1
|
Huang C, Huangfu C, Bai Z, Zhu L, Shen P, Wang N, Li G, Deng H, Ma Z, Zhou W, Gao Y. Multifunctional carbomer based ferulic acid hydrogel promotes wound healing in radiation-induced skin injury by inactivating NLRP3 inflammasome. J Nanobiotechnology 2024; 22:576. [PMID: 39300534 PMCID: PMC11411768 DOI: 10.1186/s12951-024-02789-7] [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: 05/29/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Radiation-induced skin injury is a significant adverse reaction to radiotherapy. However, there is a lack of effective prevention and treatment methods for this complication. Ferulic acid (FA) has been identified as an effective anti-radiation agent. Conventional administrations of FA limit the reaching of it on skin. We aimed to develop a novel FA hydrogel to facilitate the use of FA in radiation-induced skin injury. METHODS We cross-linked carbomer 940, a commonly used adjuvant, with FA at concentrations of 5%, 10%, and 15%. Sweep source optical coherence tomography system, a novel skin structure evaluation method, was applied to investigate the influence of FA on radiation-induced skin injury. Calcein-AM/PI staining, CCK8 assay, hemolysis test and scratch test were performed to investigate the biocompatibility of FA hydrogel. The reducibility of DPPH and ABTS radicals by FA hydrogel was also performed. HE staining, Masson staining, laser Doppler blood flow monitor, and OCT imaging system are used to evaluate the degree of skin tissue damage. Potential differentially expressed genes were screened via transcriptome analysis. RESULTS Good biocompatibility and in vitro antioxidant ability of the FA hydrogels were observed. 10% FA hydrogel presented a better mechanical stability than 5% and 15% FA hydrogel. All three concentrations of FA remarkably promoted the recovery of radiation-induced skin injury by reducing inflammation, oxidative conidiation, skin blood flow, and accelerating skin tissue reconstruction, collagen deposition. FA hydrogel greatly inhibiting the levels of NLRP3, caspase-1, IL-18, pro-IL-1β and IL-1β in vivo and vitro levels through restraining the activation of NLRP3 inflammasome. Transcriptome analysis indicated that FA might regulate wound healing via targeting immune response, inflammatory response, cell migration, angiogenesis, hypoxia response, and cell matrix adhesion. CONCLUSIONS These findings suggest that the novel FA hydrogel is a promising therapeutic method for the prevention and treatment of radiation-induced skin injury patients.
Collapse
Affiliation(s)
- Congshu Huang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Beijing, 100850, China
- Department of Traditional Chinese medicine, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Chaoji Huangfu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Beijing, 100850, China
| | - Zhijie Bai
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Beijing, 100850, China
| | - Long Zhu
- Qinghai University, No. 251 Ningda Road, Xining, 810016, China
| | - Pan Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Beijing, 100850, China
| | - Ningning Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Beijing, 100850, China
| | - Gaofu Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Beijing, 100850, China
| | - Huifang Deng
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Beijing, 100850, China
| | - Zengchun Ma
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Beijing, 100850, China.
| | - Wei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Beijing, 100850, China.
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Beijing, 100850, China.
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
2
|
Zheng Y, Gong W, Wu Z, Zhang S, Wang N, Hu Z, Shou Y, Xu T, Shen Y, Li X, Jin L, Cong W, Zhu Z. FGF21 Ameliorates Fibroblasts Activation and Systemic Sclerosis by Inhibiting CK2α/GLI2 Signaling Axis. J Invest Dermatol 2024:S0022-202X(24)02058-X. [PMID: 39182559 DOI: 10.1016/j.jid.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024]
Abstract
Systemic sclerosis is a typical fibrotic disease of unknown etiology that is characterized by abnormal fibroblast activation and excessive deposition of extracellular matrix. Unfortunately, effective therapeutic approaches are lacking. FGF21 plays a key role in mediating a variety of biological activities. However, its specific function in systemic sclerosis is unclear. In this study, we found that the expression of FGF21 was significantly downregulated in fibrotic skin tissue and in TGF-β-stimulated fibroblasts. Furthermore, our studies demonstrated that treatment with recombinant FGF21 in the skin significantly alleviated bleomycin-induced and TBRI-activated fibrosis and inhibited the activation of fibroblasts, whereas skin fibrosis was exacerbated by deletion of FGF21. Mechanistically, FGF21 inhibits the activity of CK2α and promotes the degradation of GLI2. In conclusion, these results indicate that FGF21 attenuates skin fibrosis through the CK2α/GLI2 signaling pathway and therefore may be a potential therapeutic target for systemic sclerosis.
Collapse
Affiliation(s)
- Yeyi Zheng
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China; Ningbo Key Laboratory of Skin Science, Ningbo College of Health Sciences, Ningbo, People's Republic of China
| | - Wenjie Gong
- Department of Pharmacy, Ningbo Women and Children's Hospital, Ningbo, People's Republic of China
| | - Zhaohang Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Siyi Zhang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Nan Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Zhenyu Hu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yanni Shou
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Tianpeng Xu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yingjie Shen
- School of Life Sciences, Huzhou University, Huzhou, People's Republic of China
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China; Ningbo Key Laboratory of Skin Science, Ningbo College of Health Sciences, Ningbo, People's Republic of China
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Zhongxin Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China.
| |
Collapse
|
3
|
Yin H, Staples SCR, Pickering JG. The fundamentals of fibroblast growth factor 9. Differentiation 2024; 139:100731. [PMID: 37783652 DOI: 10.1016/j.diff.2023.09.004] [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: 07/08/2023] [Revised: 09/07/2023] [Accepted: 09/17/2023] [Indexed: 10/04/2023]
Abstract
Fibroblast growth factor 9 (FGF9) was first identified during a screen for factors acting on cells of the central nervous system (CNS). Research over the subsequent two decades has revealed this protein to be a critically important and elegantly regulated growth factor. A hallmark control feature is reciprocal compartmentalization, particularly during development, with epithelium as a dominant source and mesenchyme a prime target. This mesenchyme selectivity is accomplished by the high affinity of FGF9 to the IIIc isoforms of FGFR1, 2, and 3. FGF9 is expressed widely in the embryo, including the developing heart and lungs, and more selectively in the adult, including the CNS and kidneys. Global Fgf9-null mice die shortly after birth due to respiratory failure from hypoplastic lungs. As well, their hearts are dilated and poorly vascularized, the skeleton is small, the intestine is shortened, and male-to-female sex reversal can be found. Conditional Fgf9-null mice have revealed CNS phenotypes, including ataxia and epilepsy. In humans, FGF9 variants have been found to underlie multiple synostoses syndrome 3, a syndrome characterized by multiple joint fusions. Aberrant FGF9 signaling has also been implicated in differences of sex development and cancer, whereas vascular stabilizing effects of FGF9 could benefit chronic diseases. This primer reviews the attributes of this vital growth factor.
