1
|
Gu S, Xu L, Huang B, Xiong K, Yang X, Ye J. Decoding Macrophage Dynamics: A Pathway to Understanding and Treating Inflammatory Skin Diseases. Int J Mol Sci 2025; 26:4287. [PMID: 40362523 PMCID: PMC12071885 DOI: 10.3390/ijms26094287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Psoriasis and atopic dermatitis (AD) are both chronic inflammatory skin diseases. Their pathogenesis remains incompletely understood. The polarization states of macrophages, as a crucial part of the innate immune system, are influenced by various factors such as cytokines, inflammatory mediators, and epigenetics. Research has demonstrated that macrophages play a "double-edged sword" role in the pathological process of inflammatory skin diseases: they both drive inflammation progression and participate in tissue repair. This article summarizes the roles of macrophages in the inflammatory development and tissue homeostasis of psoriasis and atopic dermatitis. It explores the impact of different factors on macrophages and inflammatory skin diseases. In conclusion, understanding the classification and plasticity of macrophages is crucial for a deeper understanding of the pathogenesis of psoriasis and AD and the development of personalized treatments.
Collapse
Affiliation(s)
- Shengliang Gu
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (S.G.); (L.X.); (B.H.)
- Yunnan Provincial Clinical Medical Centre for Traditional Chinese Medicine Project (Dermatology), Kunming 650500, China
| | - Lei Xu
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (S.G.); (L.X.); (B.H.)
| | - Bin Huang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (S.G.); (L.X.); (B.H.)
| | - Kai Xiong
- The First School of Clinical Medicine, Guizhou University of Chinese Medicine, Guiyang 550025, China;
| | - Xuesong Yang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (S.G.); (L.X.); (B.H.)
- Yunnan Provincial Clinical Medical Centre for Traditional Chinese Medicine Project (Dermatology), Kunming 650500, China
| | - Jianzhou Ye
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (S.G.); (L.X.); (B.H.)
- Yunnan Provincial Clinical Medical Centre for Traditional Chinese Medicine Project (Dermatology), Kunming 650500, China
| |
Collapse
|
2
|
Lopes AMM, Vieira JF, da Silva SFM, Murta EFC, Michelin MA. Dendritic cell immunotherapy has its antitumor action improved by the LPS in the maturation process. Clin Transl Oncol 2025:10.1007/s12094-025-03858-5. [PMID: 39979657 DOI: 10.1007/s12094-025-03858-5] [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: 10/07/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025]
Abstract
Immunotherapy with dendritic cells (DCs) in cancer patients aims to activate the immune response to eliminate neoplastic cells. The present study aimed to investigate lipopolysaccharide (LPS)-stimulated bone marrow-derived dendritic cells in investigating antitumor immune response in experimental breast cancer. For this, we submitted bone marrow pluripotent cells of Balb/c mice differentiated by GM-CSF and IL-4 to maturation with TNF-α and tumor lysate (DCs protocol) or with TNF-α, LPS, and tumor lysate (LPS/DCs protocol). Both immunotherapies were tested in 4T1 breast cancer to evaluate their impact on splenic and tumor microenvironment. We observed that DCs and LPS/DCs reduce the tumor growth rate (p < 0.0001). Besides, the LPS/DCs vaccine shows higher splenic and intratumoral T helper lymphocytes (p < 0.001). Both vaccines increased the production of IFN-γ in the tumor microenvironment (p < 0.0001). The LPS/DCs induced lower Treg lymphocytes and macrophages in the tumor microenvironment (p < 0.0001). The results allow us to conclude that bone marrow-derived dendritic cells stimulated with LPS have been shown to reduce tumor growth rate efficiently and could be better immunotherapy in breast cancer by reducing immunosuppressive cells and increasing antitumoral immune cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Angela Maria Moed Lopes
- Research Institute of Oncology (IPON), Federal University of Triângulo Mineiro (UFTM), Rua Vigário Carlos 100, Abadia, Uberaba, Minas Gerais, 38025-350, Brazil
| | - Jéssica Ferreira Vieira
- Research Institute of Oncology (IPON), Federal University of Triângulo Mineiro (UFTM), Rua Vigário Carlos 100, Abadia, Uberaba, Minas Gerais, 38025-350, Brazil
| | - Saulo Fernando Moreira da Silva
- Research Institute of Oncology (IPON), Federal University of Triângulo Mineiro (UFTM), Rua Vigário Carlos 100, Abadia, Uberaba, Minas Gerais, 38025-350, Brazil
| | - Eddie Fernando Candido Murta
- Research Institute of Oncology (IPON), Federal University of Triângulo Mineiro (UFTM), Rua Vigário Carlos 100, Abadia, Uberaba, Minas Gerais, 38025-350, Brazil
- Discipline of Gynecology and Obstetrics, Federal University of Triângulo Mineiro (UFTM), Uberaba, MG, Brazil
| | - Márcia Antoniazi Michelin
- Research Institute of Oncology (IPON), Federal University of Triângulo Mineiro (UFTM), Rua Vigário Carlos 100, Abadia, Uberaba, Minas Gerais, 38025-350, Brazil.
