1
|
Sánchez M, Mercader Ruiz J, Marijuán Pinel D, Sánchez P, Fiz N, Guadilla J, Azofra J, Beitia M, Delgado D. Increasing the concentration of plasma molecules improves the biological activity of platelet-rich plasma for tissue regeneration. Sci Rep 2025; 15:4523. [PMID: 39915642 PMCID: PMC11802898 DOI: 10.1038/s41598-025-88918-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/31/2025] [Indexed: 02/09/2025] Open
Abstract
Platelet-rich plasma (PRP) has emerged as a promising therapy in a variety of medical fields. However, it is crucial to go beyond simple platelet concentration and examine the complex molecular composition both inside and outside platelets. The present work studies the effectiveness of a novel type of PRP named 'balanced protein-concentrate plasma' (BPCP). Different growth factor (GF) levels were measured using Enzyme Linked Immunosorbent Assay (ELISA), and in addition to the increase in intra-platelet GFs found in standard PRP (sPRP), BPCP also showed a higher concentration of plasmatic protein. Furthermore, extracellular vesicle (EV) concentration was significantly higher in BPCP. Cell proliferation was higher in cells incubated with lysates derived from BPCP compared to those cultured with sPRP. Regarding cell migration capacity, it was found that the process is platelet-dependent. Finally, the anti-inflammatory effect of BPCP was evaluated by inducing an inflammatory environment in M1-type macrophages. Cytokine levels were measured by ELISA following BPCP administration, showing a significant decrease in pro-inflammatory IL-1β, IL-6 and TNF-α. In summary, although further preclinical and clinical studies are needed in order to determine the therapeutic potential of BPCP, this PRP with unique characteristics demonstrates encouraging in vitro results that could potentially enhance tissue regeneration capacity.
Collapse
Affiliation(s)
- Mikel Sánchez
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain.
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain.
| | - Jon Mercader Ruiz
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Daniel Marijuán Pinel
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Pello Sánchez
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Nicolás Fiz
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Jorge Guadilla
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Juan Azofra
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Maider Beitia
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Diego Delgado
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| |
Collapse
|
2
|
Hassanpour M, Salybkov AA, Kobayashi S, Asahara T. Anti-inflammatory Prowess of endothelial progenitor cells in the realm of biology and medicine. NPJ Regen Med 2024; 9:27. [PMID: 39349482 PMCID: PMC11442670 DOI: 10.1038/s41536-024-00365-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 08/23/2024] [Indexed: 10/02/2024] Open
Abstract
Endothelial inflammation plays a crucial role in vascular-related diseases, a leading cause of global mortality. Among various cellular players, endothelial progenitor cells (EPCs) emerge as non-differentiated endothelial cells circulating in the bloodstream. Recent evidence highlights the transformative role of EPCs in shifting from an inflammatory/immunosuppressive crisis to an anti-inflammatory/immunomodulatory response. Despite the importance of these functions, the regulatory mechanisms governing EPC activities and their physiological significance in vascular regenerative medicine remain elusive. Surprisingly, the current literature lacks a comprehensive review of EPCs' effects on inflammatory processes. This narrative review aims to fill this gap by exploring the cutting-edge role of EPCs against inflammation, from molecular intricacies to broader medical perspectives. By examining how EPCs modulate inflammatory responses, we aim to unravel their anti-inflammatory significance in vascular regenerative medicine, deepening insights into EPCs' molecular mechanisms and guiding future therapeutic strategies targeting vascular-related diseases.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Shonan Research, Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Center for Cell therapy & Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Amankeldi A Salybkov
- Shonan Research, Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Center for Cell therapy & Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Shuzo Kobayashi
- Shonan Research, Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Takayuki Asahara
- Shonan Research, Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan.
- Center for Cell therapy & Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan.
| |
Collapse
|
3
|
Ran C, Olofsgård FJ, Wellfelt K, Steinberg A, Belin AC. Elevated cytokine levels in the central nervous system of cluster headache patients in bout and in remission. J Headache Pain 2024; 25:121. [PMID: 39044165 PMCID: PMC11267889 DOI: 10.1186/s10194-024-01829-9] [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: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Cluster headache is characterized by activation of the trigeminovascular pathway with subsequent pain signalling in the meningeal vessels, and inflammation has been suggested to play a role in the pathophysiology. To further investigate inflammation in cluster headache, inflammatory markers were analysed in patients with cluster headache and controls. METHODS We performed a case-control study, collecting cerebrospinal fluid and serum samples from healthy controls, cluster headache patients in remission, active bout, and during an attack to cover the dynamic range of the cluster headache phenotype. Inflammatory markers were quantified using Target 48 OLINK cytokine panels. RESULTS Altered levels of several cytokines were found in patients with cluster headache compared to controls. CCL8, CCL13, CCL11, CXCL10, CXCL11, HGF, MMP1, TNFSF10 and TNFSF12 levels in cerebrospinal fluid were comparable in active bout and remission, though significantly higher than in controls. In serum samples, CCL11 and CXCL11 displayed decreased levels in patients. Only one cytokine, IL-13 was differentially expressed in serum during attacks. CONCLUSION AND INTERPRETATION Our data shows signs of possible neuroinflammation occurring in biological samples from cluster headache patients. Increased cerebrospinal fluid cytokine levels are detectable in active bout and during remission, indicating neuroinflammation could be considered a marker for cluster headache and is unrelated to the different phases of the disorder.
Collapse
Affiliation(s)
- Caroline Ran
- Centre for Cluster Headache, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | | - Katrin Wellfelt
- Centre for Cluster Headache, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Steinberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Andrea Carmine Belin
- Centre for Cluster Headache, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Contreras-Zentella ML, Alatriste-Contreras MG, Suárez-Cuenca JA, Hernández-Muñoz R. Gender effect of glucose, insulin/glucagon ratio, lipids, and nitrogen-metabolites on serum HGF and EGF levels in patients with diabetes type 2. Front Mol Biosci 2024; 11:1362305. [PMID: 38654922 PMCID: PMC11035728 DOI: 10.3389/fmolb.2024.1362305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Hepatocyte growth factor (HGF) exhibits potent growth-inducing properties across various tissues, while epidermal growth factor (EGF) acts as a molecular integration point for diverse stimuli. HGF plays a crucial role in hepatic metabolism, tissue repair, and offers protective effects on epithelial and non-epithelial organs, in addition to its involvement in reducing apoptosis and inflammation, underscoring its anti-inflammatory capabilities. The HGF-Met system is instrumental in hepatic metabolism and enhancing insulin sensitivity in animal diabetes models. Similarly, the EGF and its receptor tyrosine kinase family (EGFR) are critical in regulating cell growth, proliferation, migration, and differentiation in both healthy and diseased states, with EGF also contributing to insulin sensitivity. In this observational study, we aimed to identify correlations between serum levels of HGF and EGF, insulin, glucagon, glucose, and primary serum lipids in patients with type 2 diabetes mellitus (DM), taking into account the impact of gender. We noted differences in the management of glucose, insulin, and glucagon between healthy men and women, potentially due to the distinct influences of sexual hormones on the development of type 2 DM. Additionally, metabolites such as glucose, albumin, direct bilirubin, nitrites, and ammonia might influence serum levels of growth factors and hormones. In summary, our results highlight the regulatory role of insulin and glucagon in serum glucose and lipids, along with variations in HGF and EGF levels, which are affected by gender. This link is especially significant in DM, where impaired cell proliferation or repair mechanisms lead to metabolic changes. The gender-based differences in growth factors point to their involvement in the pathophysiology of the disease.
Collapse
Affiliation(s)
- Martha Lucinda Contreras-Zentella
- Departamento de Biología Celular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Martha Gabriela Alatriste-Contreras
- Departamento de Métodos Cuantitativos, División de Estudios Profesionales, Facultad de Economía, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Juan Antonio Suárez-Cuenca
- Departamento de Medicina Interna, Hospital General “Xoco”, Secretaría de Salud (SS), Mexico City, Mexico
| | - Rolando Hernández-Muñoz
- Departamento de Biología Celular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
5
|
Gong J, Ding G, Hao Z, Li Y, Deng A, Zhang C. Elucidating the mechanism of corneal epithelial cell repair: unraveling the impact of growth factors. Front Med (Lausanne) 2024; 11:1384500. [PMID: 38638937 PMCID: PMC11024251 DOI: 10.3389/fmed.2024.1384500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
The repair mechanism for corneal epithelial cell injuries encompasses migration, proliferation, and differentiation of corneal epithelial cells, and extracellular matrix remodeling of the stromal structural integrity. Furthermore, it involves the consequential impact of corneal limbal stem cells (LSCs). In recent years, as our comprehension of the mediating mechanisms underlying corneal epithelial injury repair has advanced, it has become increasingly apparent that growth factors play a pivotal role in this intricate process. These growth factors actively contribute to the restoration of corneal epithelial injuries by orchestrating responses and facilitating specific interactions at targeted sites. This article systematically summarizes the role of growth factors in corneal epithelial cell injury repair by searching relevant literature in recent years, and explores the limitations of current literature search, providing a certain scientific basis for subsequent basic research and clinical applications.