Collapse
Affiliation(s)
- Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Sabrina C R Staples
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada; Department of Medical Biophysics, Western University, London, Canada
| | - J Geoffrey Pickering
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada; Department of Medical Biophysics, Western University, London, Canada; Department of Biochemistry, Western University, London, Canada; Department of Medicine, Western University, London, Canada; London Health Sciences Centre, London, Canada.
| |
Collapse
|
4
|
Liang M, Dickel N, Györfi AH, SafakTümerdem B, Li YN, Rigau AR, Liang C, Hong X, Shen L, Matei AE, Trinh-Minh T, Tran-Manh C, Zhou X, Zehender A, Kreuter A, Zou H, Schett G, Kunz M, Distler JHW. Attenuation of fibroblast activation and fibrosis by adropin in systemic sclerosis. Sci Transl Med 2024; 16:eadd6570. [PMID: 38536934 DOI: 10.1126/scitranslmed.add6570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 02/26/2024] [Indexed: 04/05/2024]
Abstract
Fibrotic diseases impose a major socioeconomic challenge on modern societies and have limited treatment options. Adropin, a peptide hormone encoded by the energy homeostasis-associated (ENHO) gene, is implicated in metabolism and vascular homeostasis, but its role in the pathogenesis of fibrosis remains enigmatic. Here, we used machine learning approaches in combination with functional in vitro and in vivo experiments to characterize adropin as a potential regulator involved in fibroblast activation and tissue fibrosis in systemic sclerosis (SSc). We demonstrated consistent down-regulation of adropin/ENHO in skin across multiple cohorts of patients with SSc. The prototypical profibrotic cytokine TGFβ reduced adropin/ENHO expression in a JNK-dependent manner. Restoration of adropin signaling by therapeutic application of bioactive adropin34-76 peptides in turn inhibited TGFβ-induced fibroblast activation and fibrotic tissue remodeling in primary human dermal fibroblasts, three-dimensional full-thickness skin equivalents, mouse models of bleomycin-induced pulmonary fibrosis and sclerodermatous chronic graft-versus-host-disease (sclGvHD), and precision-cut human skin slices. Knockdown of GPR19, an adropin receptor, abrogated the antifibrotic effects of adropin in fibroblasts. RNA-seq demonstrated that the antifibrotic effects of adropin34-76 were functionally linked to deactivation of GLI1-dependent profibrotic transcriptional networks, which was experimentally confirmed in vitro, in vivo, and ex vivo using cultured human dermal fibroblasts, a sclGvHD mouse model, and precision-cut human skin slices. ChIP-seq confirmed adropin34-76-induced changes in TGFβ/GLI1 signaling. Our study characterizes the TGFβ-induced down-regulation of adropin/ENHO expression as a potential pathomechanism of SSc as a prototypical systemic fibrotic disease that unleashes uncontrolled activation of profibrotic GLI1 signaling.
Collapse
Affiliation(s)
- Minrui Liang
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Division of Rheumatology, Huashan Rare Disease Center, Huashan Hospital, Fudan University, 200032 Shanghai, P. R. China
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Nicholas Dickel
- Chair of Medical Informatics, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Andrea-Hermina Györfi
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Bilgesu SafakTümerdem
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Yi-Nan Li
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Aleix Rius Rigau
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Chunguang Liang
- Chair of Medical Informatics, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91058 Erlangen, Germany
- Institute of Immunology, Jena University Hospital, 07743 Jena, Germany
| | - Xuezhi Hong
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Lichong Shen
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
- Division of Rheumatology, Renji Hospital, Shanghai Jiao Tong University, 200001 Shanghai, P. R. China
| | - Alexandru-Emil Matei
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Thuong Trinh-Minh
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Cuong Tran-Manh
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Xiang Zhou
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Ariella Zehender
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Alexander Kreuter
- Department of Dermatology and Allergology, HELIOS Sankt Elisabeth Klinik Oberhausen, 46045 Oberhausen, Nordrhein-Westfalen, Germany
| | - Hejian Zou
- Division of Rheumatology, Huashan Rare Disease Center, Huashan Hospital, Fudan University, 200032 Shanghai, P. R. China
| | - Georg Schett
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Meik Kunz
- Chair of Medical Informatics, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Jörg H W Distler
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University; 40225 Düsseldorf, Germany
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
5
|
Bracken SJ, Suthers AN, DiCioccio RA, Su H, Anand S, Poe JC, Jia W, Visentin J, Basher F, Jordan CZ, McManigle WC, Li Z, Hakim FT, Pavletic SZ, Bhuiya NS, Ho VT, Horwitz ME, Chao NJ, Sarantopoulos S. Heightened TLR7 signaling primes BCR-activated B cells in chronic graft-versus-host disease for effector functions. Blood Adv 2024; 8:667-680. [PMID: 38113462 PMCID: PMC10839617 DOI: 10.1182/bloodadvances.2023010362] [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: 04/03/2023] [Revised: 11/02/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
ABSTRACT Chronic graft-versus-host disease (cGVHD) is a debilitating, autoimmune-like syndrome that can occur after allogeneic hematopoietic stem cell transplantation. Constitutively activated B cells contribute to ongoing alloreactivity and autoreactivity in patients with cGVHD. Excessive tissue damage that occurs after transplantation exposes B cells to nucleic acids in the extracellular environment. Recognition of endogenous nucleic acids within B cells can promote pathogenic B-cell activation. Therefore, we hypothesized that cGVHD B cells aberrantly signal through RNA and DNA sensors such as Toll-like receptor 7 (TLR7) and TLR9. We found that B cells from patients and mice with cGVHD had higher expression of TLR7 than non-cGVHD B cells. Using ex vivo assays, we found that B cells from patients with cGVHD also demonstrated increased interleukin-6 production after TLR7 stimulation with R848. Low-dose B-cell receptor (BCR) stimulation augmented B-cell responses to TLR7 activation. TLR7 hyperresponsiveness in cGVHD B cells correlated with increased expression and activation of the downstream transcription factor interferon regulatory factor 5. Because RNA-containing immune complexes can activate B cells through TLR7, we used a protein microarray to identify RNA-containing antigen targets of potential pathological relevance in cGVHD. We found that many of the unique targets of active cGVHD immunoglobulin G (IgG) were nucleic acid-binding proteins. This unbiased assay identified the autoantigen and known cGVHD target Ro-52, and we found that RNA was required for IgG binding to Ro-52. Herein, we find that BCR-activated B cells have aberrant TLR7 signaling responses that promote potential effector responses in cGVHD.