- Discipline of Immunology, Federal University of Triângulo Mineiro (UFTM), Uberaba, MG, Brazil.
| |
Collapse
|
3
|
Song J, Zeng L, Ye Z, Lu P, Jing C, Hu B, He H. The promoting effect of Balanus albicostatus cement protein 19k (Balcp19k) on wound healing by regulating fibroblast migration and relieving early-stage inflammation responses. Int J Biol Macromol 2025; 289:138781. [PMID: 39681245 DOI: 10.1016/j.ijbiomac.2024.138781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
The high incidence of skin wounds (e.g., burns, diabetic ulcers, venous ulcers, pressure sores, and radiation injuries) remains a significant challenge in clinical settings. Dressing materials, infused with bioactive components, are essential and extensively utilized in specific circumstances. According to traditional Chinese medicine therapeutic regimens, powders made of the flesh/shells of Balanus albicostatus were used to treat sores due to their ability to alleviate inflammatory responses. Nevertheless, information is scarce regarding the effective components and the pharmacological mechanisms involved. In this work, purified Balanus albicostatus cement protein 19k (Balcp19k) was obtained as a single-component material via an E. coli-based heterologous expression system. It had the potential to promote fibroblast proliferation and migration, which were crucial for wound healing. Immunofluorescence revealed that Balcp19k treatment promoted M2 macrophage transformation in wound tissues. Correspondingly, the expression of inflammation-related genes (e.g., IL-6, TNF-α, and IFN-γ) was significantly reduced (p < 0.05). Moreover, histological examination indicated that this treatment accelerated skin-wound healing by promoting collagen fiber deposition and angiogenesis, and restraining inflammatory cell infiltration in the full-thickness skin of mice. In conclusion, Balcp19k is a biocompatible, effective constituent found in Balanus albicostatus. This bioactive protein could be novel therapeutic material for wound healing, with ability to regulate fibroblast migration, alleviate early-stage inflammatory responses, and enhance collagen fiber deposition and angiogenesis.
Collapse
Affiliation(s)
- Junyi Song
- College of Science, National University of Defense Technology, Changsha, China
| | - Ling Zeng
- College of Science, National University of Defense Technology, Changsha, China
| | - Zijian Ye
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China; The Institute of Vascular Diseases, Central South University, Changsha, China
| | - Peng Lu
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China; The Institute of Vascular Diseases, Central South University, Changsha, China
| | - Chengyu Jing
- College of Science, National University of Defense Technology, Changsha, China
| | - Biru Hu
- College of Science, National University of Defense Technology, Changsha, China.
| | - Hao He
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China; The Institute of Vascular Diseases, Central South University, Changsha, China.
| |
Collapse
|
4
|
Zhang Q, Jiao J, Wang X, Zhang L. The role of fibroblast in chronic rhinosinusitis with nasal polyps: a key player in the inflammatory process. Expert Rev Clin Immunol 2025; 21:169-179. [PMID: 39378160 DOI: 10.1080/1744666x.2024.2414774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/31/2024] [Accepted: 10/07/2024] [Indexed: 10/10/2024]
Abstract
INTRODUCTION Fibroblasts are the primary supporting cells in connective tissue and have long been thought to contribute to chronic rhinosinusitis with nasal polyps (CRSwNP) by producing extracellular matrix (ECM), leading to fibrosis and tissue remodeling. However, recent studies have highlighted the critical role of nasal polyp-derived fibroblasts (NPDFs) in triggering and intensifying the inflammatory response in CRSwNP. AREAS COVERED This review undertook a comprehensive literature search across the PubMed database, Web of Science since 2000, offering an in-depth summary of the pivotal role of NPDFs in tissue remodeling and inflammatory responses in CRSwNP. Additionally, single-cell RNA sequencing data provides a deeper exploration of the heterogeneity and functional mechanisms of fibroblasts in CRSwNP. Consequently, these insights point to fibroblasts as promising therapeutic targets for effectively treating CRSwNP. EXPERT OPINION Current data underscore the essential role of fibroblasts in the pathogenesis of CRSwNP. Fully elucidating the specific mechanisms by which fibroblasts contribute to the disease process is crucial for developing targeted therapies. Furthermore, advancements in single-cell RNA sequencing pave the way for selectively targeting and depleting pathological fibroblast subpopulations. Despite these advancements, the clinical development of fibroblast-targeted therapies in CRSwNP remains challenging.
Collapse
Affiliation(s)
- Qinqin Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Jian Jiao
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Xiangdong Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Mukherjee S, Kumar D, Guha D. Insights of probiotics as an alternative medicine for cancer therapy, mechanism, and applications. MEDICINE IN MICROECOLOGY 2024; 22:100111. [DOI: 10.1016/j.medmic.2024.100111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
6
|
Homma S, Ogasawara T, Suga M, Nakamura Y, Takenaka K, Marshall S, Kawauchi K, Mori N, Kuroda H, Nakamura N, Miyagi Y, Masunaga A. Triple immunostaining demonstrates the possible existence of segregated-nucleus-containing atypical monocytes in human primary myelofibrosis bone marrow: a case report. J Med Case Rep 2024; 18:510. [PMID: 39472997 PMCID: PMC11524003 DOI: 10.1186/s13256-024-04844-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/03/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Segregated-nucleus-containing atypical monocytes have recently been identified in mice. Segregated-nucleus-containing atypical monocytes are thought to originate from the bone marrow and induce fibrosis in the drug-injured lung. The Lyc6c- murine monocyte subset is the counterpart to human CD14-CD16++ non-classical monocytes; however, the human counterpart to murine segregated-nucleus-containing atypical monocytes has not yet been identified. Primary myelofibrosis is a well-known disease of progressive marrow fibrosis, and atypical megakaryocytes are thought to be closely related to fibrosis in primary myelofibrosis bone marrow. However, recently, monocytes have been reported to play an important role in marrow fibrosis in primary myelofibrosis. We speculated that, if there is a human counterpart to murine segregated-nucleus-containing atypical monocytes, it would present the same markers as murine segregated-nucleus-containing atypical monocytes, such as CD14-CD16+ macrophage-1 antigen (CD11b/CD18 complex)+, MSR1+, and CEACAM1+, and it might exist in the bone marrow of patients with primary myelofibrosis. CASE PRESENTATION A 74-year-old Japanese male visited our hospital for clinical follow-up after total prostatectomy for prostatic cancer. Anemia, thrombocytosis, and elevated lactate dehydrogenase were suddenly observed in a periodic examination. CALR mutation type 2 (p.K385fs*47) was observed. The histological features of the patient's bone marrow were consistent with fibrotic primary myelofibrosis. We immunohistochemically studied the bone marrow in an attempt to identify a human counterpart to murine segregated-nucleus-containing atypical monocytes. We detected a few CD16+MSR1+CEACAM1+ cells, but not CD14+MSR1+CEACAM1+ cells, by triple immunostaining. The patient is in a good condition and does not require treatment for primary myelofibrosis. CONCLUSION There is a possibility that human segregated-nucleus-containing atypical monocytes exist in the bone marrow of primary myelofibrosis patients and might be related to marrow fibrosis.
