheikh UniversityKafrelsheikhEgypt
| | - Miyumi Seki
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - So Kuwakado
- Department of Orthopaedic Surgery, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kahona Zushi
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Junri Miyamoto
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Shoko Sawano
- Department of Food and Life Science, School of Life and Environmental ScienceAzabu UniversitySagamiharaJapan
| | - Wataru Mizunoya
- Department of Animal Science and Biotechnology, School of Veterinary MedicineAzabu UniversitySagamiharaJapan
| | - Kenshiro Ehara
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Naruha Watanabe
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Yohei Ogawa
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Hikaru Imakyure
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Reina Fujimaru
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Rika Osaki
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Kazuki Shitamitsu
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Kaoru Mizoguchi
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Tomoki Ushijima
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Takahiro Maeno
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Takashi Nakashima
- Department of Bioscience and Biotechnology, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Takahiro Suzuki
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Mako Nakamura
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| | - Judy E. Anderson
- Department of Biological Sciences, Faculty of ScienceUniversity of ManitobaWinnipegManitobaCanada
| | - Ryuichi Tatsumi
- Department of Animal and Marine Bioresource SciencesGraduate School of Agriculture, Kyushu UniversityFukuokaJapan
| |
Collapse
|
3
|
Dudek P, Talar-Wojnarowska R. Current Approach to Risk Factors and Biomarkers of Intestinal Fibrosis in Inflammatory Bowel Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:305. [PMID: 38399592 PMCID: PMC10889938 DOI: 10.3390/medicina60020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Inflammatory bowel disease (IBD), especially Crohn's disease (CD), characterized by a chronic inflammatory process and progressive intestinal tissue damage, leads to the unrestrained proliferation of mesenchymal cells and the development of bowel strictures. Complications induced by fibrosis are related to high rates of morbidity and mortality and lead to a substantial number of hospitalizations and surgical procedures, generating high healthcare costs. The development of easily obtained, reliable fibrogenesis biomarkers is essential to provide an important complementary tool to existing diagnostic and prognostic methods in IBD management, guiding decisions on the intensification of pharmacotherapy, proceeding to surgical methods of treatment and monitoring the efficacy of anti-fibrotic therapy in the future. The most promising potential markers of fibrosis include cartilage oligomeric matrix protein (COMP), hepatocyte growth factor activator (HGFA), and fibronectin isoform- extra domain A (ED-A), as well as antibodies against granulocyte macrophage colony-stimulating factor (GM-CSF Ab), cathelicidin (LL-37), or circulatory miRNAs: miR-19a-3p and miR-19b-3p. This review summarizes the role of genetic predisposition, and risk factors and serological markers potentially contributing to the pathophysiology of fibrotic strictures in the course of IBD.
Collapse
|
4
|
Elgaabari A, Imatomi N, Kido H, Nakashima T, Okuda S, Manabe Y, Sawano S, Mizunoya W, Kaneko R, Tanaka S, Maeno T, Matsuyoshi Y, Seki M, Kuwakado S, Zushi K, Daneshvar N, Nakamura M, Suzuki T, Sunagawa K, Anderson JE, Allen RE, Tatsumi R. Age-related nitration/dysfunction of myogenic stem cell activator HGF. Aging Cell 2024; 23:e14041. [PMID: 37985931 PMCID: PMC10861216 DOI: 10.1111/acel.14041] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/22/2023] Open
Abstract
Mechanical perturbation triggers activation of resident myogenic stem cells to enter the cell cycle through a cascade of events including hepatocyte growth factor (HGF) release from its extracellular tethering and the subsequent presentation to signaling-receptor c-met. Here, we show that with aging, extracellular HGF undergoes tyrosine-residue (Y) nitration and loses c-met binding, thereby disturbing muscle homeostasis. Biochemical studies demonstrated that nitration/dysfunction is specific to HGF among other major growth factors and is characterized by its locations at Y198 and Y250 in c-met-binding domains. Direct-immunofluorescence microscopy of lower hind limb muscles from three age groups of rat, provided direct in vivo evidence for age-related increases in nitration of ECM-bound HGF, preferentially stained for anti-nitrated Y198 and Y250-HGF mAbs (raised in-house) in fast IIa and IIx myofibers. Overall, findings highlight inhibitory impacts of HGF nitration on myogenic stem cell dynamics, pioneering a cogent discussion for better understanding age-related muscle atrophy and impaired regeneration with fibrosis (including sarcopenia and frailty).
Collapse
Affiliation(s)
- Alaa Elgaabari
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
- Department of Physiology, Faculty of Veterinary MedicineKafrelsheikh UniversityKafrelsheikhEgypt
| | - Nana Imatomi
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Hirochika Kido
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Takashi Nakashima
- Department of Bioscience and Biotechnology, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Shoko Okuda
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Yoshitaka Manabe
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Shoko Sawano
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
- Present address:
Department of Food and Life Science, School of Life and Environmental ScienceAzabu UniversitySagamiharaJapan
| | - Wataru Mizunoya
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
- Present address:
Department of Animal Science and Biotechnology, School of Veterinary MedicineAzabu UniversitySagamiharaJapan
| | - Ryuki Kaneko
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Sakiho Tanaka
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Takahiro Maeno
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Yuji Matsuyoshi
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Miyumi Seki
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - So Kuwakado
- Department of Orthopaedic Surgery, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kahona Zushi
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Nasibeh Daneshvar
- Department of Biological Sciences, Faculty of ScienceUniversity of ManitobaWinnipegManitobaCanada
| | - Mako Nakamura
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Takahiro Suzuki
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| | - Kenji Sunagawa
- Department of Cardiovascular Medicine, Graduate School of MedicineKyushu UniversityFukuokaJapan
| | - Judy E. Anderson
- Department of Biological Sciences, Faculty of ScienceUniversity of ManitobaWinnipegManitobaCanada
| | - Ronald E. Allen
- The School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| | - Ryuichi Tatsumi
- Department of Animal and Marine Bioresource Sciences, Graduate School of AgricultureKyushu UniversityFukuokaJapan
| |
Collapse
|
5
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:1-95. [DOI: 10.1016/b978-0-7020-8228-3.00001-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
García-Bañuelos J, Oceguera-Contreras E, Sandoval-Rodríguez A, Bastidas-Ramírez BE, Lucano-Landeros S, Gordillo-Bastidas D, Gómez-Meda BC, Santos A, Cerda-Reyes E, Armendariz-Borunda J. AdhMMP8 Vector Administration in Muscle: An Alternate Strategy to Regress Hepatic Fibrosis. Cells 2023; 12:2127. [PMID: 37681859 PMCID: PMC10486800 DOI: 10.3390/cells12172127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/10/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023] Open
Abstract
The development of several vaccines against the SARS-CoV2 virus and their application in millions of people have shown efficacy and safety in the transfer of genes to muscle turning this tissue into a protein-producing factory. Established advanced liver fibrosis, is characterized by replacement of hepatic parenchyma by tissue scar, mostly collagen type I, with increased profibrogenic and proinflammatory molecules gene expression. Matrix metalloproteinase 8 (MMP-8) is an interstitial collagen-degrading proenzyme acting preferentially on collagen type I when activated. This study was carried out to elucidate the effect of an intramuscularly delivered adenoviral vector containing proMMP-8 gene cDNA (AdhMMP8) in male Wistar rats with experimental advanced liver fibrosis induced by thioacetamide. Therapeutic effects were monitored after 1, 2, or 3 weeks of a single dose (3 × 1011 vp/kg) of AdhMMP8. Circulating and liver concentration of MMP-8 protein remained constant; hepatic fibrosis decreased up to 48%; proinflammatory and profibrogenic genes expression diminished: TNF-α 2.28-fold, IL-1 1.95-fold, Col 1A1 4-fold, TGF-β1 3-fold and CTGF 2-fold; and antifibrogenic genes expression raised, MMP-9 2.8-fold and MMP-1 10-fold. Our data proposes that the administration of AdhMMP8 in muscle is safe and effective in achieving liver fibrosis regression at a comparable extent as when the adenoviral vector is delivered systemically to reach the liver, using a minimally invasive procedure.
Collapse
Affiliation(s)
- Jesús García-Bañuelos
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Edén Oceguera-Contreras
- Laboratorio de Sistemas Biológicos, Centro Universitario de los Valles, Universidad de Guadalajara, Carretera Guadalajara-Ameca km. 45.5, Ameca 46600, Jalisco, Mexico
| | - Ana Sandoval-Rodríguez
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Blanca Estela Bastidas-Ramírez
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Silvia Lucano-Landeros
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Daniela Gordillo-Bastidas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Nuevo Leon, Mexico
| | - Belinda C. Gómez-Meda
- Instituto de Genética Humana “Dr. Enrique Corona Rivera”, Department of Molecular Biology and Genomics, Health Sciences University Center, Guadalajara 44340, Jalisco, Mexico
| | - Arturo Santos
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Nuevo Leon, Mexico
| | | | - Juan Armendariz-Borunda
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Nuevo Leon, Mexico
| |
Collapse
|
7
|
Obata Y, Abe K, Miyazaki M, Koji T, Tabata Y, Nishino T. The Transfer of the Hepatocyte Growth Factor Gene by Macrophages Ameliorates the Progression of Peritoneal Fibrosis in Mice. Int J Mol Sci 2023; 24:ijms24086951. [PMID: 37108115 PMCID: PMC10139180 DOI: 10.3390/ijms24086951] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Growing evidence indicates that hepatocyte growth factor (HGF) possesses potent antifibrotic activity. Furthermore, macrophages migrate to inflamed sites and have been linked to the progression of fibrosis. In this study, we utilized macrophages as vehicles to express and deliver the HGF gene and investigated whether macrophages carrying the HGF expression vector (HGF-M) could suppress peritoneal fibrosis development in mice. We obtained macrophages from the peritoneal cavity of mice stimulated with 3% thioglycollate and used cationized gelatin microspheres (CGMs) to produce HGF expression vector-gelatin complexes. Macrophages phagocytosed these CGMs, and gene transfer into macrophages was confirmed in vitro. Peritoneal fibrosis was induced by intraperitoneal injection of chlorhexidine gluconate (CG) for three weeks; seven days after the first CG injection, HGF-M was administered intravenously. Transplantation of HGF-M significantly suppressed submesothelial thickening and reduced type III collagen expression. Moreover, in the HGF-M-treated group, the number of α-smooth muscle actin- and TGF-β-positive cells were significantly lower in the peritoneum, and ultrafiltration was preserved. Our results indicated that the transplantation of HGF-M prevented the progression of peritoneal fibrosis and indicated that this novel gene therapy using macrophages may have potential for treating peritoneal fibrosis.
Collapse
Affiliation(s)
- Yoko Obata
- Department of Nephrology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Katsushige Abe
- Abe Diabetes Clinic, 16-13 Nakakasuga-machi, Oita 870-0039, Japan
| | | | - Takehiko Koji
- Department of Histology and Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tomoya Nishino
- Department of Nephrology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| |
Collapse
|
8
|
de Nola G, Leclercq B, Mougel A, Taront S, Simonneau C, Forneris F, Adriaenssens E, Drobecq H, Iamele L, Dubuquoy L, Melnyk O, Gherardi E, de Jonge H, Vicogne J. Dimerization of kringle 1 domain from hepatocyte growth factor/scatter factor provides a potent MET receptor agonist. Life Sci Alliance 2022; 5:5/12/e202201424. [PMID: 35905995 PMCID: PMC9348577 DOI: 10.26508/lsa.202201424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 12/22/2022] Open
Abstract
We designed and characterized a potent full MET receptor agonist consisting of two recombinantly linked HGF/SF kringle 1 domains and demonstrated its potential in epithelial tissue regeneration. Hepatocyte growth factor/scatter factor (HGF/SF) and its cognate receptor MET play several essential roles in embryogenesis and regeneration in postnatal life of epithelial organs such as the liver, kidney, lung, and pancreas, prompting a strong interest in harnessing HGF/SF-MET signalling for regeneration of epithelial organs after acute or chronic damage. The limited stability and tissue diffusion of native HGF/SF, however, which reflect the tightly controlled, local mechanism of action of the morphogen, have led to a major search of HGF/SF mimics for therapy. In this work, we describe the rational design, production, and characterization of K1K1, a novel minimal MET agonist consisting of two copies of the kringle 1 domain of HGF/SF in tandem orientation. K1K1 is highly stable and displays biological activities equivalent or superior to native HGF/SF in a variety of in vitro assay systems and in a mouse model of liver disease. These data suggest that this engineered ligand may find wide applications in acute and chronic diseases of the liver and other epithelial organs dependent of MET activation.