Collapse
Affiliation(s)
- Sonali J. Bracken
- Division of Rheumatology and Immunology, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Amy N. Suthers
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Rachel A. DiCioccio
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Hsuan Su
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Sarah Anand
- Division of Hematology and Medical Oncology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Jonathan C. Poe
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Wei Jia
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Jonathan Visentin
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
- Department of Immunology and Immunogenetics, Bordeaux University Hospital, Bordeaux, France
- UMR CNRS 5164 ImmunoConcEpT, Bordeaux University, Bordeaux, France
| | - Fahmin Basher
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Collin Z. Jordan
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham NC
| | - William C. McManigle
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham NC
| | - Zhiguo Li
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham NC
- Duke Cancer Institute, Duke University Medical Center, Durham NC
| | - Frances T. Hakim
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, MD
| | - Steven Z. Pavletic
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, MD
| | - Nazmim S. Bhuiya
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, MD
| | - Vincent T. Ho
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Mitchell E. Horwitz
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
- Duke Cancer Institute, Duke University Medical Center, Durham NC
| | - Nelson J. Chao
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
- Duke Cancer Institute, Duke University Medical Center, Durham NC
- Department of Integrated Immunobiology, Duke University School of Medicine, Durham, NC
| | - Stefanie Sarantopoulos
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
- Duke Cancer Institute, Duke University Medical Center, Durham NC
- Department of Integrated Immunobiology, Duke University School of Medicine, Durham, NC
| |
Collapse
|
6
|
Jimenez SA, Piera-Velazquez S. Cellular Transdifferentiation: A Crucial Mechanism of Fibrosis in Systemic Sclerosis. Curr Rheumatol Rev 2024; 20:388-404. [PMID: 37921216 DOI: 10.2174/0115733971261932231025045400] [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: 05/12/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 11/04/2023]
Abstract
Systemic Sclerosis (SSc) is a systemic autoimmune disease of unknown etiology with a highly complex pathogenesis that despite extensive investigation is not completely understood. The clinical and pathologic manifestations of the disease result from three distinct processes: 1) Severe and frequently progressive tissue fibrosis causing exaggerated and deleterious accumulation of interstitial collagens and other extracellular matrix molecules in the skin and various internal organs; 2) extensive fibroproliferative vascular lesions affecting small arteries and arterioles causing tissue ischemic alterations; and 3) cellular and humoral immunity abnormalities with the production of numerous autoantibodies, some with very high specificity for SSc. The fibrotic process in SSc is one of the main causes of disability and high mortality of the disease. Owing to its essentially universal presence and the severity of its clinical effects, the mechanisms involved in the development and progression of tissue fibrosis have been extensively investigated, however, despite intensive investigation, the precise molecular mechanisms have not been fully elucidated. Several recent studies have suggested that cellular transdifferentiation resulting in the phenotypic conversion of various cell types into activated myofibroblasts may be one important mechanism. Here, we review the potential role that cellular transdifferentiation may play in the development of severe and often progressive tissue fibrosis in SSc.
Collapse
Affiliation(s)
- Sergio A Jimenez
- Department of Dermatology and Cutaneous Biology, Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia 19107, USA
| | - Sonsoles Piera-Velazquez
- Department of Dermatology and Cutaneous Biology, Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia 19107, USA
| |
Collapse
|
7
|
Helm M, Schmidt M, Del Duca E, Liu Y, Mortensen LS, Loui J, Zheng Y, Binder H, Guttman-Yassky E, Cotsarelis G, Simon JC, Ferrer RA. Repurposing DPP4 Inhibition to Improve Hair Follicle Activation and Regeneration. J Invest Dermatol 2023; 143:2132-2144.e15. [PMID: 37236597 DOI: 10.1016/j.jid.2023.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023]
Abstract
Skin injury and several diseases elicit fibrosis and induce hair follicle (HF) growth arrest and loss. The resulting alopecia and disfiguration represent a severe burden for patients, both physically and psychologically. Reduction of profibrotic factors such as dipeptidyl peptidase 4 (DPP4) might be a strategy to tackle this issue. We show DPP4 overrepresentation in settings with HF growth arrest (telogen), HF loss, and nonregenerative wound areas in mouse skin and human scalp. Topical DPP4 inhibition with Food and Drug Administration/European Medicines Agency-approved sitagliptin on preclinical models of murine HF activation/regeneration results in accelerated anagen progress, whereas treatment of wounds with sitagliptin results in reduced expression of fibrosis markers, increased induction of anagen around wounds, and HF regeneration in the wound center. These effects are associated with higher expression of Wnt target Lef1, known to be required for HF anagen/HF-activation and regeneration. Sitagliptin treatment decreases profibrotic signaling in the skin, induces a differentiation trajectory of HF cells, and activates Wnt targets related to HF activation/growth but not those supporting fibrosis. Taken together, our study shows a role for DPP4 in HF biology and shows how DPP4 inhibition, currently used as oral medication to treat diabetes, could be repurposed into a topical treatment agent to potentially reverse HF loss in alopecia and after injury.
Collapse
Affiliation(s)
- Maria Helm
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Center, University Leipzig, Leipzig, Germany
| | - Maria Schmidt
- Interdisciplinary Center for Bioinformatics, University Leipzig, Leipzig, Germany
| | - Ester Del Duca
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, Mount Sinai, New York City, New York, USA
| | - Ying Liu
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, Mount Sinai, New York City, New York, USA
| | - Lena Sünke Mortensen
- Interdisciplinary Center for Bioinformatics, University Leipzig, Leipzig, Germany
| | - Juliane Loui
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Center, University Leipzig, Leipzig, Germany
| | - Ying Zheng
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hans Binder
- Interdisciplinary Center for Bioinformatics, University Leipzig, Leipzig, Germany
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, Mount Sinai, New York City, New York, USA
| | - George Cotsarelis
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jan C Simon
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Center, University Leipzig, Leipzig, Germany
| | - Rubén A Ferrer
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Center, University Leipzig, Leipzig, Germany.