Collapse
Affiliation(s)
- Shunsuke Homma
- Department of Medical Intern Center, Tokyo Women's Medical University Adachi Medical Center, Tokyo, Japan
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Toshie Ogasawara
- Department of Medicine, Tokyo Women's Medical University Adachi Medical Center, Tokyo, Japan
| | - Michie Suga
- Department of Diagnostic Pathology, Tokyo Women's Medical University Adachi Medical Center, 4-33-1, Kohoku, Adachi-ku, 123-8558, Tokyo, Japan
| | - Yoshiyasu Nakamura
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan
| | - Katsuya Takenaka
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan
- TR Company, Shin Nippon Biomedical Laboratories, Ltd., Kagoshima, Japan
| | - Shoko Marshall
- Department of Medicine, Tokyo Women's Medical University Adachi Medical Center, Tokyo, Japan
| | - Kiyotaka Kawauchi
- Department of Medicine, Tokyo Women's Medical University Adachi Medical Center, Tokyo, Japan
| | - Naoki Mori
- Department of Medicine, Tokyo Women's Medical University Adachi Medical Center, Tokyo, Japan
- Department of Hematology, International University of Health and Welfare Narita Hospital, Narita, Japan
| | - Hajime Kuroda
- Department of Diagnostic Pathology, Tokyo Women's Medical University Adachi Medical Center, 4-33-1, Kohoku, Adachi-ku, 123-8558, Tokyo, Japan
| | - Naoya Nakamura
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan
| | - Atsuko Masunaga
- Department of Diagnostic Pathology, Tokyo Women's Medical University Adachi Medical Center, 4-33-1, Kohoku, Adachi-ku, 123-8558, Tokyo, Japan.
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan.
| |
Collapse
|
7
|
Li H, Li B, Zheng Y. Role of microglia/macrophage polarisation in intraocular diseases (Review). Int J Mol Med 2024; 53:45. [PMID: 38551157 PMCID: PMC10998719 DOI: 10.3892/ijmm.2024.5369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/05/2024] [Indexed: 04/02/2024] Open
Abstract
Macrophages form a crucial component of the innate immune system, and their activation is indispensable for various aspects of immune and inflammatory processes, tissue repair, and maintenance of the balance of the body's state. Macrophages are found in all ocular tissues, spanning from the front surface, including the cornea, to the posterior pole, represented by the choroid/sclera. The neural retina is also populated by specialised resident macrophages called microglia. The plasticity of microglia/macrophages allows them to adopt different activation states in response to changes in the tissue microenvironment. When exposed to various factors, microglia/macrophages polarise into distinct phenotypes, each exhibiting unique characteristics and roles. Furthermore, extensive research has indicated a close association between microglia/macrophage polarisation and the development and reversal of various intraocular diseases. The present article provides a review of the recent findings on the association between microglia/macrophage polarisation and ocular pathological processes (including autoimmune uveitis, optic neuritis, sympathetic ophthalmia, retinitis pigmentosa, glaucoma, proliferative vitreoretinopathy, subretinal fibrosis, uveal melanoma, ischaemic optic neuropathy, retinopathy of prematurity and choroidal neovascularization). The paradoxical role of microglia/macrophage polarisation in retinopathy of prematurity is also discussed. Several studies have shown that microglia/macrophages are involved in the pathology of ocular diseases. However, it is required to further explore the relevant mechanisms and regulatory processes. The relationship between the functional diversity displayed by microglia/macrophage polarisation and intraocular diseases may provide a new direction for the treatment of intraocular diseases.
