1
|
Lee SJ, Koh A, Lee SH, Kim KW. Efficacy of epidermal growth factor in suppressing inflammation and proliferation in pterygial fibroblasts through interactions with microenvironmental M1 macrophages. Sci Rep 2024; 14:22601. [PMID: 39349715 PMCID: PMC11442942 DOI: 10.1038/s41598-024-74413-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
The protein epidermal growth factor (EGF), which plays a crucial role in promoting cell proliferation and survival, has recently demonstrated potential in reducing inflammation. In this study, we examined the impact of EGF on the anti-inflammatory and anti-proliferative properties of pterygium, a prevalent hypervascular proliferative disease affecting the ocular surface. In surgically excised tissues, markers for fibrotic and inflammatory signals, including VIM, ACTA2, FAP, MMP2, VCAM1, ICAM1, CD86, IL6, and IL1B were upregulated in the pterygium body stroma compared to the normal conjunctival stroma. EGF exerted anti-inflammatory and anti-vasculogenic effects on pterygial fibroblasts when co-cultured with M1 macrophages. Moreover, exosomes derived from EGF-preconditioned M1 macrophages suppressed the heightened inflammatory and vasculogenic signals in pterygial fibroblasts induced by exosomes from M1 macrophages. Paradoxically, the proliferation of pterygial fibroblasts was inhibited by EGF in the in vitro microenvironment with M1 macrophages, despite EGF being known as a growth factor. EGF-preconditioning of M1 macrophages rescued the increased proliferation of pterygial fibroblasts induced by exosomes from M1 macrophages. In conclusion, our findings demonstrate that EGF effectively mitigates inflammation and proliferation in pterygial fibroblasts within a microenvironment containing M1 macrophages.
Collapse
Affiliation(s)
- Soo Jin Lee
- Chung-Ang Ocular Surface Restoration via Immune-inflammation Alleviation (CORIA) Laboratory, Seoul, Republic of Korea
| | - Ahra Koh
- Chung-Ang Ocular Surface Restoration via Immune-inflammation Alleviation (CORIA) Laboratory, Seoul, Republic of Korea
- Chung-Ang University Graduate School, Seoul, Republic of Korea
| | - Seung Hyeun Lee
- Chung-Ang Ocular Surface Restoration via Immune-inflammation Alleviation (CORIA) Laboratory, Seoul, Republic of Korea
- Department of Ophthalmology, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong-si, Gyeonggi-do, Republic of Korea
| | - Kyoung Woo Kim
- Chung-Ang Ocular Surface Restoration via Immune-inflammation Alleviation (CORIA) Laboratory, Seoul, Republic of Korea.
- Chung-Ang University Graduate School, Seoul, Republic of Korea.
- Department of Ophthalmology, Chung-Ang University College of Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Dongjak-gu, Seoul, 06973, Republic of Korea.
| |
Collapse
|
2
|
Zhao R, Wang Z, Wang G, Geng J, Wu H, Liu X, Bin E, Sui J, Dai H, Tang N. Sustained amphiregulin expression in intermediate alveolar stem cells drives progressive fibrosis. Cell Stem Cell 2024; 31:1344-1358.e6. [PMID: 39096904 DOI: 10.1016/j.stem.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/05/2024] [Accepted: 07/09/2024] [Indexed: 08/05/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal fibrotic disease. Recent studies have highlighted the persistence of an intermediate state of alveolar stem cells in IPF lungs. In this study, we discovered a close correlation between the distribution pattern of intermediate alveolar stem cells and the progression of fibrotic changes. We showed that amphiregulin (AREG) expression is significantly elevated in intermediate alveolar stem cells of mouse fibrotic lungs and IPF patients. High levels of serum AREG correlate significantly with profound deteriorations in lung function in IPF patients. We demonstrated that AREG in alveolar stem cells is both required and sufficient for activating EGFR in fibroblasts, thereby driving lung fibrosis. Moreover, pharmacological inhibition of AREG using a neutralizing antibody effectively blocked the initiation and progression of lung fibrosis in mice. Our study underscores the therapeutic potential of anti-AREG antibodies in attenuating IPF progression, offering a promising strategy for treating fibrotic diseases.
Collapse
Affiliation(s)
- Rui Zhao
- Pulmongene (Beijing) Ltd., Beijing 102206, China.
| | - Zheng Wang
- National Institute of Biological Sciences, Beijing 102206, China
| | - Guowu Wang
- Pulmongene (Beijing) Ltd., Beijing 102206, China
| | - Jing Geng
- Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Huijuan Wu
- National Institute of Biological Sciences, Beijing 102206, China
| | - Ximing Liu
- National Institute of Biological Sciences, Beijing 102206, China
| | - Ennan Bin
- National Institute of Biological Sciences, Beijing 102206, China
| | - Jianhua Sui
- National Institute of Biological Sciences, Beijing 102206, China
| | - Huaping Dai
- Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Nan Tang
- National Institute of Biological Sciences, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100190, China.
| |
Collapse
|
3
|
Maddali MV, Moore AR, Sinha P, Newton CA, Kim JS, Adegunsoye A, Ma SF, Strek ME, Chen CH, Linderholm AL, Zemans RL, Moore BB, Wolters PJ, Martinez FJ, Rogers AJ, Raj R, Noth I, Oldham JM. Molecular Endotypes of Idiopathic Pulmonary Fibrosis: A Latent Class Analysis of Two Multicenter Observational Cohorts. Am J Respir Crit Care Med 2024; 210:455-464. [PMID: 38913573 PMCID: PMC11351813 DOI: 10.1164/rccm.202402-0339oc] [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: 02/12/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024] Open
Abstract
Rationale: Idiopathic pulmonary fibrosis (IPF) causes irreversible fibrosis of the lung parenchyma. Although antifibrotic therapy can slow IPF progression, treatment response is variable. There exists a critical need to develop a precision medicine approach to IPF. Objectives: To identify and validate biologically driven molecular endotypes of IPF. Methods: Latent class analysis (LCA) was independently performed in prospectively recruited discovery (n = 875) and validation (n = 347) cohorts. Twenty-five plasma biomarkers associated with fibrogenesis served as class-defining variables. The association between molecular endotype and 4-year transplant-free survival was tested using multivariable Cox regression adjusted for baseline confounders. Endotype-dependent differential treatment response to future antifibrotic exposure was then assessed in a pooled cohort of patients naive to antifibrotic therapy at the time of biomarker measurement (n = 555). Measurements and Main Results: LCA independently identified two latent classes in both cohorts (P < 0.0001). WFDC2 (WAP four-disulfide core domain protein 2) was the most important determinant of class membership across cohorts. Membership in class 2 was characterized by higher biomarker concentrations and a higher risk of death or transplant (discovery, hazard ratio [HR], 2.02; 95% confidence interval [CI], 1.64-2.48; P < 0.001; validation, HR, 1.95; 95% CI, 1.34-2.82; P < 0.001). In pooled analysis, significant heterogeneity in treatment effect was observed between endotypes (P = 0.030 for interaction), with a favorable antifibrotic response in class 2 (HR, 0.64; 95% CI, 0.45-0.93; P = 0.018) but not in class 1 (HR, 1.19; 95% CI, 0.77-1.84; P = 0.422). Conclusions: In this multicohort study, we identified two novel molecular endotypes of IPF with divergent clinical outcomes and responses to antifibrotic therapy. Pending further validation, these endotypes could enable a precision medicine approach for future IPF clinical trials.
Collapse
Affiliation(s)
- Manoj V. Maddali
- Division of Pulmonary, Allergy, and Critical Care Medicine and
- Department of Biomedical Data Science, Stanford University, Stanford, California
| | - Andrew R. Moore
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | - Pratik Sinha
- Division of Clinical and Translational Research, Washington University School of Medicine, St. Louis, Missouri
- Division of Critical Care, Department of Anesthesia, Washington University, St. Louis, Missouri
| | - Chad A. Newton
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - John S. Kim
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Ayodeji Adegunsoye
- Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois
| | - Shwu-Fan Ma
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Mary E. Strek
- Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois
| | - Ching-Hsien Chen
- Division of Pulmonary and Critical Care Medicine, University of California, Davis, Davis, California
| | - Angela L. Linderholm
- Division of Pulmonary and Critical Care Medicine, University of California, Davis, Davis, California
| | | | - Bethany B. Moore
- Division of Pulmonary and Critical Care Medicine
- Department of Microbiology and Immunology, and
| | - Paul J. Wolters
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, University of California, San Francisco, San Francisco, California; and
| | - Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Cornell University, New York, New York
| | | | - Rishi Raj
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Justin M. Oldham
- Division of Pulmonary and Critical Care Medicine
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
4
|
Kaneko T, Iwamura C, Kiuchi M, Kurosugi A, Onoue M, Matsumura T, Chiba T, Nakayama T, Kato N, Hirahara K. Amphiregulin-producing T H2 cells facilitate esophageal fibrosis of eosinophilic esophagitis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100287. [PMID: 39040657 PMCID: PMC11260569 DOI: 10.1016/j.jacig.2024.100287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/26/2024] [Accepted: 03/05/2024] [Indexed: 07/24/2024]
Abstract
Background Massive eosinophil infiltration into the esophagus is associated with subepithelial fibrosis and esophageal stricture in patients with eosinophilic esophagitis (EoE). However, the pathogenesis of esophageal fibrosis remains unclear. Objective We sought to elucidate the cellular and molecular mechanisms underlying the induction of esophageal fibrosis. Methods We established a murine model of EoE accompanied by fibrotic responses following long-term intranasal administration of house dust mite antigen. Using this murine model, we investigated the characteristics of immune cells infiltrating the fibrotic region of the inflamed esophagus using flow cytometry and histological analyses. We also analyzed the local inflammatory sites in the esophagus of patients with EoE using single-cell RNA sequencing, flow cytometry, and immunohistochemistry. Results Enhanced infiltration of both amphiregulin-producing and IL-5-producing TH2 cells was detected in the fibrotic area of the esophagus in mice subjected to repeated house dust mite exposure. Deletion of amphiregulin in CD4+ T cells ameliorates esophageal fibrosis. An analysis of human esophageal biopsy samples showed that the infiltration of amphiregulin-producing CD4+ T cells was higher in patients with EoE than in control patients. Furthermore, the number of infiltrated amphiregulin-producing CD4+ T cells was associated with the degree of esophageal fibrosis in patients with EoE. Conclusions Amphiregulin, produced by TH2 cells, contributes to esophageal fibrosis in EoE and may be a therapeutic target.
Collapse
Affiliation(s)
- Tatsuya Kaneko
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Chiaki Iwamura
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
| | - Masahiro Kiuchi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akane Kurosugi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Miki Onoue
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomoaki Matsumura
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tetsuhiro Chiba
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- AMED-CREST, AMED, 1-8-1 Inohana, Chuo-ku, Chiba, Japan
| | - Naoya Kato
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
- AMED-CREST, AMED, 1-8-1 Inohana, Chuo-ku, Chiba, Japan
| |
Collapse
|
5
|
Libra A, Sciacca E, Muscato G, Sambataro G, Spicuzza L, Vancheri C. Highlights on Future Treatments of IPF: Clues and Pitfalls. Int J Mol Sci 2024; 25:8392. [PMID: 39125962 PMCID: PMC11313529 DOI: 10.3390/ijms25158392] [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: 06/09/2024] [Revised: 07/22/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterized by irreversible scarring of lung tissue, leading to death. Despite recent advancements in understanding its pathophysiology, IPF remains elusive, and therapeutic options are limited and non-curative. This review aims to synthesize the latest research developments, focusing on the molecular mechanisms driving the disease and on the related emerging treatments. Unfortunately, several phase 2 studies showing promising preliminary results did not meet the primary endpoints in the subsequent phase 3, underlying the complexity of the disease and the need for new integrated endpoints. IPF remains a challenging condition with a complex interplay of genetic, epigenetic, and pathophysiological factors. Ongoing research into the molecular keystones of IPF is critical for the development of targeted therapies that could potentially stop the progression of the disease. Future directions include personalized medicine approaches, artificial intelligence integration, growth in genetic insights, and novel drug targets.
Collapse
Affiliation(s)
- Alessandro Libra
- Department of Clinical and Experimental Medicine, Regional Referral Center for Rare Lung Disease, Policlinico “G. Rodolico-San Marco”, University of Catania, 95123 Catania, CT, Italy; (A.L.); (E.S.); (G.M.); (L.S.)
| | - Enrico Sciacca
- Department of Clinical and Experimental Medicine, Regional Referral Center for Rare Lung Disease, Policlinico “G. Rodolico-San Marco”, University of Catania, 95123 Catania, CT, Italy; (A.L.); (E.S.); (G.M.); (L.S.)
| | - Giuseppe Muscato
- Department of Clinical and Experimental Medicine, Regional Referral Center for Rare Lung Disease, Policlinico “G. Rodolico-San Marco”, University of Catania, 95123 Catania, CT, Italy; (A.L.); (E.S.); (G.M.); (L.S.)
| | - Gianluca Sambataro
- Artroreuma s.r.l., Rheumatology Outpatient Clinic, 95030 Mascalucia, CT, Italy;
| | - Lucia Spicuzza
- Department of Clinical and Experimental Medicine, Regional Referral Center for Rare Lung Disease, Policlinico “G. Rodolico-San Marco”, University of Catania, 95123 Catania, CT, Italy; (A.L.); (E.S.); (G.M.); (L.S.)
| | - Carlo Vancheri
- Department of Clinical and Experimental Medicine, Regional Referral Center for Rare Lung Disease, Policlinico “G. Rodolico-San Marco”, University of Catania, 95123 Catania, CT, Italy; (A.L.); (E.S.); (G.M.); (L.S.)
| |
Collapse
|
6
|
Pinnarò V, Kirchberger S, Künig S, Gil Cantero S, Ciardulli MC, Della Porta G, Blüml S, Elbe-Bürger A, Bochkov V, Stöckl J. Oxidized Phospholipids Regulate Tenocyte Function via Induction of Amphiregulin in Dendritic Cells. Int J Mol Sci 2024; 25:7600. [PMID: 39062855 PMCID: PMC11277520 DOI: 10.3390/ijms25147600] [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: 06/13/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Inflammation is a driving force of tendinopathy. The oxidation of phospholipids by free radicals is a consequence of inflammatory reactions and is an important indicator of tissue damage. Here, we have studied the impact of oxidized phospholipids (OxPAPC) on the function of human tenocytes. We observed that treatment with OxPAPC did not alter the morphology, growth and capacity to produce collagen in healthy or diseased tenocytes. However, since OxPAPC is a known modulator of the function of immune cells, we analyzed whether OxPAPC-treated immune cells might influence the fate of tenocytes. Co-culture of tenocytes with immature, monocyte-derived dendritic cells treated with OxPAPC (Ox-DCs) was found to enhance the proliferation of tenocytes, particularly those from diseased tendons. Using transcriptional profiling of Ox-DCs, we identified amphiregulin (AREG), a ligand for EGFR, as a possible mediator of this proliferation enhancing effect, which we could confirm using recombinant AREG. Of note, diseased tenocytes were found to express higher levels of EGFR compared to tenocytes isolated from healthy donors and show a stronger proliferative response upon co-culture with Ox-DCs, as well as AREG treatment. In summary, we identify an AREG-EGFR axis as a mediator of a DC-tenocyte crosstalk, leading to increased tenocyte proliferation and possibly tendon regeneration.