Collapse
Affiliation(s)
- Jinjin Gong
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Ophthalmology, Jinan Second People’s Hospital, Jinan, China
| | - Gang Ding
- Department of Ophthalmology, Jinan Second People’s Hospital, Jinan, China
| | - Zhongkai Hao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Ophthalmology, Jinan Second People’s Hospital, Jinan, China
| | - Yuchun Li
- Wuxi No. 2 Chinese Medicine Hospital, Wuxi, China
| | - Aijun Deng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Chenming Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Ophthalmology, Jinan Second People’s Hospital, Jinan, China
| |
Collapse
|
6
|
Gantier M, Rispal R, Fourrier A, Ménoret S, Delbos F, Anegon I, Nguyen TH. Cryopreserved cGMP-compliant human pluripotent stem cell-derived hepatic progenitors rescue mice from acute liver failure through rapid paracrine effects on liver cells. Stem Cell Res Ther 2024; 15:71. [PMID: 38475825 DOI: 10.1186/s13287-024-03673-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Liver transplantation remains the only curative treatment for end-stage liver diseases. Unfortunately, there is a drastic organ donor shortage. Hepatocyte transplantation emerged as a viable alternative to liver transplantation. Considering their unique expansion capabilities and their potency to be driven toward a chosen cell fate, pluripotent stem cells are extensively studied as an unlimited cell source of hepatocytes for cell therapy. It has been previously shown that freshly prepared hepatocyte-like cells can cure mice from acute and chronic liver failure and restore liver function. METHODS Human PSC-derived immature hepatic progenitors (GStemHep) were generated using a new protocol with current good manufacturing practice compliant conditions from PSC amplification and hepatic differentiation to cell cryopreservation. The therapeutic potential of these cryopreserved cells was assessed in two clinically relevant models of acute liver failure, and the mode of action was studied by several analytical methods, including unbiased proteomic analyses. RESULTS GStemHep cells present an immature hepatic phenotype (alpha-fetoprotein positive, albumin negative), secrete hepatocyte growth factor and do not express major histocompatibility complex. A single dose of thawed GStemHep rescue mice from sudden death caused by acetaminophen and thioacetamide-induced acute liver failure, both in immunodeficient and immunocompetent animals in the absence of immunosuppression. Therapeutic biological effects were observed as soon as 3 h post-cell transplantation with a reduction in serum transaminases and in liver necrosis. The swiftness of the therapeutic effect suggests a paracrine mechanism of action of GStemHep leading to a rapid reduction of inflammation as well as a rapid cytoprotective effect with as a result a proteome reprograming of the host hepatocytes. The mode of action of GStemHep relie on the alleviation of inhibitory factors of liver regeneration, an increase in proliferation-promoting factors and a decrease in liver inflammation. CONCLUSIONS We generated cryopreserved and current good manufacturing practice-compliant human pluripotent stem cell-derived immature hepatic progenitors that were highly effective in treating acute liver failure through rapid paracrine effects reprogramming endogenous hepatocytes. This is also the first report highlighting that human allogeneic cells could be used as cryopreserved cells and in the absence of immunosuppression for human PSC-based regenerative medicine for acute liver failure.
Collapse
Affiliation(s)
- Malika Gantier
- GoLiver Therapeutics, 44007, Nantes, France.
- Nantes Université, Inserm, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44000, Nantes, France.
| | - Raphaël Rispal
- Nantes Université, Inserm, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44000, Nantes, France
| | | | - Séverine Ménoret
- Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016 CNRS UMS 3556, 44000, Nantes, France
| | | | - Ignacio Anegon
- Nantes Université, Inserm, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44000, Nantes, France
| | | |
Collapse
|
7
|
Insua A, Galindo-Moreno P, Miron RJ, Wang HL, Monje A. Emerging factors affecting peri-implant bone metabolism. Periodontol 2000 2024; 94:27-78. [PMID: 37904311 DOI: 10.1111/prd.12532] [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: 05/03/2023] [Revised: 08/05/2023] [Accepted: 09/10/2023] [Indexed: 11/01/2023]
Abstract
Implant dentistry has evolved to the point that standard implant osseointegration is predictable. This is attributed in part to the advancements in material sciences that have led toward improvements in implant surface technology and characteristics. Nonetheless, there remain several cases where implant therapy fails (specifically at early time points), most commonly attributed to factors affecting bone metabolism. Among these patients, smokers are known to have impaired bone metabolism and thus be subject to higher risks of early implant failure and/or late complications related to the stability of the peri-implant bone and mucosal tissues. Notably, however, emerging data have unveiled other critical factors affecting osseointegration, namely, those related to the metabolism of bone tissues. The aim of this review is to shed light on the effects of implant-related factors, like implant surface or titanium particle release; surgical-related factors, like osseodensification or implanted biomaterials; various drugs, like selective serotonin reuptake inhibitors, proton pump inhibitors, anti-hypertensives, nonsteroidal anti-inflammatory medication, and statins, and host-related factors, like smoking, diet, and metabolic syndrome on bone metabolism, and aseptic peri-implant bone loss. Despite the infectious nature of peri-implant biological complications, these factors must be surveyed for the effective prevention and management of peri-implantitis.
Collapse
Affiliation(s)
- Angel Insua
- Department of Periodontology and Oral Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Pablo Galindo-Moreno
- Department of Periodontology and Oral Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Oral Surgery and Implant Dentistry, University of Granada, Granada, Spain
| | - Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Hom-Lay Wang
- Department of Periodontology and Oral Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alberto Monje
- Department of Periodontology and Oral Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Periodontology, University of Bern, Bern, Switzerland
- Department of Periodontology, Universitat Internacional de Catalunya, Barcelona, Spain
| |
Collapse
|
8
|
Avtanski D, Stojchevski R. Significance of Adipose Tissue as an Endocrine Organ. CONTEMPORARY ENDOCRINOLOGY 2024:1-46. [DOI: 10.1007/978-3-031-72570-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Boonpethkaew S, Meephansan J, Ponnikorn S, Jumlongpim O, Juntongjin P, Chakkavittumrong P, Wongpiyabovorn J, Morita A, Komine M. Exploring the role of growth factors as potential regulators in psoriatic plaque formation. Exp Dermatol 2023; 32:1924-1934. [PMID: 37665186 DOI: 10.1111/exd.14918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023]
Abstract
Psoriasis is a chronic inflammatory skin disease in which growth activity is more prominent than inflammatory activity at the centre of lesional skin (CE skin). This growth activity is partly influenced by growth factors (GFs) that play an important role in cell growth and inflammation during the plaque development. In this study, we identified potential GFs in CE skin and predicted their regulatory functions and biological activity in mediating transcripts in the plaques. Samples of uninvolved skin (UN skin) and CE skin were biopsied from patients with psoriasis vulgaris for RNA-sequencing analysis in order to identify differentially expressed genes (DEGs). Our finding revealed that epidermal growth factor (EGF), fibroblast growth factor (FGF), platelet-derived growth factor (PDGF) and hepatocyte growth factor (HGF) signalling were enriched by CE/UN skin-derived DEGs. Additionally, several EGFR ligands, namely EGF, heparin-binding EGF like growth factor (HB-EGF), amphiregulin (AREG) and transforming growth factor (TGF)-α, as well as TGF-β1, TGF-β2, vascular endothelial growth factor-A, FGFs, PDGF-B and HGF, were predicted to be GF regulators. The regulatory pattern and biological activity of these GF regulators on mediating the CE/UN skin-derived DEGs was demonstrated. This study provides a novel hypothesis regarding the overall regulatory function of GFs, which appear to modulate the expression of the transcripts involved in inflammation and growth in the CE skin. In addition, some GFs may exert anti-inflammatory effects. Further investigations on the mechanisms underlying this regulation may contribute to a deeper understanding of psoriasis and the identification of potential therapeutic targets for patients with psoriasis.