Collapse
Affiliation(s)
- Haoran Li
- School of Opthalmology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Biao Li
- School of Opthalmology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Yanlin Zheng
- School of Opthalmology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
8
|
Saldarriaga OA, Wanninger TG, Arroyave E, Gosnell J, Krishnan S, Oneka M, Bao D, Millian DE, Kueht ML, Moghe A, Jiao J, Sanchez JI, Spratt H, Beretta L, Rao A, Burks JK, Stevenson HL. Heterogeneity in intrahepatic macrophage populations and druggable target expression in patients with steatotic liver disease-related fibrosis. JHEP Rep 2024; 6:100958. [PMID: 38162144 PMCID: PMC10757256 DOI: 10.1016/j.jhepr.2023.100958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 08/18/2023] [Accepted: 09/25/2023] [Indexed: 01/03/2024] Open
Abstract
Background & Aims Clinical trials for reducing fibrosis in steatotic liver disease (SLD) have targeted macrophages with variable results. We evaluated intrahepatic macrophages in patients with SLD to determine if activity scores or fibrosis stages influenced phenotypes and expression of druggable targets, such as CCR2 and galectin-3. Methods Liver biopsies from controls or patients with minimal or advanced fibrosis were subject to gene expression analysis using nCounter to determine differences in macrophage-related genes (n = 30). To investigate variability among individual patients, we compared additional biopsies by staining them with multiplex antibody panels (CD68/CD14/CD16/CD163/Mac387 or CD163/CCR2/galectin-3/Mac387) followed by spectral imaging and spatial analysis. Algorithms that utilize deep learning/artificial intelligence were applied to create cell cluster plots, phenotype profile maps, and to determine levels of protein expression (n = 34). Results Several genes known to be pro-fibrotic (e.g. CD206, TREM2, CD163, and ARG1) showed either no significant differences or significantly decreased with advanced fibrosis. Although marked variability in gene expression was observed in individual patients with cirrhosis, several druggable targets and their ligands (e.g. CCR2, CCR5, CCL2, CCL5, and LGALS3) were significantly increased when compared to patients with minimal fibrosis. Antibody panels identified populations that were significantly increased (e.g. Mac387+), decreased (e.g. CD14+), or enriched (e.g. interactions of Mac387) in patients that had progression of disease or advanced fibrosis. Despite heterogeneity in patients with SLD, several macrophage phenotypes and druggable targets showed a positive correlation with increasing NAFLD activity scores and fibrosis stages. Conclusions Patients with SLD have markedly varied macrophage- and druggable target-related gene and protein expression in their livers. Several patients had relatively high expression, while others were like controls. Overall, patients with more advanced disease had significantly higher expression of CCR2 and galectin-3 at both the gene and protein levels. Impact and implications Appreciating individual differences within the hepatic microenvironment of patients with SLD may be paramount to developing effective treatments. These results may explain why such a small percentage of patients have responded to macrophage-targeting therapies and provide additional support for precision medicine-guided treatment of chronic liver diseases.
Collapse
Affiliation(s)
- Omar A. Saldarriaga
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Timothy G. Wanninger
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Esteban Arroyave
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Joseph Gosnell
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Santhoshi Krishnan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
| | - Morgan Oneka
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Daniel Bao
- School of Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Daniel E. Millian
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Michael L. Kueht
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Akshata Moghe
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Jingjing Jiao
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jessica I. Sanchez
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heidi Spratt
- Department of Biostatistics and Data Science, University of Texas Medical Branch, Galveston, TX, USA
| | - Laura Beretta
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
- Departmen of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, Rice University, Ann Arbor, MI, USA
| | - Jared K. Burks
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
9
|
Anjum S, Li T, Saeed M, Ao Q. Exploring polysaccharide and protein-enriched decellularized matrix scaffolds for tendon and ligament repair: A review. Int J Biol Macromol 2024; 254:127891. [PMID: 37931866 DOI: 10.1016/j.ijbiomac.2023.127891] [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: 07/17/2023] [Revised: 10/07/2023] [Accepted: 11/02/2023] [Indexed: 11/08/2023]
Abstract
Tissue engineering (TE) has become a primary research topic for the treatment of diseased or damaged tendon/ligament (T/L) tissue. T/L injuries pose a severe clinical burden worldwide, necessitating the development of effective strategies for T/L repair and tissue regeneration. TE has emerged as a promising strategy for restoring T/L function using decellularized extracellular matrix (dECM)-based scaffolds. dECM scaffolds have gained significant prominence because of their native structure, relatively high bioactivity, low immunogenicity, and ability to function as scaffolds for cell attachment, proliferation, and differentiation, which are difficult to imitate using synthetic materials. Here, we review the recent advances and possible future prospects for the advancement of dECM scaffolds for T/L tissue regeneration. We focus on crucial scaffold properties and functions, as well as various engineering strategies employed for biomaterial design in T/L regeneration. dECM provides both the physical and mechanical microenvironments required by cells to survive and proliferate. Various decellularization methods and sources of allogeneic and xenogeneic dECM in T/L repair and regeneration are critically discussed. Additionally, dECM hydrogels, bio-inks in 3D bioprinting, and nanofibers are briefly explored. Understanding the opportunities and challenges associated with dECM-based scaffold development is crucial for advancing T/L repairs in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Shabnam Anjum
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, Shenyang 110122, China; NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Ting Li
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Mohammad Saeed
- Dr. A.P.J Abdul Kalam Technical University, Lucknow 226031, India
| | - Qiang Ao
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, Shenyang 110122, China; NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
10
|
Liang W, Huang X, Shi J. Macrophages Serve as Bidirectional Regulators and Potential Therapeutic Targets for Liver Fibrosis. Cell Biochem Biophys 2023; 81:659-671. [PMID: 37695501 DOI: 10.1007/s12013-023-01173-w] [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: 12/23/2022] [Accepted: 09/02/2023] [Indexed: 09/12/2023]
Abstract
Liver fibrosis is a dynamic pathological process in which the structure and function of the liver abnormally change due to long-term complex inflammatory reactions and chronic liver injury caused by multiple internal and external factors. Previous studies believed that the activation of hepatic stellate cells is a critical part of the occurrence and development of liver fibrosis. However, an increasing number of studies have indicated that the macrophage plays an important role as a central regulator in liver fibrosis, and it directly affects the development and recovery of liver fibrosis. Studies of macrophages and liver fibrosis in the recent 10 years will be reviewed in this paper. This review will not only clarify the molecular mechanism of liver fibrosis regulated by macrophages but also provide new strategies and methods for ameliorating and treating liver fibrosis.