| |
Collapse
|
8
|
Tilg H, Adolph TE, Tacke F. Therapeutic modulation of the liver immune microenvironment. Hepatology 2023; 78:1581-1601. [PMID: 37057876 DOI: 10.1097/hep.0000000000000386] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/14/2023] [Indexed: 04/15/2023]
Abstract
Inflammation is a hallmark of progressive liver diseases such as chronic viral or immune-mediated hepatitis, alcohol-associated liver disease, and NAFLD. Preclinical and clinical studies have provided robust evidence that cytokines and related cellular stress sensors in innate and adaptive immunity orchestrate hepatic disease processes. Unresolved inflammation and liver injury result in hepatic scarring, fibrosis, and cirrhosis, which may culminate in HCC. Liver diseases are accompanied by gut dysbiosis and a bloom of pathobionts, fueling hepatic inflammation. Anti-inflammatory strategies are extensively used to treat human immune-mediated conditions beyond the liver, while evidence for immunomodulatory therapies and cell therapy-based strategies in liver diseases is only emerging. The development and establishment of novel immunomodulatory therapies for chronic liver diseases has been dampened by several clinical challenges, such as invasive monitoring of therapeutic efficacy with liver biopsy in clinical trials and risk of DILI in several studies. Such aspects prevented advancements of novel medical therapies for chronic inflammatory liver diseases. New concepts modulating the liver immune environment are studied and eagerly awaited to improve the management of chronic liver diseases in the future.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| |
Collapse
|
9
|
Zhou X, Trinh-Minh T, Matei AE, Györfi AH, Hong X, Bergmann C, Schett G, Atkinson J, Bowcutt R, Patel J, Johnson TS, Distler JHW. Amelioration of Fibrotic Remodeling of Human 3-Dimensional Full-Thickness Skin by Transglutamase 2 Inhibition. Arthritis Rheumatol 2023; 75:1619-1627. [PMID: 37057394 DOI: 10.1002/art.42518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 02/09/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023]
Abstract
OBJECTIVE Fibrotic tissues are characterized by excessive crosslinking between extracellular matrix (ECM) proteins, rendering them more resistant to degradation. Although increased crosslinking of ECM is thought to play an important role for progression of tissue fibrosis, enhanced ECM crosslinking has not yet been targeted therapeutically in systemic sclerosis (SSc). Here, we investigated the role of transglutaminase 2 (TG2), a central crosslinking enzyme, in the activation of SSc fibroblasts. METHODS We assessed TG2 expression and activity using TG2 staining, Western blotting, and TG2 activity assays. We inhibited TG2 in fibroblasts cultured under standard 2-dimensional conditions and in a 3-dimensional full-thickness equivalent skin model using monoclonal inhibitory anti-TG2 antibodies. RESULTS TG2 expression was increased in the skin of patients with SSc compared with healthy controls, with levels particularly high in patients with SSc-associated interstitial lung disease. TG2 expression and TG2 activity were also increased in SSc dermal fibroblasts. Moreover, the levels of circulating TG2 in the plasma samples from SSc patients were increased versus samples from healthy controls. Anti-TG2 antibodies did not show consistent antifibrotic effects across different fibroblast cell lines under 2-dimensional culture conditions; however, anti-TG2 antibodies effectively reduced transforming growth factor β-induced dermal thickening, myofibroblast differentiation, and collagen accumulation in the 3-dimensional full-thickness model of human skin. CONCLUSION We provide the first evidence, to our knowledge, that inhibition of TG2 might be a potential antifibrotic approach in SSc. Our findings have translational potential as anti-TG2 antibodies are currently evaluated in a phase II clinical trial in chronic allograft injury and would thus be available for clinical studies in SSc (ClinicalTrials.gov identifier: NCT04335578).
Collapse
Affiliation(s)
- Xiang Zhou
- Clinic for Rheumatology University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, and Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany
| | - Thuong Trinh-Minh
- Clinic for Rheumatology University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, and Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany
| | - Alexandru-Emil Matei
- Clinic for Rheumatology University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, and Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany
| | - Andrea-Hermina Györfi
- Clinic for Rheumatology University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, and Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany
| | - Xuezhi Hong
- Clinic for Rheumatology University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, and Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany
| | - Christina Bergmann
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, and Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, and Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | | | | | | | | | - Jörg H W Distler
- Clinic for Rheumatology University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, and Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
10
|
Abstract
Systemic sclerosis (SSc) is a chronic immune-mediated disease characterized by microangiopathy, immune dysregulation, and progressive fibrosis of the skin and internal organs. Though not fully understood, the pathogenesis of SSc is dominated by microvascular injury, endothelial dysregulation, and immune response that are thought to be associated with fibroblast activation and related fibrogenesis. Among the main clinical subsets, diffuse SSc (dSSc) is a progressive form with rapid and disseminated skin thickening accompanied by internal organ fibrosis and dysfunction. Despite recent advances and multiple randomized clinical trials in early dSSc patients, an effective disease-modifying treatment for progressive skin fibrosis is still missing, and there is a crucial need to identify new targets for therapeutic intervention. Eotaxin-2 (CCL24) is a chemokine secreted by immune cells and epithelial cells, which promotes trafficking of immune cells and activation of pro-fibrotic cells through CCR3 receptor binding. Higher levels of CCL24 and CCR3 were found in the skin and sera of patients with SSc compared with healthy controls; elevated levels of CCL24 and CCR3 were associated with fibrosis and predictive of greater lung function deterioration. Growing evidence supports the potency of a CCL24-blocking antibody as an anti-inflammatory and anti-fibrotic modulating agent in multiple preclinical models that involve liver, skin, and lung inflammation and fibrosis. This review highlights the role of CCL24 in orchestrating immune, vascular, and fibrotic pathways, and the potential of CCL24 inhibition as a novel treatment for SSc.
Collapse
Affiliation(s)
| | | | - Alexandra Balbir-Gurman
- Rheumatology Institute, Rambam Health Care Campus, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
11
|
Sun C, Bai M, Jia Y, Tian X, Guo Y, Xu X, Guo Z. mRNA sequencing reveals the distinct gene expression and biological functions in cardiac fibroblasts regulated by recombinant fibroblast growth factor 2. PeerJ 2023; 11:e15736. [PMID: 37483983 PMCID: PMC10362857 DOI: 10.7717/peerj.15736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/20/2023] [Indexed: 07/25/2023] Open
Abstract
After myocardial injury, cardiac fibroblasts (CFs) differentiate into myofibroblasts, which express and secrete extracellular matrix (ECM) components for myocardial repair, but also promote myocardial fibrosis. Recombinant fibroblast growth factor 2 (FGF2) protein drug with low molecular weight can promote cell survival and angiogenesis, and it was found that FGF2 could inhibit the activation of CFs, suggesting FGF2 has great potential in myocardial repair. However, the regulatory role of FGF2 on CFs has not been fully elucidated. Here, we found that recombinant FGF2 significantly suppressed the expression of alpha smooth muscle actin (α-SMA) in CFs. Through RNA sequencing, we analyzed mRNA expression in CFs and the differently expressed genes regulated by FGF2, including 430 up-regulated genes and 391 down-regulated genes. Gene ontology analysis revealed that the differentially expressed genes were strongly enriched in multiple biological functions, including ECM organization, cell adhesion, actin filament organization and axon guidance. The results of gene set enrichment analysis (GSEA) show that ECM organization and actin filament organization are down-regulated, while axon guidance is up-regulated. Further cellular experiments indicate that the regulatory functions of FGF2 are consistent with the findings of the gene enrichment analysis. This study provides valuable insights into the potential therapeutic role of FGF2 in treating cardiac fibrosis and establishes a foundation for further research to uncover the underlying mechanisms of CFs gene expression regulated by FGF2.