Collapse
Affiliation(s)
- Suphagan Boonpethkaew
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
- Thammasat University, Chonburi, Thailand
| | - Jitlada Meephansan
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Saranyoo Ponnikorn
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
- Thammasat University, Chonburi, Thailand
| | - Onjira Jumlongpim
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Premjit Juntongjin
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Panlop Chakkavittumrong
- Division of Dermatology, Department of Internal Medicine, Thammasat University, Pathum Thani, Thailand
| | - Jongkonnee Wongpiyabovorn
- Center of Excellence in Immunology and Immune-Mediated Disease, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Mayumi Komine
- Department of Dermatology, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
10
|
Sun K, Li X, Scherer PE. Extracellular Matrix (ECM) and Fibrosis in Adipose Tissue: Overview and Perspectives. Compr Physiol 2023; 13:4387-4407. [PMID: 36715281 PMCID: PMC9957663 DOI: 10.1002/cphy.c220020] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fibrosis in adipose tissue is a major driver of obesity-related metabolic dysregulation. It is characterized by an overaccumulation of extracellular matrix (ECM) during unhealthy expansion of adipose tissue in response to over nutrition. In obese adipose-depots, hypoxia stimulates multiple pro-fibrotic signaling pathways in different cell populations, thereby inducing the overproduction of the ECM components, including collagens, noncollagenous proteins, and additional enzymatic components of ECM synthesis. As a consequence, local fibrosis develops. The result of fibrosis-induced mechanical stress not only triggers cell necrosis and inflammation locally in adipose tissue but also leads to system-wide lipotoxicity and insulin resistance. A better understanding of the mechanisms underlying the obesity-induced fibrosis will help design therapeutic approaches to reduce or reverse the pathological changes associated with obese adipose tissue. Here, we aim to summarize the major advances in the field, which include newly identified fibrotic factors, cell populations that contribute to the fibrosis in adipose tissue, as well as novel mechanisms underlying the development of fibrosis. We further discuss the potential therapeutic strategies to target fibrosis in adipose tissue for the treatment of obesity-linked metabolic diseases and cancer. © 2023 American Physiological Society. Compr Physiol 13:4387-4407, 2023.
Collapse
Affiliation(s)
- Kai Sun
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xin Li
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Philipp E. Scherer
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
11
|
Suppression of lipopolysaccharide-induced corneal opacity by hepatocyte growth factor. Sci Rep 2022; 12:494. [PMID: 35017561 PMCID: PMC8752742 DOI: 10.1038/s41598-021-04418-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/22/2021] [Indexed: 12/21/2022] Open
Abstract
Keratitis induced by bacterial toxins, including lipopolysaccharide (LPS), is a major cause of corneal opacity and vision loss. Our previous study demonstrates hepatocyte growth factor (HGF) promotes epithelial wound healing following mechanical corneal injury. Here, we investigated whether HGF has the capacity to suppress infectious inflammatory corneal opacity using a new model of LPS-induced keratitis. Keratitis, induced by two intrastromal injections of LPS on day 1 and 4 in C57BL/6 mice, resulted in significant corneal opacity for up to day 10. Following keratitis induction, corneas were topically treated with 0.1% HGF or PBS thrice daily for 5 days. HGF-treated mice showed a significantly smaller area of corneal opacity compared to PBS-treated mice, thus improving corneal transparency. Moreover, HGF treatment resulted in suppression of α-SMA expression, compared to PBS treatment. HGF-treated corneas showed normalized corneal structure and reduced expression of pro-inflammatory cytokine, demonstrating that HGF restores corneal architecture and immune quiescence in corneas with LPS-induced keratitis. These findings offer novel insight into the potential application of HGF-based therapies for the prevention and treatment of infection-induced corneal opacity.
Collapse
|
12
|
Chen CL, Kao CC, Yang MH, Fan GY, Cherng JH, Tsao CW, Wu ST, Cha TL, Meng E. A Novel Intravesical Dextrose Injection Improves Lower Urinary Tract Symptoms on Interstitial Cystitis/Bladder Pain Syndrome. Front Pharmacol 2022; 12:755615. [PMID: 34975473 PMCID: PMC8715092 DOI: 10.3389/fphar.2021.755615] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS) is a painful recurrent condition characterized by the discomfort of the bladder, and current treatment options have limited effectiveness. Prolotherapy is a well-known treatment that involves the injection of non-biologic solutions to reduce pain and/or promote proliferation of soft tissue, and dextrose is the most common injectate. This study investigated the effects of dextrose prolotherapy in a rat model of IC/BPS and patients with IC/BPS. We used cyclophosphamide to induce IC/BPS in rats, and intravesical instillation of 10% dextrose solution was performed. After 1 week, we conducted a urodynamic test, bladder staining, and ECM-related gene expression analysis to examine the treatment’s efficacy. We found that dextrose treatment could recover the instability of the bladder, reduce frequent urination, and improve the glycosaminoglycan layer regeneration and the bladder wall thickness along with a significant intense expression of CD44 receptors. Furthermore, we enrolled 29 IC/BPS patients with previous hyaluronic acid/Botox treatment for more than 6 months with remained unchanged condition. In this study, they received intravesical injections of 10% dextrose solution followed by assessments for up to 12 weeks. Patient characteristics and a 3-day voiding diary before treatment were recorded. Patient responses were examined using IC/BPS-related questionnaires. Moreover, expressions of growth factors and cytokines were analyzed. The results demonstrated that dextrose prolotherapy in patients with IC/BPS reduced the frequency of treatment over time, with the mean number of treatments being 3.03 ± 1.52, and significantly reduced the incidence of nocturia and questionnaire scores associated with symptoms. Dextrose prolotherapy significantly enhanced EGF level and, in contrast, reduced the level of HGF, PIGF-1, and VEGF-D after several weeks following treatment. The cytokine analysis showed that the expressions of IL-12p70 and IL-10 were significantly up-regulated after dextrose prolotherapy in IC/BPS patients. The levels of most growth factors and cytokines in IC/BPS patients had no significant difference and showed a similar tendency as time progressed when compared to healthy controls. Overall, the alteration of growth factors and cytokines exhibited safe treatment and potential stimulation of tissue remodeling. In summary, our study demonstrated that dextrose prolotherapy is a promising treatment strategy for IC/BPS disease management.
Collapse
Affiliation(s)
- Chin-Li Chen
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Chang Kao
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Hsin Yang
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Gang-Yi Fan
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Juin-Hong Cherng
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Wei Tsao
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Sheng-Tang Wu
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tai-Lung Cha
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - En Meng
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department and Graduate Institute of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
13
|
HGF can reduce accumulation of inflammation and regulate glucose homeostasis in T2D mice. J Physiol Biochem 2021; 77:613-624. [PMID: 34363605 DOI: 10.1007/s13105-021-00828-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 07/09/2021] [Indexed: 10/20/2022]
Abstract
Hepatocyte growth factor (HGF) has been studied as a protective factor for the survival of islet β cells and regulatory glucose transport and metabolism in many studies. The addition of exogenous HGF to cells or mice is the most common way to study HGF, but the persistence and stability of the administered HGF are unclear. In this experiment, wild-type C57BL6 (WT) mice and HGF-overexpressing transgenic (HGF-Tg) mice were divided into a normal diet (ND) group and an HFD group. The HGF protein level in the liver, kidney, spleen, pancreas, and VAT of HGF-Tg-ND mice was upregulated compared to that of WT-ND mice, and it was also upregulated in HGF-Tg-HFD mice compared to that in WT-HFD mice. In the ND group, though the HGF-Tg-ND mice showed higher fasted blood glucose levels and larger integrated density (IOD) of glucagon-positive cells than WT-ND mice, we found that HGF-Tg-ND mice can still maintain normal glucose tolerance based on an intraperitoneal glucose tolerance test (IPGTT) and an intraperitoneal insulin tolerance test (IPITT). In the HFD group, the HGF-Tg-HFD mice showed insulin sensitivity in IPGTT and IPITT and had larger areas and higher IOD values of islet β cells and smaller areas and IOD values of islet α cells than the WT-HFD mice. HGF-Tg-HFD mice had lower level of serum insulin than WT-HFD mice. The HGF-Tg-HFD mice showed inhibited accumulation of CD4+ T cells, CD8+ T cells, Ly6G+ neutrophils, and F4/80+ macrophages in the blood and tissues and protected liver and kidney functions. Oil Red O-stained liver sections revealed that WT-HFD mice had larger areas and higher IOD values of Oil Red O-positive cells than HGF-Tg-HFD mice, and WT-HFD mice had higher score of NASH. PAS-stained kidney sections found WT-HFD has higher mesangial area/glomerular area × 100% than HGF-Tg-HFD mice. Comparative analyses demonstrated that HGF reduces the proportions of inflammatory cells in the blood and tissues, and protects liver and kidney tissues by regulating glucose homeostasis of type 2 diabetic mice.
Collapse
|
14
|
Jeske R, Yuan X, Fu Q, Bunnell BA, Logan TM, Li Y. In Vitro Culture Expansion Shifts the Immune Phenotype of Human Adipose-Derived Mesenchymal Stem Cells. Front Immunol 2021; 12:621744. [PMID: 33777002 PMCID: PMC7988085 DOI: 10.3389/fimmu.2021.621744] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem or stromal cells (hMSCs) are known for their potential in regenerative medicine due to their differentiation abilities, secretion of trophic factors, and regulation of immune responses in damaged tissues. Due to the limited quantity of hMSCs typically isolated from bone marrow, other tissue sources, such as adipose tissue-derived mesenchymal stem cells (hASCs), are considered a promising alternative. However, differences have been observed for hASCs in the context of metabolic characteristics and response to in vitro culture stress compared to bone marrow derived hMSCs (BM-hMSCs). In particular, the relationship between metabolic homeostasis and stem cell functions, especially the immune phenotype and immunomodulation of hASCs, remains unknown. This study thoroughly assessed the changes in metabolism, redox cycles, and immune phenotype of hASCs during in vitro expansion. In contrast to BM-hMSCs, hASCs did not respond to culture stress significantly during expansion as limited cellular senescence was observed. Notably, hASCs exhibited the increased secretion of pro-inflammatory cytokines and the decreased secretion of anti-inflammatory cytokines after extended culture expansion. The NAD+/NADH redox cycle and other metabolic characteristics associated with aging were relatively stable, indicating that hASC functional decline may be regulated through an alternative mechanism rather than NAD+/Sirtuin aging pathways as observed in BM-hMSCs. Furthermore, transcriptome analysis by mRNA-sequencing revealed the upregulation of genes for pro-inflammatory cytokines/chemokines and the downregulation of genes for anti-inflammatory cytokines for hASCs at high passage. Proteomics analysis indicated key pathways (e.g., tRNA charging, EIF2 signaling, protein ubiquitination pathway) that may be associated with the immune phenotype shift of hASCs. Together, this study advances our understanding of the metabolism and senescence of hASCs and may offer vital insights for the biomanufacturing of hASCs for clinical use.