Collapse
Affiliation(s)
- Giovanni de Nola
- Department of Molecular Medicine, University of Pavia, Unit of Immunology and General Pathology Section, Pavia, Italy
| | - Bérénice Leclercq
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019, UMR 9017, CIIL, Center for Infection and Immunity of Lille, Lille, France
| | - Alexandra Mougel
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019, UMR 9017, CIIL, Center for Infection and Immunity of Lille, Lille, France
| | - Solenne Taront
- University of Lille, Inserm, CHU Lille, U1286, INFINITE, Institute for Translational Research in Inflammation, Lille, France
| | - Claire Simonneau
- Roche Pharmaceutical Research and Early Development (pRED), Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Federico Forneris
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Eric Adriaenssens
- University of Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020, UMR 1277, Canther, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Hervé Drobecq
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019, UMR 9017, CIIL, Center for Infection and Immunity of Lille, Lille, France
| | - Luisa Iamele
- Department of Molecular Medicine, University of Pavia, Unit of Immunology and General Pathology Section, Pavia, Italy
| | - Laurent Dubuquoy
- University of Lille, Inserm, CHU Lille, U1286, INFINITE, Institute for Translational Research in Inflammation, Lille, France
| | - Oleg Melnyk
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019, UMR 9017, CIIL, Center for Infection and Immunity of Lille, Lille, France
| | - Ermanno Gherardi
- Department of Molecular Medicine, University of Pavia, Unit of Immunology and General Pathology Section, Pavia, Italy
| | - Hugo de Jonge
- Department of Molecular Medicine, University of Pavia, Unit of Immunology and General Pathology Section, Pavia, Italy
| | - Jérôme Vicogne
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019, UMR 9017, CIIL, Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
9
|
Filliol A, Saito Y, Nair A, Dapito DH, Yu LX, Ravichandra A, Bhattacharjee S, Affo S, Fujiwara N, Su H, Sun Q, Savage TM, Wilson-Kanamori JR, Caviglia JM, Chin L, Chen D, Wang X, Caruso S, Kang JK, Amin AD, Wallace S, Dobie R, Yin D, Rodriguez-Fiallos OM, Yin C, Mehal A, Izar B, Friedman RA, Wells RG, Pajvani UB, Hoshida Y, Remotti HE, Arpaia N, Zucman-Rossi J, Karin M, Henderson NC, Tabas I, Schwabe RF. Opposing roles of hepatic stellate cell subpopulations in hepatocarcinogenesis. Nature 2022; 610:356-365. [PMID: 36198802 PMCID: PMC9949942 DOI: 10.1038/s41586-022-05289-6] [Citation(s) in RCA: 185] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/30/2022] [Indexed: 01/21/2023]
Abstract
Hepatocellular carcinoma (HCC), the fourth leading cause of cancer mortality worldwide, develops almost exclusively in patients with chronic liver disease and advanced fibrosis1,2. Here we interrogated functions of hepatic stellate cells (HSCs), the main source of liver fibroblasts3, during hepatocarcinogenesis. Genetic depletion, activation or inhibition of HSCs in mouse models of HCC revealed their overall tumour-promoting role. HSCs were enriched in the preneoplastic environment, where they closely interacted with hepatocytes and modulated hepatocarcinogenesis by regulating hepatocyte proliferation and death. Analyses of mouse and human HSC subpopulations by single-cell RNA sequencing together with genetic ablation of subpopulation-enriched mediators revealed dual functions of HSCs in hepatocarcinogenesis. Hepatocyte growth factor, enriched in quiescent and cytokine-producing HSCs, protected against hepatocyte death and HCC development. By contrast, type I collagen, enriched in activated myofibroblastic HSCs, promoted proliferation and tumour development through increased stiffness and TAZ activation in pretumoural hepatocytes and through activation of discoidin domain receptor 1 in established tumours. An increased HSC imbalance between cytokine-producing HSCs and myofibroblastic HSCs during liver disease progression was associated with increased HCC risk in patients. In summary, the dynamic shift in HSC subpopulations and their mediators during chronic liver disease is associated with a switch from HCC protection to HCC promotion.
Collapse
Affiliation(s)
- Aveline Filliol
- Department of Medicine, Columbia University, New York, NY, USA
| | - Yoshinobu Saito
- Department of Medicine, Columbia University, New York, NY, USA
| | - Ajay Nair
- Department of Medicine, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Dianne H Dapito
- Department of Medicine, Columbia University, New York, NY, USA
| | - Le-Xing Yu
- Department of Medicine, Columbia University, New York, NY, USA
| | - Aashreya Ravichandra
- Department of Medicine, Columbia University, New York, NY, USA
- Klinikum Rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | | | - Silvia Affo
- Department of Medicine, Columbia University, New York, NY, USA
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Naoto Fujiwara
- Liver Tumor Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hua Su
- Department of Pharmacology, School of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Qiuyan Sun
- Department of Medicine, Columbia University, New York, NY, USA
| | - Thomas M Savage
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - John R Wilson-Kanamori
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Jorge M Caviglia
- Department of Medicine, Columbia University, New York, NY, USA
- Department of Health and Nutrition Sciences, Brooklyn College, City University of New York, New York, NY, USA
| | - LiKang Chin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biomedical Engineering, Widener University, Chester, PA, USA
| | - Dongning Chen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University, New York, NY, USA
| | - Stefano Caruso
- Functional Genomics of Solid Tumors Laboratory, Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Jin Ku Kang
- Department of Medicine, Columbia University, New York, NY, USA
- Institute of Human Nutrition, Columbia University, New York, NY, USA
| | - Amit Dipak Amin
- Department of Medicine, Columbia University, New York, NY, USA
| | - Sebastian Wallace
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Ross Dobie
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Deqi Yin
- Department of Medicine, Columbia University, New York, NY, USA
| | | | - Chuan Yin
- Department of Medicine, Columbia University, New York, NY, USA
- Department of Gastroenterology, Changzheng Hospital, Shanghai, China
| | - Adam Mehal
- Department of Medicine, Columbia University, New York, NY, USA
| | - Benjamin Izar
- Department of Medicine, Columbia University, New York, NY, USA
| | - Richard A Friedman
- Biomedical Informatics Shared Resource, Herbert Irving Comprehensive Cancer Center, and Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
| | - Rebecca G Wells
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, NY, USA
- Institute of Human Nutrition, Columbia University, New York, NY, USA
| | - Yujin Hoshida
- Liver Tumor Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Helen E Remotti
- Department of Pathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Nicholas Arpaia
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jessica Zucman-Rossi
- Functional Genomics of Solid Tumors Laboratory, Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Michael Karin
- Department of Pharmacology, School of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Ira Tabas
- Department of Medicine, Columbia University, New York, NY, USA
- Institute of Human Nutrition, Columbia University, New York, NY, USA
- Department of Pathology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology, Columbia University, New York, NY, USA
| | - Robert F Schwabe
- Department of Medicine, Columbia University, New York, NY, USA.
- Institute of Human Nutrition, Columbia University, New York, NY, USA.
| |
Collapse
|
10
|
Yamazaki K, Igarashi-Takeuchi H, Numabe Y. Hepatocyte growth factor exhibits anti-fibrotic effects in an in vitro model of nifedipine-induced gingival overgrowth. J Oral Sci 2022; 64:99-104. [PMID: 34980825 DOI: 10.2334/josnusd.21-0419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
PURPOSE The aim of this study was to establish an in vitro model of nifedipine-induced gingival overgrowth and characterize the anti-fibrotic effect of hepatocyte growth factor (HGF) using this model. METHODS Human gingival fibroblasts were cultured-treated with 0.1, 1, or 10 µg/mL nifedipine or 10 ng/mL IL-1β + 0.1, 1, or 10 µg/mL nifedipine (0.1N, 1N, 10N, IL + 0.1N, IL + 1N, IL + 10N). Cell proliferation and levels of type I collagen, TGF-β1, CCN2/CTGF, and α-SMA were measured 48 h after the simultaneous addition of 10 and 50 ng/mL HGF (10 and 50HGF) along with IL-1β and nifedipine. Type I collagen was measured after administration of anti-HGF neutralizing antibody. RESULTS Significant increases in type I collagen, TGF-β1, and CCN2/CTGF were observed after treatment in the 1N and IL + 0.1N groups. Levels of type I collagen and CCN2/CTGF differed significantly between the IL + 0.1N group and the IL + 0.1N + 50HGF group. Production of type I collagen increased significantly following addition of anti-HGF antibody. CONCLUSION This study demonstrated the establishment of an in vitro model of nifedipine-induced gingival overgrowth by showing increased collagen levels. Experiments using this model suggested that HGF exerts anti-fibrotic effects.
Collapse
Affiliation(s)
- Kei Yamazaki
- Department of Periodontology, School of Life Dentistry at Tokyo, The Nippon Dental University
| | - Hiroko Igarashi-Takeuchi
- Department of Periodontology, School of Life Dentistry at Tokyo, The Nippon Dental University.,Core Research Facilities for Basic Science, Research Center for Medical Science, The Jikei University School of Medicine
| | - Yukihiro Numabe
- Department of Periodontology, School of Life Dentistry at Tokyo, The Nippon Dental University
| |
Collapse
|
11
|
ArefNezhad R, Motedayyen H, Roghani-Shahraki H. Do cytokines associate periodontitis with metabolic disorders? An overview of current documents. Endocr Metab Immune Disord Drug Targets 2022; 22:778-786. [PMID: 35043774 DOI: 10.2174/1871530322666220119112026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/28/2021] [Accepted: 11/26/2021] [Indexed: 11/22/2022]
Abstract
Periodontitis is an oral chronic inflammatory condition affecting the adult population worldwide. Many microorganisms act as an initiator for induction of inflammatory immune responses, which participate in the destruction of connective tissue surrounding the teeth and thereby result in tooth loss. Cytokines may have indispensable roles in its pathogenesis through enhancing inflammatory and immune responses. Cytokines can affect functions of some cells of different tissues, such as the cells of the pancreas, liver, and adipose tissues. There is evidence that periodontitis is associated with metabolic disorders, like liver cirrhosis, obesity, and diabetes mellitus. Hence, this review was focused on determining how cytokines can participate in the correlation of periodontitis with metabolic disorders.
Collapse
Affiliation(s)
- Reza ArefNezhad
- Halal Research Center of IRI, FDA, Tehran, Iran
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Motedayyen
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | | |
Collapse
|
12
|
Moghassemi S, Dadashzadeh A, Azevedo RB, Feron O, Amorim CA. Photodynamic cancer therapy using liposomes as an advanced vesicular photosensitizer delivery system. J Control Release 2021; 339:75-90. [PMID: 34562540 DOI: 10.1016/j.jconrel.2021.09.024] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 09/16/2021] [Accepted: 09/18/2021] [Indexed: 12/26/2022]
Abstract
The multidisciplinary field of photodynamic therapy (PDT) is a combination of photochemistry and photophysics sciences, which has shown tremendous potential for cancer therapy application. PDT employs a photosensitizing agent (PS) and light to form cytotoxic reactive oxygen species and subsequently oxidize light-exposed tissue. Despite numerous advantages of PDT and enormous progress in this field, common PSs are still far from ideal treatment because of their poor permeability, non-specific phototoxicity, side effects, hydrophobicity, weak bioavailability, and tendency to self-aggregation. To circumvent these limitations, PS can be encapsulated in liposomes, an advanced drug delivery system that has demonstrated the ability to enhance drug permeability into biological membranes and loading both hydrophobic and lipophilic agents. Moreover, liposomes can also be coated by targeting agents to improve delivery efficiency. The present review aims to summarize the principles of PDT, various PS generations, PS-loaded nanoparticles, liposomes, and their impact on PDT, then discuss recent photodynamic cancer therapy strategies using liposomes as PS-loaded vectors, and highlight future possibilities and perspectives.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Ricardo Bentes Azevedo
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil
| | - Olivier Feron
- Pôle de Pharmacologie et thérapeutique, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
13
|
Huang W, Han N, Du L, Wang M, Chen L, Tang H. A narrative review of liver regeneration-from models to molecular basis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1705. [PMID: 34988214 PMCID: PMC8667151 DOI: 10.21037/atm-21-5234] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/04/2021] [Indexed: 12/11/2022]
Abstract
Objective To elucidate the characteristics of different liver regeneration animal models, understand the activation signals and mechanisms related to liver regeneration, and obtain a more comprehensive conception of the entire liver regeneration process. Background Liver regeneration is one of the most enigmatic and fascinating phenomena of the human organism. Despite suffering significant injuries, the liver still can continue to perform its complex functions through the regeneration system. Although advanced topics on liver regeneration have been proposed; unfortunately, complete regeneration of the liver has not been achieved until now. Therefore, increasing understanding of the liver regenerative process can help improve our treatment of liver failure. It will provide a new sight for the treatment of patients with liver injury in the clinic. Methods Literatures on liver regeneration animal models and involved basic research on molecular mechanisms were retrieved to analyze the characteristics of different models and those related to molecular basis. Conclusions The process of liver regeneration is complex and intricate, consisting of various and interactive pathways. There is sufficient evidence to demonstrate that liver regeneration is similar between humans and rodents. At the same time, many of the same cytokines, growth factors, and signaling pathways are relevant. There are many gaps in our current knowledge. Understanding of this knowledge will provide more supportive clinical treatment strategies, including small-scale liver transplantation and high-quality regenerative process after surgical resection, and offer possible targets to treat the dysregulation of regeneration that occurs in chronic hepatic diseases and tumors. Current research work, such as the use of animal models as in vivo vectors for high-quality human hepatocytes, represents a unique and significant cutting edge in the field of liver regeneration.