Collapse
Affiliation(s)
- Changye Sun
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Mengru Bai
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yangyang Jia
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiangqin Tian
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yonglong Guo
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xinhui Xu
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
12
|
Li L, Shapiro RL, Joo MK, Josyula A, Hsueh HT, Gutierrez OB, Halpert G, Akshintala V, Chen H, Curtis S, Better M, Davison C, Hu H, Almario JAN, Steinway SN, Hunt K, Del Sesto RE, Izzi J, Salimian KJ, Ensign LM, Selaru FM. Injectable, Drug-Eluting Nanocrystals Prevent Fibrosis and Stricture Formation In Vivo. Gastroenterology 2023; 164:937-952.e13. [PMID: 36657529 PMCID: PMC10151160 DOI: 10.1053/j.gastro.2023.01.006] [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: 07/08/2022] [Revised: 12/07/2022] [Accepted: 01/10/2023] [Indexed: 01/21/2023]
Abstract
BACKGROUND & AIMS Tissue fibrosis results from uncontrolled healing responses leading to excessive mesenchymal cell activation and collagen and other extracellular matrix deposition. In the gastrointestinal tract, fibrosis leads to narrowing of the lumen and stricture formation. A drug treatment to prevent fibrosis and strictures in the gastrointestinal tract would be transformational for patient care. We aimed to develop a stricture treatment with the following characteristics and components: a small molecule with strong antifibrotic effects that is delivered locally at the site of the stricture to ensure correct lesional targeting while protecting the systemic circulation, and that is formulated with sustained-release properties to act throughout the wound healing processes. METHODS A high-throughput drug screening was performed to identify small molecules with antifibrotic properties. Next, we formulated an antifibrotic small molecule for sustained release and tested its antifibrotic potential in 3 animal models of fibrosis. RESULTS Sulconazole, a US Food and Drug Administration-approved drug for fungal infections, was found to have strong antifibrotic properties. Sulconazole was formulated as sulconazole nanocrystals for sustained release. We found that sulconazole nanocrystals provided superior or equivalent fibrosis prevention with less frequent dosing in mouse models of skin and intestinal tissue fibrosis. In a patient-like swine model of bowel stricture, a single injection of sulconazole nanocrystals prevented stricture formation. CONCLUSIONS The current data lay the foundation for further studies to improve the management of a range of diseases and conditions characterized by tissue fibrosis.
Collapse
Affiliation(s)
- Ling Li
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Rachel L Shapiro
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Min Kyung Joo
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aditya Josyula
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Henry T Hsueh
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Olaya Brewer Gutierrez
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Gilad Halpert
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Venkata Akshintala
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Haiming Chen
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Samuel Curtis
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marina Better
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Charlotte Davison
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Haijie Hu
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Jose Antonio Navarro Almario
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Steven N Steinway
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Kelton Hunt
- Department of Chemistry and Biochemistry, Utah Tech University, St George, Utah
| | - Rico E Del Sesto
- Department of Chemistry and Biochemistry, Utah Tech University, St George, Utah
| | - Jessica Izzi
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, Maryland
| | - Kevan J Salimian
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Laura M Ensign
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Gynecology and Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Medicine, Division of Infectious Diseases, The Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Sidney Kimmel Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Florin M Selaru
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland; Department of Oncology, Sidney Kimmel Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland; The Institute for Nanobiotechnology, The Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
13
|
The Molecular Mechanisms of Systemic Sclerosis-Associated Lung Fibrosis. Int J Mol Sci 2023; 24:ijms24032963. [PMID: 36769282 PMCID: PMC9917655 DOI: 10.3390/ijms24032963] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Systemic sclerosis (SSc), also known as scleroderma, is an autoimmune disorder that affects the connective tissues and has the highest mortality rate among the rheumatic diseases. One of the hallmarks of SSc is fibrosis, which may develop systemically, affecting the skin and virtually any visceral organ in the body. Fibrosis of the lungs leads to interstitial lung disease (ILD), which is currently the leading cause of death in SSc. The identification of effective treatments to stop or reverse lung fibrosis has been the main challenge in reducing SSc mortality and improving patient outcomes and quality of life. Thus, understanding the molecular mechanisms, altered pathways, and their potential interactions in SSc lung fibrosis is key to developing potential therapies. In this review, we discuss the diverse molecular mechanisms involved in SSc-related lung fibrosis to provide insights into the altered homeostasis state inherent to this fatal disease complication.
Collapse
|
14
|
Odell ID, Steach H, Gauld SB, Reinke-Breen L, Karman J, Carr TL, Wetter JB, Phillips L, Hinchcliff M, Flavell RA. Epiregulin is a dendritic cell-derived EGFR ligand that maintains skin and lung fibrosis. Sci Immunol 2022; 7:eabq6691. [PMID: 36490328 PMCID: PMC9840167 DOI: 10.1126/sciimmunol.abq6691] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immune cells are fundamental regulators of extracellular matrix (ECM) production by fibroblasts and have important roles in determining extent of fibrosis in response to inflammation. Although much is known about fibroblast signaling in fibrosis, the molecular signals between immune cells and fibroblasts that drive its persistence are poorly understood. We therefore analyzed skin and lung samples of patients with diffuse cutaneous systemic sclerosis, an autoimmune disease that causes debilitating fibrosis of the skin and internal organs. Here, we define a critical role of epiregulin-EGFR signaling between dendritic cells and fibroblasts to maintain elevated ECM production and accumulation in fibrotic tissue. We found that epiregulin expression marks an inducible state of DC3 dendritic cells triggered by type I interferon and that DC3-derived epiregulin activates EGFR on fibroblasts, driving a positive feedback loop through NOTCH signaling. In mouse models of skin and lung fibrosis, epiregulin was essential for persistence of fibrosis in both tissues, which could be abrogated by epiregulin genetic deficiency or a neutralizing antibody. Therapeutic administration of epiregulin antibody reversed fibrosis in patient skin and lung explants, identifying it as a previously unexplored biologic drug target. Our findings reveal epiregulin as a crucial immune signal that maintains skin and lung fibrosis in multiple diseases and represents a promising antifibrotic target.
Collapse
Affiliation(s)
- Ian D. Odell
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Holly Steach
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | | | | - Monique Hinchcliff
- Department of Internal Medicine, Section of Rheumatology, Allergy & Immunology, Yale School of Medicine, New Haven, CT, USA
| | - Richard A. Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
15
|
Jiang M, Wang J, Shen Y, Zhu J, Liu Z, Gong W, Yu Y, Zhang S, Zhou X, He S, Song Y, Zhu Z, Jin L, Cong W. Ribosomal S6 Protein Kinase 2 Aggravates the Process of Systemic Scleroderma. J Invest Dermatol 2022; 142:3175-3183.e5. [PMID: 35853487 DOI: 10.1016/j.jid.2022.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 01/05/2023]
Abstract
Systemic sclerosis is a complex process of pathogenesis, and the contributions of inherited genes, infections, and chemicals remain largely unknown. In this study, we showed that p90 ribosomal S6 protein kinase 2 (RSK2) was selectively upregulated in fibrotic skin and fibroblasts treated with the profibrotic cytokine TGF-β. Moreover, knockout of Rsk2 specifically in skin fibroblasts or pharmacological inhibition of RSK2 attenuated skin fibrosis in a mouse model. Mechanistically, RSK2 directly interacted with glycogen synthase kinase 3β in vivo and in vitro and thereby induced phosphorylation of glycogen synthase kinase 3β at Ser9 to inhibit ubiquitination and degradation of GLI1, which promoted fibroblast differentiation and skin fibrosis. Consequently, RSK2 plays an important role in the dermal skin of systemic sclerosis. These findings provided a potential therapeutic target for systemic sclerosis.