Collapse
Affiliation(s)
- Veronica Pinnarò
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria; (V.P.); (S.K.); (S.G.C.)
| | | | - Sarojinidevi Künig
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria; (V.P.); (S.K.); (S.G.C.)
| | - Sara Gil Cantero
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria; (V.P.); (S.K.); (S.G.C.)
| | - Maria Camilla Ciardulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy; (M.C.C.); (G.D.P.)
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy; (M.C.C.); (G.D.P.)
| | - Stephan Blüml
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria;
| | | | - Valery Bochkov
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, 8010 Graz, Austria;
| | - Johannes Stöckl
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria; (V.P.); (S.K.); (S.G.C.)
| |
Collapse
|
7
|
Osakabe Y, Taniguchi Y, Hamada Ode K, Shimamura Y, Inotani S, Nishikawa H, Matsumoto T, Horino T, Fujimoto S, Terada Y. Clinical significance of amphiregulin in patients with chronic kidney disease. Clin Exp Nephrol 2024; 28:421-430. [PMID: 38402497 DOI: 10.1007/s10157-023-02445-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 12/02/2023] [Indexed: 02/26/2024]
Abstract
BACKGROUND Amphiregulin (AREG) is a ligand of epidermal growth factor receptor (EGFR), which plays an important role in injury-induced kidney fibrosis. However, the clinical significance of serum soluble AREG in chronic kidney disease (CKD) is unclear. In this study, we elucidated the clinical significance of serum soluble AREG in CKD by analyzing the association of serum soluble AREG levels with renal function and other clinical parameters in patients with CKD. METHODS In total, 418 Japanese patients with CKD were enrolled, and serum samples were collected for the determination of soluble AREG and creatinine (Cr) levels, and other clinical parameters. Additionally, these parameters were evaluated after 2 and 3 years. Moreover, immunohistochemical assay was performed ate AREG expression in the kidney tissues of patients with CKD. RESULTS Soluble AREG levels were positively correlated with serum Cr (p < 0.0001). Notably, initial AREG levels were positively correlated with changes in renal function (ΔCr) after 2 (p < 0.0001) and 3 years (P = 0.048). Additionally, soluble AREG levels were significantly higher (p < 0.05) in patients with diabetic nephropathy or primary hypertension. Moreover, AREG was highly expressed in renal tubular cells in patients with advanced CKD, but only weakly expressed in patients with preserved renal function. CONCLUSION Serum soluble AREG levels were significantly correlated with renal function, and changes in renal function after 2 and 3 years, indicating that serum soluble AREG levels might serve as a biomarker of renal function and renal prognosis in CKD.
Collapse
Affiliation(s)
- Yuki Osakabe
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan.
| | - Yoshinori Taniguchi
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Kazu Hamada Ode
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Yoshiko Shimamura
- Department of Dialysis, Kochi Memorial Hospital, Shiromi-cho, Kochi, Kochi, 780-0824, Japan
| | - Satoshi Inotani
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Hirofumi Nishikawa
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Tatsuki Matsumoto
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Taro Horino
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Shimpei Fujimoto
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Yoshio Terada
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| |
Collapse
|
8
|
Koga R, Maehara T, Aoyagi R, Munemura R, Murakami Y, Doi A, Kono M, Yamamoto H, Niiro H, Kiyoshima T, Tanabe M, Nakano T, Matsukuma Y, Kawano M, Stone JH, Pillai S, Nakamura S, Kawano S. Granzyme K- and amphiregulin-expressing cytotoxic T cells and activated extrafollicular B cells are potential drivers of IgG4-related disease. J Allergy Clin Immunol 2024; 153:1095-1112. [PMID: 38092138 DOI: 10.1016/j.jaci.2023.11.916] [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: 09/05/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND IgG4-related disease (IgG4-RD), an example of a type I immune disease, is an immune-mediated fibrotic disorder characterized by dysregulated resolution of severe inflammation and wound healing. However, truly dominant or pathognomonic autoantibodies related to IgG4-RD are not identified. OBJECTIVE We sought to perform single-cell RNA sequencing and T-cell receptor and B-cell receptor sequencing to obtain a comprehensive, unbiased view of tissue-infiltrating T and B cells. METHODS We performed unbiased single-cell RNA-sequencing analysis for the transcriptome and T-cell receptor sequencing and B-cell receptor sequencing on sorted CD3+ T or CD19+ B cells from affected tissues of patients with IgG4-RD. We also conducted quantitative analyses of CD3+ T-cell and CD19+ B-cell subsets in 68 patients with IgG4-RD and 30 patients with Sjögren syndrome. RESULTS Almost all clonally expanded T cells in these lesions were either Granzyme K (GZMK)-expressing CD4+ cytotoxic T cells or GZMK+CD8+ T cells. These GZMK-expressing cytotoxic T cells also expressed amphiregulin and TGF-β but did not express immune checkpoints, and the tissue-infiltrating CD8+ T cells were phenotypically heterogeneous. MKI67+ B cells and IgD-CD27-CD11c-CXCR5- double-negative 3 B cells were clonally expanded and infiltrated affected tissue lesions. GZMK+CD4+ cytotoxic T cells colocalized with MKI67+ B cells in the extrafollicular area from affected tissue sites. CONCLUSIONS The above-mentioned cells likely participate in T-B collaborative events, suggesting possible avenues for targeted therapies. Our findings were validated using orthogonal approaches, including multicolor immunofluorescence and the use of comparator disease groups, to support the central role of cytotoxic CD4+ and CD8+ T cells expressing GZMK, amphiregulin, and TGF-β in the pathogenesis of inflammatory fibrotic disorders.
Collapse
Affiliation(s)
- Risako Koga
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takashi Maehara
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan; Dento-craniofacial Development and Regeneration Research Center, Faculty of Dental Science, Kyushu University, Kyushu, Japan.
| | - Ryuichi Aoyagi
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ryusuke Munemura
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuka Murakami
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | | | - Michihito Kono
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hidetaka Yamamoto
- Graduate School of Medicine, Dentistry & Pharmaceutical Science, Okayama University, Okayama, Japan
| | - Hiroaki Niiro
- Department of Medical Education, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tamotsu Kiyoshima
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Mika Tanabe
- Department of Ophthalmology, Graduate School of Medicine Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuta Matsukuma
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mitsuhiro Kawano
- Division of Rheumatology, Department of Internal Medicine, Kanazawa University Hospital, Kanazawa, Japan
| | - John H Stone
- Division of Rheumatology, Allergy, and Immunology, Harvard Medical School, Boston, Mass
| | - Shiv Pillai
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Seiji Nakamura
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shintaro Kawano
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
9
|
Jin R, Forbes CM, Miller NL, Lafin J, Strand DW, Case T, Cates JM, Liu Q, Ramirez-Solano M, Mohler JL, Matusik RJ. Transcriptomic analysis of benign prostatic hyperplasia identifies critical pathways in prostatic overgrowth and 5-alpha reductase inhibitor resistance. Prostate 2024; 84:441-459. [PMID: 38168866 DOI: 10.1002/pros.24661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/06/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND The medical therapy of prostatic symptoms (MTOPS) trial randomized men with symptoms of benign prostatic hyperplasia (BPH) and followed response of treatment with a 5α-reductase inhibitor (5ARI), an alpha-adrenergic receptor antagonist (α-blocker), the combination of 5ARI and α-blocker or no medical therapy (none). Medical therapy reduced risk of clinical progression by 66% but the reasons for nonresponse or loss of therapeutic response in some patients remains unresolved. Our previous work showed that prostatic glucocorticoid levels are increased in 5ARI-treated patients and that glucocorticoids can increased branching of prostate epithelia in vitro. To understand the transcriptomic changes associated with 5ARI treatment, we performed bulk RNA sequencing of BPH and control samples from patients who received 5ARI versus those that did not. Deconvolution analysis was performed to estimate cellular composition. Bulk RNA sequencing was also performed on control versus glucocorticoid-treated prostate epithelia in 3D culture to determine underlying transcriptomic changes associated with branching morphogenesis. METHOD Surgical BPH (S-BPH) tissue was defined as benign prostatic tissue collected from the transition zone (TZ) of patients who failed medical therapy while control tissue termed Incidental BPH (I-BPH) was obtained from the TZ of men undergoing radical prostatectomy for low-volume/grade prostatic adenocarcinoma confined to the peripheral zone. S-BPH patients were divided into four subgroups: men on no medical therapy (none: n = 7), α-blocker alone (n = 10), 5ARI alone (n = 6) or combination therapy (α-blocker and 5ARI: n = 7). Control I-BPH tissue was from men on no medical therapy (none: n = 8) or on α-blocker (n = 6). A human prostatic cell line in 3D culture that buds and branches was used to identify genes involved in early prostatic growth. Snap-frozen prostatic tissue taken at the time of surgery and 3D organoids were used for RNA-seq analysis. Bulk RNAseq data were deconvoluted using CIBERSORTx. Differentially expressed genes (DEG) that were statistically significant among S-BPH, I-BPH, and during budding and branching of organoids were used for pathway analysis. RESULTS Transcriptomic analysis between S-BPH (n = 30) and I-BPH (n = 14) using a twofold cutoff (p < 0.05) identified 377 DEG (termed BPH377) and a cutoff < 0.05 identified 3377 DEG (termed BPH3377). Within the S-BPH, the subgroups none and α-blocker were compared to patients on 5ARI to reveal 361 DEG (termed 5ARI361) that were significantly changed. Deconvolution analysis of bulk RNA seq data with a human prostate single cell data set demonstrated increased levels of mast cells, NK cells, interstitial fibroblasts, and prostate luminal cells in S-BPH versus I-BPH. Glucocorticoid (GC)-induced budding and branching of benign prostatic cells in 3D culture was compared to control organoids to identify early events in prostatic morphogenesis. GC induced 369 DEG (termed GC359) in 3D culture. STRING analysis divided the large datasets into 20-80 genes centered around a hub. In general, biological processes induced in BPH supported growth and differentiation such as chromatin modification and DNA repair, transcription, cytoskeleton, mitochondrial electron transport, ubiquitination, protein folding, and cholesterol synthesis. Identified signaling pathways were pooled to create a list of DEG that fell into seven hubs/clusters. The hub gene centrality was used to name the network including AP-1, interleukin (IL)-6, NOTCH1 and NOTCH3, NEO1, IL-13, and HDAC/KDM. All hubs showed connections to inflammation, chromatin structure, and development. The same approach was applied to 5ARI361 giving multiple networks, but the EGF and sonic hedgehog (SHH) hub was of particular interest as a developmental pathway. The BPH3377, 5ARI363, and GC359 lists were compared and 67 significantly changed DEG were identified. Common genes to the 3D culture included an IL-6 hub that connected to genes identified in BPH hubs that defined AP1, IL-6, NOTCH, NEO1, IL-13, and HDAC/KDM. CONCLUSIONS Reduction analysis of BPH and 3D organoid culture uncovered networks previously identified in prostatic development as being reinitiated in BPH. Identification of these pathways provides insight into the failure of medical therapy for BPH and new therapeutic targets for BPH/LUTS.
Collapse
Affiliation(s)
- Renjie Jin
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Connor M Forbes
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Urology Department, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Nicole L Miller
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John Lafin
- Department of Urology, University of Texas, Southwestern, Dallas, Texas, USA
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Douglas W Strand
- Department of Urology, University of Texas, Southwestern, Dallas, Texas, USA
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas Case
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin M Cates
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marisol Ramirez-Solano
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James L Mohler
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Robert J Matusik
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
10
|
VanSlyke JK, Boswell BA, Musil LS. Tonic ErbB signaling underlies TGFβ-induced activation of ERK and is required for lens cell epithelial to myofibroblast transition. Mol Biol Cell 2024; 35:ar35. [PMID: 38170570 PMCID: PMC10916858 DOI: 10.1091/mbc.e23-07-0294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/01/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Fibrosis is a major, but incompletely understood, component of many diseases. The most common vision-disrupting complication of cataract surgery involves differentiation of residual lens cells into myofibroblasts. In serum-free primary cultures of lens epithelial cells (DCDMLs), inhibitors of either ERK or of ErbB signaling prevent TGFβ from upregulating both early (fibronectin) and late (αSMA) markers of myofibroblast differentiation. TGFβ stimulates ERK in DCDMLs within 1.5 h. Kinase inhibitors of ErbBs, but not of several other growth factor receptors in lens cells, reduce phospho ERK to below basal levels in the absence or presence of TGFβ. This effect is attributable to constitutive ErbB activity playing a major role in regulating the basal levels pERK. Additional studies support a model in which TGFβ-generated reactive oxygen species serve to indirectly amplify ERK signaling downstream of tonically active ErbBs to mediate myofibroblast differentiation. ERK activity is in turn essential for expression of ErbB1 and ErbB2, major inducers of ERK signaling. By mechanistically linking TGFβ, ErbB, and ERK signaling to myofibroblast differentiation, our data elucidate a new role for ErbBs in fibrosis and reveal a novel mode by which TGFβ directs lens cell fate.