Collapse
Affiliation(s)
- Wei Huang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.,Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Ning Han
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.,Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Lingyao Du
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.,Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.,Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Liyu Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.,Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.,Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Zhou M, Hou Y, Wu J, Li G, Cao P, Chen W, Hu L, Gan D. miR-93-5p promotes insulin resistance to regulate type 2 diabetes progression in HepG2 cells by targeting HGF. Mol Med Rep 2021; 23:329. [PMID: 33760164 PMCID: PMC7974269 DOI: 10.3892/mmr.2021.11968] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022] Open
Abstract
Insulin resistance is a common feature of type 2 diabetes mellitus (T2DM). However, the mechanisms underlying insulin resistance are not completely understood. The present study aimed to investigate the effect of microRNA (miR)-93-5p on insulin resistance in T2DM cells. Human hepatocellular carcinoma (HCC; HepG2) cells were cultured in medium with high glucose content (30 mM glucose) to establish an in vitro insulin-resistant cell model (IR group). Glucose consumption and glycogen synthesis assays were performed to assess glucose consumption and glycogen synthesis, respectively. By performing immunoprecipitation assays, the abundance of the Met-insulin receptor complex was detected in HepG2 cells. miR-93-5p and hepatocyte growth factor (HGF) mRNA expression levels were measured via reverse transcription-quantitative PCR, and HGF protein expression levels were measured via western blotting. A dual-luciferase reporter assay was conducted to investigate the interaction between miR-93-5p and HGF. Cell Counting Kit-8, BrdU and caspase-3 activity assays were performed to evaluate cell viability, proliferation and apoptosis, respectively, in insulin-resistant HepG2 cells following transfection with small interfering RNA-HGF, HGF overexpression vector, miR-93-5p mimic or miR-93-5p inhibitor. The results demonstrated that miR-93-5p expression was significantly increased and HGF expression was significantly decreased in HCC tissues isolated from patients with or without T2DM compared with adjacent healthy tissues isolated from patients without T2DM. Compared with the IR group, miR-93-5p overexpression significantly increased cell proliferation, glucose consumption and glycogen synthesis, but significantly inhibited apoptosis in insulin-resistant HepG2 cells. By contrast, compared with the IR group, HGF overexpression significantly inhibited cell proliferation, glucose consumption and glycogen synthesis, but significantly enhanced cell apoptosis in insulin-resistant HepG2 cells. Following co-transfection with HGF overexpression vector and miR-93-5p mimic, miR-93-5p mimic-mediated induction of HepG2 cell proliferation, glucose consumption and glycogen synthesis in insulin-resistant HepG2 cells was inhibited. Collectively, the results of the present study indicated that miR-93-5p enhanced insulin resistance to regulate T2DM progression in HepG2 cells by targeting HGF.
Collapse
Affiliation(s)
- Man Zhou
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Yilin Hou
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Jun Wu
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Guangli Li
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Ping Cao
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Wan Chen
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Lingli Hu
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Dingyun Gan
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
15
|
Xiang Y, Qin Z, Yang Y, Fisher GJ, Quan T. Age-related elevation of HGF is driven by the reduction of fibroblast size in a YAP/TAZ/CCN2 axis-dependent manner. J Dermatol Sci 2021; 102:36-46. [PMID: 33648801 DOI: 10.1016/j.jdermsci.2021.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 02/03/2021] [Accepted: 02/12/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Aged human skin is primarily attributable to the loss of collagen. Hepatocyte growth factor (HGF) acts as an anti-fibrotic factor by suppression of collagen production. In aged human skin, HGF is elevated in dermal fibroblasts and thus contributes to dermal aging (thin dermis) by suppression of collagen production. OBJECTIVE We aimed to investigate the underlying mechanisms of age-related elevation of HGF expression. METHODS Collagen fibrils in the aged skin dermis are fragmented and disorganized, which impairs collagen-fibroblast interaction, resulting in reduced fibroblast spreading and size. To explore the connection between reduced dermal fibroblast size and age-related elevation of HGF expression, we manipulate dermal fibroblast size, and cell-size dependent regulation of HGF was investigated by laser capture microdissection, immunostaining, capillary electrophoresis immunoassay, and quantitative RT-PCR. RESULTS We found that reduced fibroblast size is responsible for age-related elevation of HGF expression. Further investigation indicated that cell size-dependent upregulation of HGF expression was mediated by impeded YAP/TAZ nuclear translocation and their target gene, CCN2. Conversely, restoration of dermal fibroblast size rapidly reversed cell-size-dependent upregulation of HGF in a YAP/TAZ-dependent manner. Finally, we confirmed that elevated HGF expression is accompanied by the reduced expression of YAP/TAZ and CCN2 in the aged human skin in vivo. CONCLUSION Age-related elevation of HGF is driven by the reduction of fibroblast size in a YAP/TAZ/CCN2 axis-dependent manner. These data reveal a novel mechanism by which reduction of fibroblast size upregulates HGF expression, which in turn contributes to loss of collagen, a prominent feature of aged human skin.
Collapse
Affiliation(s)
- Yaping Xiang
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Zhaoping Qin
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yan Yang
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gary J Fisher
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
16
|
Miceli V, Bulati M, Iannolo G, Zito G, Gallo A, Conaldi PG. Therapeutic Properties of Mesenchymal Stromal/Stem Cells: The Need of Cell Priming for Cell-Free Therapies in Regenerative Medicine. Int J Mol Sci 2021; 22:763. [PMID: 33466583 PMCID: PMC7828743 DOI: 10.3390/ijms22020763] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are multipotent adult stem cells that support homeostasis during tissue regeneration. In the last decade, cell therapies based on the use of MSCs have emerged as a promising strategy in the field of regenerative medicine. Although these cells possess robust therapeutic properties that can be applied in the treatment of different diseases, variables in preclinical and clinical trials lead to inconsistent outcomes. MSC therapeutic effects result from the secretion of bioactive molecules affected by either local microenvironment or MSC culture conditions. Hence, MSC paracrine action is currently being explored in several clinical settings either using a conditioned medium (CM) or MSC-derived exosomes (EXOs), where these products modulate tissue responses in different types of injuries. In this scenario, MSC paracrine mechanisms provide a promising framework for enhancing MSC therapeutic benefits, where the composition of secretome can be modulated by priming of the MSCs. In this review, we examine the literature on the priming of MSCs as a tool to enhance their therapeutic properties applicable to the main processes involved in tissue regeneration, including the reduction of fibrosis, the immunomodulation, the stimulation of angiogenesis, and the stimulation of resident progenitor cells, thereby providing new insights for the therapeutic use of MSCs-derived products.
Collapse
Affiliation(s)
- Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), 90127 Palermo, Italy; (M.B.); (G.I.); (G.Z.); (A.G.); (P.G.C.)
| | | | | | | | | | | |
Collapse
|
17
|
Allen JN, Dey A, Cai J, Zhang J, Tian Y, Kennett M, Ma Y, Liang TJ, Patterson AD, Hankey-Giblin PA. Metabolic Profiling Reveals Aggravated Non-Alcoholic Steatohepatitis in High-Fat High-Cholesterol Diet-Fed Apolipoprotein E-Deficient Mice Lacking Ron Receptor Signaling. Metabolites 2020; 10:metabo10080326. [PMID: 32796650 PMCID: PMC7464030 DOI: 10.3390/metabo10080326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/29/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) represents the progressive sub-disease of non-alcoholic fatty liver disease that causes chronic liver injury initiated and sustained by steatosis and necroinflammation. The Ron receptor is a tyrosine kinase of the Met proto-oncogene family that potentially has a beneficial role in adipose and liver-specific inflammatory responses, as well as glucose and lipid metabolism. Since its discovery two decades ago, the Ron receptor has been extensively investigated for its differential roles on inflammation and cancer. Previously, we showed that Ron expression on tissue-resident macrophages limits inflammatory macrophage activation and promotes a repair phenotype, which can retard the progression of NASH in a diet-induced mouse model. However, the metabolic consequences of Ron activation have not previously been investigated. Here, we explored the effects of Ron receptor activation on major metabolic pathways that underlie the development and progression of NASH. Mice lacking apolipoprotein E (ApoE KO) and double knockout (DKO) mice that lack ApoE and Ron were maintained on a high-fat high-cholesterol diet for 18 weeks. We observed that, in DKO mice, the loss of ligand-dependent Ron signaling aggravated key pathological features in steatohepatitis, including steatosis, inflammation, oxidation stress, and hepatocyte damage. Transcriptional programs positively regulating fatty acid (FA) synthesis and uptake were upregulated in the absence of Ron receptor signaling, whereas lipid disposal pathways were downregulated. Consistent with the deregulation of lipid metabolism pathways, the DKO animals exhibited increased accumulation of FAs in the liver and decreased level of bile acids. Altogether, ligand-dependent Ron receptor activation provides protection from the deregulation of major metabolic pathways that initiate and aggravate non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Joselyn N. Allen
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Adwitia Dey
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Jingwei Cai
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Jingtao Zhang
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Yuan Tian
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Mary Kennett
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Yanling Ma
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, The National Institutes of Health, Bethesda, MD 20814, USA; (Y.M.); (T.J.L.)
| | - T. Jake Liang
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, The National Institutes of Health, Bethesda, MD 20814, USA; (Y.M.); (T.J.L.)
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
- Correspondence: (A.D.P.); (P.A.H.-G.); Tel.: +1-814-867-4565; (A.D.P.); +1-814-863-0128 (P.A.H.-G.)
| | - Pamela A. Hankey-Giblin
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
- Correspondence: (A.D.P.); (P.A.H.-G.); Tel.: +1-814-867-4565; (A.D.P.); +1-814-863-0128 (P.A.H.-G.)
| |
Collapse
|
18
|
Ma J, Yan X, Lin Y, Tan Q. Hepatocyte Growth Factor Secreted from Human Adipose-Derived Stem Cells Inhibits Fibrosis in Hypertrophic Scar Fibroblasts. Curr Mol Med 2020; 20:558-571. [PMID: 31903876 DOI: 10.2174/1566524020666200106095745] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 11/22/2022]
Abstract
AIMS To study the effect of Adipose-derived stem cells (ADSCs) on fibrosis of hypertrophic scar-derived fibroblasts (HSFs) and its concrete mechanism. BACKGROUND ADSCs have been reported to reduce collagen production and fibroblast proliferation in co-culture experiments. Conditioned medium from adipose-derived stem cells (ADSCs-CM) has successfully inhibited fibrosis by decreasing the expression of collagen type І (Col1) and α-smooth muscle actin (α-SMA) in rabbit ear scar models. Hepatocyte growth factor (HGF), the primary growth factor in ADSCs-CM, has been shown to reverse fibrosis in various fibrotic diseases. OBJECTIVE To test the hypothesis that ADSCs inhibit fibrosis of HSFs through the secretion of HGF. METHODS HSFs were treated with DMEM containing 0%, 10%, 50% and 100% concentration of ADSCs-CM. The effect of ADSCs-CM on the viability was determined by cell viability assay, and the collagen production in HSFs was examined by Sirius red staining. Expression and secretion of fibrosis and degradation proteins were detected separately. After measuring the concentration of HGF in ADSCs-CM, the same number of HSFs were treated with 50% ADSCs-CM or HGF. HGF activity in ADSCs-CM was neutralized with a goat anti-human HGF antibody. RESULTS The results demonstrated that ADSCs-CM dose-dependently decreased cell viability, expression of fibrosis molecules, and tissue inhibitor of metalloproteinases-1 (TIMP-1), and significantly increased matrix metalloproteinase-1 (MMP-1) expression in HSFs. Collagen production and the ratio of collagen type І and type III (Col1/Col3) were also suppressed by ADSCs-CM in a dose-dependent manner. When HSFs were cultured with either 50% ADSCs-CM or HGF (1 ng/ml), a similar trend was observed in gene expression and protein secretion. Adding an HGF antibody to both groups returned protein expression and secretion to basal levels but did not significantly affect the fibrosis factors in the control group. CONCLUSION Our findings revealed that adipose-derived stem cell-secreted HGF effectively inhibits fibrosis-related factors and regulates extracellular matrix (ECM) remodeling in hypertrophic scar fibroblasts.
Collapse
Affiliation(s)
- Ji Ma
- 1Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210008, China
| | - Xin Yan
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Yue Lin
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Qian Tan
- 1Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210008, China
| |
Collapse
|
19
|
Choi JH, Loarca L, De Hoyos-Vega JM, Dadgar N, Loutherback K, Shah VH, Stybayeva G, Revzin A. Microfluidic confinement enhances phenotype and function of hepatocyte spheroids. Am J Physiol Cell Physiol 2020; 319:C552-C560. [PMID: 32697600 DOI: 10.1152/ajpcell.00094.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A number of cell culture approaches have been described for maintenance of primary hepatocytes. Forming hepatocytes into three-dimensional (3-D) spheroids is one well-accepted method for extending epithelial phenotype of these cells. Our laboratory has previously observed enhanced function of two-dimensional (2-D, monolayer) hepatocyte cultures in microfluidic devices due to increased production of several hepato-inductive growth factors, including hepatocyte growth factor (HGF). In the present study, we wanted to test a hypothesis that culturing hepatocyte spheroids (3-D) in microfluidic devices will also result in enhanced phenotype and function. To test this hypothesis, we fabricated devices with small and large volumes. Both types of devices included a microstructured floor containing arrays of pyramidal wells to promote assembly of hepatocytes into spheroids with individual diameters of ~100 µm. The hepatocyte spheroids were found to be more functional, as evidenced by higher level of albumin synthesis, bile acid production, and hepatic enzyme expression, in low-volume compared with large-volume devices. Importantly, high functionality of spheroid cultures correlated with elevated levels of HGF secretion. Although decay of hepatic function (albumin secretion) was observed over the course 3 wk, this behavior could be abrogated by inhibiting TGF-β1 signaling. With TGF-β1 inhibitor, microfluidic hepatocyte spheroid cultures maintained high and stable levels of albumin synthesis over the course of 4 wk. To further highlight utility of this culture platform for liver disease modeling, we carried out alcohol injury experiments in microfluidic devices and tested protective effects of interleukin-22: a potential therapy for alcoholic hepatitis.