Collapse
Affiliation(s)
- Mengying Jiang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Jianan Wang
- Department of Pharmacy, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No.2 Hospital), Ningbo, China
| | - Yingjie Shen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Junjie Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Zhili Liu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Wenjie Gong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Ying Yu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Siyi Zhang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Xuan Zhou
- Ningbo First Hospital, Ningbo, China
| | - Shengqu He
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Yonghuan Song
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongxin Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
16
|
Sun C, Tian X, Jia Y, Yang M, Li Y, Fernig DG. Functions of exogenous FGF signals in regulation of fibroblast to myofibroblast differentiation and extracellular matrix protein expression. Open Biol 2022; 12:210356. [PMID: 36102060 PMCID: PMC9471990 DOI: 10.1098/rsob.210356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Fibroblasts are widely distributed cells found in most tissues and upon tissue injury, they are able to differentiate into myofibroblasts, which express abundant extracellular matrix (ECM) proteins. Overexpression and unordered organization of ECM proteins cause tissue fibrosis in damaged tissue. Fibroblast growth factor (FGF) family proteins are well known to promote angiogenesis and tissue repair, but their activities in fibroblast differentiation and fibrosis have not been systematically reviewed. Here we summarize the effects of FGFs in fibroblast to myofibroblast differentiation and ECM protein expression and discuss the underlying potential regulatory mechanisms, to provide a basis for the clinical application of recombinant FGF protein drugs in treatment of tissue damage.
Collapse
Affiliation(s)
- Changye Sun
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Xiangqin Tian
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Yangyang Jia
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Mingming Yang
- Department of Cardiology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, People's Republic of China
| | - Yong Li
- Department of Biochemistry, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - David G Fernig
- Department of Biochemistry, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| |
Collapse
|
17
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
18
|
Fu M, Peng D, Lan T, Wei Y, Wei X. Multifunctional regulatory protein connective tissue growth factor (CTGF): A potential therapeutic target for diverse diseases. Acta Pharm Sin B 2022; 12:1740-1760. [PMID: 35847511 PMCID: PMC9279711 DOI: 10.1016/j.apsb.2022.01.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/22/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
Connective tissue growth factor (CTGF), a multifunctional protein of the CCN family, regulates cell proliferation, differentiation, adhesion, and a variety of other biological processes. It is involved in the disease-related pathways such as the Hippo pathway, p53 and nuclear factor kappa-B (NF-κB) pathways and thus contributes to the developments of inflammation, fibrosis, cancer and other diseases as a downstream effector. Therefore, CTGF might be a potential therapeutic target for treating various diseases. In recent years, the research on the potential of CTGF in the treatment of diseases has also been paid more attention. Several drugs targeting CTGF (monoclonal antibodies FG3149 and FG3019) are being assessed by clinical or preclinical trials and have shown promising outcomes. In this review, the cellular events regulated by CTGF, and the relationships between CTGF and pathogenesis of diseases are systematically summarized. In addition, we highlight the current researches, focusing on the preclinical and clinical trials concerned with CTGF as the therapeutic target.
Collapse
|
19
|
Vichaikul S, Gurrea-Rubio M, Amin MA, Campbell PL, Wu Q, Mattichak MN, Brodie WD, Palisoc PJ, Ali M, Muraoka S, Ruth JH, Model EN, Rohraff DM, Hervoso JL, Mao-Draayer Y, Fox DA, Khanna D, Sawalha AH, Tsou PS. Inhibition of histone readers bromodomain extra-terminal proteins alleviates skin fibrosis in experimental models of scleroderma. JCI Insight 2022; 7:150871. [PMID: 35349485 PMCID: PMC9090238 DOI: 10.1172/jci.insight.150871] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 03/24/2022] [Indexed: 11/24/2022] Open
Abstract
Binding of the bromodomain and extraterminal domain proteins (BETs) to acetylated histone residues is critical for gene transcription. We sought to determine the antifibrotic efficacy and potential mechanisms of BET inhibition in systemic sclerosis (SSc). Blockade of BETs was done using a pan-BET inhibitor, JQ1; BRD2 inhibitor, BIC1; or BRD4 inhibitors AZD5153 or ARV825. BET inhibition, specifically BRD4 blockade, showed antifibrotic effects in an animal model of SSc and in patient-derived diffuse cutaneous SSc (dcSSc) fibroblasts. Transcriptome analysis of JQ1-treated dcSSc fibroblasts revealed differentially expressed genes related to extracellular matrix, cell cycle, and calcium (Ca2+) signaling. The antifibrotic effect of BRD4 inhibition was mediated at least in part by downregulation of Ca2+/calmodulin–dependent protein kinase II α and reduction of intracellular Ca2+ concentrations. On the basis of these results, we propose targeting Ca2+ pathways or BRD4 as potentially novel therapeutic approaches for progressive tissue fibrosis.
Collapse
Affiliation(s)
- Sirapa Vichaikul
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - Mikel Gurrea-Rubio
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - M. Asif Amin
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - Phillip L. Campbell
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - Qi Wu
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Megan N. Mattichak
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - William D. Brodie
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - Pamela J. Palisoc
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - Mustafa Ali
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - Sei Muraoka
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - Jeffrey H. Ruth
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - Ellen N. Model
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - Dallas M. Rohraff
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - Jonatan L. Hervoso
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - David A. Fox
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
| | - Dinesh Khanna
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
- University of Michigan Scleroderma Program, Ann Arbor, Michigan, USA
| | - Amr H. Sawalha
- Division of Rheumatology, Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Rheumatology and Clinical Immunology; Department of Medicine
- Lupus Center of Excellence; and
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Pei-Suen Tsou
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, and
- University of Michigan Scleroderma Program, Ann Arbor, Michigan, USA
| |
Collapse
|
20
|
Chen Q, Chu L, Li X, Li H, Zhang Y, Cao Q, Zhuang Q. Investigation of an FGFR-Signaling-Related Prognostic Model and Immune Landscape in Head and Neck Squamous Cell Carcinoma. Front Cell Dev Biol 2022; 9:801715. [PMID: 35237609 PMCID: PMC8882630 DOI: 10.3389/fcell.2021.801715] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/29/2021] [Indexed: 12/11/2022] Open
Abstract
Background: There is accumulating evidence on the clinical importance of the fibroblast growth factor receptor (FGFR) signal, hypoxia, and glycolysis in the immune microenvironment of head and neck squamous cell carcinoma (HNSCC), yet reliable prognostic signatures based on the combination of the fibrosis signal, hypoxia, and glycolysis have not been systematically investigated. Herein, we are committed to establish a fibrosis–hypoxia–glycolysis–related prediction model for the prognosis and related immune infiltration of HNSCC. Methods: Fibrotic signal status was estimated with microarray data of a discovery cohort from the TCGA database using the UMAP algorithm. Hypoxia, glycolysis, and immune-cell infiltration scores were imputed using the ssGSEA algorithm. Cox regression with the LASSO method was applied to define prognostic genes and develop a fibrosis–hypoxia–glycolysis–related gene signature. Immunohistochemistry (IHC) was conducted to identify the expression of specific genes in the prognostic model. Protein expression of several signature genes was evaluated in HPA. An independent cohort from the GEO database was used for external validation. Another scRNA-seq data set was used to clarify the related immune infiltration of HNSCC. Results: Six genes, including AREG, THBS1, SEMA3C, ANO1, IGHG2, and EPHX3, were identified to construct a prognostic model for risk stratification, which was mostly validated in the independent cohort. Multivariate analysis revealed that risk score calculated by our prognostic model was identified as an independent adverse prognostic factor (p < .001). Activated B cells, immature B cells, activated CD4+ T cells, activated CD8+ T cells, effector memory CD8+ T cells, MDSCs, and mast cells were identified as key immune cells between high- and low-risk groups. IHC results showed that the expression of SEMA3C, IGHG2 were slightly higher in HNSCC tissue than normal head and neck squamous cell tissue. THBS1, ANO1, and EPHX3 were verified by IHC in HPA. By using single-cell analysis, FGFR-related genes and highly expressed DEGs in low-survival patients were more active in monocytes than in other immune cells. Conclusion: A fibrosis–hypoxia–glycolysis–related prediction model provides risk estimation for better prognoses to patients diagnosed with HNSCC.