Collapse
Affiliation(s)
- Judy K. VanSlyke
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239
| | - Bruce A. Boswell
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239
| | - Linda S. Musil
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239
| |
Collapse
|
11
|
Mucciolo G, Araos Henríquez J, Jihad M, Pinto Teles S, Manansala JS, Li W, Ashworth S, Lloyd EG, Cheng PSW, Luo W, Anand A, Sawle A, Piskorz A, Biffi G. EGFR-activated myofibroblasts promote metastasis of pancreatic cancer. Cancer Cell 2024; 42:101-118.e11. [PMID: 38157863 DOI: 10.1016/j.ccell.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 10/10/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis. Cancer-associated fibroblasts (CAFs) are recognized potential therapeutic targets, but poor understanding of these heterogeneous cell populations has limited the development of effective treatment strategies. We previously identified transforming growth factor beta (TGF-β) as a main driver of myofibroblastic CAFs (myCAFs). Here, we show that epidermal growth factor receptor/Erb-B2 receptor (EGFR/ERBB2) signaling is induced by TGF-β in myCAFs through an autocrine process mediated by amphiregulin. Inhibition of this EGFR/ERBB2-signaling network in PDAC organoid-derived cultures and mouse models differentially impacts distinct CAF subtypes, providing insights into mechanisms underpinning their heterogeneity. Remarkably, EGFR-activated myCAFs promote PDAC metastasis in mice, unmasking functional significance in myCAF heterogeneity. Finally, analyses of other cancer datasets suggest that these processes might operate in other malignancies. These data provide functional relevance to myCAF heterogeneity and identify a candidate target for preventing tumor invasion in PDAC.
Collapse
Affiliation(s)
- Gianluca Mucciolo
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Joaquín Araos Henríquez
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Muntadher Jihad
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Sara Pinto Teles
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Judhell S Manansala
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Wenlong Li
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Sally Ashworth
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Eloise G Lloyd
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Priscilla S W Cheng
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Weike Luo
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Akanksha Anand
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Ashley Sawle
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Anna Piskorz
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Giulia Biffi
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK.
| |
Collapse
|
12
|
Panahipour L, Abbasabadi AO, Wagner A, Kratochwill K, Pichler M, Gruber R. Bone Allograft Acid Lysates Change the Genetic Signature of Gingival Fibroblasts. Int J Mol Sci 2023; 24:16181. [PMID: 38003371 PMCID: PMC10671348 DOI: 10.3390/ijms242216181] [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: 10/11/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Bone allografts are widely used as osteoconductive support to guide bone regrowth. Bone allografts are more than a scaffold for the immigrating cells as they maintain some bioactivity of the original bone matrix. Yet, it remains unclear how immigrating cells respond to bone allografts. To this end, we have evaluated the response of mesenchymal cells exposed to acid lysates of bone allografts (ALBA). RNAseq revealed that ALBA has a strong impact on the genetic signature of gingival fibroblasts, indicated by the increased expression of IL11, AREG, C11orf96, STC1, and GK-as confirmed by RT-PCR, and for IL11 and STC1 by immunoassays. Considering that transforming growth factor-β (TGF-β) is stored in the bone matrix and may have caused the expression changes, we performed a proteomics analysis, TGF-β immunoassay, and smad2/3 nuclear translocation. ALBA neither showed detectable TGF-β nor was the lysate able to induce smad2/3 translocation. Nevertheless, the TGF-β receptor type I kinase inhibitor SB431542 significantly decreased the expression of IL11, AREG, and C11orf96, suggesting that other agonists than TGF-β are responsible for the robust cell response. The findings suggest that IL11, AREG, and C11orf96 expression in mesenchymal cells can serve as a bioassay reflecting the bioactivity of the bone allografts.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
| | - Azarakhsh Oladzad Abbasabadi
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
| | - Anja Wagner
- Core Facility Proteomics, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (K.K.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Klaus Kratochwill
- Core Facility Proteomics, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (K.K.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
13
|
Yao HC, Zhu Y, Lu HY, Ju HM, Xu SQ, Qiao Y, Wei SJ. Type 2 innate lymphoid cell-derived amphiregulin regulates type II alveolar epithelial cell transdifferentiation in a mouse model of bronchopulmonary dysplasia. Int Immunopharmacol 2023; 122:110672. [PMID: 37480752 DOI: 10.1016/j.intimp.2023.110672] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/03/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a common complication in preterm infants characterized by alveolar growth arrest. Interleukin (IL)-33 and type 2 innate lymphoid cell (ILC2) affect type II alveolar epithelial cell (AECII) differentiation in BPD mice and may cause increased lung epithelial-mesenchymal transition (EMT). Amphiregulin (AREG) can be produced by ILC2 and is associated with tissue repair. However, the action mechanism of AREG produced by ILC2 to alveolar development in BPD is unclear. In this study, we aimed to demonstrate the role and mechanism of AREG in influencing AECII transdifferentiation in the lung tissue of BPD mice. The effects of ILC2-derived AREG on AECII transdifferentiation were verified in vivo and in vitro, and the role of IL-33 on ILC2-derived AREG in AECII transdifferentiation in BPD mice and a preliminary investigation of the role of AREG's receptor-epidermal growth factor receptor (EGFR) on AECII transdifferentiation. The results showed that neonatal mice developed severe lung injury after hyperoxia, and IL-33 induced AREG production via ILC2 affected normal AECII differentiation and promoted EMT. In addition, the blockade of EGFR was found to alleviate the impaired AECII differentiation under hyperoxia in an in vitro study. In summary, our study demonstrates that AREG secreted by ILC2 affects AECII transdifferentiation in BPD mice, which provides a new idea for the clinical treatment of BPD.
Collapse
Affiliation(s)
- Hui-Ci Yao
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yue Zhu
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Hong-Yan Lu
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Hui-Min Ju
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Su-Qing Xu
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yu Qiao
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Shan-Jie Wei
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
14
|
Wang J, Zhao X, Wan YY. Intricacies of TGF-β signaling in Treg and Th17 cell biology. Cell Mol Immunol 2023; 20:1002-1022. [PMID: 37217798 PMCID: PMC10468540 DOI: 10.1038/s41423-023-01036-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Balanced immunity is pivotal for health and homeostasis. CD4+ helper T (Th) cells are central to the balance between immune tolerance and immune rejection. Th cells adopt distinct functions to maintain tolerance and clear pathogens. Dysregulation of Th cell function often leads to maladies, including autoimmunity, inflammatory disease, cancer, and infection. Regulatory T (Treg) and Th17 cells are critical Th cell types involved in immune tolerance, homeostasis, pathogenicity, and pathogen clearance. It is therefore critical to understand how Treg and Th17 cells are regulated in health and disease. Cytokines are instrumental in directing Treg and Th17 cell function. The evolutionarily conserved TGF-β (transforming growth factor-β) cytokine superfamily is of particular interest because it is central to the biology of both Treg cells that are predominantly immunosuppressive and Th17 cells that can be proinflammatory, pathogenic, and immune regulatory. How TGF-β superfamily members and their intricate signaling pathways regulate Treg and Th17 cell function is a question that has been intensely investigated for two decades. Here, we introduce the fundamental biology of TGF-β superfamily signaling, Treg cells, and Th17 cells and discuss in detail how the TGF-β superfamily contributes to Treg and Th17 cell biology through complex yet ordered and cooperative signaling networks.
Collapse
Affiliation(s)
- Junying Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xingqi Zhao
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yisong Y Wan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
15
|
Chen S, Chen Q, Zhang X, Shen Y, Shi X, Dai X, Yi S. Schwann cell-derived amphiregulin enhances nerve regeneration via supporting the proliferation and migration of Schwann cells and the elongation of axons. J Neurochem 2023; 166:678-691. [PMID: 37439370 DOI: 10.1111/jnc.15916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/14/2023]
Abstract
Peripheral nerves have limited regeneration ability following nerve injury. Applying growth factors with neurotrophic roles is beneficial for accelerating peripheral nerve regeneration. Here we show that after rat sciatic nerve injury, growth factor amphiregulin (AREG) is upregulated in Schwann cells of sciatic nerves. Elevated AREG stimulates the proliferation and migration of Schwann cells by activating ERK1/2 cascade. Schwann cell-secreted AREG further facilitates the outgrowth of neurites and the elongation of injured axons. Administration of AREG to injured sciatic nerves stimulates the proliferation of Schwann cells to replace lost cell population, encourages the migration of Schwann cells to form cell cords, and facilitates the regrowth of axons. Overall, our results identify AREG as an important neurotrophic factor and thus provide a promising therapeutic avenue towards peripheral nerve injury.
Collapse
Affiliation(s)
- Sailing Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Qianqian Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Xiaojiao Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Yinying Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Xinyu Shi
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Xiu Dai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| |
Collapse
|
16
|
Xue Y, Mei H, Chen Y, Griffin JD, Liu Q, Weisberg E, Yang J. Repurposing clinically available drugs and therapies for pathogenic targets to combat SARS-CoV-2. MedComm (Beijing) 2023; 4:e254. [PMID: 37193304 PMCID: PMC10183156 DOI: 10.1002/mco2.254] [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: 12/02/2022] [Revised: 02/11/2023] [Accepted: 03/07/2023] [Indexed: 05/18/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has affected a large portion of the global population, both physically and mentally. Current evidence suggests that the rapidly evolving coronavirus subvariants risk rendering vaccines and antibodies ineffective due to their potential to evade existing immunity, with enhanced transmission activity and higher reinfection rates that could lead to new outbreaks across the globe. The goal of viral management is to disrupt the viral life cycle as well as to relieve severe symptoms such as lung damage, cytokine storm, and organ failure. In the fight against viruses, the combination of viral genome sequencing, elucidation of the structure of viral proteins, and identifying proteins that are highly conserved across multiple coronaviruses has revealed many potential molecular targets. In addition, the time- and cost-effective repurposing of preexisting antiviral drugs or approved/clinical drugs for these targets offers considerable clinical advantages for COVID-19 patients. This review provides a comprehensive overview of various identified pathogenic targets and pathways as well as corresponding repurposed approved/clinical drugs and their potential against COVID-19. These findings provide new insight into the discovery of novel therapeutic strategies that could be applied to the control of disease symptoms emanating from evolving SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Yiying Xue
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Husheng Mei
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical ScienceChinese Academy of SciencesHefeiChina
- University of Science and Technology of ChinaHefeiAnhuiChina
| | - Yisa Chen
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - James D. Griffin
- Department of Medical Oncology, Dana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medicine, Harvard Medical SchoolBostonMassachusettsUSA
| | - Qingsong Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical ScienceChinese Academy of SciencesHefeiChina
- University of Science and Technology of ChinaHefeiAnhuiChina
- Hefei Cancer HospitalChinese Academy of SciencesHefeiChina
| | - Ellen Weisberg
- Department of Medical Oncology, Dana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medicine, Harvard Medical SchoolBostonMassachusettsUSA
| | - Jing Yang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical ScienceChinese Academy of SciencesHefeiChina
| |
Collapse
|
17
|
Saul S, Karim M, Ghita L, Huang PT, Chiu W, Durán V, Lo CW, Kumar S, Bhalla N, Leyssen P, Alem F, Boghdeh NA, Tran DH, Cohen CA, Brown JA, Huie KE, Tindle C, Sibai M, Ye C, Khalil AM, Martinez-Sobrido L, Dye JM, Pinsky BA, Ghosh P, Das S, Solow-Cordero DE, Jin J, Wikswo JP, Jochmans D, Neyts J, Jonghe SD, Narayanan A, Einav S. Anticancer pan-ErbB inhibitors reduce inflammation and tissue injury and exert broad-spectrum antiviral effects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2021.05.15.444128. [PMID: 34159337 PMCID: PMC8219101 DOI: 10.1101/2021.05.15.444128] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Targeting host factors exploited by multiple viruses could offer broad-spectrum solutions for pandemic preparedness. Seventeen candidates targeting diverse functions emerged in a screen of 4,413 compounds for SARS-CoV-2 inhibitors. We demonstrated that lapatinib and other approved inhibitors of the ErbB family receptor tyrosine kinases suppress replication of SARS-CoV-2, Venezuelan equine encephalitis virus (VEEV), and other emerging viruses with a high barrier to resistance. Lapatinib suppressed SARS-CoV-2 entry and later stages of the viral life cycle and showed synergistic effect with the direct-acting antiviral nirmatrelvir. We discovered that ErbB1, 2 and 4 bind SARS-CoV-2 S1 protein and regulate viral and ACE2 internalization, and they are required for VEEV infection. In human lung organoids, lapatinib protected from SARS-CoV-2-induced activation of ErbB-regulated pathways implicated in non-infectious lung injury, pro-inflammatory cytokine production, and epithelial barrier injury. Lapatinib suppressed VEEV replication, cytokine production and disruption of the blood-brain barrier integrity in microfluidic-based human neurovascular units, and reduced mortality in a lethal infection murine model. We validated lapatinib-mediated inhibition of ErbB activity as an important mechanism of antiviral action. These findings reveal regulation of viral replication, inflammation, and tissue injury via ErbBs and establish a proof-of-principle for a repurposed, ErbB-targeted approach to combat emerging viruses.
Collapse
|
18
|
Leiby KL, Yuan Y, Ng R, Raredon MSB, Adams TS, Baevova P, Greaney AM, Hirschi KK, Campbell SG, Kaminski N, Herzog EL, Niklason LE. Rational engineering of lung alveolar epithelium. NPJ Regen Med 2023; 8:22. [PMID: 37117221 PMCID: PMC10147714 DOI: 10.1038/s41536-023-00295-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 04/06/2023] [Indexed: 04/30/2023] Open
Abstract
Engineered whole lungs may one day expand therapeutic options for patients with end-stage lung disease. However, the feasibility of ex vivo lung regeneration remains limited by the inability to recapitulate mature, functional alveolar epithelium. Here, we modulate multimodal components of the alveolar epithelial type 2 cell (AEC2) niche in decellularized lung scaffolds in order to guide AEC2 behavior for epithelial regeneration. First, endothelial cells coordinate with fibroblasts, in the presence of soluble growth and maturation factors, to promote alveolar scaffold population with surfactant-secreting AEC2s. Subsequent withdrawal of Wnt and FGF agonism synergizes with tidal-magnitude mechanical strain to induce the differentiation of AEC2s to squamous type 1 AECs (AEC1s) in cultured alveoli, in situ. These results outline a rational strategy to engineer an epithelium of AEC2s and AEC1s contained within epithelial-mesenchymal-endothelial alveolar-like units, and highlight the critical interplay amongst cellular, biochemical, and mechanical niche cues within the reconstituting alveolus.