Collapse
Affiliation(s)
- Jong Hoon Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Lorena Loarca
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Jose M De Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Neda Dadgar
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Kevin Loutherback
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
20
|
Xu H, Fan GK. The Role of Cytokines in Modulating Vocal Fold Fibrosis: A Contemporary Review. Laryngoscope 2020; 131:139-145. [PMID: 32293731 DOI: 10.1002/lary.28507] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/11/2019] [Accepted: 12/29/2019] [Indexed: 01/21/2023]
Abstract
OBJECTIVES Vocal fold (VF) scarring and laryngeal stenosis are a significant clinical challenge. Excessive scar formation causes low voice quality or even life-threatening obstructions. Cytokines are thought to modulate multiple steps of the establishment of VF fibrosis, but there is no systematic report regarding their role in modulating VF fibrosis. This review aims to investigate the role of cytokines in modulating vocal fold fibrosis. STUDY DESIGN Literature review. METHODS This review searched for all relevant peer publications in English for the period 2009 to 2019 in the PubMed database using search terms: "laryngeal stenosis," "vocal fold scarring," and "cytokines." A thorough investigation of the methods and results of the reviewed studies was performed. RESULTS Comprehensive research in various studies, including analyses of prostaglandin E2 (PGE2), granulocyte-macrophage colony-stimulating factor (GM-CSF), hepatocyte growth factor (HGF), basic fibroblast growth factor (bFGF), transforming growth factor-β3 (TGF-β3), and interleukin-10 (IL-10), supports cytokine therapy for VF scarring and laryngeal stenosis to some extent. A few clinical studies on this topic support the conclusion that HGF and bFGF can be selected as effective drugs, and no serious side effects were found. CONCLUSIONS This review describes the potential of cytokines for modulating the process of VF fibrogenesis, although cytokines are still an unproven treatment method. As no ideal drugs exist, cytokines may be considered the candidate treatment for preventing VF fibrogenesis. Laryngoscope, 131:139-145, 2021.
Collapse
Affiliation(s)
- Haoyuan Xu
- Department of Otolaryngology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guo-Kang Fan
- Department of Otolaryngology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
21
|
Saad KM, Shaker ME, Shaaban AA, Abdelrahman RS, Said E. The c-Met inhibitor capmatinib alleviates acetaminophen-induced hepatotoxicity. Int Immunopharmacol 2020; 81:106292. [PMID: 32062076 DOI: 10.1016/j.intimp.2020.106292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 02/08/2023]
Abstract
Acetaminophen (APAP)-induced hepatotoxicity comes among the most frequent humans' toxicities caused by drugs. So far, therapeutic interventions for such type of drug-induced toxicity are still limited. In the current study, we examined the influence of capmatinib (Cap), a novel c-Met inhibitor, on APAP-induced hepatotoxicity in mice when administered 2 h prior, 2 h post and 4 h post APAP-challenge. The results revealed that Cap administration significantly attenuated APAP-induced liver injury when administered only 2 h prior and post APAP-administration. Cap hepatoprotective effect was mediated by lowering the excessive formation of lipid peroxidation and nitrosative stress products caused by APAP. Besides, Cap attenuated APAP-induced overproduction and release of proinflammatory mediators like TNF-α, IL-1β, IL-17A, IL-6, and MCP-1. Cap treatment also led to avoidance of APAP-subsequent repair by abating APAP-induced elevation of hepatic IL-22 and PCNA expressions. In conclusion, c-Met receptor inhibition may be a potential strategy for alleviating APAP-hepatotoxicity, especially when administered in the early phase of intoxication.
Collapse
Affiliation(s)
- Kareem M Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed E Shaker
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacology, Faculty of Pharmacy, Jouf University, Sakaka 2014, Saudi Arabia.
| | - Ahmed A Shaaban
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Aqaba University of Technology, Aqaba 77110, Jordan
| | - Rehab S Abdelrahman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taibah University, Al Madinah Al-Munawwarah 30001, Saudi Arabia
| | - Eman Said
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
22
|
Molecular Engineering Strategies Tailoring the Apoptotic Response to a MET Therapeutic Antibody. Cancers (Basel) 2020; 12:cancers12030741. [PMID: 32245152 PMCID: PMC7140090 DOI: 10.3390/cancers12030741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/11/2020] [Indexed: 12/13/2022] Open
Abstract
The MET oncogene encodes a tyrosine kinase receptor involved in the control of a complex network of biological responses that include protection from apoptosis and stimulation of cell growth during embryogenesis, tissue regeneration, and cancer progression. We previously developed an antagonist antibody (DN30) inducing the physical removal of the receptor from the cell surface and resulting in suppression of the biological responses to MET. In its bivalent form, the antibody displayed a residual agonist activity, due to dimerization of the lingering receptors, and partial activation of the downstream signaling cascade. The balance between the two opposing activities is variable in different biological systems and is hardly predictable. In this study, we generated and characterized two single-chain antibody fragments derived from DN30, sharing the same variable regions but including linkers different in length and composition. The two engineered molecules bind MET with high affinity but induce different biological responses. One behaves as a MET-antagonist, promoting programmed cell death in MET “addicted” cancer cells. The other acts as a hepatocyte growth factor (HGF)-mimetic, protecting normal cells from doxorubicin-induced apoptosis. Thus, by engineering the same receptor antibody, it is possible to generate molecules enhancing or inhibiting apoptosis either to kill cancer cells or to protect healthy tissues from the injuries of chemotherapy.
Collapse
|
23
|
Peng L, Agogo GO, Guo J, Yan M. Substance P and fibrotic diseases. Neuropeptides 2019; 76:101941. [PMID: 31256921 DOI: 10.1016/j.npep.2019.101941] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023]
Abstract
Substance P (SP) is an undecapeptide encoding the tachykinin 1 (TAC1) gene and belongs to the tachykinin family. SP is widely distributed in the central nervous system and the peripheral nervous system. SP is also produced by nonneuronal cells, such as inflammatory cells and endothelial cells. The biological activities of SP are mainly regulated through the high-affinity neurokinin 1 receptor (NK-1R). The SP/NK-1R system plays an important role in the molecular bases of many human pathophysiologic processes, such as pain, infectious and inflammatory diseases, and cancer. In addition, this system has been implicated in fibrotic diseases and processes such as wound healing, myocardial fibrosis, bowel fibrosis, myelofibrosis, renal fibrosis, and lung fibrosis. Recently, studies have shown that SP plays an important role in liver fibrosis and that NK-1R antagonists can inhibit the progression of fibrosis. NK-1R receptor antagonists could provide clinical solutions for fibrotic diseases. This review summarizes the structure and function of SP and its involvement in fibrotic diseases.
Collapse
Affiliation(s)
- Lei Peng
- Department of Gastroenterology, The Second Hospital of Shandong University, Jinan, Shandong Province, China.
| | - George O Agogo
- Department of Internal Medicine, Medical School of Yale University, New Haven, CT 06511, USA.
| | - Jianqiang Guo
- Department of Gastroenterology, The Second Hospital of Shandong University, Jinan, Shandong Province, China.
| | - Ming Yan
- Department of Hepatology and Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
24
|
Moon SH, Lee CM, Park SH, Jin Nam M. Effects of hepatocyte growth factor gene-transfected mesenchymal stem cells on dimethylnitrosamine-induced liver fibrosis in rats. Growth Factors 2019; 37:105-119. [PMID: 31452434 DOI: 10.1080/08977194.2019.1652399] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Nowadays, transplantation of human mesenchymal stem cells (MSCs) has emerged as a potential cellular therapy for liver cirrhosis. Hepatocyte growth factor (HGF) plays an important role in the regeneration of the liver. The objective of the study was to investigate the therapeutic effect of HGF-transfected human umbilical cord blood-derived MSCs on dimethylnitrosamine (DMN)-induced liver fibrosis in rats. HGF-transfected MSCs were transplanted into rats with DMN-induced liver fibrosis. H2O2-induced cytotoxicity, apoptosis and intracellular reactive oxygen species were reduced in HGF-transfected MSCs in HGF-transfected MSCs. Pro-apoptotic proteins, such as cleaved poly (ADP-ribose) polymerase and cleaved caspase-3, were decreased in HGF-transfected MSCs. Biochemical analysis showed that the levels of aspartate aminotransferase and alanine aminotransferase were decreased after transplantation of HGF-transfected MSCs in rat fibrosis. Trichrome staining showed that HGF-transfected MSCs reduced liver damage. Taken together, our study indicated that HGF-transfected MSCs have therapeutic effects on DMN-induced liver fibrosis in rats.
Collapse
Affiliation(s)
- Soung Hoon Moon
- Department of Biological Science, Gachon University, Seongnam, Republic of Korea
| | - Chang Min Lee
- Department of Biological Science, Gachon University, Seongnam, Republic of Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, Republic of Korea
| | - Myeong Jin Nam
- Department of Biological Science, Gachon University, Seongnam, Republic of Korea
- HanCell Inc, Seongnam, Republic of Korea
| |
Collapse
|
25
|
Almalé L, García-Álvaro M, Martínez-Palacián A, García-Bravo M, Lazcanoiturburu N, Addante A, Roncero C, Sanz J, de la O López M, Bragado P, Mikulits W, Factor VM, Thorgeirsson SS, Casal JI, Segovia JC, Rial E, Fabregat I, Herrera B, Sánchez A. c-Met Signaling Is Essential for Mouse Adult Liver Progenitor Cells Expansion After Transforming Growth Factor-β-Induced Epithelial-Mesenchymal Transition and Regulates Cell Phenotypic Switch. Stem Cells 2019; 37:1108-1118. [PMID: 31108004 DOI: 10.1002/stem.3038] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/08/2019] [Accepted: 04/29/2019] [Indexed: 01/10/2023]
Abstract
Adult hepatic progenitor cells (HPCs)/oval cells are bipotential progenitors that participate in liver repair responses upon chronic injury. Recent findings highlight HPCs plasticity and importance of the HPCs niche signals to determine their fate during the regenerative process, favoring either fibrogenesis or damage resolution. Transforming growth factor-β (TGF-β) and hepatocyte growth factor (HGF) are among the key signals involved in liver regeneration and as component of HPCs niche regulates HPCs biology. Here, we characterize the TGF-β-triggered epithelial-mesenchymal transition (EMT) response in oval cells, its effects on cell fate in vivo, and the regulatory effect of the HGF/c-Met signaling. Our data show that chronic treatment with TGF-β triggers a partial EMT in oval cells based on coexpression of epithelial and mesenchymal markers. The phenotypic and functional profiling indicates that TGF-β-induced EMT is not associated with stemness but rather represents a step forward along hepatic lineage. This phenotypic transition confers advantageous traits to HPCs including survival, migratory/invasive and metabolic benefit, overall enhancing the regenerative potential of oval cells upon transplantation into a carbon tetrachloride-damaged liver. We further uncover a key contribution of the HGF/c-Met pathway to modulate the TGF-β-mediated EMT response. It allows oval cells expansion after EMT by controlling oxidative stress and apoptosis, likely via Twist regulation, and it counterbalances EMT by maintaining epithelial properties. Our work provides evidence that a coordinated and balanced action of TGF-β and HGF are critical for achievement of the optimal regenerative potential of HPCs, opening new therapeutic perspectives. Stem Cells 2019;37:1108-1118.
Collapse
Affiliation(s)
- Laura Almalé
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - María García-Álvaro
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Adoración Martínez-Palacián
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - María García-Bravo
- Cell Differentiation and Cytometry Unit, Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Advanced Therapies Mixed Unit, CIEMAT/IIS Fundación Jiménez Díaz, Madrid, Spain
| | - Nerea Lazcanoiturburu
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Annalisa Addante
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Cesáreo Roncero
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Julián Sanz
- Department of Pathology, Hospital Clínico San Carlos, Madrid, Spain
| | - María de la O López
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Paloma Bragado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Wolfgang Mikulits
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Valentina M Factor
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Snorri S Thorgeirsson
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.,Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - J Ignacio Casal
- Department of Functional Proteomics, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - José-Carlos Segovia
- Cell Differentiation and Cytometry Unit, Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Advanced Therapies Mixed Unit, CIEMAT/IIS Fundación Jiménez Díaz, Madrid, Spain
| | - Eduardo Rial
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Blanca Herrera
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Aránzazu Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
26
|
MET/HGF Co-Targeting in Pancreatic Cancer: A Tool to Provide Insight into the Tumor/Stroma Crosstalk. Int J Mol Sci 2018; 19:ijms19123920. [PMID: 30544501 PMCID: PMC6321305 DOI: 10.3390/ijms19123920] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 02/07/2023] Open
Abstract
The ‘onco-receptor’ MET (Hepatocyte Growth Factor Receptor) is involved in the activation of the invasive growth program that is essential during embryonic development and critical for wound healing and organ regeneration during adult life. When aberrantly activated, MET and its stroma-secreted ligand HGF (Hepatocyte Growth Factor) concur to tumor onset, progression, and metastasis in solid tumors, thus representing a relevant target for cancer precision medicine. In the vast majority of tumors, wild-type MET behaves as a ‘stress-response’ gene, and relies on ligand stimulation to sustain cancer cell ‘scattering’, invasion, and protection form apoptosis. Moreover, the MET/HGF axis is involved in the crosstalk between cancer cells and the surrounding microenvironment. Pancreatic cancer (namely, pancreatic ductal adenocarcinoma, PDAC) is an aggressive malignancy characterized by an abundant stromal compartment that is associated with early metastases and resistance to conventional and targeted therapies. Here, we discuss the role of the MET/HGF axis in tumor progression and dissemination considering as a model pancreatic cancer, and provide a proof of concept for the application of dual MET/HGF inhibition as an adjuvant therapy in pancreatic cancer patients.