Collapse
Affiliation(s)
- Qi Chen
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Ling Chu
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xinyu Li
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Hao Li
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, China
| | - Ying Zhang
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingtai Cao
- Hunan Normal University School of Medicine, Changsha, China
| | - Quan Zhuang
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, China
- Research Center of National Health Ministry on Transplantation Medicine, Changsha, China
- *Correspondence: Quan Zhuang,
| |
Collapse
|
21
|
Fearon AE, Slabber CF, Kuklin A, Bachofner M, Tortola L, Pohlmeier L, Pantasis S, Hornemann T, Chen L, Kopf M, Werner S. Fibroblast growth factor receptor 3 in hepatocytes protects from toxin-induced liver injury and fibrosis. iScience 2021; 24:103143. [PMID: 34646985 PMCID: PMC8497853 DOI: 10.1016/j.isci.2021.103143] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/21/2021] [Accepted: 09/14/2021] [Indexed: 12/13/2022] Open
Abstract
The liver's remarkable regenerative capacity is orchestrated by several growth factors and cytokines. Fibroblast growth factor receptor 3 (Fgfr3) is frequently overexpressed in hepatocellular carcinoma and promotes cancer aggressiveness, whereas its role in liver homeostasis, repair and regeneration is unknown. We show here that Fgfr3 is expressed by hepatocytes in the healthy liver. Its major ligand, Fgf9, is mainly expressed by non-parenchymal cells and upregulated upon injury. Mice lacking Fgfr3 in hepatocytes exhibit increased tissue necrosis after acute toxin treatment and more excessive fibrosis after long-term injury. This was not a consequence of immunological alterations in the non-injured liver as revealed by comprehensive flow cytometry analysis. Rather, loss of Fgfr3 altered the expression of metabolic and pro-fibrotic genes in hepatocytes. These results identify a paracrine Fgf9-Fgfr3 signaling pathway that protects from toxin-induced cell death and the resulting liver fibrosis and suggests a potential use of FGFR3 ligands for therapeutic purposes. Fgfr3 is important for hepatocyte survival following CCl4-induced liver injury Fgfr3 in hepatocytes regulates expression of metabolic and pro-fibrotic genes Fgfr3 protects from extensive fibrosis after chronic CCl4 treatment Fgf receptors have unique, but also overlapping functions in hepatocytes
Collapse
Affiliation(s)
- Abbie E Fearon
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Coenraad F Slabber
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Andrii Kuklin
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Marc Bachofner
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Luigi Tortola
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Lea Pohlmeier
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Sophia Pantasis
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zurich, 8092 Zurich, Switzerland
| | - Lin Chen
- Center of Bone Metabolism and Repair, Department of Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| |
Collapse
|
22
|
Cho SK, Chong BF. SnapshotDx Quiz: October 2021. J Invest Dermatol 2021. [DOI: 10.1016/j.jid.2021.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
23
|
Györfi AH, Matei AE, Fuchs M, Liang C, Rigau AR, Hong X, Zhu H, Luber M, Bergmann C, Dees C, Ludolph I, Horch RE, Distler O, Wang J, Bengsch B, Schett G, Kunz M, Distler JH. Engrailed 1 coordinates cytoskeletal reorganization to induce myofibroblast differentiation. J Exp Med 2021; 218:e20201916. [PMID: 34259830 PMCID: PMC8288503 DOI: 10.1084/jem.20201916] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 04/05/2021] [Accepted: 05/24/2021] [Indexed: 12/18/2022] Open
Abstract
Transforming growth factor-β (TGFβ) is a key mediator of fibroblast activation in fibrotic diseases, including systemic sclerosis. Here we show that Engrailed 1 (EN1) is reexpressed in multiple fibroblast subpopulations in the skin of SSc patients. We characterize EN1 as a molecular amplifier of TGFβ signaling in myofibroblast differentiation: TGFβ induces EN1 expression in a SMAD3-dependent manner, and in turn, EN1 mediates the profibrotic effects of TGFβ. RNA sequencing demonstrates that EN1 induces a profibrotic gene expression profile functionally related to cytoskeleton organization and ROCK activation. EN1 regulates gene expression by modulating the activity of SP1 and other SP transcription factors, as confirmed by ChIP-seq experiments for EN1 and SP1. Functional experiments confirm the coordinating role of EN1 on ROCK activity and the reorganization of cytoskeleton during myofibroblast differentiation, in both standard fibroblast culture systems and in vitro skin models. Consistently, mice with fibroblast-specific knockout of En1 demonstrate impaired fibroblast-to-myofibroblast transition and are partially protected from experimental skin fibrosis.