Collapse
Affiliation(s)
- Wei Liang
- Clinical Medical Research Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China.
| | - Xianing Huang
- Guangxi International Travel Healthcare Centre (Port Clinic of Nanning Customs District), Nanning, 530021, Guangxi, China
| | - Jingjing Shi
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
11
|
Zheng Y, Ji S, Li X, Wen L. Qijia rougan formula ameliorates ECM deposition in hepatic fibrosis by regulating the JAK1/STAT6-microRNA-23a feedback loop in macrophage M2 polarization. Biomed Pharmacother 2023; 168:115794. [PMID: 37922651 DOI: 10.1016/j.biopha.2023.115794] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/15/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Hepatic fibrosis is the critical pathological stage in the progression of chronic liver disease to cirrhosis and hepatocellular carcinoma (HCC). However, no approved anti-hepatic fibrosis drugs are available currently. Qijia Rougan Formula (QRF) is a traditional Chinese medicine (TCM) with significant clinical efficacy on hepatic fibrosis. It was derived from Sanjiasan, a famous decoction documented in the Book of Treatise on the Pestilence in the Ming Dynasty of China. However, the underlying regulatory mechanisms remain elusive. This study further confirmed the therapeutic effects of QRF on hepatic fibrosis and dissected its underlying molecular mechanisms from the perspective of macrophage M2 polarization, one of the critical events in hepatic fibrosis. Experimentally, QRF significantly improved extracellular matrix (ECM) deposition and fibrosis in the liver of model rats. QRF diminished the proportion of M2 macrophages, decreased the levels of TGF-β, PDGFB and IL-10, and regulated the expression of p-JAK1, p-STAT6, JAK1 and microRNA-23a both in vitro and in vivo. Collectively, it was confirmed that QRF effectively improves liver function and hepatocyte damage, and reduces ECM deposition. QRF ameliorates hepatic fibrosis by regulating JAK1/STAT6-microRNA-23a negative feedback loop to inhibit macrophage M2 polarization and thus reduce ECM deposition. Our study illustrates the potential of QRF for hepatic fibrosis therapy, suggesting that QRF is a promising anti-hepatic fibrosis drug candidate.
Collapse
Affiliation(s)
- Yanfeng Zheng
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shaoxiu Ji
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xia Li
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Li Wen
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
12
|
Li R, Feng D, Han S, Zhai X, Yu X, Fu Y, Jin F. Macrophages and fibroblasts in foreign body reactions: How mechanical cues drive cell functions? Mater Today Bio 2023; 22:100783. [PMID: 37701130 PMCID: PMC10494263 DOI: 10.1016/j.mtbio.2023.100783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Biomaterials, when implanted in the human body, can induce a series of cell- and cytokine-related reactions termed foreign body reactions (FBRs). In the progression of FBRs, macrophages regulate inflammation and healing by polarizing to either a pro-inflammatory or pro-healing phenotype and recruit fibroblasts by secreting cytokines. Stimulated by the biomaterials, fibrotic capsule is formed eventually. The implant, along with its newly formed capsule, introduces various mechanical cues that influence cellular functions. Mechanosensing proteins, such as integrins or ion channels, transduce extracellular mechanical signals into cytoplasm biochemical signals in response to mechanical stimuli. Consequently, the morphology, migration mode, function, and polarization state of the cells are affected. Modulated by different intracellular signaling pathways and their crosstalk, the expression of fibrotic genes increases with fibroblast activation and fibroblast to myofibroblast transition under stiff or force stimuli. However, summarized in most current studies, the outcomes of macrophage polarization in the effect of different mechanical cues are inconsistent. The underlying mechanisms should be investigated with more advanced technology and considering more interfering aspects. Further research is needed to determine how to modulate the progression of fibrotic capsule formation in FBR artificially.
Collapse
Affiliation(s)
- Rihan Li
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Dongdong Feng
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Siyuan Han
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Xiaoyue Zhai
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, Liaoning, 110000, China
| | - Xinmiao Yu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Yuanyuan Fu
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, Liaoning, 110000, China
| | - Feng Jin
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| |
Collapse
|
13
|
Ha JG, Cho HJ. Unraveling the Role of Epithelial Cells in the Development of Chronic Rhinosinusitis. Int J Mol Sci 2023; 24:14229. [PMID: 37762530 PMCID: PMC10531804 DOI: 10.3390/ijms241814229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
The pathophysiology of CRS is multifactorial and complex yet needs to be completed. Recent evidence emphasizes the crucial part played by epithelial cells in the development of CRS. The epithelial cells act as physical barriers and play crucial roles in host defense, including initiating and shaping innate and adaptive immune responses. This review aims to present a comprehensive understanding of the significance of nasal epithelial cells in CRS. New research suggests that epithelial dysfunction plays a role in developing CRS through multiple mechanisms. This refers to issues with a weakened barrier function, disrupted mucociliary clearance, and irregular immune responses. When the epithelial barrier is compromised, it can lead to the passage of pathogens and allergens, triggering inflammation in the body. Furthermore, impaired mucociliary clearance can accumulate pathogens and secretions of inflammatory mediators, promoting chronic inflammation. Epithelial cells can release cytokines and chemokines, which attract and activate immune cells. This can result in an imbalanced immune response that continues to cause inflammation. The interaction between nasal epithelial cells and various immune cells leads to the production of cytokines and chemokines, which can either increase or decrease inflammation. By comprehending the role of epithelial cells in CRS, we can enhance our understanding of the disease's pathogenesis and explore new therapeutics.