Collapse
Affiliation(s)
- Richard Jeske
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Xuegang Yuan
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States.,The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, United States
| | - Qin Fu
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Bruce A Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Timothy M Logan
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL, United States
| | - Yan Li
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States.,Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
15
|
Dregalla RC, Herrera JA, Donner EJ. Soluble factors differ in platelets derived from separate niches: a pilot study comparing the secretome of peripheral blood and bone marrow platelets. Cytotherapy 2021; 23:677-682. [PMID: 33678599 DOI: 10.1016/j.jcyt.2021.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/30/2020] [Accepted: 01/11/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND AIMS Platelet-rich plasma (PRP) and bone marrow aspirate are commonly used in orthobiologics for their anti-inflammatory, anabolic/regenerative and immunomodulatory characteristics via platelet degranulation and cell secretions. Although platelets are derived from megakaryocytes in the bone marrow, no attention has been paid to the potential benefits of bone marrow platelets and whether their contents differ from aging platelets in peripheral blood. METHODS In the present study, leukocyte-poor peripheral blood-derived platelets in plasma (LPP) and leukocyte-poor bone marrow platelets in plasma (BMP) were prepared from six donors, activated with calcium chloride, incubated and sampled at day 0, day 3 and day 6. LPP and BMP are platelet preparations intended to evaluate the respective platelet secretomes but are not classified as conventional PRPs, as they are not concentrated to the extent necessary to meet the qualifying criteria. At each time point, 15 growth and immunomodulatory factors were quantitated in LPP and BMP: platelet-derived growth factor AA, basic fibroblast growth factor/fibroblast growth factor 2, granulocyte-macrophage colony-stimulating factor, hepatocyte growth factor, macrophage colony-stimulating factor, stem cell factor, vascular endothelial growth factor, tumor necrosis factor alpha, IL-1β, interferon gamma, IL-4, IL-10, IL-1 receptor antagonist protein, IL-12p40 and arginase-1. RESULTS The results illustrate that platelets derived from bone marrow have a unique secretome profile compared with those derived from peripheral blood, with significant differences in anti-inflammatory cytokines, which are associated with monocyte polarization. CONCLUSIONS Ultimately, bone marrow-derived platelets may be useful as a stand-alone orthobiologic or as an effective adjuvant to autologous cell therapies where anti-inflammatory and anabolic processes are desired, especially with respect to monocyte function.
Collapse
Affiliation(s)
- Ryan C Dregalla
- Elite Regenerative Stem Cell Specialists, LLC, Johnstown, Colorado, USA; R&D Regenerative Laboratory Resources, LLC, Johnstown, Colorado, USA.
| | - Jessica A Herrera
- Elite Regenerative Stem Cell Specialists, LLC, Johnstown, Colorado, USA; R&D Regenerative Laboratory Resources, LLC, Johnstown, Colorado, USA
| | - Edward J Donner
- Elite Regenerative Stem Cell Specialists, LLC, Johnstown, Colorado, USA; R&D Regenerative Laboratory Resources, LLC, Johnstown, Colorado, USA; Colorado Spine Institute, PLLC, Johnstown, Colorado, USA
| |
Collapse
|
16
|
Wani K, AlHarthi H, Alghamdi A, Sabico S, Al-Daghri NM. Role of NLRP3 Inflammasome Activation in Obesity-Mediated Metabolic Disorders. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:E511. [PMID: 33435142 PMCID: PMC7826517 DOI: 10.3390/ijerph18020511] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/22/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
NLRP3 inflammasome is one of the multimeric protein complexes of the nucleotide-binding domain, leucine-rich repeat (NLR)-containing pyrin and HIN domain family (PYHIN). When activated, NLRP3 inflammasome triggers the release of pro-inflammatory interleukins (IL)-1β and IL-18, an essential step in innate immune response; however, defective checkpoints in inflammasome activation may lead to autoimmune, autoinflammatory, and metabolic disorders. Among the consequences of NLRP3 inflammasome activation is systemic chronic low-grade inflammation, a cardinal feature of obesity and insulin resistance. Understanding the mechanisms involved in the regulation of NLRP3 inflammasome in adipose tissue may help in the development of specific inhibitors for the treatment and prevention of obesity-mediated metabolic diseases. In this narrative review, the current understanding of NLRP3 inflammasome activation and regulation is highlighted, including its putative roles in adipose tissue dysfunction and insulin resistance. Specific inhibitors of NLRP3 inflammasome activation which can potentially be used to treat metabolic disorders are also discussed.
Collapse
Affiliation(s)
| | | | | | | | - Nasser M. Al-Daghri
- Biochemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (K.W.); (H.A.); (A.A.); (S.S.)
| |
Collapse
|
17
|
Park GW, Heo J, Kang JY, Yang JW, Kim JS, Kwon KD, Yu BC, Lee SJ. Topical cell-free conditioned media harvested from adipose tissue-derived stem cells promote recovery from corneal epithelial defects caused by chemical burns. Sci Rep 2020; 10:12448. [PMID: 32709896 PMCID: PMC7381646 DOI: 10.1038/s41598-020-69020-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Corneal chemical burns can lead to blindness following serious complications. As most of these complications are caused by failure of reepithelization during the acute phase, treatment at this stage is critical. Although there have been some studies on corneal injury recovery using adipose tissue-derived stem cells (ADSCs), none has reported the effect of topical cell-free conditioned culture media (CM) derived from ADSCs on corneal epithelial regeneration. Here, the best conditions for CM were selected and used for in vitro and in vivo experiments. Corneal burn in rats was induced using 100% alcohol. The chosen CM was administered to corneal burn rats (CM-treated [CT] group) four times a day for three days and this group was compared with the normal control and corneal burn (CB) groups. Biomicroscopic fluorescence images and the actual physical corneas were taken over time and used for analysis. mRNA levels of hepatocyte growth factor and epidermal growth factor (EGF) were significantly increased, whereas those of vascular endothelial growth factor, interleukin (IL)-1β, IL-6, IL-10, and matrix metalloproteinase-9 were significantly decreased in the CT group compared with those in the CB group. The numbers of proliferating cell nuclear antigen- and zonular occludens-1-positive cells in the CT group were significantly higher than those in the CB group. The macrophage-infiltrating corneas in the CT group expressed significantly more of the M2 marker arginase than corneas in the CB group. Optimal CM (× 0.5 concentration) treatment significantly accelerated the migration of corneal epithelial cells and induced upregulation of the expression of IL-6, EGF, and C-X-C chemokine receptor type 4 mRNAs. Overall, in this study, topical administration of cell-free CM promoted regeneration of the corneal epithelium after induction of chemical burns.
Collapse
Affiliation(s)
| | - Jeonghoon Heo
- Department of Molecular Biology and Immunology, College of Medicine, Kosin University, Busan, Korea
| | | | - Ji Won Yang
- Stem Bank Company, Busan, Korea
- Department of Ophthalmology, College of Medicine, Kosin University, #34 Amnam-dong, Suh-ku, Busan, 602-702, Korea
| | - Jong Sik Kim
- Department of Anatomy, College of Medicine, Kosin University, Busan, Korea
| | - Ki Dong Kwon
- Department of Ophthalmology, College of Medicine, Kosin University, #34 Amnam-dong, Suh-ku, Busan, 602-702, Korea
| | - Byung Chul Yu
- Department of Preventive Medicine, College of Medicine, Kosin University, Busan, Korea
| | - Sang Joon Lee
- Department of Ophthalmology, College of Medicine, Kosin University, #34 Amnam-dong, Suh-ku, Busan, 602-702, Korea.
| |
Collapse
|
18
|
Sanada F, Fujikawa T, Shibata K, Taniyama Y, Rakugi H, Morishita R. Therapeutic Angiogenesis Using HGF Plasmid. Ann Vasc Dis 2020; 13:109-115. [PMID: 32595785 PMCID: PMC7315247 DOI: 10.3400/avd.ra.20-00035] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 03/04/2020] [Indexed: 12/27/2022] Open
Abstract
Hepatocyte growth factor (HGF) is secreted from stromal and mesenchymal cells, and its receptor cMet is expressed on various types of cells such as smooth muscle cells, fibroblast, and endothelial cells. HGF stimulates epithelial and endothelial cell proliferation, motility, and morphogenesis in a paracrine and autocrine manner, organizing multistep of angiogenesis in many organs. In addition, HGF is recognized as a potent anti-inflammatory and anti-fibrotic growth factor, which has been proved in several animal studies, including neointimal hyperplasia and acute myocardial infarction model in rodent. Thus, as compared to other angiogenic growth factors, HGF exerts multiple effects on ischemic tissues, accompanied by the regression of tissue inflammation and fibrosis. These data suggest the therapeutic potential of the HGF for peripheral artery disease as it being accompanied with chronic tissue inflammation and fibrosis. In the present narrative review, the pleiotropic action of the HGF that differentiates it from other angiogenic growth factors is discussed first, and later, outcomes of the human clinical study with gene therapy are overviewed.