Collapse
|
27
|
Salum GM, Bader El Din NG, Ibrahim MK, Dawood RM, Farouk S, El Awady MK. Correlation Between TGF-β1 and c-MET Expression in HCV Genotype 4-Induced Liver Fibrosis. J Interferon Cytokine Res 2018; 38:552-558. [DOI: 10.1089/jir.2018.0096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Ghada M. Salum
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| | - Noha G. Bader El Din
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| | - Marwa K. Ibrahim
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| | - Reham M. Dawood
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| | - Sally Farouk
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| | - Mostafa K. El Awady
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| |
Collapse
|
28
|
Kawai Y, Kishimoto Y, Sogami T, Suzuki R, Tsuji T, Hiwatashi N, Tateya I, Kanemaru SI, Nakamura T, Omori K, Hirano S. Characterization of aged rat vocal fold fibroblasts. Laryngoscope 2018; 129:E94-E101. [PMID: 30450675 DOI: 10.1002/lary.27464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVES/HYPOTHESIS To elucidate the aging physiology of the vocal folds, we examined the characters of aged vocal fold fibroblasts (VFFs) in various conditions. STUDY DESIGN In vitro study. METHODS VFFs from young (12-week-old) and aged (19-month-old) Sprague-Dawley rats were compared. Proliferative capacity, ratio of myofibroblast to fibroblast, myofibroblast function, and extracellular matrix production were examined in the following conditions: naïve, basic fibroblast growth factor (bFGF) supplemented, and hepatocyte growth factor (HGF) supplemented. RESULTS Aged VFFs demonstrated reduced proliferation by cell counting, though the ratio of Ki-67-positive cells showed no difference. Aged VFFs exhibited an increased expression of α-smooth muscle actin (α-SMA); however, they demonstrated no enhanced contractile ability in a gel contraction assay. Type I collagen protein was increased age dependently, accompanied with decreased Mmp1 and unchanged Col1a1 transcription. Type I collagen protein and α-SMA represented quite similar reduction patterns to bFGF or HGF administration. CONCLUSIONS The following possible characteristics of aged VFFs were implied: long duration of mitosis, increased myofibroblast population size with certain dysfunctions, reduced type I collagen turnover, and correlation between α-SMA expression and type I collagen metabolism. Further investigations of these features will help to clarify presbyphonia's pathology and establish treatment strategies. LEVEL OF EVIDENCE NA Laryngoscope, 129:E94-E101, 2019.
Collapse
Affiliation(s)
- Yoshitaka Kawai
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yo Kishimoto
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tohru Sogami
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryo Suzuki
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takuya Tsuji
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nao Hiwatashi
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ichiro Tateya
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shin-Ichi Kanemaru
- Department of Otolaryngology-Head and Neck Surgery, Kitano Hospital, Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Tatsuo Nakamura
- Department of Bioartificial Organs, Institute for Frontier Medical Science, Kyoto University, Kyoto, Japan
| | - Koichi Omori
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigeru Hirano
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
29
|
Papaccio F, Della Corte CM, Viscardi G, Di Liello R, Esposito G, Sparano F, Ciardiello F, Morgillo F. HGF/MET and the Immune System: Relevance for Cancer Immunotherapy. Int J Mol Sci 2018; 19:E3595. [PMID: 30441809 PMCID: PMC6274701 DOI: 10.3390/ijms19113595] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/09/2018] [Accepted: 11/11/2018] [Indexed: 12/20/2022] Open
Abstract
An overactivation of hepatocyte growth factor (HGF)/mesenchymal-epithelial transition factor (MET) axis promotes tumorigenesis and tumor progression in various cancer types. Research data recently evidenced that HGF/MET signaling is also involved also in the immune response, mainly modulating dendritic cells functions. In general, the pathway seems to play an immunosuppressive role, thus hypothesizing that it could constitute a mechanism of primary and acquired resistance to cancer immunotherapy. Recently, some approaches are being developed, including drug design and cell therapy to combine MET and programmed cell death receptor-1 (PD-1)/programmed cell death receptor-ligand 1 (PD-L1) inhibition. This approach could represent a new weapon in cancer therapy in the future.
Collapse
Affiliation(s)
- Federica Papaccio
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Carminia Maria Della Corte
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Giuseppe Viscardi
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Raimondo Di Liello
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Giovanna Esposito
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Francesca Sparano
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Fortunato Ciardiello
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Floriana Morgillo
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| |
Collapse
|
30
|
c-Met Signaling Protects from Nonalcoholic Steatohepatitis- (NASH-) Induced Fibrosis in Different Liver Cell Types. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6957497. [PMID: 30538805 PMCID: PMC6260421 DOI: 10.1155/2018/6957497] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/08/2018] [Accepted: 09/19/2018] [Indexed: 12/28/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) is the most common chronic, progressive liver disease in Western countries. The significance of cellular interactions of the HGF/c-Met axis in different liver cell subtypes and its relation to the oxidative stress response remains unclear so far. Hence, the present study is aimed at investigating the role of c-Met and the interaction with the oxidative stress response during NASH development in mice and humans. Conditional c-Met knockout (KO) lines (LysCre for Kupffer cells/macrophages, GFAPCre for α-SMA+ and CK19+ cells and MxCre for bone marrow-derived immune cells) were fed chow and either methionine-choline-deficient diet (MCD) for 4 weeks or high-fat diet (HFD) for 24 weeks. Mice lacking c-Met either in Kupffer cells, α-SMA+ and CK19+ cells, or bone marrow-derived immune cells displayed earlier and faster progressing steatohepatitis during dietary treatments. Severe fatty liver degeneration and histomorphological changes were accompanied by an increased infiltration of immune cells and a significant upregulation of inflammatory cytokine expression reflecting an earlier initiation of steatohepatitis development. In addition, animals with a cell-type-specific deletion of c-Met exhibited a strong generation of reactive oxygen species (ROS) by dihydroethidium (hydroethidine) (DHE) staining showing a significant increase in the oxidative stress response especially in LysCre/c-Metmut and MxCre/c-Metmut animals. All these changes finally lead to earlier and stronger fibrosis progression with strong accumulation of collagen within liver tissue of mice deficient for c-Met in different liver cell types. The HGF/c-Met signaling pathway prevents from steatosis development and has a protective function in the progression to steatohepatitis and fibrosis. It conveys an antifibrotic role independent on which cell type c-Met is missing (Kupffer cells/macrophages, α-SMA+ and CK19+ cells, or bone marrow-derived immune cells). These results highlight a global protective capacity of c-Met in NASH development and progression.
Collapse
|
31
|
Panahi Y, Azimi A, Naderi M, Jadidi K, Sahebkar A. An analytical enrichment-based review of structural genetic studies on keratoconus. J Cell Biochem 2018; 120:4748-4756. [PMID: 30260013 DOI: 10.1002/jcb.27764] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 09/06/2018] [Indexed: 12/19/2022]
Abstract
Keratoconus is a progressive bilateral corneal protrusion that leads to irregular astigmatism and impairment of vision. Keratoconus is an etiologically heterogeneous corneal dystrophy and both environmental and genetic factors play a role in its etiopathogenesis. In this analytical review, we have studied all the genes that are structurally associated with keratoconus and have tried to explain the function of each gene and its association with other eye disorders in a concise way. In addition, using gene set enrichment analysis, it was attempted to find the most important impaired metabolic pathways in keratoconus. Several genetic studies have been carried out on keratoconus and several genes have been identified as risk factors involved in the etiology of the disease. In the current study, 16 studies, including nine association studies, five genome-wide association studies, one linkage study, and one meta-analysis, were reviewed and based on the 19 genes found, enrichment was performed and the most important metabolic pathways involved in the disease were identified. The enrichment results indicated that the two pathways, interleukin 1 processing and assembly of collagen fibrils, are significantly associated with the disease. Obviously, the results of this study, in addition to providing information about the genes involved in the disease, can provide an integrated insight into the gene-based etiology of keratoconus and therapeutic opportunities thereof.
Collapse
Affiliation(s)
- Yunes Panahi
- Chemical Injuries Research Center, System Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Azimi
- Department of Ophthalmology, Poostchi Eye Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mostafa Naderi
- Chemical Injuries Research Center, System Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Khosrow Jadidi
- Chemical Injuries Research Center, System Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic inflammation Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
32
|
Gupta S, Fink MK, Ghosh A, Tripathi R, Sinha PR, Sharma A, Hesemann NP, Chaurasia SS, Giuliano EA, Mohan RR. Novel Combination BMP7 and HGF Gene Therapy Instigates Selective Myofibroblast Apoptosis and Reduces Corneal Haze In Vivo. Invest Ophthalmol Vis Sci 2018; 59:1045-1057. [PMID: 29490341 PMCID: PMC5822743 DOI: 10.1167/iovs.17-23308] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose We tested the potential of bone morphogenic protein 7 (BMP7) and hepatocyte growth factor (HGF) combination gene therapy to treat preformed corneal fibrosis using established rabbit in vivo and human in vitro models. Methods Eighteen New Zealand White rabbits were used. Corneal fibrosis was produced by alkali injury. Twenty-four hours after scar formation, cornea received topically either balanced salt solution (BSS; n = 6), polyethylenimine-conjugated gold nanoparticle (PEI2-GNP)-naked plasmid (n = 6) or PEI2-GNP plasmids expressing BMP7 and HGF genes (n = 6). Donor human corneas were used to obtain primary human corneal fibroblasts and myofibroblasts for mechanistic studies. Gene therapy effects on corneal fibrosis and ocular safety were evaluated by slit-lamp microscope, stereo microscopes, quantitative real-time PCR, immunofluorescence, TUNEL, modified MacDonald-Shadduck scoring system, and Draize tests. Results PEI2-GNP–mediated BMP7+HGF gene therapy significantly decreased corneal fibrosis in live rabbits in vivo (Fantes scale was 0.6 in BMP7+HGF-treated eyes compared to 3.3 in −therapy group; P < 0.001). Corneas that received BMP7+HGF demonstrated significantly reduced mRNA levels of profibrotic genes: α-SMA (3.2-fold; P < 0.01), fibronectin (2.3-fold, P < 0.01), collagen I (2.1-fold, P < 0.01), collagen III (1.6-fold, P < 0.01), and collagen IV (1.9-fold, P < 0.01) compared to the −therapy corneas. Furthermore, BMP7+HGF-treated corneas showed significantly fewer myofibroblasts compared to the −therapy controls (83%; P < 0.001). The PEI2-GNP introduced >104 gene copies per microgram DNA of BMP7 and HGF genes. The recombinant HGF rendered apoptosis in corneal myofibroblasts but not in fibroblasts. Localized topical BMP7+HGF therapy showed no ocular toxicity. Conclusions Localized topical BMP7+HGF gene therapy treats corneal fibrosis and restores transparency in vivo mitigating excessive healing and rendering selective apoptosis in myofibroblasts.