Collapse
Affiliation(s)
- Andrea-Hermina Györfi
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Alexandru-Emil Matei
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Maximilian Fuchs
- Medical Informatics, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Chunguang Liang
- Medical Informatics, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Aleix Rius Rigau
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Xuezhi Hong
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Honglin Zhu
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Markus Luber
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Christina Bergmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Clara Dees
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Ingo Ludolph
- Department of Plastic and Hand Surgery, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Raymund E. Horch
- Department of Plastic and Hand Surgery, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Oliver Distler
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital of Zurich, Zurich, Switzerland
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, P.R. China
- Human Phenome Institute, Fudan University, Shanghai, P.R. China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, P.R. China
| | - Bertram Bengsch
- Department of Medicine II: Gastroenterology, Hepatology, Endocrinology, and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, Freiburg, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Meik Kunz
- Medical Informatics, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Jörg H.W. Distler
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
24
|
Bektik E, Sun Y, Dennis AT, Sakon P, Yang D, Deschênes I, Fu JD. Inhibition of CREB-CBP Signaling Improves Fibroblast Plasticity for Direct Cardiac Reprogramming. Cells 2021; 10:cells10071572. [PMID: 34206684 PMCID: PMC8307124 DOI: 10.3390/cells10071572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/09/2021] [Accepted: 06/17/2021] [Indexed: 01/14/2023] Open
Abstract
Direct cardiac reprogramming of fibroblasts into induced cardiomyocytes (iCMs) is a promising approach but remains a challenge in heart regeneration. Efforts have focused on improving the efficiency by understanding fundamental mechanisms. One major challenge is that the plasticity of cultured fibroblast varies batch to batch with unknown mechanisms. Here, we noticed a portion of in vitro cultured fibroblasts have been activated to differentiate into myofibroblasts, marked by the expression of αSMA, even in primary cell cultures. Both forskolin, which increases cAMP levels, and TGFβ inhibitor SB431542 can efficiently suppress myofibroblast differentiation of cultured fibroblasts. However, SB431542 improved but forskolin blocked iCM reprogramming of fibroblasts that were infected with retroviruses of Gata4, Mef2c, and Tbx5 (GMT). Moreover, inhibitors of cAMP downstream signaling pathways, PKA or CREB-CBP, significantly improved the efficiency of reprogramming. Consistently, inhibition of p38/MAPK, another upstream regulator of CREB-CBP, also improved reprogramming efficiency. We then investigated if inhibition of these signaling pathways in primary cultured fibroblasts could improve their plasticity for reprogramming and found that preconditioning of cultured fibroblasts with CREB-CBP inhibitor significantly improved the cellular plasticity of fibroblasts to be reprogrammed, yielding ~2-fold more iCMs than untreated control cells. In conclusion, suppression of CREB-CBP signaling improves fibroblast plasticity for direct cardiac reprogramming.
Collapse
Affiliation(s)
- Emre Bektik
- Department of Physiology, Cell Biology College of Medicine, Ohio State University, 333 W 10th Avenue, Columbus, OH 43210, USA; (E.B.); (D.Y.); (I.D.)
- Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA;
| | - Yu Sun
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, OH 44109, USA; (Y.S.); (A.T.D.)
| | - Adrienne T. Dennis
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, OH 44109, USA; (Y.S.); (A.T.D.)
| | - Phraew Sakon
- Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA;
| | - Dandan Yang
- Department of Physiology, Cell Biology College of Medicine, Ohio State University, 333 W 10th Avenue, Columbus, OH 43210, USA; (E.B.); (D.Y.); (I.D.)
| | - Isabelle Deschênes
- Department of Physiology, Cell Biology College of Medicine, Ohio State University, 333 W 10th Avenue, Columbus, OH 43210, USA; (E.B.); (D.Y.); (I.D.)
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, OH 44109, USA; (Y.S.); (A.T.D.)
| | - Ji-Dong Fu
- Department of Physiology, Cell Biology College of Medicine, Ohio State University, 333 W 10th Avenue, Columbus, OH 43210, USA; (E.B.); (D.Y.); (I.D.)
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, OH 44109, USA; (Y.S.); (A.T.D.)
- Correspondence: ; Tel.: +1-(614)-685-0657
| |
Collapse
|
25
|
SnapshotDx Quiz: June 2021. J Invest Dermatol 2021. [DOI: 10.1016/j.jid.2021.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
26
|
Seitz T, Hellerbrand C. Role of fibroblast growth factor signalling in hepatic fibrosis. Liver Int 2021; 41:1201-1215. [PMID: 33655624 DOI: 10.1111/liv.14863] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022]
Abstract
Fibrotic remodelling is a highly conserved protective response to tissue injury and it is essential for the maintenance of structural and functional tissue integrity. Also hepatic fibrosis can be considered as a wound-healing response to liver injury, reflecting a balance between liver repair and scar formation. In contrast, pathological fibrosis corresponds to impaired wound healing. Usually, the liver regenerates after acute injury. However, if the damaging mechanisms persist, the liver reacts with progressive and uncontrolled accumulation of extracellular matrix proteins. Eventually, excessive fibrosis can lead to cirrhosis and hepatic failure. Furthermore, cirrhosis is the major risk factor for the development of hepatocellular cancer (HCC). Therefore, hepatic fibrosis is the most critical pathological factor that determines the morbidity and mortality of patients with chronic liver disease. Still, no effective anti-fibrogenic therapies exist, despite the very high medical need. The regulation of fibroblast growth factor (FGF) signalling is a prerequisite for adequate wound healing, repair and homeostasis in various tissues and organs. The FGF family comprises 22 proteins that can be classified into paracrine, intracrine and endocrine factors. Most FGFs signal through transmembrane tyrosine kinase FGF receptors (FGFRs). Although FGFRs are promising targets for the treatment of HCC, the expression and function of FGFR-ligands in hepatic fibrosis is still poorly understood. This review summarizes the latest advances in our understanding of FGF signalling in hepatic fibrosis. Furthermore, the potential of FGFs as targets for the treatment of hepatic fibrosis and remaining challenges for the field are discussed.
Collapse
Affiliation(s)
- Tatjana Seitz
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
27
|
De Pieri A, Korman BD, Jüngel A, Wuertz-Kozak K. Engineering Advanced In Vitro Models of Systemic Sclerosis for Drug Discovery and Development. Adv Biol (Weinh) 2021; 5:e2000168. [PMID: 33852183 PMCID: PMC8717409 DOI: 10.1002/adbi.202000168] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/19/2022]
Abstract
Systemic sclerosis (SSc) is a complex multisystem disease with the highest case-specific mortality among all autoimmune rheumatic diseases, yet without any available curative therapy. Therefore, the development of novel therapeutic antifibrotic strategies that effectively decrease skin and organ fibrosis is needed. Existing animal models are cost-intensive, laborious and do not recapitulate the full spectrum of the disease and thus commonly fail to predict human efficacy. Advanced in vitro models, which closely mimic critical aspects of the pathology, have emerged as valuable platforms to investigate novel pharmaceutical therapies for the treatment of SSc. This review focuses on recent advancements in the development of SSc in vitro models, sheds light onto biological (e.g., growth factors, cytokines, coculture systems), biochemical (e.g., hypoxia, reactive oxygen species) and biophysical (e.g., stiffness, topography, dimensionality) cues that have been utilized for the in vitro recapitulation of the SSc microenvironment, and highlights future perspectives for effective drug discovery and validation.
Collapse
Affiliation(s)
- Andrea De Pieri
- Dr. A. De Pieri, Prof. K. Wuertz-Kozak, Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY, 14623, USA
| | - Benjamin D Korman
- Prof. B. D. Korman, Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Astrid Jüngel
- Prof. A. Jüngel, Center of Experimental Rheumatology, University Clinic of Rheumatology, Balgrist University Hospital, University Hospital Zurich, Zurich, 8008, Switzerland
- Prof. A. Jüngel, Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Zurich, 8008, Switzerland
| | - Karin Wuertz-Kozak
- Dr. A. De Pieri, Prof. K. Wuertz-Kozak, Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY, 14623, USA
- Prof. K. Wuertz-Kozak, Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), Munich, 81547, Germany
| |
Collapse
|
28
|
Clarke J. FGF signalling enters the SSc arena. Nat Rev Rheumatol 2020; 16:665. [PMID: 33116278 DOI: 10.1038/s41584-020-00536-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|