Collapse
Affiliation(s)
- Fumihiro Sanada
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tatsuya Fujikawa
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kana Shibata
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshiaki Taniyama
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
19
|
Hu ZP, Fang XL, Sheng B, Guo Y, Yu YQ. Melatonin inhibits macrophage infiltration and promotes plaque stabilization by upregulating anti-inflammatory HGF/c-Met system in the atherosclerotic rabbit: USPIO-enhanced MRI assessment. Vascul Pharmacol 2020; 127:106659. [DOI: 10.1016/j.vph.2020.106659] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/06/2020] [Accepted: 02/14/2020] [Indexed: 01/08/2023]
|
20
|
Chen H, Erndt-Marino J, Diaz-Rodriguez P, Kulwatno J, Jimenez-Vergara AC, Thibeault SL, Hahn MS. In vitro evaluation of anti-fibrotic effects of select cytokines for vocal fold scar treatment. J Biomed Mater Res B Appl Biomater 2019; 107:1056-1067. [PMID: 30184328 PMCID: PMC7011756 DOI: 10.1002/jbm.b.34198] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/30/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023]
Abstract
Scarring of the vocal fold lamina propria (LP) can cause considerable voice disorders due to reduced pliability in scar tissue, attributed in part to abnormal extracellular matrix (ECM) deposition produced by the fibrotic vocal fold fibroblast (fVFF). Cytokines with anti-fibrotic potential have been investigated to limit abnormal LP ECM, but are limited by the need for repeat injections. Moreover, the potentially significant role played by activated macrophages (AMOs) is usually not considered even though the interaction between AMO and fibrotic fibroblasts is known to regulate scar formation across different tissues. AMO are also regulated by cytokines that are used for LP scar removal, but little is known about AMO behaviors in response to these cytokines within the context of LP scar. In the present study, we evaluated anti-fibrotic effects of hepatocyte growth factor (HGF), interleukin-10 (IL-10) and interleukin-6 (IL-6) in a 3D, in vitro fVFF-AMO co-culture system using poly(ethylene glycol) diacrylate (PEGDA) hydrogels. Data from all cytokines was synthesized into a heat-map that enabled assessment of specific associations between AMO and fVFF phenotypes. Cumulatively, our results indicated that both HGF and IL-10 are potentially anti-fibrotic (reduction in fibrotic markers and enhancement in normal, anti-fibrotic VFF markers), while IL-6 displays more complex, marker specific effects. Possible associations between AMO and fVFF phenotypes were found and may highlight a potential desirable macrophage phenotype. These data support the therapeutic potential of HGF and IL-10 for LP scar treatment, and shed light on future strategies aimed at targeting specific AMO phenotypes. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1056-1067, 2019.
Collapse
Affiliation(s)
- Hongyu Chen
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Josh Erndt-Marino
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | | | - Jonathan Kulwatno
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | | | - Susan L Thibeault
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
21
|
Oliveira AG, Araújo TG, Carvalho BDM, Rocha GZ, Santos A, Saad MJA. The Role of Hepatocyte Growth Factor (HGF) in Insulin Resistance and Diabetes. Front Endocrinol (Lausanne) 2018; 9:503. [PMID: 30214428 PMCID: PMC6125308 DOI: 10.3389/fendo.2018.00503] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 08/10/2018] [Indexed: 12/12/2022] Open
Abstract
In obesity, insulin resistance (IR) and diabetes, there are proteins and hormones that may lead to the discovery of promising biomarkers and treatments for these metabolic disorders. For example, these molecules may impair the insulin signaling pathway or provide protection against IR. Thus, identifying proteins that are upregulated in IR states is relevant to the diagnosis and treatment of the associated disorders. It is becoming clear that hepatocyte growth factor (HGF) is an important component of the pathophysiology of IR, with increased levels in most common IR conditions, including obesity. HGF has a role in the metabolic flux of glucose in different insulin sensitive cell types; plays a key role in β-cell homeostasis; and is capable of modulating the inflammatory response. In this review, we discuss how, and to what extent HGF contributes to IR and diabetes pathophysiology, as well as its role in cancer which is more prevalent in obesity and diabetes. Based on the current literature and knowledge, it is clear that HGF plays a central role in these metabolic disorders. Thus, HGF levels could be employed as a biomarker for disease status/progression, and HGF/c-Met signaling pathway modulators could effectively regulate IR and treat diabetes.
Collapse
Affiliation(s)
- Alexandre G. Oliveira
- Department of Internal Medicine, State University of Campinas, Campinas, Brazil
- Department of Physical Education, Institute of Biosciences, São Paulo State University (UNESP), Rio Claro, Brazil
- *Correspondence: Alexandre G. Oliveira
| | - Tiago G. Araújo
- Department of Internal Medicine, State University of Campinas, Campinas, Brazil
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil
| | - Bruno de Melo Carvalho
- Department of Internal Medicine, State University of Campinas, Campinas, Brazil
- Institute of Biological Sciences, University of Pernambuco, Recife, Brazil
| | - Guilherme Z. Rocha
- Department of Internal Medicine, State University of Campinas, Campinas, Brazil
| | - Andrey Santos
- Department of Internal Medicine, State University of Campinas, Campinas, Brazil
| | - Mario J. A. Saad
- Department of Internal Medicine, State University of Campinas, Campinas, Brazil
- Mario J. A. Saad
| |
Collapse
|
22
|
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: 13] [Impact Index Per Article: 1.9] [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
|
23
|
Shrivastava P, Naoghare PK, Gandhi D, Devi SS, Krishnamurthi K, Bafana A, Kashyap SM, Chakrabarti T. Application of cell-based assays for toxicity characterization of complex wastewater matrices: Possible applications in wastewater recycle and reuse. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2017; 142:555-566. [PMID: 28482324 DOI: 10.1016/j.ecoenv.2017.04.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 06/07/2023]
Abstract
Exposure to pre-concentrated inlet or outlet STP wastewater extracts at different concentrations (0.001% to 1%) induced dose-dependent toxicity in MCF-7 cells, whereas drinking water extracts did not induce cytotoxicity in cells treated. GC-MS analysis revealed the occurrence of xenobiotic compounds (Benzene, Phthalate, etc.) in inlet/outlet wastewater extracts. Cells exposed to inlet/outlet extract showed elevated levels of reactive oxygen species (ROS: inlet: 186.58%, p<0.05, outlet, 147.8%, p<0.01) and loss of mitochondrial membrane potential (Δψm: inlet, 74.91%, p<0.01; outlet, 86.70%, p<0.05) compared to the control. These concentrations induced DNA damage (Tail length: inlet: 34.4%, p<0.05, outlet, 26.7%, p<0.05) in treated cells compared to the control (Tail length: 7.5%). Cell cycle analysis displayed drastic reduction in the G1 phase in treated cells (inlet, G1:45.0%; outlet, G1:58.3%) compared to the control (G1:67.3%). Treated cells showed 45.18% and 28.0% apoptosis compared to the control (1.2%). Drinking water extracts did not show any significant alterations with respect to ROS, Δψm, DNA damage, cell cycle and apoptosis compared to the control. Genes involved in cell cycle and apoptosis were found to be differentially expressed in cells exposed to inlet/outlet extracts. Herein, we propose cell-based toxicity assays to evaluate the efficacies of wastewater treatment and recycling processes.