Collapse
Affiliation(s)
- Suneel Gupta
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States.,One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri Columbia, Missouri, United States
| | - Michael K Fink
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States.,One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri Columbia, Missouri, United States
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | - Ratnakar Tripathi
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States.,One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri Columbia, Missouri, United States
| | - Prashant R Sinha
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States.,One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri Columbia, Missouri, United States
| | - Ajay Sharma
- Chapman University School of Pharmacy, Irvine, California, United States
| | - Nathan P Hesemann
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States.,Mason Eye Institute, University of Missouri School of Medicine, Columbia, Missouri, United States
| | - Shyam S Chaurasia
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States.,One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri Columbia, Missouri, United States
| | - Elizabeth A Giuliano
- One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri Columbia, Missouri, United States
| | - Rajiv R Mohan
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States.,One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri Columbia, Missouri, United States.,Mason Eye Institute, University of Missouri School of Medicine, Columbia, Missouri, United States
| |
Collapse
|
33
|
Bell EJ, Decker PA, Tsai MY, Pankow JS, Hanson NQ, Wassel CL, Larson NB, Cohoon KP, Budoff MJ, Polak JF, Stein JH, Bielinski SJ. Hepatocyte growth factor is associated with progression of atherosclerosis: The Multi-Ethnic Study of Atherosclerosis (MESA). Atherosclerosis 2018; 272:162-167. [PMID: 29609131 PMCID: PMC5908230 DOI: 10.1016/j.atherosclerosis.2018.03.040] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 02/16/2018] [Accepted: 03/22/2018] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND AIMS Hepatocyte growth factor (HGF) has previously been associated with risk of stroke, coronary heart disease, and atherosclerosis. We hypothesized that higher circulating HGF is associated with greater progression of measures of atherosclerosis: coronary artery calcium (CAC) and carotid plaque. METHODS Participants aged 45-84 years from the prospective cohort study Multi-Ethnic Study of Atherosclerosis had HGF measured at baseline (between 2000 and 2002) and were followed for progression of atherosclerosis for up to 12 years. CAC was measured at all five exams using the Agatston method. Mixed-effects models were used to examine the association of HGF and CAC progression among 6695 participants with available data. Relative risk regression was used to assess the association between HGF and new or additional carotid plaque between exams 1 and 5 in 3400 participants with available data. All point estimates were adjusted for potential confounding variables. RESULTS Each standard deviation higher HGF at baseline was associated with 2.9 Agatston units/year greater CAC progression (95% CI: 1.6-4.2, p < 0.0001), and the magnitude of this association differed by race/ethnicity (p value for interaction by race = 0.003). Each standard deviation higher HGF at baseline was associated with a 4% higher risk of new or additional carotid plaque (95% CI: 1.01-1.08, p = 0.005). CONCLUSIONS Higher levels of HGF were significantly associated with greater progression of atherosclerosis in this large and diverse population. Circulating HGF continues to show promise as a potential clinical biomarker for cardiovascular disease.
Collapse
Affiliation(s)
- Elizabeth J Bell
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Paul A Decker
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Michael Y Tsai
- Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Naomi Q Hanson
- Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Christina L Wassel
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Colchester, VT, USA
| | - Nicholas B Larson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Kevin P Cohoon
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | | | | | - James H Stein
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Suzette J Bielinski
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
34
|
Graupp M, Rinner B, Frisch MT, Weiss G, Fuchs J, Sundl M, El-Heliebi A, Moser G, Kamolz LP, Karbiener M, Gugatschka M. Towards an in vitro fibrogenesis model of human vocal fold scarring. Eur Arch Otorhinolaryngol 2018. [PMID: 29520499 PMCID: PMC5893733 DOI: 10.1007/s00405-018-4922-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background Vocal fold (VF) scarring remains a therapeutic dilemma and challenge in modern laryngology. To facilitate corresponding research, we aimed to establish an in vitro fibrogenesis model employing human VF fibroblasts (hVFF) and the principles of macromolecular crowding (MMC). Methods Fibrogenesis was promoted by addition of transforming growth factor-β1 to standard medium and medium containing inert macromolecules (MMC). Hepatocyte growth factor (HGF) and Botox type A were tested for their antifibrotic properties in various doses. Experiments were analyzed with respect to the biosynthesis of collagen, fibronectin, and α-smooth muscle actin using immunofluorescence, silver stain and western blot. Results MMC led to favourable enhanced deposition of collagen and other extracellular matrix components, reflecting fibrotic conditions. Low doses of HGF were able to dampen profibrotic effects. This could not be observed for higher HGF concentrations. Botox type A did not show any effects. Conclusion Based on the principles of MMC we could successfully establish a laryngeal fibrogenesis model employing hVFF. Our finding of dose-dependent HGF effects is important before going into clinical trials in humans and has never been shown before. Our model provides a novel option to screen various potential antifibrotic compounds under standardized conditions in a short time. Electronic supplementary material The online version of this article (10.1007/s00405-018-4922-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M Graupp
- Department of Phoniatrics, ENT University Hospital Graz, Medical University of Graz, Auenbruggerplatz 26, 8036, Graz, Austria
| | - B Rinner
- Division of Biomedical Research, Core Facility Alternative Biomodels and Preclinical Imaging, Medical University of Graz, Graz, Austria
| | - M T Frisch
- Division of Biomedical Research, Core Facility Alternative Biomodels and Preclinical Imaging, Medical University of Graz, Graz, Austria
| | - G Weiss
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - J Fuchs
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - M Sundl
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - A El-Heliebi
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - G Moser
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - L P Kamolz
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - M Karbiener
- Department of Phoniatrics, ENT University Hospital Graz, Medical University of Graz, Auenbruggerplatz 26, 8036, Graz, Austria.
| | - M Gugatschka
- Department of Phoniatrics, ENT University Hospital Graz, Medical University of Graz, Auenbruggerplatz 26, 8036, Graz, Austria
| |
Collapse
|
35
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function, and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2018:1-87. [DOI: 10.1016/b978-0-7020-6697-9.00001-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
36
|
Kumar R, Jain AG, Rashid MU, Ali S, Khetpal N, Hussain I, Ahmad S. HGFR and FGR2: Their Roles in Progression and Metastasis of Esophageal Cancer. ROLE OF TYROSINE KINASES IN GASTROINTESTINAL MALIGNANCIES 2018:1-14. [DOI: 10.1007/978-981-13-1486-5_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
37
|
Miyagi H, Thomasy SM, Russell P, Murphy CJ. The role of hepatocyte growth factor in corneal wound healing. Exp Eye Res 2018; 166:49-55. [PMID: 29024692 PMCID: PMC5831200 DOI: 10.1016/j.exer.2017.10.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/19/2017] [Accepted: 10/08/2017] [Indexed: 11/28/2022]
Abstract
Hepatocyte growth factor (HGF) is a glycoprotein produced by mesenchymal cells and operates as a key molecule for tissue generation and renewal. During corneal injury, HGF is primarily secreted by stromal fibroblasts and promotes epithelial wound healing in a paracrine manner. While this mesenchymal-epithelial interaction is well characterized in various organs and the cornea, the role of HGF in corneal stromal and endothelial wound healing is understudied. In addition, HGF has been shown to play an anti-fibrotic role by inhibiting myofibroblast generation and subsequent production of a disorganized extracellular matrix and tissue fibrosis. Therefore, HGF represents a potential therapeutic tool in numerous organs in which myofibroblasts are responsible for tissue scarring. Corneal fibrosis can be a devastating sequela of injury and can result in corneal opacification and retrocorneal membrane formation leading to severe vision loss. In this article, we concisely review the available literature regarding the role of HGF in corneal wound healing. We highlight the influence of HGF on cellular behaviors in each corneal layer. Additionally, we suggest the possibility that HGF may represent a therapeutic tool for interrupting dysregulated corneal repair processes to improve patient outcomes.
Collapse
Affiliation(s)
- Hidetaka Miyagi
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, 1 Shields Ave., Davis, CA, 95616, USA; Department of Ophthalmology and Visual Sciences, Graduate School of Biomedical Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, 7348551, Japan.
| | - Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, 1 Shields Ave., Davis, CA, 95616, USA; Department of Ophthalmology & Vision Science, School of Medicine, UC Davis Medical Center, 2315 Stockton Blvd, Sacramento, CA, 95817, USA.
| | - Paul Russell
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, 1 Shields Ave., Davis, CA, 95616, USA.
| | - Christopher J Murphy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, 1 Shields Ave., Davis, CA, 95616, USA; Department of Ophthalmology & Vision Science, School of Medicine, UC Davis Medical Center, 2315 Stockton Blvd, Sacramento, CA, 95817, USA.
| |
Collapse
|
38
|
Yang YM, Fukui M, Wang Z, Miao F, Karriker MJ, Seki E. Interventional Potential of Recombinant Feline Hepatocyte Growth Factor in a Mouse Model of Non-alcoholic Steatohepatitis. Front Endocrinol (Lausanne) 2018; 9:378. [PMID: 30083132 PMCID: PMC6064873 DOI: 10.3389/fendo.2018.00378] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/22/2018] [Indexed: 12/12/2022] Open
Abstract
Background and Aims: Hepatocyte growth factor (HGF) is a multifunctional pleiotropic protein involved in tissue regeneration, protection, angiogenesis, anti-inflammatory and anti-fibrotic responses, and tumorigenesis, through binding to its receptor MET. Recombinant HGF protein has been shown to mitigate various liver disease models, such as alcohol-induced liver injury, hepatic ischemia-reperfusion injury, and fibrosis. This study aimed to investigate the anti-inflammatory, anti-fibrotic, and anti-lipogenic effects of exogenous administration of feline HGF on a non-alcoholic steatohepatitis (NASH) mouse model. Methods: Wild-type C57BL/6 mice were fed a choline-deficient amino acid defined (CDAA) diet for 3 weeks to create the mouse model of NASH, which displays hepatic steatosis, inflammation, injury, and very mild fibrosis. One mg/kg of recombinant feline HGF was administered intravenously daily in the last 7 days of the total 3 weeks of CDAA diet feeding. Then, hepatic steatosis, inflammation, injury, and fibrogenic gene expression was examined. Results: After 3 weeks of a CDAA diet-feeding, the vehicle-treated mice exhibited evident deposition of lipid droplets in hepatocytes, inflammatory cell infiltration, and hepatocyte ballooning along with increased serum ALT levels whereas recombinant HGF-treated mice showed reduced hepatic steatosis, inflammation, and ballooned hepatocytes with a reduction of serum ALT levels. Recombinant HGF administration promoted hepatocyte proliferation. Increased hepatic lipid accumulation was accompanied by elevated expression of lipogenesis genes Fasn and Dgat1 in vehicle-treated mice. In HGF-treated mice, these genes were reduced with a decrease of lipid accumulation in the liver. Consistent with the anti-inflammatory property of HGF, augmented macrophage infiltration and upregulation of chemokines, Cxcl1, Ccl2, and Ccl5 in the CDAA diet fed mice, were suppressed by the addition of the HGF treatment. Finally, we examined the fibrotic response. The vehicle-treated mice had mild fibrosis with upregulation of Col1a1, Acta2, Timp1, Tgfb1, and Serpine1 expression. Recombinant HGF treatment significantly suppressed fibrogenic gene expression and collagen deposition in the liver. Conclusion: Recombinant feline HGF treatment suppressed the progression of NASH in a CDAA diet feeding mouse model.This suggests that recombinant HGF protein has therapeutic potential for NASH.
Collapse
Affiliation(s)
- Yoon Mee Yang
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Masato Fukui
- Veterinary Medical Center–San Diego, University of California, San Diego, San Diego, CA, United States
| | - Zhijun Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Fiona Miao
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Margo J. Karriker
- Veterinary Medical Center–San Diego, University of California, San Diego, San Diego, CA, United States
| | - Ekihiro Seki
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Division of Gastroenterology, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Ekihiro Seki
| |
Collapse
|
39
|
Dally J, Khan JS, Voisey A, Charalambous C, John HL, Woods EL, Steadman R, Moseley R, Midgley AC. Hepatocyte Growth Factor Mediates Enhanced Wound Healing Responses and Resistance to Transforming Growth Factor-β₁-Driven Myofibroblast Differentiation in Oral Mucosal Fibroblasts. Int J Mol Sci 2017; 18:ijms18091843. [PMID: 28837064 PMCID: PMC5618492 DOI: 10.3390/ijms18091843] [Citation(s) in RCA: 32] [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: 08/02/2017] [Revised: 08/21/2017] [Accepted: 08/23/2017] [Indexed: 01/07/2023] Open
Abstract
Oral mucosal wounds are characterized by rapid healing with minimal scarring, partly attributable to the "enhanced" wound healing properties of oral mucosal fibroblasts (OMFs). Hepatocyte growth factor (HGF) is a pleiotropic growth factor, with potential key roles in accelerating healing and preventing fibrosis. HGF can exist as full-length or truncated (HGF-NK), NK1 and NK2 isoforms. As OMFs display elevated HGF expression compared to dermal fibroblasts (DFs), this study investigated the extent to which HGF mediates the preferential cellular functions of OMFs, and the influence of pro-fibrotic, transforming growth factor-β₁ (TGF-β₁) on these responses. Knockdown of HGF expression in OMFs by short-interfering RNA (siHGF) significantly inhibited OMF proliferative and migratory responses. Supplementation with exogenous TGF-β₁ also significantly inhibited proliferation and migration, concomitant with significantly down-regulated HGF expression. In addition, knockdown abrogated OMF resistance to TGF-β₁-driven myofibroblast differentiation, as evidenced by increased α-smooth muscle actin (α-SMA) expression, F-actin reorganisation, and stress fibre formation. Responses were unaffected in siHGF-transfected DFs. OMFs expressed significantly higher full-length HGF and NK1 levels compared to patient-matched DFs, whilst NK2 expression was similar in both OMFs and DFs. Furthermore, NK2 was preferentially expressed over NK1 in DFs. TGF-β₁ supplementation significantly down-regulated full-length HGF and NK1 expression by OMFs, while NK2 was less affected. This study demonstrates the importance of HGF in mediating "enhanced" OMF cellular function. We also propose that full-length HGF and HGF-NK1 convey desirable wound healing properties, whilst fibroblasts preferentially expressing more HGF-NK2 readily undergo TGF-β₁-driven differentiation into myofibroblasts.
Collapse
Affiliation(s)
- Jordanna Dally
- Stem Cells, Wound Repair & Regeneration, Oral & Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK.