Collapse
Affiliation(s)
- Katherine L Leiby
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Yale School of Medicine, New Haven, CT, USA
| | - Yifan Yuan
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Ronald Ng
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Micha Sam Brickman Raredon
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Yale School of Medicine, New Haven, CT, USA
| | - Taylor S Adams
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Pavlina Baevova
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Allison M Greaney
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Karen K Hirschi
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Stuart G Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Naftali Kaminski
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Erica L Herzog
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Laura E Niklason
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
19
|
Welz L, Aden K. Fibrosis and Inflammation in Inflammatory Bowel Disease-More Than 2 Sides of the Same Coin? Gastroenterology 2023; 164:19-21. [PMID: 36341736 DOI: 10.1053/j.gastro.2022.10.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 10/25/2022] [Indexed: 02/03/2023]
Affiliation(s)
- Lina Welz
- Institute of Clinical Molecular Biology and, Department of Internal Medicine I, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Konrad Aden
- Institute of Clinical Molecular Biology and, Department of Internal Medicine I, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
20
|
Zhao X, Yang W, Yu T, Yu Y, Cui X, Zhou Z, Yang H, Yu Y, Bilotta AJ, Yao S, Xu J, Zhou J, Yochum GS, Koltun WA, Portolese A, Zeng D, Xie J, Pinchuk IV, Zhang H, Cong Y. Th17 Cell-Derived Amphiregulin Promotes Colitis-Associated Intestinal Fibrosis Through Activation of mTOR and MEK in Intestinal Myofibroblasts. Gastroenterology 2023; 164:89-102. [PMID: 36113570 PMCID: PMC9772145 DOI: 10.1053/j.gastro.2022.09.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND & AIMS Intestinal fibrosis is a significant complication of Crohn's disease (CD). Gut microbiota reactive Th17 cells are crucial in the pathogenesis of CD; however, how Th17 cells induce intestinal fibrosis is still not completely understood. METHODS In this study, T-cell transfer model with wild-type (WT) and Areg-/- Th17 cells and dextran sulfate sodium (DSS)-induced chronic colitis model in WT and Areg-/- mice were used. CD4+ T-cell expression of AREG was determined by quantitative reverse-transcriptase polymerase chain reaction and enzyme-linked immunosorbent assay. The effect of AREG on proliferation/migration/collagen expression in human intestinal myofibroblasts was determined. AREG expression was assessed in healthy controls and patients with CD with or without intestinal fibrosis. RESULTS Although Th1 and Th17 cells induced intestinal inflammation at similar levels when transferred into Tcrβxδ-/- mice, Th17 cells induced more severe intestinal fibrosis. Th17 cells expressed higher levels of AREG than Th1 cells. Areg-/- mice developed less severe intestinal fibrosis compared with WT mice on DSS insults. Transfer of Areg-/- Th17 cells induced less severe fibrosis in Tcrβxδ-/- mice compared with WT Th17 cells. Interleukin (IL)6 and IL21 promoted AREG expression in Th17 cells by activating Stat3. Stat3 inhibitor suppressed Th17-induced intestinal fibrosis. AREG promoted human intestinal myofibroblast proliferation, motility, and collagen I expression, which was mediated by activating mammalian target of rapamycin and MEK. AREG expression was increased in intestinal CD4+ T cells in fibrotic sites compared with nonfibrotic sites from patients with CD. CONCLUSIONS These findings reveal that Th17-derived AREG promotes intestinal fibrotic responses in experimental colitis and human patients with CD. Thereby, AREG might serve as a potential therapeutic target for fibrosis in CD.
Collapse
Affiliation(s)
- Xiaojing Zhao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas; Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenjing Yang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas; Sealy Center for Microbiome Research, University of Texas Medical Branch, Galveston, Texas
| | - Tianming Yu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas; Sealy Center for Microbiome Research, University of Texas Medical Branch, Galveston, Texas
| | - Yu Yu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Xiufang Cui
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Zhou
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Hui Yang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Yanbo Yu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Anthony J Bilotta
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Suxia Yao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Jimin Xu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas
| | - Gregory S Yochum
- Department of Biochemistry and Molecular Biology, Pennsylvania State Milton S. Hershey Medical Center, Hershey, Pennsylvania; Department of Surgery, Pennsylvania State Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Walter A Koltun
- Department of Surgery, Pennsylvania State Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Austin Portolese
- Department of Surgery, Pennsylvania State Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Defu Zeng
- Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California
| | - Jingwu Xie
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, Indiana
| | - Iryna V Pinchuk
- Division of Gastroenterology, Department of Medicine, Pennsylvania State Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Hongjie Zhang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas; Sealy Center for Microbiome Research, University of Texas Medical Branch, Galveston, Texas.
| |
Collapse
|
21
|
Martin JG, Azuelos I. Fibrogenic Effects of Heparin-Binding Epidermal Growth Factor-like Growth Factor: Myeloid or Epithelial Origin? Am J Respir Cell Mol Biol 2022; 67:611-612. [PMID: 36154637 PMCID: PMC9743182 DOI: 10.1165/rcmb.2022-0348ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- James G. Martin
- McGill University Health Center Research InstituteMcGill UniversityMontreal, Quebec, Canada,Department of MedicineMcGill UniversityMontreal, Quebec, Canada
| | - Ilan Azuelos
- McGill University Health Center Research InstituteMcGill UniversityMontreal, Quebec, Canada,Department of MedicineMcGill UniversityMontreal, Quebec, Canada
| |
Collapse
|
22
|
Chen Z, Lan H, Liao Z, Huang J, Jian X, Hu J, Liao H. Regulatory T cells-centered regulatory networks of skeletal muscle inflammation and regeneration. Cell Biosci 2022; 12:112. [PMID: 35869487 PMCID: PMC9308315 DOI: 10.1186/s13578-022-00847-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 07/08/2022] [Indexed: 11/10/2022] Open
Abstract
As the understanding of skeletal muscle inflammation is increasingly clarified, the role of Treg cells in the treatment of skeletal muscle diseases has attracted more attention in recent years. A consensus has been reached that the regulation of Treg cells is the key to completing the switch of inflammation and repair of skeletal muscle, whose presence directly determine the repairing quality of the injured skeletal muscle. However, the functioning process of Treg cells remains unreported, thereby making it necessary to summarize the current role of Treg cells in skeletal muscle. In this review, the characteristics, origins, and cellular kinetics of these Treg cells are firstly described; Then, the relationship between Treg cells and muscle satellite cells (MuSCs), conventional T cells (Tconv) is discussed (the former is involved in the entire repair and regeneration process, while the latter matters considerably in causing most skeletal muscle autoimmune diseases); Next, focus is placed on the control of Treg cells on the phenotypic switch of macrophages, which is the key to the switch of inflammation; Finally, factors regulating the functional process of Treg cells are analyzed, and a regulatory network centered on Treg cells is summarized. The present study summarizes the cell-mediated interactions in skeletal muscle repair over the past decade, and elucidates the central role of regulatory T cells in this process, so that other researchers can more quickly and comprehensively understand the development and direction of this very field. It is believed that the hereby proposed viewpoints and problems can provide fresh visions for the latecomers.
Collapse
|
23
|
Huang P, Zhang J, Duan W, Jiao J, Leng A, Qu J. Plant polysaccharides with anti-lung injury effects as a potential therapeutic strategy for COVID-19. Front Pharmacol 2022; 13:982893. [DOI: 10.3389/fphar.2022.982893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
When coronavirus disease 2019 (COVID-19) develops into the severe phase, lung injury, acute respiratory distress syndrome, and/or respiratory failure could develop within a few days. As a result of pulmonary tissue injury, pathomorphological changes usually present endothelial dysfunction, inflammatory cell infiltration of the lung interstitium, defective gas exchange, and wall leakage. Consequently, COVID-19 may progress to tremendous lung injury, ongoing lung failure, and death. Exploring the treatment drugs has important implications. Recently, the application of traditional Chinese medicine had better performance in reducing fatalities, relieving symptoms, and curtailing hospitalization. Through constant research and study, plant polysaccharides may emerge as a crucial resource against lung injury with high potency and low side effects. However, the absence of a comprehensive understanding of lung-protective mechanisms impedes further investigation of polysaccharides. In the present article, a comprehensive review of research into plant polysaccharides in the past 5 years was performed. In total, 30 types of polysaccharides from 19 kinds of plants have shown lung-protective effects through the pathological processes of inflammation, oxidative stress, apoptosis, autophagy, epithelial–mesenchymal transition, and immunomodulation by mediating mucin and aquaporins, macrophage, endoplasmic reticulum stress, neutrophil, TGF-β1 pathways, Nrf2 pathway, and other mechanisms. Moreover, the deficiencies of the current studies and the future research direction are also tentatively discussed. This research provides a comprehensive perspective for better understanding the mechanism and development of polysaccharides against lung injury for the treatment of COVID-19.
Collapse
|
24
|
Maranatha D, Hasan H, Bakhtiar A, Widyoningroem A, Aryati. Association of TNF-α, TGF-β1, amphiregulin, IL-2, and EGFR WITH pulmonary fibrosis in COVID-19. J Infect Public Health 2022; 15:1072-1075. [PMID: 36088825 PMCID: PMC9398556 DOI: 10.1016/j.jiph.2022.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/04/2022] [Accepted: 08/17/2022] [Indexed: 01/06/2023] Open
Abstract
Pulmonary fibrosis is a well-recognized sequela associated with coronavirus disease 2019 (COVID-19), however the mechanism is yet to be clearly understood. The study was designed to evaluate the association of TNF-α, TGF- β1, amphiregulin, IL-2, and EGFR with pulmonary fibrosis after COVID-19 pneumonia. Non-severe, severe, and critical COVID-19 pneumonia patients were included in this study after the patients agreed and gave written informed consent. Blood samples were analyzed with the ELISA method for cytokine examination. The non-contrast chest CT scan was performed after patients were discharged from hospital. Seventy-nine patients with a mean age of 54 years (57 % men, 43 % women) were fully evaluated. Pulmonary fibrosis was found in 74 patients (93.7 %). Serum levels of TGF-β1 60.55 pg/mL (11.42-2001.16), TNF-α 13.31 pg/mL (3.54-200.32), EGFR 14.9 pg/mL(6.4-53.6), IL-2 12.41 pg/mL(11-14.13), amphiregulin 156.5 pg/mL (21.7-1234). Serum levels of TNF-α increase according to the severity of clinical classification. A significant association between serum levels of TGF-β1, TNF- α, and pulmonary fibrosis with rs-0.247, p = 0.027; rs 0.259, p = 0.046 was found. According to this study, TNF-α and TGF-β1 potentially participate in the process of pulmonary fibrosis in COVID-19.
Collapse
Affiliation(s)
- Daniel Maranatha
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Airlangga - Dr. Soetomo Academic Hospital, Indonesia.
| | - Helmia Hasan
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Airlangga - Dr. Soetomo Academic Hospital, Indonesia
| | - Arief Bakhtiar
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Airlangga - Dr. Soetomo Academic Hospital, Indonesia
| | - Anita Widyoningroem
- Department of Radiology, Faculty of Medicine, Universitas Airlangga - Dr. Soetomo Academic Hospital, Indonesia
| | - Aryati
- Department of Clinical Pathology, Faculty of Medicine, Universitas Airlangga - Dr. Soetomo Academic Hospital, Indonesia
| |
Collapse
|
25
|
Jaeger B, Schupp JC, Plappert L, Terwolbeck O, Artysh N, Kayser G, Engelhard P, Adams TS, Zweigerdt R, Kempf H, Lienenklaus S, Garrels W, Nazarenko I, Jonigk D, Wygrecka M, Klatt D, Schambach A, Kaminski N, Prasse A. Airway basal cells show a dedifferentiated KRT17 highPhenotype and promote fibrosis in idiopathic pulmonary fibrosis. Nat Commun 2022; 13:5637. [PMID: 36163190 PMCID: PMC9513076 DOI: 10.1038/s41467-022-33193-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 09/07/2022] [Indexed: 11/10/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal disease with limited treatment options. In this study, we focus on the properties of airway basal cells (ABC) obtained from patients with IPF (IPF-ABC). Single cell RNA sequencing (scRNAseq) of bronchial brushes revealed extensive reprogramming of IPF-ABC towards a KRT17high PTENlow dedifferentiated cell type. In the 3D organoid model, compared to ABC obtained from healthy volunteers, IPF-ABC give rise to more bronchospheres, de novo bronchial structures resembling lung developmental processes, induce fibroblast proliferation and extracellular matrix deposition in co-culture. Intratracheal application of IPF-ABC into minimally injured lungs of Rag2-/- or NRG mice causes severe fibrosis, remodeling of the alveolar compartment, and formation of honeycomb cyst-like structures. Connectivity MAP analysis of scRNAseq of bronchial brushings suggested that gene expression changes in IPF-ABC can be reversed by SRC inhibition. After demonstrating enhanced SRC expression and activity in these cells, and in IPF lungs, we tested the effects of saracatinib, a potent SRC inhibitor previously studied in humans. We demonstrate that saracatinib modified in-vitro and in-vivo the profibrotic changes observed in our 3D culture system and novel mouse xenograft model.
Collapse
Affiliation(s)
- Benedikt Jaeger
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- German Center for Lung Research, BREATH, Hannover, Germany
| | - Jonas Christian Schupp
- German Center for Lung Research, BREATH, Hannover, Germany
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Pulmonology, Hannover Medical School, Hannover, Germany
| | - Linda Plappert
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- German Center for Lung Research, BREATH, Hannover, Germany
| | - Oliver Terwolbeck
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- German Center for Lung Research, BREATH, Hannover, Germany
| | - Nataliia Artysh
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- German Center for Lung Research, BREATH, Hannover, Germany
- Department of Pulmonology, Hannover Medical School, Hannover, Germany
| | - Gian Kayser
- Institute of Surgical Pathology, University Medical Center, Freiburg, Germany
| | - Peggy Engelhard
- Department of Pneumology, University Medical Center, Freiburg, Germany
| | - Taylor Sterling Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Henning Kempf
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Stefan Lienenklaus
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Wiebke Garrels
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Irina Nazarenko
- Institute for Infection Prevention and Hospital Epidemiology, Medical Center - University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Danny Jonigk
- German Center for Lung Research, BREATH, Hannover, Germany
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Justus Liebig University, Gießen, Germany
| | - Denise Klatt
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Antje Prasse
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany.