Collapse
Affiliation(s)
- Jong-Gyun Ha
- Department of Otorhinolaryngology—Head and Neck Surgery, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong 14353, Republic of Korea;
| | - Hyung-Ju Cho
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- The Airway Mucus Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
14
|
Abbaszadeh M, Karimi M, Rajaei S. The landscape of non-coding RNAs in the immunopathogenesis of Endometriosis. Front Immunol 2023; 14:1223828. [PMID: 37675122 PMCID: PMC10477370 DOI: 10.3389/fimmu.2023.1223828] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/02/2023] [Indexed: 09/08/2023] Open
Abstract
Endometriosis is a complex disorder that is characterized by the abnormal growth of endometrial-like tissue outside the uterus. It is associated with chronic inflammation, severe pelvic pain, infertility, and significantly reduced quality of life. Although the exact mechanism of endometriosis remains unknown, inflammation and altered immunity are considered key factors in the immunopathogenesis of the disorder. Disturbances of immune responses result in reduced clearance of regurgitated endometrial cells, which elicits oxidative stress and progression of inflammation. Proinflammatory mediators could affect immune cells' recruitment, fate, and function. Reciprocally, the activation of immune cells can promote inflammation. Aberrant expression of non-coding RNA (ncRNA) in patient and animal lesions could be suggestive of their role in endometriosis establishment. The engagement of these RNAs in regulating diverse biological processes, including inflammatory responses and activation of inflammasomes, altered immunity, cell proliferation, migration, invasion, and angiogenesis are widespread and far-reaching. Therefore, ncRNAs can be identified as a determining candidate regulating the inflammatory responses and immune system. This review aims in addition to predict the role of ncRNAs in the immunopathogenesis of endometriosis through regulating inflammation and altered immunity based on previous studies, it presents a comprehensive view of inflammation role in the pathogenesis of endometriosis.
Collapse
Affiliation(s)
| | | | - Samira Rajaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Moon S, Hong J, Go S, Kim BS. Immunomodulation for Tissue Repair and Regeneration. Tissue Eng Regen Med 2023; 20:389-409. [PMID: 36920675 PMCID: PMC10219918 DOI: 10.1007/s13770-023-00525-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/11/2023] [Accepted: 01/26/2023] [Indexed: 03/16/2023] Open
Abstract
Various immune cells participate in repair and regeneration following tissue injury or damage, orchestrating tissue inflammation and regeneration processes. A deeper understanding of the immune system's involvement in tissue repair and regeneration is critical for the development of successful reparatory and regenerative strategies. Here we review recent technologies that facilitate cell-based and biomaterial-based modulation of the immune systems for tissue repair and regeneration. First, we summarize the roles of various types of immune cells in tissue repair. Second, we review the principle, examples, and limitations of regulatory T (Treg) cell-based therapy, a representative cell-based immunotherapy. Finally, we discuss biomaterial-based immunotherapy strategies that aim to modulate immune cells using various biomaterials for tissue repair and regeneration.
Collapse
Affiliation(s)
- Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Institute of Chemical Processes, Institute of Engineering Research, BioMAX, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
16
|
Gunatilaka A, Zhang S, Tan WSD, G Stewart A. Anti-fibrotic strategies and pulmonary fibrosis. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 98:179-224. [PMID: 37524487 DOI: 10.1016/bs.apha.2023.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) results from the dysregulated process of injury and repair, which promotes scarring of the lung tissue and deposition of collagen-rich extracellular matrix (ECM) components, that make the lung unphysiologically stiff. IPF presents a serious concern as its pathogenesis remains elusive, and current anti-fibrotic treatments are only effective in slowing rather than halting disease progression. The IPF disease pathogenesis is incompletely defined, complex and incorporates interplay between different fibrogenesis signaling pathways. Preclinical IPF experimental models used to validate drug candidates present significant limitations in modeling IPF pathobiology, with their limited time frame, simplicity and inaccurate representation of the disease and the mechanical influences of IPF. Potentially more accurate mimetic disease models that capture the cell-cell and cell-matrix interaction, such as 3D cultures, organoids and precision-cut lung slices (PCLS), may yield more meaningful clinical predictions for drug candidates. Recent advances in developing anti-fibrotic compounds have positioned drug towards targeting components of the fibrogenesis signaling pathway of IPF or the extracellular microenvironment. The major goals in this area of research focus on finding ways to reverse or halt the disease progression by utilizing more disease-relevant experimental models to improve the qualification of potential drug targets for treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Avanka Gunatilaka
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, Australia; ARC Centre for Personalised Therapeutics Technologies, The University of Melbourne, Parkville, VIC, Australia
| | - Stephanie Zhang
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, Australia
| | - Wan Shun Daniel Tan
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, Australia
| | - Alastair G Stewart
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, Australia; ARC Centre for Personalised Therapeutics Technologies, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