- Cardiff Institute of Tissue Engineering & Repair (CITER), Cardiff University, Cardiff CF10 3AX, UK.
| | - Jabur S Khan
- Stem Cells, Wound Repair & Regeneration, Oral & Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK.
- Cardiff Institute of Tissue Engineering & Repair (CITER), Cardiff University, Cardiff CF10 3AX, UK.
| | - Alex Voisey
- Stem Cells, Wound Repair & Regeneration, Oral & Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK.
- Wales Kidney Research Unit (WKRU), Systems Immunity Research Institute, Division of Infection and Immunity, College of Biomedical & Life Sciences, Cardiff University, Cardiff CF14 4XN, UK.
| | - Chrisandrea Charalambous
- Stem Cells, Wound Repair & Regeneration, Oral & Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK.
- Wales Kidney Research Unit (WKRU), Systems Immunity Research Institute, Division of Infection and Immunity, College of Biomedical & Life Sciences, Cardiff University, Cardiff CF14 4XN, UK.
| | - Hannah L John
- Stem Cells, Wound Repair & Regeneration, Oral & Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK.
- Wales Kidney Research Unit (WKRU), Systems Immunity Research Institute, Division of Infection and Immunity, College of Biomedical & Life Sciences, Cardiff University, Cardiff CF14 4XN, UK.
| | - Emma L Woods
- Stem Cells, Wound Repair & Regeneration, Oral & Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK.
- Cardiff Institute of Tissue Engineering & Repair (CITER), Cardiff University, Cardiff CF10 3AX, UK.
| | - Robert Steadman
- Cardiff Institute of Tissue Engineering & Repair (CITER), Cardiff University, Cardiff CF10 3AX, UK.
- Wales Kidney Research Unit (WKRU), Systems Immunity Research Institute, Division of Infection and Immunity, College of Biomedical & Life Sciences, Cardiff University, Cardiff CF14 4XN, UK.
| | - Ryan Moseley
- Stem Cells, Wound Repair & Regeneration, Oral & Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK.
- Cardiff Institute of Tissue Engineering & Repair (CITER), Cardiff University, Cardiff CF10 3AX, UK.
| | - Adam C Midgley
- Cardiff Institute of Tissue Engineering & Repair (CITER), Cardiff University, Cardiff CF10 3AX, UK.
- Wales Kidney Research Unit (WKRU), Systems Immunity Research Institute, Division of Infection and Immunity, College of Biomedical & Life Sciences, Cardiff University, Cardiff CF14 4XN, UK.
| |
Collapse
|
40
|
Qin Z, Worthen CA, Quan T. Cell-size-dependent upregulation of HGF expression in dermal fibroblasts: Impact on human skin connective tissue aging. J Dermatol Sci 2017; 88:289-297. [PMID: 28826691 DOI: 10.1016/j.jdermsci.2017.08.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/05/2017] [Accepted: 08/02/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND Aged human skin is primarily attributable to loss of collagen, the main structural component of skin. Hepatocyte growth factor (HGF) acts as an anti-fibrotic factor by suppression of collagen production. It is not known whether HGF is involved in age-related collagen deficit in human skin. OBJECTIVE The objective of this study was to investigate the expression of HGF in human skin, and the underlying mechanisms of age-related elevation of HGF expression. METHODS The expression of HGF in young (25±5years, six subjects) and aged (75±6years, six subjects) human skin was determined by laser capture microdissection (LCM) coupled real-time PCR and immunohistology. The underlying mechanisms of age-related elevation of HGF were investigated by reducing dermal fibroblast size, which is a prominent feature of aged skin fibroblast in vivo. RESULTS HGF is predominantly expressed in human skin dermal fibroblasts, the major cells responsible for collagen production, and is significantly elevated in aged human skin in vivo. Mechanistically, reduced fibroblast size, which is a prominent feature of aged skin fibroblasts in vivo, is responsible for age-related elevation of HGF expression. Cell-size-dependent upregulation of HGF expression is driven by increased c-Jun and impaired TGF-β signaling. Restoration of fibroblast size normalizes increased c-Jun expression and impaired TGF-β signaling, and thus reversed the elevated HGF expression. Finally, we confirmed that application of retinoid (ROL), which has been shown to improve aged human skin, significantly reduced elevated HGF mRNA expression in aged human skin in vivo (78±4years, six subjects). CONCLUSION These data reveal a novel mechanism by which reduction of fibroblast size upregulates HGF expression, which in turn contributes to loss of collagen, a prominent feature of aged skin.
Collapse
Affiliation(s)
- Zhaoping Qin
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Christal A Worthen
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
41
|
Panel of three novel serum markers predicts liver stiffness and fibrosis stages in patients with chronic liver disease. PLoS One 2017; 12:e0173506. [PMID: 28301573 PMCID: PMC5354278 DOI: 10.1371/journal.pone.0173506] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/21/2017] [Indexed: 02/06/2023] Open
Abstract
Latest data suggest that placental growth factor (PLGF), growth differentiation factor-15 (GDF-15) and hepatic growth factor (HGF) are involved in hepatic fibrogenesis. Diagnostic performance of these markers for non-invasive liver fibrosis prediction was evaluated based on liver histology and stiffness. In total 834 patients were recruited. Receiver-operating-characteristics were used to define cut-offs for markers correlating to fibrosis stages. Odds-ratios were calculated for the presence/absence of fibrosis/cirrhosis and confirmed in the sub-group of patients phenotyped by elastography only. Logistic and uni- and multivariate regression analyses were used to test for association of markers with liver fibrosis stages and for independent prediction of liver histology and stiffness. Marker concentrations correlated significantly (P<0.001) with histology and stiffness. Cut-offs for liver fibrosis (≥F2) were PLGF = 20.20 pg/ml, GDF15 = 1582.76 pg/ml and HGF = 2598.00 pg/ml. Logistic regression confirmed an increase of ORs from 3.6 over 33.0 to 108.4 with incremental (1–3) markers positive for increased liver stiffness (≥12.8kPa; all P<0.05). Subgroup analysis revealed associations with advanced fibrosis for HCV (three markers positive: OR = 59.9, CI 23.4–153.4, P<0.001) and non-HCV patients (three markers positive: OR = 144, CI 59–3383, P<0.001). Overall, serum markers identified additional 50% of patients at risk for advanced fibrosis presenting with low elastography results. In conclusion, this novel combination of markers reflects the presence of significant liver fibrosis detected by elastography and histology and may also identify patients at risk presenting with low elastography values.
Collapse
|
42
|
Lam BQ, Dai L, Qin Z. The role of HGF/c-MET signaling pathway in lymphoma. J Hematol Oncol 2016; 9:135. [PMID: 27923392 PMCID: PMC5141645 DOI: 10.1186/s13045-016-0366-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/01/2016] [Indexed: 01/15/2023] Open
Abstract
Inappropriate activation of c-mesenchymal-epithelial transition (MET), the receptor tyrosine kinase (RTK) for hepatocyte growth factor (HGF), has been implicated in tumorigenesis and represented a promising therapeutic target for developing anticancer agents. In contrast to other solid tumors, there are limited data describing the functional role of HGF/c-MET signaling pathway in lymphoma. In the current review, we summarize recent findings about the expression, cellular mechanisms/functions, and therapeutic application of HGF/c-MET in different types of lymphoma, especially B cell lymphoma, T and NK cell lymphoma, and Hodgkin lymphoma. We also discuss the existing problems and future directions about studying the HGF/c-MET pathway in lymphoma cells.
Collapse
Affiliation(s)
- Bao Quoc Lam
- Departments of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, Suite 902, 1700 Tulane Ave., New Orleans, LA, 70112, USA
| | - Lu Dai
- Departments of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, Suite 902, 1700 Tulane Ave., New Orleans, LA, 70112, USA.,Department of Oncology, Research Center for Translational Medicine and Key Laboratory of Arrhythmias, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhiqiang Qin
- Departments of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, Suite 902, 1700 Tulane Ave., New Orleans, LA, 70112, USA. .,Department of Oncology, Research Center for Translational Medicine and Key Laboratory of Arrhythmias, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
43
|
Fiore E, Malvicini M, Bayo J, Peixoto E, Atorrasagasti C, Sierra R, Rodríguez M, Gómez Bustillo S, García MG, Aquino JB, Mazzolini G. Involvement of hepatic macrophages in the antifibrotic effect of IGF-I-overexpressing mesenchymal stromal cells. Stem Cell Res Ther 2016; 7:172. [PMID: 27876093 PMCID: PMC5120504 DOI: 10.1186/s13287-016-0424-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/07/2016] [Accepted: 10/18/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Cirrhosis is a major health problem worldwide and new therapies are needed. Hepatic macrophages (hMø) have a pivotal role in liver fibrosis, being able to act in both its promotion and its resolution. It is well-known that mesenchymal stromal cells (MSCs) can modulate the immune/inflammatory cells. However, the effects of MSCs over hMø in the context of liver fibrosis remain unclear. We previously described evidence of the antifibrotic effects of in vivo applying MSCs, which were enhanced by forced overexpression of insulin-like growth factor 1 (AdIGF-I-MSCs). The aim of this work was to analyze the effect of MSCs on hMø behavior in the context of liver fibrosis resolution. METHODS Fibrosis was induced in BALB/c mice by chronic administration of thioacetamide (8 weeks). In vivo gene expression analyses, in vitro experiments using hMø isolated from the nonparenchymal liver cells fraction, and in vivo experiments with depletion of Mø were performed. RESULTS One day after treatment, hMø from fibrotic livers of MSCs-treated animals showed reduced pro-inflammatory and pro-fibrogenic gene expression profiles. These shifts were more pronounced in AdIGF-I-MSCs condition. This group showed a significant upregulation in the expression of arginase-1 and a higher downregulation of iNOS expression thus suggesting decreased levels of oxidative stress. An upregulation in IGF-I and HGF expression was observed in hMø from AdIGF-I-MSCs-treated mice suggesting a restorative phenotype in these cells. Factors secreted by hMø, preconditioned with MSCs supernatant, caused a reduction in the expression levels of hepatic stellate cells pro-fibrogenic and activation markers. Interestingly, hMø depletion abrogated the therapeutic effect achieved with AdIGF-I-MSCs therapy. Expression profile analyses for cell cycle markers were performed on fibrotic livers after treatment with AdIGF-I-MSCs and showed a significant regulation in genes related to DNA synthesis and repair quality control, cell cycle progression, and DNA damage/cellular stress compatible with early induction of pro-regenerative and hepatoprotective mechanisms. Moreover, depletion of hMø abrogated such effects on the expression of the most highly regulated genes. CONCLUSIONS Our results indicate that AdIGF-I-MSCs are able to induce a pro-fibrotic to resolutive phenotype shift on hepatic macrophages, which is a key early event driving liver fibrosis amelioration.
Collapse
Affiliation(s)
- Esteban Fiore
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Mariana Malvicini
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas) Godoy Cruz 2290, Buenos Aires, Argentina
| | - Juan Bayo
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Estanislao Peixoto
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Catalina Atorrasagasti
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas) Godoy Cruz 2290, Buenos Aires, Argentina
| | - Romina Sierra
- Developmental Biology and Regenerative Medicine Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Marcelo Rodríguez
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Sofia Gómez Bustillo
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Mariana G. García
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas) Godoy Cruz 2290, Buenos Aires, Argentina
| | - Jorge B. Aquino
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas) Godoy Cruz 2290, Buenos Aires, Argentina
- Developmental Biology and Regenerative Medicine Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Guillermo Mazzolini
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas) Godoy Cruz 2290, Buenos Aires, Argentina
| |
Collapse
|
44
|
Ductular reaction-on-a-chip: Microfluidic co-cultures to study stem cell fate selection during liver injury. Sci Rep 2016; 6:36077. [PMID: 27796316 PMCID: PMC5086854 DOI: 10.1038/srep36077] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 10/06/2016] [Indexed: 01/09/2023] Open
Abstract
Liver injury modulates local microenvironment, triggering production of signals that instruct stem cell fate choices. In this study, we employed a microfluidic co-culture system to recreate important interactions in the liver stem cell niche, those between adult hepatocytes and liver progenitor cells (LPCs). We demonstrate that pluripotent stem cell-derived LPCs choose hepatic fate when cultured next to healthy hepatocytes but begin biliary differentiation program when co-cultured with injured hepatocytes. We connect this fate selection to skewing in production of hepatocyte growth factor (HGF) and transforming growth factor (TGF)-β1 caused by injury. Significantly, biliary fate selection of LPCs was not observed in the absence of hepatocytes nor did it happen in the presence of TGF-β inhibitors. Our study demonstrates that microfluidic culture systems may offer an interesting new tool for dissecting cellular interactions leading to aberrant stem cell differentiation during injury.