Collapse
Affiliation(s)
- Preeti Shrivastava
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Pravin K Naoghare
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India.
| | - Deepa Gandhi
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - S Saravana Devi
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Kannan Krishnamurthi
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Amit Bafana
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Sanjay M Kashyap
- Analytical Instrumentation Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | | |
Collapse
|
24
|
Omoto M, Suri K, Amouzegar A, Li M, Katikireddy KR, Mittal SK, Chauhan SK. Hepatocyte Growth Factor Suppresses Inflammation and Promotes Epithelium Repair in Corneal Injury. Mol Ther 2017; 25:1881-1888. [PMID: 28502469 DOI: 10.1016/j.ymthe.2017.04.020] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/11/2017] [Accepted: 04/17/2017] [Indexed: 10/19/2022] Open
Abstract
Corneal injuries are among the major causes of ocular morbidity and vision impairment. Optimal epithelial wound healing is critical for the integrity and transparency of the cornea after injury. Hepatocyte growth factor (HGF) is a mitogen and motility factor that primarily regulates epithelial cell function. Herein, we investigate the effect of HGF on proliferation of corneal epithelial cells (CECs) in inflamed conditions both in vitro and in vivo. We demonstrate that HGF not only promotes CEC proliferation in homeostatic conditions but also reverses the anti-proliferative effect of the inflammatory environment on these cells. Furthermore, using a mouse model of ocular injury, we show that HGF treatment suppresses ocular inflammation and actively augments CEC proliferation, leading to improved and accelerated corneal epithelial repair. These findings have potential translational implications and could provide a framework for the development of novel HGF-based therapies for corneal epithelial defects.
Collapse
Affiliation(s)
- Masahiro Omoto
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kunal Suri
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Afsaneh Amouzegar
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Mingshun Li
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Kishore R Katikireddy
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Sharad K Mittal
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Fabersani E, Abeijon-Mukdsi MC, Ross R, Medina R, González S, Gauffin-Cano P. Specific Strains of Lactic Acid Bacteria Differentially Modulate the Profile of Adipokines In Vitro. Front Immunol 2017; 8:266. [PMID: 28348560 PMCID: PMC5346559 DOI: 10.3389/fimmu.2017.00266] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/23/2017] [Indexed: 12/12/2022] Open
Abstract
Obesity induces local/systemic inflammation accompanied by increases in macrophage infiltration into adipose tissue and production of inflammatory cytokines, chemokines, and hormones. Previous studies have shown that probiotics could improve the intestinal dysbiosis induced by metabolic diseases such as obesity, diabetes, and metabolic syndrome. Microorganisms could (directly or indirectly) affect adipokine levels due to their capacity to induce translocation of several intestinal microbial antigens into systemic circulation, which could lead to metabolic endotoxemia or produce immunomodulation in different organs. The aim of the present study was to select non-inflammatory lactic acid bacteria (LAB) strains with the capacity to modulate adipokine secretion by the adipose tissue. We wish to elucidate the role of potential probiotic strains in the regulation of the cross talking between immune cells such as macrophages and adipose cells. Mouse macrophage cell line RAW 264.7 was used for evaluating the ability of 14 LAB strains to induce cytokine production. The LAB strains were chosen based on their previously studied beneficial properties in health. Then, in murine adipocyte culture and macrophage–adipocyte coculture, we determined the ability of these strains to induce cytokines and leptin secretion. Tumor necrosis factor alpha, interleukin 6 (IL-6), IL-10, monocyte chemoattractant protein-1, and leptin levels were measured in cell supernatants. We also performed the detection and quantification of leptin receptor (Ob-Rb) expression in macrophage cell lines stimulated by these LAB strains. Differential secretion profile of cytokines in macrophage cells induced by LAB strains was observed. Also, the levels of Ob-Rb expression diverged among different LAB strains. In LAB-stimulated coculture cells (adipocytes and macrophages), we observed differential production of leptin and cytokines. Furthermore, we detected lower production levels in single culture than cocultured cells. The principal component analysis showed an association between the four clusters of strains established according to their inflammatory profiles and leptin adipocyte production and leptin receptor expression in macrophages. We conclude that coculture is the most appropriate system for selecting strains with the ability to modulate adipokine secretion. The use of microorganisms with low and medium inflammatory properties and ability to modulate leptin levels could be a strategy for the treatment of some metabolic diseases associated with dysregulation of immune response.
Collapse
Affiliation(s)
- Emanuel Fabersani
- Centro de Referencia para Lactobacilos (CERELA) - CONICET , Tucumán , Argentina
| | - María Claudia Abeijon-Mukdsi
- Centro de Referencia para Lactobacilos (CERELA) - CONICET, Tucumán, Argentina; Facultad Ciencias de la Salud, Universidad del Norte Santo Tomás de Aquino (UNSTA), Tucumán, Argentina
| | - Romina Ross
- Facultad Ciencias de la Salud, Universidad del Norte Santo Tomás de Aquino (UNSTA), Tucumán, Argentina; Universidad Nacional de Tucumán (UNT), Tucumán, Argentina
| | - Roxana Medina
- Centro de Referencia para Lactobacilos (CERELA) - CONICET, Tucumán, Argentina; Universidad Nacional de Tucumán (UNT), Tucumán, Argentina
| | | | - Paola Gauffin-Cano
- Centro de Referencia para Lactobacilos (CERELA) - CONICET, Tucumán, Argentina; Facultad Ciencias de la Salud, Universidad del Norte Santo Tomás de Aquino (UNSTA), Tucumán, Argentina
| |
Collapse
|
26
|
Muratsu J, Iwabayashi M, Sanada F, Taniyama Y, Otsu R, Rakugi H, Morishita R. Hepatocyte Growth Factor Prevented High-Fat Diet-Induced Obesity and Improved Insulin Resistance in Mice. Sci Rep 2017; 7:130. [PMID: 28273932 PMCID: PMC5427909 DOI: 10.1038/s41598-017-00199-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/14/2017] [Indexed: 01/01/2023] Open
Abstract
Obesity and its associated chronic inflammation in adipose tissue initiate insulin resistance, which is related to several pathologies including hypertension and atherosclerosis. Previous reports demonstrated that circulating hepatocyte growth factor (HGF) level was associated with obesity and type 2 diabetes. However, its precise role in obesity and related-pathology is unclear. In this experiment, cardiac-specific over-expression of human HGF in mice (HGF-Tg mice) which showed 4–5 times higher serum HGF levels than wild-type mice were used. While body weight in wild-type mice fed with high fat diet (HFD) for 14 weeks was significantly increased accompanied with insulin resistance, HGF-Tg mice prevented body weight gain and insulin resistance. The accumulation of macrophages and elevated levels of inflammatory mediators in adipose tissue were significantly inhibited in HGF-Tg mice as compared to wild-type mice. The HFD-induced obesity in wild-type mice treated with HGF-neutralizing antibody showed an exacerbated response to the glucose tolerance test. These gain-of-function and loss-of-function studies demonstrated that the elevated HGF level induced by HFD have protective role against obesity and insulin resistance.
Collapse
Affiliation(s)
- Jun Muratsu
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.,Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Masaaki Iwabayashi
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Fumihiro Sanada
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yoshiaki Taniyama
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan. .,Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - Rei Otsu
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Hiromi Rakugi
- Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
27
|
Nakano M, Tamura Y, Yamato M, Kume S, Eguchi A, Takata K, Watanabe Y, Kataoka Y. NG2 glial cells regulate neuroimmunological responses to maintain neuronal function and survival. Sci Rep 2017; 7:42041. [PMID: 28195192 PMCID: PMC5307324 DOI: 10.1038/srep42041] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 01/06/2017] [Indexed: 11/10/2022] Open
Abstract
NG2-expressing neural progenitor cells (i.e., NG2 glial cells) maintain their proliferative and migratory activities even in the adult mammalian central nervous system (CNS) and produce myelinating oligodendrocytes and astrocytes. Although NG2 glial cells have been observed in close proximity to neuronal cell bodies in order to receive synaptic inputs, substantive non-proliferative roles of NG2 glial cells in the adult CNS remain unclear. In the present study, we generated NG2-HSVtk transgenic rats and selectively ablated NG2 glial cells in the adult CNS. Ablation of NG2 glial cells produced defects in hippocampal neurons due to excessive neuroinflammation via activation of the interleukin-1 beta (IL-1β) pro-inflammatory pathway, resulting in hippocampal atrophy. Furthermore, we revealed that the loss of NG2 glial cell-derived hepatocyte growth factor (HGF) exacerbated these abnormalities. Our findings suggest that NG2 glial cells maintain neuronal function and survival via the control of neuroimmunological function.