- German Center for Lung Research, BREATH, Hannover, Germany.
- Department of Pulmonology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
26
|
Glaubitz J, Wilden A, Golchert J, Homuth G, Völker U, Bröker BM, Thiele T, Lerch MM, Mayerle J, Aghdassi AA, Weiss FU, Sendler M. In mouse chronic pancreatitis CD25 +FOXP3 + regulatory T cells control pancreatic fibrosis by suppression of the type 2 immune response. Nat Commun 2022; 13:4502. [PMID: 35922425 PMCID: PMC9349313 DOI: 10.1038/s41467-022-32195-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 07/20/2022] [Indexed: 12/19/2022] Open
Abstract
Chronic pancreatitis (CP) is characterized by chronic inflammation and the progressive fibrotic replacement of exocrine and endocrine pancreatic tissue. We identify Treg cells as central regulators of the fibroinflammatory reaction by a selective depletion of FOXP3-positive cells in a transgenic mouse model (DEREG-mice) of experimental CP. In Treg-depleted DEREG-mice, the induction of CP results in a significantly increased stroma deposition, the development of exocrine insufficiency and significant weight loss starting from day 14 after disease onset. In CP, FOXP3+CD25+ Treg cells suppress the type-2 immune response by a repression of GATA3+ T helper cells (Th2), GATA3+ innate lymphoid cells type 2 (ILC2) and CD206+ M2-macrophages. A suspected pathomechanism behind the fibrotic tissue replacement may involve an observed dysbalance of Activin A expression in macrophages and of its counter regulator follistatin. Our study identified Treg cells as key regulators of the type-2 immune response and of organ remodeling during CP. The Treg/Th2 axis could be a therapeutic target to prevent fibrosis and preserve functional pancreatic tissue. The function of T regulatory cells in the tissue fibrosis in chronic pancreatitis is not fully understood. Here the authors use a mouse model of chronic pancreatitis to show that Treg cells reduce IL-4 mediated chronic inflammation in the pancreas associated with M2-like macrophages in vivo.
Collapse
Affiliation(s)
- Juliane Glaubitz
- Department of Medicine A, University Medicine, University of Greifswald, Greifswald, Germany
| | - Anika Wilden
- Department of Medicine A, University Medicine, University of Greifswald, Greifswald, Germany
| | - Janine Golchert
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Barbara M Bröker
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine, Greifswald, Germany
| | - Thomas Thiele
- Department of Transfusion Medicine, Institute of Immunology and Transfusion Medicine, University Medicine, Greifswald, Germany
| | - Markus M Lerch
- Department of Medicine A, University Medicine, University of Greifswald, Greifswald, Germany
| | - Julia Mayerle
- Department of Medicine A, University Medicine, University of Greifswald, Greifswald, Germany.,Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Ali A Aghdassi
- Department of Medicine A, University Medicine, University of Greifswald, Greifswald, Germany
| | - Frank U Weiss
- Department of Medicine A, University Medicine, University of Greifswald, Greifswald, Germany
| | - Matthias Sendler
- Department of Medicine A, University Medicine, University of Greifswald, Greifswald, Germany.
| |
Collapse
|
27
|
Chait M, Yilmaz MM, Shakil S, Ku AW, Dogra P, Connors TJ, Szabo PA, Gray JI, Wells SB, Kubota M, Matsumoto R, Poon MM, Snyder ME, Baldwin MR, Sims PA, Saqi A, Farber DL, Weisberg SP. Immune and epithelial determinants of age-related risk and alveolar injury in fatal COVID-19. JCI Insight 2022; 7:157608. [PMID: 35446789 PMCID: PMC9228710 DOI: 10.1172/jci.insight.157608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/20/2022] [Indexed: 01/08/2023] Open
Abstract
Respiratory failure in COVID-19 is characterized by widespread disruption of the lung’s alveolar gas exchange interface. To elucidate determinants of alveolar lung damage, we performed epithelial and immune cell profiling in lungs from 24 COVID-19 autopsies and 43 uninfected organ donors ages 18–92 years. We found marked loss of type 2 alveolar epithelial (T2AE) cells and increased perialveolar lymphocyte cytotoxicity in all fatal COVID-19 cases, even at early stages before typical patterns of acute lung injury are histologically apparent. In lungs from uninfected organ donors, there was also progressive loss of T2AE cells with increasing age, which may increase susceptibility to COVID-19–mediated lung damage in older individuals. In the fatal COVID-19 cases, macrophage infiltration differed according to the histopathological pattern of lung injury. In cases with acute lung injury, we found accumulation of CD4+ macrophages that expressed distinctly high levels of T cell activation and costimulation genes and strongly correlated with increased extent of alveolar epithelial cell depletion and CD8+ T cell cytotoxicity. Together, our results show that T2AE cell deficiency may underlie age-related COVID-19 risk and initiate alveolar dysfunction shortly after infection, and we define immune cell mediators that may contribute to alveolar injury in distinct pathological stages of fatal COVID-19.
Collapse
Affiliation(s)
- Michael Chait
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, United States of America
| | - Mine M Yilmaz
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, United States of America
| | - Shanila Shakil
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, United States of America
| | - Amy W Ku
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, United States of America
| | - Pranay Dogra
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, United States of America
| | - Thomas J Connors
- Department of Pediatrics, Columbia University Irving Medical Center, New York, United States of America
| | - Peter A Szabo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, United States of America
| | - Joshua I Gray
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, United States of America
| | - Steven B Wells
- Department of Systems Biology, Columbia University Irving Medical Center, New York, United States of America
| | - Masaru Kubota
- Department of Surgery, Columbia University Irving Medical Center, New York, United States of America
| | - Rei Matsumoto
- Department of Surgery, Columbia University Irving Medical Center, New York, United States of America
| | - Maya Ml Poon
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, United States of America
| | - Mark E Snyder
- Department of Medicine, University of Pittsburgh, Pittsburgh, United States of America
| | - Matthew R Baldwin
- Department of Medicine, Columbia University Iring Medical Ceter, New York, United States of America
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, United States of America
| | - Anjali Saqi
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, United States of America
| | - Donna L Farber
- Department of Surgery, Columbia University Irving Medical Center, New York, United States of America
| | - Stuart P Weisberg
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, United States of America
| |
Collapse
|
28
|
Abstract
In this review article, we will first provide a brief overview of the ErbB receptor-ligand system and its importance in developmental and physiological processes. We will then review the literature regarding the role of ErbB receptors and their ligands in the maladaptive remodeling of lung tissue, with special emphasis on idiopathic pulmonary fibrosis (IPF). Here we will focus on the pathways and cellular processes contributing to epithelial-mesenchymal miscommunication seen in this pathology. We will also provide an overview of the in vivo studies addressing the efficacy of different ErbB signaling inhibitors in experimental models of lung injury and highlight how such studies may contribute to our understanding of ErbB biology in the lung. Finally, we will discuss what we learned from clinical applications of the ErbB1 signaling inhibitors in cancer in order to advance clinical trials in IPF.
Collapse
|
29
|
Son B, Kim TR, Park JH, Yun SI, Choi H, Choi JW, Jeon C, Park HO. SAMiRNA Targeting Amphiregulin Alleviate Total-Body-Irradiation-Induced Renal Fibrosis. Radiat Res 2022; 197:471-479. [PMID: 35148406 DOI: 10.1667/rade-21-00220.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/05/2022] [Indexed: 11/12/2022]
Abstract
Fibrosis is a serious unintended side effect of radiation therapy. In this study, we aimed to investigate whether amphiregulin (AREG) plays a critical role in fibrosis development after total-body irradiation (TBI). We found that the expression of AREG and fibrotic markers, such as α-smooth muscle actin (α-SMA) and collagen type I alpha 1 (COL1α1), was elevated in the kidneys of 6 Gy TBI mice. Expression of AREG and α-SMA was mainly elevated in the proximal and distal tubules of the kidney in response to TBI, which was confirmed by immunofluorescence staining. Knockdown of Areg mRNA using self-assembled-micelle inhibitory RNA (SAMiRNA) significantly reduced the expression of fibrotic markers, including α-SMA and COL1α1, and inflammatory regulators. Finally, intravenous injections of SAMiRNA targeting mouse Areg mRNA (SAMiRNA-mAREG) diminished radiation-induced collagen accumulation in the renal cortex and medulla. Taken together, the results of the present study suggest that blocking of AREG signaling via SAMiRNA-mAREG treatment could be a promising therapeutic approach to alleviate radiation-induced kidney fibrosis.
Collapse
Affiliation(s)
- Beomseok Son
- siRNAgen Therapeutics, Daejeon 34302, Republic of Korea
| | - Tae Rim Kim
- siRNAgen Therapeutics, Daejeon 34302, Republic of Korea
| | - Jun Hong Park
- siRNAgen Therapeutics, Daejeon 34302, Republic of Korea
| | - Sung-Il Yun
- siRNAgen Therapeutics, Daejeon 34302, Republic of Korea
| | - Hanjoo Choi
- siRNAgen Therapeutics, Daejeon 34302, Republic of Korea
| | - Ji Woo Choi
- siRNAgen Therapeutics, Daejeon 34302, Republic of Korea
| | | | - Han-Oh Park
- siRNAgen Therapeutics, Daejeon 34302, Republic of Korea
| |
Collapse
|
30
|
Wang L, Zhu W, Sun R, Liu J, Ma Q, Zhang B, Shi Y. Network Pharmacology Prediction and Molecular Docking-Based Strategy to Discover the Potential Pharmacological Mechanism of Wen-Yu-Jin against Pulmonary Fibrosis in a Mouse Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:7753508. [PMID: 35186103 PMCID: PMC8853792 DOI: 10.1155/2022/7753508] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a devastating lung disease, resulting in gas exchange dysfunction until death. The two drugs approved by the FDA, pirfenidone and nintedanib, have obvious side effects. Wen-yu-jin (WYJ), one of the commonly used herbs in China, can treat respiratory diseases. The potential effects and the underlying mechanism of WYJ against PF are unclear. PURPOSE Employing network pharmacology, molecular docking, and in vivo and in vitro experiments to explore the potential effects and underlying mechanisms of WYJ in the treatment of PF. METHODS Ultra-high pressure liquid chromatography combined with linear ion trap-orbital tandem mass spectrometry (UHPLC-LTQ-orbital trap) was used to identify compounds of WYJ. We got PF-related targets and WYJ compounds-related targets from public databases and further completed critical targets exploration, network construction, and pathway analysis by network pharmacology. Molecular docking predicted binding activity of WYJ compounds and critical targets. Based on the above results, in vivo and in vitro experiments validated the potential effects and mechanisms of WYJ against PF. RESULTS 23 major compositions of WYJ were identified based on UHPLC-LTQ-Orbitrap. According to the results of network pharmacology, STAT3, SRC, IL6, MAPK1, AKT1, EGFR, MAPK8, MAPK14, and IL1B are critical therapeutic targets. Molecular docking results showed that most of the compounds have good binding activities with critical targets. The results of in vivo and in vitro experiments showed that WYJ alleviated the process of fibrosis by targeting MAPK and STAT3 pathways. CONCLUSION Network pharmacology, molecular docking, and in vivo and in vitro experiments showed the potential effects and mechanisms of WYJ against PF, which provides a theoretical basis for the treatment of WYJ with PF.
Collapse
Affiliation(s)
- Lu Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Wenxiang Zhu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
| | - Rui Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qihong Ma
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Binbin Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
31
|
Zha Y, Li Y, Ge Z, Wang J, Jiao Y, Zhang J, Zhang S. ADAMTS8 Promotes Cardiac Fibrosis Partly Through Activating EGFR Dependent Pathway. Front Cardiovasc Med 2022; 9:797137. [PMID: 35224040 PMCID: PMC8866452 DOI: 10.3389/fcvm.2022.797137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022] Open
Abstract
Myocardial infarction or pressure overload leads to cardiac fibrosis, the leading cause of heart failure. ADAMTS8 (A disintegrin and metalloproteinase with thrombospondin motifs 8) has been reported to be involved in many fibrosis-related diseases. However, the specific role of ADAMTS8 in cardiac fibrosis caused by myocardial infarction or pressure overload is yet unclear. The present study aimed to explore the function of ADAMTS8 in cardiac fibrosis and its underlying mechanism. ADAMTS8 expression was significantly increased in patients with dilated cardiomyopathy; its expression myocardial infarction and TAC rat models was also increased, accompanied by increased expression of α-SMA and Collagen1. Adenovirus-mediated overexpression of ADAMTS8 through cardiac in situ injection aggravated cardiac fibrosis and impaired cardiac function in the myocardial infarction rat model. Furthermore, in vitro studies revealed that ADAMTS8 promoted the activation of cardiac fibroblasts; ADAMTS8 acted as a paracrine mediator allowing for cardiomyocytes and fibroblasts to communicate indirectly. Our findings showed that ADAMTS8 could damage the mitochondrial function of cardiac fibroblasts and then activate the PI3K-Akt pathway and MAPK pathways, promoting up-regulation of YAP expression, with EGFR upstream of this pathway. This study systematically revealed the pro-fibrosis effect of ADAMTS8 in cardiac fibrosis and explored its potential role as a therapeutic target for the treatment of cardiac fibrosis and heart failure.