Collapse
|
45
|
Sakamoto K, Khai NC, Wang Y, Irie R, Takamatsu H, Matsufuji H, Kosai KI. Heparin-binding epidermal growth factor-like growth factor and hepatocyte growth factor inhibit cholestatic liver injury in mice through different mechanisms. Int J Mol Med 2016; 38:1673-1682. [PMID: 27779646 PMCID: PMC5117744 DOI: 10.3892/ijmm.2016.2784] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 09/02/2016] [Indexed: 12/23/2022] Open
Abstract
In contrast to hepatocyte growth factor (HGF), the therapeutic potential and pathophysiologic roles of heparin-binding epidermal growth factor-like growth factor (HB-EGF) in liver diseases remain relatively unknown. To address the lack of effective pharmacologic treatments for cholestatic liver injuries, as well as to clarify the biologic features of these growth factors, we explored the effects of HB-EGF and HGF in mice with cholestatic liver injury induced by bile duct ligation (BDL). The mice were assessed 3, 5 and/or 14 days after BDL (acute, subacute and/or chronic phases, respectively) and intravenous injection of adenoviral vector expressing LacZ (control), HB-EGF, HGF, or HB-EGF and HGF. HB-EGF, HGF, or a combination of the growth factors exerted potent antioncotic (antinecrotic), antiapoptotic, anticholestatic, and regenerative effects on hepatocytes in vivo, whereas no robust antiapoptotic or regenerative effects were detected in interlobular bile ducts. Based on serum transaminase levels, the acute protective effects of HB-EGF on hepatocytes were greater than those of HGF. On the other hand, liver fibrosis and cholestasis during the chronic phase were more potently inhibited by HGF compared with HB-EGF. Compared with either growth factor alone, combining HB-EGF and HGF produced greater anticholestatic and regenerative effects during the chronic phase. Taken together, these findings suggest that HB-EGF and HGF inhibited BDL-induced cholestatic liver injury, predominantly by exerting acute cytoprotective and chronic antifibrotic effects, respectively; combining the growth factors enhanced the anticholestatic effects and liver regeneration during the chronic phase. Our results contribute to a better understanding of the pathophysiologic roles of HB-EGF and HGF, as well as to the development of novel effective therapies for cholestatic liver injuries.
Collapse
Affiliation(s)
- Kouichi Sakamoto
- Department of Gene Therapy and Regenerative Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Ngin Cin Khai
- Department of Gene Therapy and Regenerative Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Yuqing Wang
- Department of Gene Therapy and Regenerative Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Rie Irie
- Department of Gene Therapy and Regenerative Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Hideo Takamatsu
- Department of Pediatric Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Hiroshi Matsufuji
- Department of Pediatric Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Ken-Ichiro Kosai
- Department of Gene Therapy and Regenerative Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| |
Collapse
|
46
|
Stella GM, Gentile A, Baderacchi A, Meloni F, Milan M, Benvenuti S. Ockham's razor for the MET-driven invasive growth linking idiopathic pulmonary fibrosis and cancer. J Transl Med 2016; 14:256. [PMID: 27590450 PMCID: PMC5010719 DOI: 10.1186/s12967-016-1008-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 08/16/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) identifies a specific lung disorder characterized by chronic, progressive fibrosing interstitial pneumonia of unknown etiology, which lacks effective treatment. According to the current pathogenic perspective, the aberrant proliferative events in IPF resemble those occurring during malignant transformation. MAIN BODY Receptor tyrosine kinases (RTK) are known to be key players in cancer onset and progression. It has been demonstrated that RTK expression is sometimes also altered and even druggable in IPF. One example of an RTK-the MET proto-oncogene-is a key regulator of invasive growth. This physiological genetic program supports embryonic development and post-natal organ regeneration, as well as cooperating in the evolution of cancer metastasis when aberrantly activated. Growing evidence sustains that MET activation may collaborate in maintaining tissue plasticity and the regenerative potential that characterizes IPF. CONCLUSION The present work aims to elucidate-by applying the logic of simplicity-the bio-molecular mechanisms involved in MET activation in IPF. This clarification is crucial to accurately design MET blockade strategies within a fully personalized approach to IPF.
Collapse
Affiliation(s)
- Giulia M. Stella
- Pneumology Unit, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, Piazzale Golgi 19, 27100 Pavia, Italy
- Investigational Clinical Oncology (INCO), IRCCS Candiolo Cancer Institute-FPO, Candiolo, 20060 Turin, Italy
| | - Alessandra Gentile
- Experimental Clinical Molecular Oncology (ECMO), IRCCS Candiolo Cancer Institute-FPO, Candiolo, 20060 Turin, Italy
| | - Alice Baderacchi
- Investigational Clinical Oncology (INCO), IRCCS Candiolo Cancer Institute-FPO, Candiolo, 20060 Turin, Italy
| | - Federica Meloni
- Pneumology Unit, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, Piazzale Golgi 19, 27100 Pavia, Italy
| | - Melissa Milan
- Experimental Clinical Molecular Oncology (ECMO), IRCCS Candiolo Cancer Institute-FPO, Candiolo, 20060 Turin, Italy
| | - Silvia Benvenuti
- Experimental Clinical Molecular Oncology (ECMO), IRCCS Candiolo Cancer Institute-FPO, Candiolo, 20060 Turin, Italy
| |
Collapse
|
47
|
Kobayashi Y, Kamimura K, Abe H, Yokoo T, Ogawa K, Shinagawa-Kobayashi Y, Goto R, Inoue R, Ohtsuka M, Miura H, Kanefuji T, Suda T, Tsuchida M, Aoyagi Y, Zhang G, Liu D, Terai S. Effects of Fibrotic Tissue on Liver-targeted Hydrodynamic Gene Delivery. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e359. [PMID: 27574785 PMCID: PMC5023407 DOI: 10.1038/mtna.2016.63] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/07/2016] [Indexed: 02/07/2023]
Abstract
Hydrodynamic gene delivery is a common method for gene transfer to the liver of small animals, and its clinical applicability in large animals has been demonstrated. Previous studies focused on functional analyses of therapeutic genes in animals with normal livers and little, however, is known regarding its effectiveness and safety in animals with liver fibrosis. Therefore, this study aimed to examine the effects of liver fibrosis on hydrodynamic gene delivery efficiency using a rat liver fibrosis model. We demonstrated for the first time, using pCMV-Luc plasmid, that this procedure is safe and that the amount of fibrotic tissue in the liver decreases gene delivery efficiency, resulting in decrease in luciferase activity depending on the volume of fibrotic tissue in the liver and the number of hepatocytes that are immunohistochemically stained positive for transgene product. We further demonstrate that antifibrotic gene therapy with matrix metalloproteinase-13 gene reduces liver fibrosis and improves efficiency of hydrodynamic gene delivery. These results demonstrate the negative effects of fibrotic tissue on hydrodynamic gene delivery and its recovery by appropriate antifibrotic therapy.
Collapse
Affiliation(s)
- Yuji Kobayashi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Hiroyuki Abe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Takeshi Yokoo
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Kohei Ogawa
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Yoko Shinagawa-Kobayashi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Ryo Goto
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Ryosuke Inoue
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Masato Ohtsuka
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, School of Medicine, Tokai University, Isehara, Kanagawa, Japan
- The Institute of Medical Sciences, Tokai University, Isehara, Kanagawa Japan
| | - Hiromi Miura
- Department of Regenerative Medicine, Basic Medical Science, School of Medicine, Tokai University, Isehara, Kanagawa, Japan
| | - Tsutomu Kanefuji
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Takeshi Suda
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Masanori Tsuchida
- Division of Thoracic and Cardiovascular Surgery, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Yutaka Aoyagi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Guisheng Zhang
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, USA
| | - Dexi Liu
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, USA
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| |
Collapse
|
48
|
Hirano S, Bless DM, Massey RJ, Hartig GK, Ford CN. Morphological and Functional Changes of Human Vocal Fold Fibroblasts with Hepatocyte Growth Factor. Ann Otol Rhinol Laryngol 2016; 112:1026-33. [PMID: 14703105 DOI: 10.1177/000348940311201206] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Fibroblasts produce extracellular matrix and play an important role in wound healing and scarring. Hepatocyte growth factor (HGF) has strong antifibrotic activity, and has been suggested to have therapeutic potential for treatment of fibrotic diseases. In the present in vitro study, morphological and functional changes of human vocal fold fibroblasts with HGF were examined by transmission electron microscopy and enzyme-linked immunosorbent assay to help clarify the potential use of HGF in the prevention or treatment of vocal fold scarring. The HGF stimulated the production of hyaluronic acid (HA) and decreased the production of collagen type I from the fibroblasts in Reinke's space (FbRS), whereas fibronectin production was not affected. The HGF also changed the shape of the FbRS from an oval shape toward a spindle and stellate shape, and developed Golgi apparatus (GA) and rough endoplasmic reticulum (rER) in the FbRS. The fibroblasts in the macula flava (FbMF) presented with much more production of HA and collagen type I than did FbRS, and were more frequently formed in a stellate shape with well-developed GA and rER. The HGF decreased the production of collagen type I from the FbMF, but barely affected the FbMF in terms of the shape of the cells, the development of GA and rER, or the production of HA. These results were interpreted to suggest that the FbMF are not as susceptible to HGF as are FbRS. On the contrary, HGF appeared to activate the FbRS and modify the function. The increased HA and decreased collagen type I production from the FbRS suggest that HGF may be useful in the prevention or treatment of fibrotic vocal fold scarring.
Collapse
Affiliation(s)
- Shigeru Hirano
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | | | |
Collapse
|
49
|
Libetta C, Esposito P, Martinelli C, Grosjean F, Gregorini M, Rampino T, Dal Canton A. Hepatocyte growth factor (HGF) and hemodialysis: physiopathology and clinical implications. Clin Exp Nephrol 2016; 20:371-378. [PMID: 26676905 DOI: 10.1007/s10157-015-1211-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/02/2015] [Indexed: 02/07/2023]
Abstract
Hepatocyte growth factor (HGF) is a pleiotropic cytokine which exerts a variety of effects on several cells, being involved in the regulation of many biological processes, such as inflammation, tissue repair, morphogenesis, angiogenesis, tumour propagation, immunomodulation of viral infections and cardio-metabolic activities. Patients undergoing regular hemodialysis (HD) present elevated levels of HGF, mainly due to the leukocyte activation associated with HD treatment. High HGF levels might account for specific clinical features of HD patients, i.e. mild liver damage in course of HCV-infection and high cardiovascular risk profile. Moreover, in patients with acute kidney injury, the induction of HGF may represent a crucial step to promote renal recovery, which can have important prognostic consequences in the short and long-term. In this review we discuss the mechanisms underlying HGF production in HD patients, the role of HGF in this particular patient population and the potential clinical implications derived from the study of HGF in HD patients.
Collapse
Affiliation(s)
- Carmelo Libetta
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Piazzale Golgi 2, 27100, Pavia, Italy
| | - Pasquale Esposito
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Piazzale Golgi 2, 27100, Pavia, Italy.
| | - Claudia Martinelli
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Piazzale Golgi 2, 27100, Pavia, Italy
| | - Fabrizio Grosjean
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Piazzale Golgi 2, 27100, Pavia, Italy
| | - Marilena Gregorini
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Piazzale Golgi 2, 27100, Pavia, Italy
| | - Teresa Rampino
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Piazzale Golgi 2, 27100, Pavia, Italy
| | - Antonio Dal Canton
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Piazzale Golgi 2, 27100, Pavia, Italy
| |
Collapse
|
50
|
Mungunsukh O, Lee YH, Bottaro DP, Day RM. The hepatocyte growth factor isoform NK2 activates motogenesis and survival but not proliferation due to lack of Akt activation. Cell Signal 2016; 28:1114-23. [PMID: 27224506 DOI: 10.1016/j.cellsig.2016.05.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/16/2016] [Indexed: 12/14/2022]
Abstract
Hepatocyte growth factor (HGF) is a pleiotrophic factor involved in cellular proliferation, migration and morphogenesis. HGF is required for normal tissue and organ development during embryogenesis, but in the adult HGF has been demonstrated to drive normal tissue repair and inhibit fibrotic remodeling. HGF has two naturally occurring human isoforms as a result of alternative splicing, NK1 and NK2. While NK1 has been defined as an agonist for HGF receptor, Met, NK2 is defined as a partial Met antagonist. Furthermore, under conditions of fibrotic remodeling, NK2 is still expressed while full length HGF is suppressed. Furthermore, the mechanism by which NK2 partially signals through Met is not completely understood. Here, we investigated the mitogenic, motogenic, and anti-apoptotic activities of NK2 compared with full length HGF in primary human bronchial epithelial cells (BEpC) and bovine pulmonary artery endothelial cells (PAEC). In human BEpC, NK2 partial activated Met, inducing Met phosphorylation at Y1234/1235 in the tyrosine-kinase domain but not at Y1349 site in the multifunctional docking domain. Partial phosphorylation of Met by NK2 resulted in activation of MAPK and STAT3, but not AKT. This correlated with motogenesis and survival in a MAPK-dependent manner, but not cell proliferation. Overexpression of a constitutively active AKT complemented NK2 signaling, allowing NK2 to induce cell proliferation. These data indicate that NK2 and HGF drive motogenic and anti-apoptotic signaling but only HGF drives cell proliferation by activating AKT-pathway signaling. These results have implications for the biological consequences of differential regulation of the two isoforms under pro-fibrotic conditions.
Collapse
Affiliation(s)
- Ognoon Mungunsukh
- The Uniformed Services University of the Health Sciences, Department of Pharmacology, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Young H Lee
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Donald P Bottaro
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Regina M Day
- The Uniformed Services University of the Health Sciences, Department of Pharmacology, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| |
Collapse
|