17
|
Saldarriaga OA, Krishnan S, Wanninger TG, Oneka M, Rao A, Bao D, Arroyave E, Gosnell J, Kueht M, Moghe A, Millian D, Jiao J, Sanchez JI, Spratt H, Beretta L, Stevenson HL. Patients with fibrosis from non-alcoholic steatohepatitis have heterogeneous intrahepatic macrophages and therapeutic targets. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.16.23285924. [PMID: 36865099 PMCID: PMC9980226 DOI: 10.1101/2023.02.16.23285924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Background and Aims In clinical trials for reducing fibrosis in NASH patients, therapeutics that target macrophages have had variable results. We evaluated intrahepatic macrophages in patients with non-alcoholic steatohepatitis to determine if fibrosis influenced phenotypes and expression of CCR2 and Galectin-3. Approach & Results We used nCounter to analyze liver biopsies from well-matched patients with minimal (n=12) or advanced (n=12) fibrosis to determine which macrophage-related genes would be significantly different. Known therapy targets (e.g., CCR2 and Galectin-3) were significantly increased in patients with cirrhosis.However, several genes (e.g., CD68, CD16, and CD14) did not show significant differences, and CD163, a marker of pro-fibrotic macrophages was significantly decreased with cirrhosis. Next, we analyzed patients with minimal (n=6) or advanced fibrosis (n=5) using approaches that preserved hepatic architecture by multiplex-staining with anti-CD68, Mac387, CD163, CD14, and CD16. Spectral data were analyzed using deep learning/artificial intelligence to determine percentages and spatial relationships. This approach showed patients with advanced fibrosis had increased CD68+, CD16+, Mac387+, CD163+, and CD16+CD163+ populations. Interaction of CD68+ and Mac387+ populations was significantly increased in patients with cirrhosis and enrichment of these same phenotypes in individuals with minimal fibrosis correlated with poor outcomes. Evaluation of a final set of patients (n=4) also showed heterogenous expression of CD163, CCR2, Galectin-3, and Mac387, and significant differences were not dependent on fibrosis stage or NAFLD activity. Conclusions Approaches that leave hepatic architecture intact, like multispectral imaging, may be paramount to developing effective treatments for NASH. In addition, understanding individual differences in patients may be required for optimal responses to macrophage-targeting therapies.
Collapse
|
18
|
Role of Nasal Fibroblasts in Airway Remodeling of Chronic Rhinosinusitis: The Modulating Functions Reexamined. Int J Mol Sci 2023; 24:ijms24044017. [PMID: 36835423 PMCID: PMC9965487 DOI: 10.3390/ijms24044017] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/19/2023] Open
Abstract
Chronic rhinosinusitis (CRS) is a multifactorial inflammatory disease of the nose and sinuses that affects more than 10% of the adult population worldwide. Currently, CRS is classified into endotypes according to the inflammatory response (Th1, Th2, and Th17) or the distribution of immune cells in the mucosa (eosinophilic and non-eosinophilic). CRS induces mucosal tissue remodeling. Extracellular matrix (ECM) accumulation, fibrin deposition, edema, immune cell infiltration, and angiogenesis are observed in the stromal region. Conversely, epithelial-to-mesenchymal transition (EMT), goblet cell hyperplasia, and increased epithelial permeability, hyperplasia, and metaplasia are found in the epithelium. Fibroblasts synthesize collagen and ECM, which create a structural skeleton of tissue and play an important role in the wound-healing process. This review discusses recent knowledge regarding the modulation of tissue remodeling by nasal fibroblasts in CRS.
Collapse
|
19
|
Sueyoshi Y, Niwa A, Nishikawa Y, Isogai N. The significance of nanofiber polyglycolic acid for promoting tissue repair in a rat subcutaneous implantation model. J Biomed Mater Res B Appl Biomater 2023; 111:16-25. [PMID: 35833260 DOI: 10.1002/jbm.b.35128] [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: 03/24/2022] [Revised: 06/03/2022] [Accepted: 06/22/2022] [Indexed: 11/08/2022]
Abstract
Among various biomaterials, we focused on nanofiber-based polyglycolic acid (PGA) fabric and examined the dynamics of cells that migrate within the non-woven fabric after implantation. The efficacy of nano-PGA as a tissue reinforcement in the process of subcutaneous tissue repair was immunohistochemically investigated. Two types of clinically available PGA non-woven sheet (nano-PGA: fiber diameter = 2.0 μm, conventional PGA: fiber diameter = 14.2 μm) were used and subcutaneously implanted in rats. Samples were collected 3 days, and 1, 2, 3, and 4 weeks after the implantation to perform histological and immunohistochemical (CD68, CD163, α-SMA, Type I collagen, CD34, MCP-1, IL-6, TNF-α, TGF-β, VEGF, IgG) examinations to assess the expression of molecules related to inflammation or tissue repair. Immunohistochemical analysis in nano-PGA revealed that the intensity and positive cells (CD68, MCP-1, IL-6, TNF-α) significantly increased which indicated an early inflammatory response. This was followed by phagocytosis of nano-PGA with foreign body giant cells and CD68+ macrophages. Finally, the number of proliferating cells (CD163, α-SMA, TGF-β) and angiogenesis (CD34, VEGF) for tissue repair promoted the formation of collagen fibers (type I collagen). Unlike nano-PGA, implantation of conventional PGA sheet resulted in a prolonged inflammatory response and was characterized by the presence of discontinuous collagen fibers with many foreign body giant cells, which did not lead to tissue repair. Nano-PGA sheets demonstrated a better tissue compatibility compared with conventional PGA by inducing early polarization to M2 phenotype macrophages, which triggered subsequent angiogenesis and tissue repair in the subcutaneous tissue.