Collapse
Affiliation(s)
- Yafang Zha
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanyan Li
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhuowang Ge
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Wang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuheng Jiao
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiayan Zhang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Song Zhang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Song Zhang
| |
Collapse
|
32
|
Cheng WH, Kao SY, Chen CL, Yuliani FS, Lin LY, Lin CH, Chen BC. Amphiregulin induces CCN2 and fibronectin expression by TGF-β through EGFR-dependent pathway in lung epithelial cells. Respir Res 2022; 23:381. [PMID: 36578010 PMCID: PMC9797108 DOI: 10.1186/s12931-022-02285-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/08/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Airway fibrosis is one of the pathological characteristics of severe asthma. Transforming growth factor (TGF)-β has been known to promote epithelial-mesenchymal transition formation and to play a role in the progression of tissue fibrosis. Cellular communication network factor 2 (CCN2) and fibronectin (FN) are well-known markers of EMT and fibrosis. However, whether AREG is involved in TGF-β-induced CCN2 and FN expression in human lung epithelial cells is unknown. METHODS AREG and FN were analyzed by immunofluorescence staining on ovalbumin-challenged mice. CCN2 and FN expression were evaluated in human lung epithelial (A459) cells following TGF or AREG treatment for the indicated times. Secreted AREG from A549 cells was detected by ELISA. Cell migration was observed by a wound healing assay. Chromatin immunoprecipitation was used to detect the c-Jun binding to the CCN2 promoter. RESULTS AREG and FN expression colocalized in lung tissues from mice with ovalbumin-induced asthma by immunofluorescence staining. Moreover, TGF-β caused the release of AREG from A549 cells into the medium. Smad3 siRNA down-regulated AREG expression. AREG also stimulated CCN2 and FN expression, JNK and c-Jun phosphorylation, and cell migration in A549 cells. AREG small interfering (si) RNA inhibited TGF-β-induced expression of CCN2, FN, and cell migration. Furthermore, AREG-induced CCN2 and FN expression were inhibited by EGFR siRNA, a JNK inhibitor (SP600125), and an activator protein-1 (AP-1) inhibitor (curcumin). EGFR siRNA attenuated AREG-induced JNK and c-Jun phosphorylation. Moreover, SP600125 downregulated AREG-induced c-Jun phosphorylation. CONCLUSION These results suggested that AREG mediates the TGF-β-induced EMT in human lung epithelial cells through EGFR/JNK/AP-1 activation. Understanding the role of AREG in the EMT could foster the development of therapeutic strategies for airway remodeling in severe asthma.
Collapse
Affiliation(s)
- Wun-Hao Cheng
- grid.412896.00000 0000 9337 0481School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031 Taiwan ,grid.412896.00000 0000 9337 0481Respiratory Therapy, Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shih-Ya Kao
- grid.412896.00000 0000 9337 0481School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031 Taiwan
| | - Chia-Ling Chen
- grid.412896.00000 0000 9337 0481School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031 Taiwan
| | - Fara Silvia Yuliani
- grid.412896.00000 0000 9337 0481International Graduate Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan ,grid.8570.a0000 0001 2152 4506Department of Pharmacology and Therapy, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Lee-Yuan Lin
- grid.412896.00000 0000 9337 0481School of Medicine, Collage of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Huang Lin
- grid.412896.00000 0000 9337 0481Gradual Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031 Taiwan
| | - Bing-Chang Chen
- grid.412896.00000 0000 9337 0481School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031 Taiwan ,grid.412896.00000 0000 9337 0481Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
33
|
Kim HY, Kim TR, Kim SH, Kim IH, Lim JO, Park JH, Yun S, Lee IC, Park HO, Kim JC. Four-Week Repeated Intravenous Dose Toxicity of Self-Assembled-Micelle Inhibitory RNA-Targeting Amphiregulin in Mice. Int J Toxicol 2021; 40:453-465. [PMID: 34286615 DOI: 10.1177/10915818211031241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The present study investigated the potential subchronic toxicity of self-assembled-micelle inhibitory RNA-targeting amphiregulin (SAMiRNA-AREG) in mice. The test reagent was administered once-daily by intravenous injection for 4 weeks at 0, 100, 200, or 300 mg/kg/day doses. Additional recovery groups (vehicle control and high dose groups) were observed for a 2-week recovery period. During the test period, mortality, clinical signs, body weight, food consumption, ophthalmology, urinalysis, hematology, serum biochemistry, gross pathology, organ weight, and histopathology were examined. An increase in the percentages of basophil and large unstained cells was observed in the 200 and 300 mg/kg/day groups of both sexes. In addition, the absolute and relative weights of the spleen were higher in males given 300 mg/kg/day relative to the concurrent controls. However, these findings were considered of no toxicological significance because the changes were minimal, were not accompanied by other relevant results (eg, correlating microscopic changes), and were not observed at the end of the 2-week recovery period indicating recovery of the findings. Based on the results, SAMiRNA-AREG did not cause treatment-related adverse effects at dose levels of up to 300 mg/kg/day in mice after 4-week repeated intravenous doses. Under these conditions, the no-observed-adverse-effect level of the SAMiRNA-AREG was ≥300 mg/kg/day in both sexes and no target organs were identified.
Collapse
Affiliation(s)
- Hyeon-Young Kim
- Jeonbuk Branch Institute, 443298Korea Institute of Toxicology, Jeongeup, Republic of Korea
- College of Veterinary Medicine, 34931Chonnam National University, Gwangju, Republic of Korea
| | - Tae Rim Kim
- 65404siRNAgen Therapeutics and Bioneer Corporation, Daejeon, Republic of Korea
| | - Sung-Hwan Kim
- Jeonbuk Branch Institute, 443298Korea Institute of Toxicology, Jeongeup, Republic of Korea
| | - In-Hyeon Kim
- Jeonbuk Branch Institute, 443298Korea Institute of Toxicology, Jeongeup, Republic of Korea
- College of Veterinary Medicine, 34931Chonnam National University, Gwangju, Republic of Korea
| | - Je-Oh Lim
- College of Veterinary Medicine, 34931Chonnam National University, Gwangju, Republic of Korea
| | - Jun Hong Park
- 65404siRNAgen Therapeutics and Bioneer Corporation, Daejeon, Republic of Korea
| | - Sungil Yun
- 65404siRNAgen Therapeutics and Bioneer Corporation, Daejeon, Republic of Korea
| | - In-Chul Lee
- Functional Biomaterial Research Center, 54679Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Han-Oh Park
- 65404siRNAgen Therapeutics and Bioneer Corporation, Daejeon, Republic of Korea
| | - Jong-Choon Kim
- College of Veterinary Medicine, 34931Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
34
|
Serum Amphiregulin and Heparin-Binding Epidermal Growth Factor as Biomarkers in Patients with Idiopathic Inflammatory Myopathy. J Clin Med 2021; 10:jcm10163730. [PMID: 34442026 PMCID: PMC8396848 DOI: 10.3390/jcm10163730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 01/27/2023] Open
Abstract
Background. The epidermal growth factors amphiregulin (AREG) and heparin-binding epidermal growth factor (HB-EGF) are implicated in the pathogenesis of several autoimmune diseases, but their clinical and pathological roles in idiopathic inflammatory myopathy (IIM) are unclear. Methods. Serum AREG and HB-EGF levels were measured by ELISA in patients with IIM (n = 37), systemic sclerosis (n = 17), and rheumatoid arthritis (n = 10), and for seven age- and sex-matched healthy controls (HCs). Associations between serum AREG or HB-EGF levels and the clinical parameters were analyzed. Results. Serum AREG levels in IIM patients were significantly elevated compared to those in HCs (median, 20.7 and 10.7 pg/mL, respectively; p = 0.025). In particular, serum AREG levels in IIM patients with interstitial lung disease (ILD) were higher than those of HCs (22.4 pg/mL, p = 0.027). The disease duration in patients with elevated serum AREG levels was significantly shorter compared to those who had normal serum AREG levels (7 and 21 months, respectively; p = 0.0012). Serum HB-EGF levels were significantly increased in IIM patients with elevated CK levels (136.2 pg/mL; p = 0.020) and patients with anti-Mi-2 antibody (183.7 pg/mL; p = 0.045) compared to those in HCs (74.9 pg/mL). Conclusion. These results suggested that AREG could be a promising biomarker associated with early-phase IIM-related ILD, and that HB-EGF expression was associated with muscle injury and regeneration in IIM.
Collapse
|
35
|
Stancil IT, Michalski JE, Davis-Hall D, Chu HW, Park JA, Magin CM, Yang IV, Smith BJ, Dobrinskikh E, Schwartz DA. Pulmonary fibrosis distal airway epithelia are dynamically and structurally dysfunctional. Nat Commun 2021; 12:4566. [PMID: 34315881 PMCID: PMC8316442 DOI: 10.1038/s41467-021-24853-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 07/06/2021] [Indexed: 01/06/2023] Open
Abstract
The airway epithelium serves as the interface between the host and external environment. In many chronic lung diseases, the airway is the site of substantial remodeling after injury. While, idiopathic pulmonary fibrosis (IPF) has traditionally been considered a disease of the alveolus and lung matrix, the dominant environmental (cigarette smoking) and genetic (gain of function MUC5B promoter variant) risk factor primarily affect the distal airway epithelium. Moreover, airway-specific pathogenic features of IPF include bronchiolization of the distal airspace with abnormal airway cell-types and honeycomb cystic terminal airway-like structures with concurrent loss of terminal bronchioles in regions of minimal fibrosis. However, the pathogenic role of the airway epithelium in IPF is unknown. Combining biophysical, genetic, and signaling analyses of primary airway epithelial cells, we demonstrate that healthy and IPF airway epithelia are biophysically distinct, identifying pathologic activation of the ERBB-YAP axis as a specific and modifiable driver of prolongation of the unjammed-to-jammed transition in IPF epithelia. Furthermore, we demonstrate that this biophysical state and signaling axis correlates with epithelial-driven activation of the underlying mesenchyme. Our data illustrate the active mechanisms regulating airway epithelial-driven fibrosis and identify targets to modulate disease progression.
Collapse
Affiliation(s)
- Ian T Stancil
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jacob E Michalski
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Duncan Davis-Hall
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, USA
| | - Hong Wei Chu
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Jin-Ah Park
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Chelsea M Magin
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ivana V Yang
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Bradford J Smith
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Division of Pediatric Pulmonary and Sleep Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Evgenia Dobrinskikh
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David A Schwartz
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
36
|
Ye Z, Hu Y. TGF‑β1: Gentlemanly orchestrator in idiopathic pulmonary fibrosis (Review). Int J Mol Med 2021; 48:132. [PMID: 34013369 PMCID: PMC8136122 DOI: 10.3892/ijmm.2021.4965] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/29/2021] [Indexed: 01/09/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a worldwide disease characterized by the chronic and irreversible decline of lung function. Currently, there is no drug to successfully treat the disease except for lung transplantation. Numerous studies have been devoted to the study of the fibrotic process of IPF and findings showed that transforming growth factor‑β1 (TGF‑β1) plays a central role in the development of IPF. TGF‑β1 promotes the fibrotic process of IPF through various signaling pathways, including the Smad, MAPK, and ERK signaling pathways. There are intersections between these signaling pathways, which provide new targets for researchers to study new drugs. In addition, TGF‑β1 can affect the fibrosis process of IPF by affecting oxidative stress, epigenetics and other aspects. Most of the processes involved in TGF‑β1 promote IPF, but TGF‑β1 can also inhibit it. This review discusses the role of TGF‑β1 in IPF.
Collapse
Affiliation(s)
- Zhimin Ye
- Department of Pathology, Basic Medical School, Central South University, Changsha, Hunan 410006, P.R. China
| | - Yongbin Hu
- Department of Pathology, Basic Medical School, Central South University, Changsha, Hunan 410006, P.R. China
| |
Collapse
|
37
|
Purcaru OS, Artene SA, Barcan E, Silosi CA, Stanciu I, Danoiu S, Tudorache S, Tataranu LG, Dricu A. The Interference between SARS-CoV-2 and Tyrosine Kinase Receptor Signaling in Cancer. Int J Mol Sci 2021; 22:4830. [PMID: 34063231 PMCID: PMC8124491 DOI: 10.3390/ijms22094830] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 01/08/2023] Open
Abstract
Cancer and viruses have a long history that has evolved over many decades. Much information about the interplay between viruses and cell proliferation and metabolism has come from the history of clinical cases of patients infected with virus-induced cancer. In addition, information from viruses used to treat some types of cancer is valuable. Now, since the global coronavirus pandemic erupted almost a year ago, the scientific community has invested countless time and resources to slow down the infection rate and diminish the number of casualties produced by this highly infectious pathogen. A large percentage of cancer cases diagnosed are strongly related to dysregulations of the tyrosine kinase receptor (TKR) family and its downstream signaling pathways. As such, many therapeutic agents have been developed to strategically target these structures in order to hinder certain mechanisms pertaining to the phenotypic characteristics of cancer cells such as division, invasion or metastatic potential. Interestingly, several authors have pointed out that a correlation between coronaviruses such as the SARS-CoV-1 and -2 or MERS viruses and dysregulations of signaling pathways activated by TKRs can be established. This information may help to accelerate the repurposing of clinically developed anti-TKR cancer drugs in COVID-19 management. Because the need for treatment is critical, drug repurposing may be an advantageous choice in the search for new and efficient therapeutic compounds. This approach would be advantageous from a financial point of view as well, given that the resources used for research and development would no longer be required and can be potentially redirected towards other key projects. This review aims to provide an overview of how SARS-CoV-2 interacts with different TKRs and their respective downstream signaling pathway and how several therapeutic agents targeted against these receptors can interfere with the viral infection. Additionally, this review aims to identify if SARS-CoV-2 can be repurposed to be a potential viral vector against different cancer types.
Collapse
Affiliation(s)
- Oana-Stefana Purcaru
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 710204 Craiova, Romania; (O.-S.P.); (S.-A.A.); (E.B.); (A.D.)
| | - Stefan-Alexandru Artene
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 710204 Craiova, Romania; (O.-S.P.); (S.-A.A.); (E.B.); (A.D.)
| | - Edmond Barcan
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 710204 Craiova, Romania; (O.-S.P.); (S.-A.A.); (E.B.); (A.D.)
| | - Cristian Adrian Silosi
- Department of Surgery, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 710204 Craiova, Romania;
| | - Ilona Stanciu
- “Victor Babeş” Clinical Hospital of Infectious Diseases and Pneumophtisiology, Craiova, Str. Calea Bucuresti, nr. 126, 200525 Craiova, Romania;
| | - Suzana Danoiu
- Department of Physiopathology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 710204 Craiova, Romania;
| | - Stefania Tudorache
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy Craiova, 710204 Craiova, Romania;
| | - Ligia Gabriela Tataranu
- Department of Neurosurgery, “Bagdasar-Arseni” Emergency Hospital, Soseaua Berceni 12, 041915 Bucharest, Romania
| | - Anica Dricu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 710204 Craiova, Romania; (O.-S.P.); (S.-A.A.); (E.B.); (A.D.)
| |
Collapse
|
38
|
Buechler MB, Fu W, Turley SJ. Fibroblast-macrophage reciprocal interactions in health, fibrosis, and cancer. Immunity 2021; 54:903-915. [PMID: 33979587 DOI: 10.1016/j.immuni.2021.04.021] [Citation(s) in RCA: 176] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023]
Abstract
Fibroblasts and macrophages are present in all tissues, and mounting evidence supports that these cells engage in direct communication to influence the overall tissue microenvironment and affect disease outcomes. Here, we review the current understanding of the molecular mechanisms that underlie fibroblast-macrophage interactions in health, fibrosis, and cancer. We present an integrated view of fibroblast-macrophage interactions that is centered on the CSF1-CSF1R axis and discuss how additional molecular programs linking these cell types can underpin disease onset, progression, and resolution. These programs may be tissue and context dependent, affected also by macrophage and fibroblast origin and state, as seen most clearly in cancer. Continued efforts to understand these cells and the means by which they interact may provide therapeutic approaches for the treatment of fibrosis and cancer.