Collapse
Affiliation(s)
- Masayuki Nakano
- Cellular Function Imaging Team, Center for Life Science Technologies, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan.,Department of Physiology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Yasuhisa Tamura
- Cellular Function Imaging Team, Center for Life Science Technologies, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan.,Multi-Modal Microstructure Analysis Unit, RIKEN CLST-JEOL Collaboration Center, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Masanori Yamato
- Cellular Function Imaging Team, Center for Life Science Technologies, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan.,Multi-Modal Microstructure Analysis Unit, RIKEN CLST-JEOL Collaboration Center, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Satoshi Kume
- Cellular Function Imaging Team, Center for Life Science Technologies, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan.,Multi-Modal Microstructure Analysis Unit, RIKEN CLST-JEOL Collaboration Center, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Asami Eguchi
- Cellular Function Imaging Team, Center for Life Science Technologies, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan.,Multi-Modal Microstructure Analysis Unit, RIKEN CLST-JEOL Collaboration Center, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Kumi Takata
- Cellular Function Imaging Team, Center for Life Science Technologies, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Yasuyoshi Watanabe
- Department of Physiology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan.,Pathophysiological and Health Science Team, Center for Life Science Technologies, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Yosky Kataoka
- Cellular Function Imaging Team, Center for Life Science Technologies, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan.,Department of Physiology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan.,Multi-Modal Microstructure Analysis Unit, RIKEN CLST-JEOL Collaboration Center, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| |
Collapse
|
28
|
Warnke I, Jocken JWE, Schoop R, Toepfer C, Goralczyk R, Schwager J. Combinations of bio-active dietary constituents affect human white adipocyte function in-vitro. Nutr Metab (Lond) 2016; 13:84. [PMID: 27895698 PMCID: PMC5117626 DOI: 10.1186/s12986-016-0143-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 11/09/2016] [Indexed: 01/14/2023] Open
Abstract
Background Specific bio-active dietary compounds modulate numerous metabolic processes in adipose tissue (AT), including pre-adipocyte proliferation and differentiation. AT dysfunction, rather than an increased fat mass per se, is strongly associated with the development of insulin resistance and is characterized by impaired adipogenesis, hypertrophic adipocytes, inflammation, and impairments in substrate metabolism. A better understanding of mechanisms underlying AT dysfunction may provide new strategies for the treatment of obesity-associated metabolic diseases. Here we evaluated the role of (all-E)-lycopene (Lyc), eicosapentaenoic acid (EPA) or trans-resveratrol (Res) and combinations thereof on human white adipocyte function. Methods In-vitro differentiating human pre-adipocytes were treated with EPA, Lyc and Res or their combinations for 14 days. The effects on intracellular lipid droplet (LD) accumulation, secreted anti- and pro-inflammatory cyto-/adipokines (e.g. adiponectin, IL-6, IL-8/CXCL-8 and MCP-1/CCL2) and on gene expression of markers of adipocyte differentiation and substrate metabolism (e.g. PPAR-gamma, C/EBP-alpha, GLUT-4, FAS, ATGL, HSL, and PLIN-1) were measured by fluorescent microscopy (Cellomics™), multi-parametric LiquiChip® technology and quantitative RT-PCR, respectively. Results Treatment of differentiating adipocytes for 14 days with the combination of Lyc/Res and EPA/Res resulted in significantly inhibited LD formation (~ -25 and -20%, respectively) compared to the effects of the single compounds. These morphological changes were accompanied by increased mRNA levels of the adipogenic marker PPAR-gamma and the lipase ATGL and by decreased expression levels of lipogenic markers (LPL, FAS, GLUT-4) and the LD-covering protein PLIN-1. In addition, a blunted adipocyte secretion of pro-inflammatory cytokines (IL-6 and MCP-1) and adiponectin was observed following treatment with these compounds. Conclusion The combination of the dietary bio-actives Lyc and EPA with Res might influence adipocyte function by affecting the balance between adipogenic, lipogenic and lipolytic gene expression, resulting in a reduced LD storage and a less inflammatory secretion profile. Taken together, our results indicate that combinations of dietary compounds may be beneficial for the prevention and treatment of metabolic disorders via effects on human white adipocyte function. Electronic supplementary material The online version of this article (doi:10.1186/s12986-016-0143-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ines Warnke
- DSM Nutritional Products Ltd., Department of Human Nutrition and Health, CH-4002 Basel, Switzerland
| | - Johan W E Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Rotraut Schoop
- DSM Nutritional Products Ltd., Department of Human Nutrition and Health, CH-4002 Basel, Switzerland
| | - Christine Toepfer
- DSM Nutritional Products Ltd., Department of Human Nutrition and Health, CH-4002 Basel, Switzerland
| | - Regina Goralczyk
- DSM Nutritional Products Ltd., Department of Human Nutrition and Health, CH-4002 Basel, Switzerland
| | - Joseph Schwager
- DSM Nutritional Products Ltd., Department of Human Nutrition and Health, CH-4002 Basel, Switzerland
| |
Collapse
|
29
|
Ates I, Ozkayar N, Ates H, Karakulak UN, Kursun O, Topcuoglu C, Inan B, Yilmaz N. Elevated circulating sST2 associated with subclinical atherosclerosis in newly diagnosed primary hypertension. Hypertens Res 2016; 39:513-518. [DOI: 10.1038/hr.2016.16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 12/22/2015] [Accepted: 12/28/2015] [Indexed: 12/15/2022]
|
30
|
Ilangumaran S, Villalobos-Hernandez A, Bobbala D, Ramanathan S. The hepatocyte growth factor (HGF)–MET receptor tyrosine kinase signaling pathway: Diverse roles in modulating immune cell functions. Cytokine 2016; 82:125-39. [PMID: 26822708 DOI: 10.1016/j.cyto.2015.12.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 12/11/2015] [Accepted: 12/12/2015] [Indexed: 12/14/2022]
|
31
|
Biwer LA, D'souza KM, Abidali A, Tu D, Siniard AL, DeBoth M, Huentelman M, Hale TM. Time course of cardiac inflammation during nitric oxide synthase inhibition in SHR: impact of prior transient ACE inhibition. Hypertens Res 2015; 39:8-18. [PMID: 26490086 DOI: 10.1038/hr.2015.107] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 07/13/2015] [Accepted: 08/25/2015] [Indexed: 11/09/2022]
Abstract
We have previously demonstrated that angiotensin-converting enzyme (ACE) inhibition with enalapril produces persistent effects that protect against future nitric oxide synthase (NOS) inhibitor (L-arginine methyl ester, L-NAME)-induced cardiac dysfunction and outer wall collagen deposition in spontaneously hypertensive rats (SHR). In the present study, we dissect the cytokine/chemokine release profile during NOS inhibition, its correlation to pathological cardiac remodeling and the impact of transient ACE inhibition on these effects. Adult male SHR were treated with enalapril (E+L) or tap water (C+L) for 2 weeks followed by a 2-week washout period. Rats were then subjected to 0, 3, 7 or 10 days of L-NAME treatment. The temporal response to NOS inhibition was evaluated by measuring arterial pressure, cardiac remodeling and cytokine/chemokine levels. L-NAME equivalently increased blood pressure and myocardial and vascular injury in C+L and E+L rats. However, pulse pressure (PP) was only transiently altered in C+L rats. The levels of several inflammatory mediators were increased during L-NAME treatment. However, interleukin-6 (IL-6) and IL-10 and monocyte chemoattractant protein-1 were uniquely increased in C+L hearts; whereas IL-4 and fractalkine were only elevated in E+L hearts. By days 7 and 10 of L-NAME treatment, there was a significant increase in the cardiac density of macrophages and proliferating cells, respectively only in C+L rats. Although myocardial injury was similar in both treatment groups, PP was not changed and there was a distinct cardiac chemokine/cytokine signature in rats previously treated with enalapril that may be related to the lack of proliferative response and macrophage infiltration in these hearts.
Collapse
Affiliation(s)
- Lauren A Biwer
- Department of Basic Medical Sciences, University of Arizona, College of Medicine-Phoenix, Phoenix AZ, USA
| | - Karen M D'souza
- Department of Basic Medical Sciences, University of Arizona, College of Medicine-Phoenix, Phoenix AZ, USA
| | - Ali Abidali
- Department of Basic Medical Sciences, University of Arizona, College of Medicine-Phoenix, Phoenix AZ, USA
| | - Danni Tu
- Department of Basic Medical Sciences, University of Arizona, College of Medicine-Phoenix, Phoenix AZ, USA
| | - Ashley L Siniard
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Matthew DeBoth
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Matthew Huentelman
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Taben M Hale
- Department of Basic Medical Sciences, University of Arizona, College of Medicine-Phoenix, Phoenix AZ, USA
| |
Collapse
|
32
|
Rodríguez A, Ezquerro S, Méndez-Giménez L, Becerril S, Frühbeck G. Revisiting the adipocyte: a model for integration of cytokine signaling in the regulation of energy metabolism. Am J Physiol Endocrinol Metab 2015; 309:E691-714. [PMID: 26330344 DOI: 10.1152/ajpendo.00297.2015] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/24/2015] [Indexed: 02/08/2023]
Abstract
Adipose tissue constitutes an extremely active endocrine organ with a network of signaling pathways enabling the organism to adapt to a wide range of different metabolic challenges, such as starvation, stress, infection, and short periods of gross energy excess. The functional pleiotropism of adipose tissue relies on its ability to synthesize and release a huge variety of hormones, cytokines, complement and growth factors, extracellular matrix proteins, and vasoactive factors, collectively termed adipokines. Obesity is associated with adipose tissue dysfunction leading to the onset of several pathologies including type 2 diabetes, dyslipidemia, nonalcoholic fatty liver, or hypertension, among others. The mechanisms underlying the development of obesity and its associated comorbidities include the hypertrophy and/or hyperplasia of adipocytes, adipose tissue inflammation, impaired extracellular matrix remodeling, and fibrosis together with an altered secretion of adipokines. Recently, the potential role of brown and beige adipose tissue in the protection against obesity has been also recognized. In contrast to white adipocytes, which store energy in the form of fat, brown and beige fat cells display energy-dissipating capacity through the promotion of triacylglycerol clearance, glucose disposal, and generation of heat for thermogenesis. Identification of the morphological and molecular changes in white, beige, and brown adipose tissue during weight gain is of utmost relevance for the identification of pharmacological targets for the treatment of obesity and its associated metabolic diseases.