Collapse
Affiliation(s)
- Matthew B Buechler
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA.
| | - Wenxian Fu
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA.
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA.
| |
Collapse
|
39
|
Mucke HA. Patent highlights December 2020-January 2021. Pharm Pat Anal 2021; 10:103-110. [PMID: 34003025 DOI: 10.4155/ppa-2021-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/23/2021] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research and development.
Collapse
|
40
|
Kim HY, Kim TR, Kim SH, Kim IH, Ko Y, Yun S, Lee IC, Park HO, Kim JC. Genotoxicity evaluation of self-assembled-micelle inhibitory RNA-targeting amphiregulin (SAMiRNA-AREG), a novel siRNA nanoparticle for the treatment of fibrotic disease. Drug Chem Toxicol 2021; 45:2109-2115. [PMID: 33906534 DOI: 10.1080/01480545.2021.1908003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The self-assembled-micelle inhibitory RNA-targeting amphiregulin (SAMiRNA-AREG) is a novel small-interfering RNA (siRNA) nanoparticle that is used for treatment of pulmonary fibrosis. We investigated the potential genotoxicity of SAMiRNA-AREG based on the guidelines published by the Organization for Economic Cooperation and Development. In the bacterial reverse mutation assay (Ames test), SAMiRNA-AREG did not induce mutations in Salmonella typhimurium TA100, TA1535, TA98, and TA1537 and Escherichia coli WP2uvrA at concentrations of up to 3000 μg/plate with or without metabolic activation. The SAMiRNA-AREG (concentrations up to 500 μg/mL) did not induce chromosomal aberrations in cultured Chinese hamster lung cells with or without metabolic activation. In the in vivo mouse bone marrow micronucleus assay, the SAMiRNA-AREG (concentrations up to 300 mg/kg body weight) did not affect the proportions of polychromatic erythrocytes and total erythrocytes, nor did it increase the number of micronucleated polychromatic erythrocytes in ICR mice. Collectively, these results suggest that SAMiRNA-AREG is safe with regard to genotoxicity such as mutagenesis or clastogenesis under the present experimental conditions. These results might support the safety of SAMiRNA-AREG as a potential therapeutic agent for pharmaceutical development.
Collapse
Affiliation(s)
- Hyeon-Young Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, Republic of Korea.,College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Tae Rim Kim
- siRNAgen therapeutics and Bioneer Corporation, Daejeon, Republic of Korea
| | - Sung-Hwan Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, Republic of Korea
| | - In-Hyeon Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, Republic of Korea.,College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Youngho Ko
- siRNAgen therapeutics and Bioneer Corporation, Daejeon, Republic of Korea
| | - Sungil Yun
- siRNAgen therapeutics and Bioneer Corporation, Daejeon, Republic of Korea
| | - In-Chul Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Han-Oh Park
- siRNAgen therapeutics and Bioneer Corporation, Daejeon, Republic of Korea
| | - Jong-Choon Kim
- College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
41
|
Kim TR, Kim HY, Kim IH, Kim KC, Ko Y, Park JH, Yun S, Lee IC, Kim SH, Park HO. Safety pharmacology of self-assembled-micelle inhibitory RNA-targeting amphiregulin (SAMiRNA-AREG), a novel siRNA nanoparticle platform. Toxicol Rep 2021; 8:839-845. [PMID: 33912399 PMCID: PMC8065257 DOI: 10.1016/j.toxrep.2021.03.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 01/07/2023] Open
Abstract
The present safety pharmacology core battery studies (neurobehavior, respiratory, cardiovascular system, and human ether a-go-go (hERG) channel current) investigated the potential harmful effects of self-assembled-micelle inhibitory RNA-targeting amphiregulin (SAMiRNA-AREG). The SAMiRNA-AREG was administered by single intravenous injection at up to 300 mg/kg and 100 mg/kg in mice and monkeys, respectively. The hERG assay was performed in Chinese hamster ovary (CHO) cells at SAMiRNA-AREG concentrations of up to 200 μg/mL. In the evaluation on neurobehavior, a transient decrease in body temperature was found at 0.5 h (30 min) post-dose at both sexes in mice, with a single 300 mg/kg dose of SAMiRNA-AREG. However, these effects had returned to normal at 1 h post-dose. In the evaluation on hERG channel current, there were statistically significant differences in the inhibition of peak hERG potassium channel current between the 20, 100, and 200 μg/mL SAMiRNA-AREG treatment groups and the vehicle control group. However, these effects were less potent than that of E-4031, a positive control article. For the respiratory and cardiovascular systems, no treatment-related changes were observed in mice or monkeys. Thus, under these experimental conditions, these studies suggest that SAMiRNA-AREG showed no adverse effects on the neurobehavior, respiratory, and cardiovascular function.
Collapse
Affiliation(s)
- Tae Rim Kim
- siRNAgen Therapeutics and Bioneer Corporation, Daejeon, 34302, Republic of Korea
| | - Hyeon-Young Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - In-Hyeon Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Ki Cheon Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Youngho Ko
- siRNAgen Therapeutics and Bioneer Corporation, Daejeon, 34302, Republic of Korea
| | - Jun Hong Park
- siRNAgen Therapeutics and Bioneer Corporation, Daejeon, 34302, Republic of Korea
| | - Sungil Yun
- siRNAgen Therapeutics and Bioneer Corporation, Daejeon, 34302, Republic of Korea
| | - In-Chul Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea
| | - Sung-Hwan Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Han-Oh Park
- siRNAgen Therapeutics and Bioneer Corporation, Daejeon, 34302, Republic of Korea
| |
Collapse
|
42
|
Pathophysiology of Lung Disease and Wound Repair in Cystic Fibrosis. PATHOPHYSIOLOGY 2021; 28:155-188. [PMID: 35366275 PMCID: PMC8830450 DOI: 10.3390/pathophysiology28010011] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive, life-threatening condition affecting many organs and tissues, the lung disease being the chief cause of morbidity and mortality. Mutations affecting the CF Transmembrane Conductance Regulator (CFTR) gene determine the expression of a dysfunctional protein that, in turn, triggers a pathophysiological cascade, leading to airway epithelium injury and remodeling. In vitro and in vivo studies point to a dysregulated regeneration and wound repair in CF airways, to be traced back to epithelial CFTR lack/dysfunction. Subsequent altered ion/fluid fluxes and/or signaling result in reduced cell migration and proliferation. Furthermore, the epithelial-mesenchymal transition appears to be partially triggered in CF, contributing to wound closure alteration. Finally, we pose our attention to diverse approaches to tackle this defect, discussing the therapeutic role of protease inhibitors, CFTR modulators and mesenchymal stem cells. Although the pathophysiology of wound repair in CF has been disclosed in some mechanisms, further studies are warranted to understand the cellular and molecular events in more details and to better address therapeutic interventions.
Collapse
|
43
|
Son SS, Hwang S, Park JH, Ko Y, Yun SI, Lee JH, Son B, Kim TR, Park HO, Lee EY. In vivo silencing of amphiregulin by a novel effective Self-Assembled-Micelle inhibitory RNA ameliorates renal fibrosis via inhibition of EGFR signals. Sci Rep 2021; 11:2191. [PMID: 33500443 PMCID: PMC7838194 DOI: 10.1038/s41598-021-81726-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
Amphiregulin (AREG) is a transmembrane glycoprotein recently implicated in kidney fibrosis. Previously, we reported that the AREG-targeting Self-Assembled-Micelle inhibitory RNA (SAMiRNA-AREG) alleviated fibrosis by stably silencing the AREG gene, and reduced the side effects of conventional siRNA treatment of pulmonary fibrosis. However, the therapeutic effect of SAMiRNA-AREG in renal fibrosis has not been studied until now. We used two animal models of renal fibrosis generated by a unilateral ureteral obstruction (UUO) and an adenine diet (AD) to investigate whether SAMiRNA-AREG inhibited renal fibrosis. To investigate the delivery of SAMiRNA-AREG to the kidney, Cy5-labeled SAMiRNA-AREG was injected into UUO- and AD-induced renal fibrosis models. In both kidney disease models, SAMiRNA-AREG was delivered primarily to the damaged kidney. We also confirmed the protective effect of SAMiRNA-AREG in renal fibrosis models. SAMiRNA-AREG markedly decreased the UUO- and AD-induced AREG mRNA expression. Furthermore, the mRNA expression of fibrosis markers, including α-smooth muscle actin, fibronectin, α1(I) collagen, and α1(III) collagen in the UUO and AD-induced kidneys, was diminished in the SAMiRNA-AREG-treated mice. The transcription of inflammatory markers (tumor necrosis factor-α and monocyte chemoattractant protein-1) and adhesion markers (vascular cell adhesion molecule 1 and intercellular adhesion molecule 1) was attenuated. The hematoxylin and eosin, Masson's trichrome, and immunohistochemical staining results showed that SAMiRNA-AREG decreased renal fibrosis, AREG expression, and epidermal growth factor receptor (EGFR) phosphorylation in the UUO- and AD-induced models. Moreover, we studied the effects of SAMiRNA-AREG in response to TGF-β1 in mouse and human proximal tubule cells, and mouse fibroblasts. TGF-β1-induced extracellular matrix production and myofibroblast differentiation were attenuated by SAMiRNA-AREG. Finally, we confirmed that upregulated AREG in the UUO or AD models was mainly localized in the distal tubules. In conclusion, SAMiRNA-AREG represents a novel siRNA therapeutic for renal fibrosis by suppressing EGFR signals.
Collapse
Affiliation(s)
- Seung Seob Son
- siRNAgen Therapeutics, Daejeon, 34302, Republic of Korea
| | - Soohyun Hwang
- siRNAgen Therapeutics, Daejeon, 34302, Republic of Korea
| | - Jun Hong Park
- siRNAgen Therapeutics, Daejeon, 34302, Republic of Korea
| | - Youngho Ko
- siRNAgen Therapeutics, Daejeon, 34302, Republic of Korea
| | - Sung-Il Yun
- Bioneer Corporation, 8-11 Munpyeongseo-ro, Daedeok-gu, Daejeon, 34302, Republic of Korea
| | - Ji-Hye Lee
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Cheonan, 31151, Republic of Korea
| | - Beomseok Son
- siRNAgen Therapeutics, Daejeon, 34302, Republic of Korea
| | - Tae Rim Kim
- siRNAgen Therapeutics, Daejeon, 34302, Republic of Korea
| | - Han-Oh Park
- siRNAgen Therapeutics, Daejeon, 34302, Republic of Korea.
- Bioneer Corporation, 8-11 Munpyeongseo-ro, Daedeok-gu, Daejeon, 34302, Republic of Korea.
| | - Eun Young Lee
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, 31 Soonchunhyang 6-gil, Cheonan, 31151, Republic of Korea.
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
- BK21 FOUR Project, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
| |
Collapse
|
44
|
Weisberg E, Parent A, Yang PL, Sattler M, Liu Q, Liu Q, Wang J, Meng C, Buhrlage SJ, Gray N, Griffin JD. Repurposing of Kinase Inhibitors for Treatment of COVID-19. Pharm Res 2020; 37:167. [PMID: 32778962 PMCID: PMC7417114 DOI: 10.1007/s11095-020-02851-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022]
Abstract
The outbreak of COVID-19, the pandemic disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spurred an intense search for treatments by the scientific community. In the absence of a vaccine, the goal is to target the viral life cycle and alleviate the lung-damaging symptoms of infection, which can be life-threatening. There are numerous protein kinases associated with these processes that can be inhibited by FDA-approved drugs, the repurposing of which presents an alluring option as they have been thoroughly vetted for safety and are more readily available for treatment of patients and testing in clinical trials. Here, we characterize more than 30 approved kinase inhibitors in terms of their antiviral potential, due to their measured potency against key kinases required for viral entry, metabolism, or reproduction. We also highlight inhibitors with potential to reverse pulmonary insufficiency because of their anti-inflammatory activity, cytokine suppression, or antifibrotic activity. Certain agents are projected to be dual-purpose drugs in terms of antiviral activity and alleviation of disease symptoms, however drug combination is also an option for inhibitors with optimal pharmacokinetic properties that allow safe and efficacious co-administration with other drugs, such as antiviral agents, IL-6 blocking agents, or other kinase inhibitors.
Collapse
Affiliation(s)
- Ellen Weisberg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. .,Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Alexander Parent
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Priscilla L Yang
- Department of Cancer Cell Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA.,Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Qingsong Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Qingwang Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Jinhua Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Chengcheng Meng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sara J Buhrlage
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Nathanael Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - James D Griffin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
45
|
Elbadawy M, Yamanaka M, Goto Y, Hayashi K, Tsunedomi R, Hazama S, Nagano H, Yoshida T, Shibutani M, Ichikawa R, Nakahara J, Omatsu T, Mizutani T, Katayama Y, Shinohara Y, Abugomaa A, Kaneda M, Yamawaki H, Usui T, Sasaki K. Efficacy of primary liver organoid culture from different stages of non-alcoholic steatohepatitis (NASH) mouse model. Biomaterials 2020; 237:119823. [PMID: 32044522 DOI: 10.1016/j.biomaterials.2020.119823] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/21/2020] [Accepted: 01/24/2020] [Indexed: 12/12/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) is associated with liver fibrosis and cirrhosis, which eventually leads to hepatocellular carcinoma. Although several animal models were developed to understand the mechanisms of NASH pathogenesis and progression, it remains obscure. A 3D organoid culture system can recapitulate organ structures and maintain gene expression profiles of original tissues. We therefore tried to generate liver organoids from different degrees [defined as mild (NASH A), moderate (NASH B) and severe (NASH C)] of methionine- and choline-deficient diet-induced NASH model mice and analyzed the difference of their architecture, cell components, organoid-forming efficacy, and gene expression profiles. Organoids from each stage of NASH model mice were successfully generated. Interestingly, epithelial-mesenchymal transition was observed in NASH C organoids. Expression of Collagen I and an activated hepatic stellite cell marker, α-sma was upregulated in the liver organoids from NASH B and C mice. The analysis of RNA sequencing revealed that several novel genes were upregulated in all NASH liver organoids. These results suggest that our generated liver organoids from different stages of NASH diseased mice might become a useful tool for in vitro studies of the molecular mechanism of NASH development and also for identifying novel biomarkers for early diagnosis of NASH disease.