Collapse
Affiliation(s)
- Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; and Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Silvia Ezquerro
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Leire Méndez-Giménez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; and Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; and Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; and Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| |
Collapse
|
33
|
Deng W, Chen W, Zhang Z, Huang S, Kong W, Sun Y, Tang X, Yao G, Feng X, Chen W, Sun L. Mesenchymal stem cells promote CD206 expression and phagocytic activity of macrophages through IL-6 in systemic lupus erythematosus. Clin Immunol 2015. [PMID: 26209923 DOI: 10.1016/j.clim.2015.07.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Human umbilical cord-derived mesenchymal stem cells (UCMSCs) show therapeutic effects on systemic lupus erythematosus (SLE). Deficiency in functional polarization and phagocytosis in macrophages has been suggested in the pathogenesis of SLE. We found that macrophages from B6.MRL-Fas(lpr) mice exhibited lower level of CD206, the marker for alternatively activated macrophage (AAM, also called M2). In addition, the phagocytic activity of B6.MRL-Fas(lpr) macrophages was also decreased. UCMSC transplantation improved the proportion of CD206(+) macrophages and their phagocytic activity in B6.MRL-Fas(lpr) mice. Importantly, macrophages from SLE patients also showed lower expression of CD206 and reduced phagocytic activity, which were corrected by being co-cultured with UCMSCs in vitro and in SLE patients receiving UCMSC transplantation. Mechanistically, we demonstrated that IL-6 was required for the up-regulation of CD206 expression and phagocytic activity of UCMSC-treated SLE macrophages. Our results indicate that UCMSCs alleviate SLE through promoting CD206 expression and phagocytic activity of macrophages in an IL-6 dependent manner.
Collapse
Affiliation(s)
- Wei Deng
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Weiwei Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Zhuoya Zhang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Saisai Huang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Wei Kong
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Yue Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Genhong Yao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Xuebing Feng
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - WanJun Chen
- Mucosal Immunology Section, OPCB, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-2190, USA
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China.
| |
Collapse
|
34
|
Hepatocyte growth factor: A regulator of inflammation and autoimmunity. Autoimmun Rev 2014; 14:293-303. [PMID: 25476732 DOI: 10.1016/j.autrev.2014.11.013] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 11/25/2014] [Indexed: 12/12/2022]
Abstract
Hepatocyte growth factor (HGF) is a pleiotropic cytokine that has been extensively studied over several decades, but was only recently recognized as a key player in mediating protection of many types of inflammatory and autoimmune diseases. HGF was reported to prevent and attenuate disease progression by influencing multiple pathophysiological processes involved in inflammatory and immune response, including cell migration, maturation, cytokine production, antigen presentation, and T cell effector function. In this review, we discuss the actions and mechanisms of HGF in inflammation and immunity and the therapeutic potential of this factor for the treatment of inflammatory and autoimmune diseases.
Collapse
|
35
|
Tsai CC, Huang SC, Tai MH, Chien CCC, Huang CC, Hsu YC. Hepatoma-derived growth factor upregulation is correlated with prognostic factors of early-stage cervical adenocarcinoma. Int J Mol Sci 2014; 15:21492-504. [PMID: 25421244 PMCID: PMC4264238 DOI: 10.3390/ijms151121492] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/13/2014] [Accepted: 10/28/2014] [Indexed: 01/29/2023] Open
Abstract
Hepatoma-derived growth factor (HDGF) is a unique nuclear/growth factor that plays an important role in the progression of different types of cancer. A total of 63 patients with early-stage cervical adenocarcinoma (Cx) were enrolled in this retrospective study. The expression of HDGF was significantly increased compared with adjacent non-tumor tissue samples (p < 0.001). Moreover, elevated nuclear HDGF levels were correlated with lymph-vascular space invasion (LVSI; p < 0.05), lymph node metastasis (LNM; p < 0.001), recurrence (p < 0.001) and advanced grade (AG; p < 0.001). The growth of cervical cancer cells (Hela cells) was enhanced by HDGF treatment. The HDGF mRNA and protein level were significantly higher in malignant cervical cancer cells compared with primary ones. By adenovirus gene delivery, HDGF overexpression enhanced, whereas HDGF knockdown perturbed the tumorigenic behaviors of cervical cancer cells. HDGF overexpression is common in early-stage cervical adenocarcinoma and is involved in the carcinogenesis of cervical adenocarcinoma. Cytoplasmic HDGF expression is strongly correlated with pelvic lymph node metastasis and recurrence, indicating that HDGF may serve as a novel prognostic marker for patients with Cx.
Collapse
Affiliation(s)
- Ching-Chou Tsai
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Kaohsiung and College of Medicine, Chang Gung University, Kaohsiung 83301, Taiwan.
| | - Shun-Chen Huang
- Department of Pathology, Chang Gung Memorial Hospital, Kaohsiung and College of Medicine, Chang Gung University, Kaohsiung 83301, Taiwan.
| | - Ming Hong Tai
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan.
| | - Chan-Chao Chang Chien
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Kaohsiung and College of Medicine, Chang Gung University, Kaohsiung 83301, Taiwan.
| | - Chao-Cheng Huang
- Department of Pathology, Chang Gung Memorial Hospital, Kaohsiung and College of Medicine, Chang Gung University, Kaohsiung 83301, Taiwan.
| | - Yi-Chiang Hsu
- Graduate Institute of Medical Science, College of Health Sciences, Chang Jung Christian University, Tainan 71101, Taiwan.
| |
Collapse
|
36
|
Li J, Chanda D, Shiri-Sverdlov R, Neumann D. MSP: an emerging player in metabolic syndrome. Cytokine Growth Factor Rev 2014; 26:75-82. [PMID: 25466635 DOI: 10.1016/j.cytogfr.2014.10.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 10/22/2014] [Indexed: 12/15/2022]
Abstract
MSP (Macrophage Stimulating Protein; also known as Hepatocyte Growth Factor-like protein (HGFL) and MST1) is a secreted protein and the ligand for transmembrane receptor tyrosine kinase Recepteur d'Origine Nantais (RON; also known as MST1R). Since its discovery, MSP has been demonstrated to play a key role in regulating inflammation in the peripheral tissues of multiple disease models. Recent evidences also point toward a beneficial role of MSP in the regulation of hepatic lipid and glucose metabolism, thereby implicating MSP as a crucial regulator in maintaining metabolic homeostasis while simultaneously suppressing inflammatory processes. In this review, we discuss the recent advances that demonstrate the significance of MSP in metabolic syndrome and build a strong case supporting its therapeutic potential.
Collapse
Affiliation(s)
- Jieyi Li
- Department of Molecular Genetics, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Dipanjan Chanda
- Department of Molecular Genetics, CARIM School for Cardiovascular Diseases, Maastricht University, 6200 MD Maastricht, The Netherlands.
| | - Ronit Shiri-Sverdlov
- Department of Molecular Genetics, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Dietbert Neumann
- Department of Molecular Genetics, CARIM School for Cardiovascular Diseases, Maastricht University, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
37
|
The Hepatocyte Growth Factor (HGF)/Met Axis: A Neglected Target in the Treatment of Chronic Myeloproliferative Neoplasms? Cancers (Basel) 2014; 6:1631-69. [PMID: 25119536 PMCID: PMC4190560 DOI: 10.3390/cancers6031631] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/04/2014] [Accepted: 08/04/2014] [Indexed: 12/17/2022] Open
Abstract
Met is the receptor of hepatocyte growth factor (HGF), a cytoprotective cytokine. Disturbing the equilibrium between Met and its ligand may lead to inappropriate cell survival, accumulation of genetic abnormalities and eventually, malignancy. Abnormal activation of the HGF/Met axis is established in solid tumours and in chronic haematological malignancies, including myeloma, acute myeloid leukaemia, chronic myelogenous leukaemia (CML), and myeloproliferative neoplasms (MPNs). The molecular mechanisms potentially responsible for the abnormal activation of HGF/Met pathways are described and discussed. Importantly, inCML and in MPNs, the production of HGF is independent of Bcr-Abl and JAK2V617F, the main molecular markers of these diseases. In vitro studies showed that blocking HGF/Met function with neutralizing antibodies or Met inhibitors significantly impairs the growth of JAK2V617F-mutated cells. With personalised medicine and curative treatment in view, blocking activation of HGF/Met could be a useful addition in the treatment of CML and MPNs for those patients with high HGF/MET expression not controlled by current treatments (Bcr-Abl inhibitors in CML; phlebotomy, hydroxurea, JAK inhibitors in MPNs).
Collapse
|