Collapse
Affiliation(s)
- Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan; Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, 13736, Moshtohor, Toukh, Elqaliobiya, Egypt
| | - Megumi Yamanaka
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Yuta Goto
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Kimika Hayashi
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Ryouichi Tsunedomi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shoichi Hazama
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan; Department of Translational Research and Developmental Therapeutics Against Cancer, Yamaguchi University School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Ryo Ichikawa
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Junta Nakahara
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Tsutomu Omatsu
- Research and Education Center for Prevention of Global Infectious Disease of Animals, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Tetsuya Mizutani
- Research and Education Center for Prevention of Global Infectious Disease of Animals, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Yukie Katayama
- Research and Education Center for Prevention of Global Infectious Disease of Animals, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Yuta Shinohara
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan; Pet Health & Food Division, Iskara Industry CO., LTD, 1-14-2, Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Amira Abugomaa
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, 35-1, Higashi 23 Ban-cho, Towada, Aomori, 034-8628, Japan
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan.
| | - Kazuaki Sasaki
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| |
Collapse
|
46
|
Raggi F, Cangelosi D, Becherini P, Blengio F, Morini M, Acquaviva M, Belli ML, Panizzon G, Cervo G, Varesio L, Eva A, Bosco MC. Transcriptome analysis defines myocardium gene signatures in children with ToF and ASD and reveals disease-specific molecular reprogramming in response to surgery with cardiopulmonary bypass. J Transl Med 2020; 18:21. [PMID: 31924244 PMCID: PMC6954611 DOI: 10.1186/s12967-020-02210-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 01/03/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Tetralogy of Fallot (ToF) and Atrial Septal Defects (ASD) are the most common types of congenital heart diseases and a major cause of childhood morbidity and mortality. Cardiopulmonary bypass (CPB) is used during corrective cardiac surgery to support circulation and heart stabilization. However, this procedure triggers systemic inflammatory and stress response and consequent increased risk of postoperative complications. The aim of this study was to define the molecular bases of ToF and ASD pathogenesis and response to CPB and identify new potential biomarkers. METHODS Comparative transcriptome analysis of right atrium specimens collected from 10 ToF and 10 ASD patients was conducted before (Pre-CPB) and after (Post-CPB) corrective surgery. Total RNA isolated from each sample was individually hybridized on Affymetrix HG-U133 Plus Array Strips containing 38,500 unique human genes. Differences in the gene expression profiles and functional enrichment/network analyses were assessed using bioinformatic tools. qRT-PCR analysis was used to validate gene modulation. RESULTS Pre-CPB samples showed significant differential expression of a total of 72 genes, 28 of which were overexpressed in ToF and 44 in ASD. According to Gene Ontology annotation, the mostly enriched biological processes were represented by matrix organization and cell adhesion in ToF and by muscle development and contractility in ASD specimens. GSEA highlighted the specific enrichment of hypoxia gene sets in ToF samples, pointing to a role for hypoxia in disease pathogenesis. The post-CPB myocardium exhibited significant alterations in the expression profile of genes related to transcription regulation, growth/apoptosis, inflammation, adhesion/matrix organization, and oxidative stress. Among them, only 70 were common to the two disease groups, whereas 110 and 24 were unique in ToF and ASD, respectively. Multiple functional interactions among differentially expressed gene products were predicted by network analysis. Interestingly, gene expression changes in ASD samples followed a consensus hypoxia profile. CONCLUSION Our results provide a comprehensive view of gene reprogramming in right atrium tissues of ToF and ASD patients before and after CPB, defining specific molecular pathways underlying disease pathophysiology and myocardium response to CPB. These findings have potential translational value because they identify new candidate prognostic markers and targets for tailored cardioprotective post-surgical therapies.
Collapse
Affiliation(s)
- Federica Raggi
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Davide Cangelosi
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Pamela Becherini
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.,Department of Internal Medicine, University of Genova, Genova, Italy
| | - Fabiola Blengio
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.,INSERM U955 Equipe 16, Creteil, France
| | - Martina Morini
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Massimo Acquaviva
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.,Immunobiology of Neurological Disorders Unit, Institute of Experimental Neurology INSPE, Ospedale San Raffaele, Milano, Italy
| | - Maria Luisa Belli
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.,Cytomorphology Laboratory, Heamo-Onco-TMO Department, IRCSS Istituto Giannina Gaslini, Genova, Italy
| | - Giuseppe Panizzon
- Department of Cardiology, IRCSS Istituto Giannina Gaslini, Genova, Italy
| | - Giuseppe Cervo
- Department of Cardiology, IRCSS Istituto Giannina Gaslini, Genova, Italy
| | - Luigi Varesio
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Alessandra Eva
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Maria Carla Bosco
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.
| |
Collapse
|
47
|
Sun B, Liu X, Zheng X, Wang C, Meng Q, Sun H, Shu X, Liu K, Sun X, Li Y, Ma X. Novel Pyrimidines as Multitarget Protein Tyrosine Kinase Inhibitors for the Treatment of Idiopathic Pulmonary Fibrosis (IPF). ChemMedChem 2019; 15:182-187. [PMID: 31755225 DOI: 10.1002/cmdc.201900606] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/17/2019] [Indexed: 12/11/2022]
Abstract
A new class of pyrimidine derivatives were identified as potent protein tyrosine kinase (PTK) inhibitors for the treatment of idiopathic pulmonary fibrosis (IPF). Most of these small-molecule inhibitors displayed strong enzymatic activity against BTK and JAK3 kinases at concentrations lower than 10 nM. The representative compound N-(3-((5-chloro-2-(4-((1-morpholino)acetylamino)phenylamino)-4-pyrimidinyl)amino)phenyl)acrylamide (6 a) also exhibited high inhibitory potency toward both BTK and JAK kinase families, as well as ErbB4, at a concentration of 10 nM, achieving rates of inhibition higher than 57 %. Additionally, in vivo biological evaluations showed that 6 a can remarkably decrease the severity of IPF disease. All these investigations suggested that the multi-PTK inhibitor 6 a may serve as a promising agent for the treatment of IPF.
Collapse
Affiliation(s)
- Bo Sun
- Department Institute of Respiratory Diseases, Department of Hematology, No. 222 Zhongshan Road, Dalian, 116022, China
| | - Xiaowen Liu
- College of Pharmacy, College of Basic Medical Science, No. 9, West section of Lvshun South Road, Dalian, Liaoning Provence, 116044, China
| | - Xu Zheng
- College of Pharmacy, College of Basic Medical Science, No. 9, West section of Lvshun South Road, Dalian, Liaoning Provence, 116044, China
| | - Changyuan Wang
- College of Pharmacy, College of Basic Medical Science, No. 9, West section of Lvshun South Road, Dalian, Liaoning Provence, 116044, China
| | - Qiang Meng
- College of Pharmacy, College of Basic Medical Science, No. 9, West section of Lvshun South Road, Dalian, Liaoning Provence, 116044, China
| | - Huijun Sun
- College of Pharmacy, College of Basic Medical Science, No. 9, West section of Lvshun South Road, Dalian, Liaoning Provence, 116044, China
| | - Xiaohong Shu
- College of Pharmacy, College of Basic Medical Science, No. 9, West section of Lvshun South Road, Dalian, Liaoning Provence, 116044, China
| | - Kexin Liu
- College of Pharmacy, College of Basic Medical Science, No. 9, West section of Lvshun South Road, Dalian, Liaoning Provence, 116044, China
| | - Xiuli Sun
- Department Institute of Respiratory Diseases, Department of Hematology, No. 222 Zhongshan Road, Dalian, 116022, China
| | - Yanxia Li
- Department Institute of Respiratory Diseases, Department of Hematology, No. 222 Zhongshan Road, Dalian, 116022, China
| | - Xiaodong Ma
- College of Pharmacy, College of Basic Medical Science, No. 9, West section of Lvshun South Road, Dalian, Liaoning Provence, 116044, China
| |
Collapse
|
48
|
Shi Y, Tao M, Wang Y, Zang X, Ma X, Qiu A, Zhuang S, Liu N. Genetic or pharmacologic blockade of enhancer of zeste homolog 2 inhibits the progression of peritoneal fibrosis. J Pathol 2019; 250:79-94. [PMID: 31579944 DOI: 10.1002/path.5352] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/01/2019] [Accepted: 09/24/2019] [Indexed: 02/06/2023]
Abstract
Dysregulation of histone methyltransferase enhancer of zeste homolog 2 (EZH2) has been implicated in the pathogenesis of many cancers. However, the role of EZH2 in peritoneal fibrosis remains unknown. We investigated EZH2 expression in peritoneal dialysis (PD) patients and assessed its role in peritoneal fibrosis in cultured human peritoneal mesothelial cells (HPMCs) and murine models of peritoneal fibrosis induced by chlorhexidine gluconate (CG) or high glucose peritoneal dialysis fluid (PDF) by using 3-deazaneplanocin A (3-DZNeP), and EZH2 conditional knockout mice. An abundance of EZH2 was detected in the peritoneum of patients with PD associated peritonitis and the dialysis effluent of long-term PD patients, which was positively correlated with expression of TGF-β1, vascular endothelial growth factor, and IL-6. EZH2 was found highly expressed in the peritoneum of mice following injury by CG or PDF. In both mouse models, treatment with 3-DZNeP attenuated peritoneal fibrosis and inhibited activation of several profibrotic signaling pathways, including TGF-β1/Smad3, Notch1, epidermal growth factor receptor and Src. EZH2 inhibition also inhibited STAT3 and nuclear factor-κB phosphorylation, and reduced lymphocyte and macrophage infiltration and angiogenesis in the injured peritoneum. 3-DZNeP effectively improved high glucose PDF-associated peritoneal dysfunction by decreasing the dialysate-to-plasma ratio of blood urea nitrogen and increasing the ratio of dialysate glucose at 2 h after PDF injection to initial dialysate glucose. Moreover, delayed administration of 3-DZNeP inhibited peritoneal fibrosis progression, reversed established peritoneal fibrosis and reduced expression of tissue inhibitor of metalloproteinase 2, and matrix metalloproteinase-2 and -9. Finally, EZH2-KO mice exhibited less peritoneal fibrosis than EZH2-WT mice. In HPMCs, treatment with EZH2 siRNA or 3-DZNeP suppressed TGF-β1-induced upregulation of α-SMA and Collagen I and preserved E-cadherin. These results indicate that EZH2 is a key epigenetic regulator that promotes peritoneal fibrosis. Targeting EZH2 may have the potential to prevent and treat peritoneal fibrosis. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yingfeng Shi
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Min Tao
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Yi Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Xiujuan Zang
- Department of Nephrology, Shanghai Songjiang District Central Hospital, Shanghai, PR China
| | - Xiaoyan Ma
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Andong Qiu
- School of Life Science and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai, PR China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China.,Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, USA
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China
| |
Collapse
|
49
|
Kefaloyianni E, Keerthi Raja MR, Schumacher J, Muthu ML, Krishnadoss V, Waikar SS, Herrlich A. Proximal Tubule-Derived Amphiregulin Amplifies and Integrates Profibrotic EGF Receptor Signals in Kidney Fibrosis. J Am Soc Nephrol 2019; 30:2370-2383. [PMID: 31676723 DOI: 10.1681/asn.2019030321] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/15/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Sustained activation of EGF receptor (EGFR) in proximal tubule cells is a hallmark of progressive kidney fibrosis after AKI and in CKD. However, the molecular mechanisms and particular EGFR ligands involved are unknown. METHODS We studied EGFR activation in proximal tubule cells and primary tubular cells isolated from injured kidneys in vitro. To determine in vivo the role of amphiregulin, a low-affinity EGFR ligand that is highly upregulated with injury, we used ischemia-reperfusion injury or unilateral ureteral obstruction in mice with proximal tubule cell-specific knockout of amphiregulin. We also injected soluble amphiregulin into knockout mice with proximal tubule cell-specific deletion of amphiregulin's releasing enzyme, the transmembrane cell-surface metalloprotease, a disintegrin and metalloprotease-17 (ADAM17), and into ADAM17 hypomorphic mice. RESULTS Yes-associated protein 1 (YAP1)-dependent upregulation of amphiregulin transcript and protein amplifies amphiregulin signaling in a positive feedback loop. YAP1 also integrates signals of other moderately injury-upregulated, low-affinity EGFR ligands (epiregulin, epigen, TGFα), which also require soluble amphiregulin and YAP1 to induce sustained EGFR activation in proximal tubule cells in vitro. In vivo, soluble amphiregulin injection sufficed to reverse protection from fibrosis after ischemia-reperfusion injury in ADAM17 hypomorphic mice; injected soluble amphiregulin also reversed the corresponding protective proximal tubule cell phenotype in injured proximal tubule cell-specific ADAM17 knockout mice. Moreover, the finding that proximal tubule cell-specific amphiregulin knockout mice were protected from fibrosis after ischemia-reperfusion injury or unilateral ureteral obstruction demonstrates that amphiregulin was necessary for the development of fibrosis. CONCLUSIONS Our results identify amphiregulin as a key player in injury-induced kidney fibrosis and suggest therapeutic or diagnostic applications of soluble amphiregulin in kidney disease.
Collapse
Affiliation(s)
- Eirini Kefaloyianni
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - Manikanda Raja Keerthi Raja
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - Julian Schumacher
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - Muthu Lakshmi Muthu
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - Vaishali Krishnadoss
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - Sushrut S Waikar
- Renal Division, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts
| | - Andreas Herrlich
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| |
Collapse
|
50
|
Hashimoto T, Satoh T, Karasuyama H, Yokozeki H. Amphiregulin from Basophils Amplifies Basophil-Mediated Chronic Skin Inflammation. J Invest Dermatol 2019; 139:1834-1837.e2. [DOI: 10.1016/j.jid.2019.02.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 02/20/2019] [Accepted: 02/24/2019] [Indexed: 11/16/2022]
|