Collapse
Affiliation(s)
- Yu Sueyoshi
- Department of Plastic and Reconstructive Surgery, Kindai University Faculty of Medicine, Osaka-sayama, Japan
| | - Atsuko Niwa
- Department of Plastic and Reconstructive Surgery, Kindai University Faculty of Medicine, Osaka-sayama, Japan
| | - Yuki Nishikawa
- Department of Plastic and Reconstructive Surgery, Kindai University Faculty of Medicine, Osaka-sayama, Japan
| | - Noritaka Isogai
- Department of Plastic and Reconstructive Surgery, Kindai University Faculty of Medicine, Osaka-sayama, Japan
| |
Collapse
|
20
|
Xiang H, Yu H, Zhou Q, Wu Y, Ren J, Zhao Z, Tao X, Dong D. Macrophages: A rising star in immunotherapy for chronic pancreatitis. Pharmacol Res 2022; 185:106508. [DOI: 10.1016/j.phrs.2022.106508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 10/10/2022] [Indexed: 11/29/2022]
|
21
|
Zhang X, Cao Y, Pan D, Yao X, Wang F, Zhang G, Luo Y. Antifibrotic pyridine-containing monoterpene alkaloids from Caryopteris glutinosa. PHYTOCHEMISTRY 2022; 203:113378. [PMID: 36007661 DOI: 10.1016/j.phytochem.2022.113378] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/10/2022] [Accepted: 08/14/2022] [Indexed: 06/15/2023]
Abstract
Three undescribed dimeric pyridine-containing alkaloids, caryopterisines C - E, and four unreported cyclopenta[c]pyridine-derived alkaloids, caryopterisines F - I, were identified from Caryopteris glutinosa Rehder (Lamiaceae), together with two known monoterpene alkaloids. Caryopterisine C, featuring with an unprecedented 6/5/6/6/5 pentacyclic rings scaffold, may biosynthetically stem from a Diels-Alder reaction of two cyclopenta[c]pyridine-containing monomers and a following aromatization rearrangement reaction. Caryopterisines D and E, possessing an unprecedented 6/6/6/6/5 fused rings framework, may originate from a same Diels-Alder reaction of two monomers and subsequent aromatization arrangement, Baeyer-Villiger oxidation, and a set of tailoring reactions. Caryopterisine C showed strong inhibition on collagen accumulation in NIH3T3 cells (IC50 = 14.26 ± 1.46 μM). Caryopterisines G and I reduce collagen accumulation with IC50 values 88.91 ± 0.95 μM and 33.09 ± 1.38 μM, respectively. The Western blotting of the transforming growth factor-β-activated signaling pathways revealed that caryopterisine C inhibits collagen expression and accumulation via suppression of the phosphorylation of ERK1/2, P38, and SMAD2/3. The present works indicate caryopterisine C is a potential lead compound for the development of antifibrotic drugs.
Collapse
Affiliation(s)
- Xuejian Zhang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China
| | - Yu Cao
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dabo Pan
- Department of Medical Technology, Qiandongnan Vocational & Technical College for Nationalities, Kaili, 556000, China
| | - Xiaojun Yao
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Fei Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China
| | - Guolin Zhang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China.
| | - Yinggang Luo
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China.
| |
Collapse
|
22
|
Bomb K, Pradhan L, Zhang Q, Jarai BM, Bhattacharjee A, Burris DL, Kloxin AM, Fromen CA. Destructive fibrotic teamwork: how both microenvironment stiffness and profibrotic interleukin 13 impair alveolar macrophage phenotype and function. Biomater Sci 2022; 10:5689-5706. [PMID: 36018297 PMCID: PMC9632634 DOI: 10.1039/d2bm00828a] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The pulmonary fibrotic microenvironment is characterized by increased stiffness of lung tissue and enhanced secretion of profibrotic soluble cues contributing to a feedback loop that leads to dysregulated wound healing and lung failure. Pinpointing the individual and tandem effects of profibrotic stimuli in impairing immune cell response remains difficult and is needed for improved therapeutic strategies. We utilized a statistical design of experiment (DOE) to investigate how microenvironment stiffness and interleukin 13 (IL13), a profibrotic soluble factor linked with disease severity, contribute to the impaired macrophage response commonly observed in pulmonary fibrosis. We used engineered bioinspired hydrogels of different stiffness, ranging from healthy to fibrotic lung tissue, and cultured murine alveolar macrophages (MH-S cells) with or without IL13 to quantify cell response and analyze independent and synergistic effects. We found that, while both stiffness and IL13 independently influence macrophage morphology, phenotype, phagocytosis and efferocytosis, these factors work synergistically to exacerbate impaired macrophage phenotype and efferocytosis. These unique findings provide insights into how macrophages in fibrotic conditions are not as effective in clearing debris, contributing to fibrosis initiation/progression, and more broadly inform how underlying drivers of fibrosis modulate immune cell response to facilitate therapeutic strategies.
Collapse
Affiliation(s)
- Kartik Bomb
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | - Lina Pradhan
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | - Qi Zhang
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | - Bader M Jarai
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | | | - David L Burris
- Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - April M Kloxin
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
- Material Science and Engineering, University of Delaware, Newark, DE, USA
| | - Catherine A Fromen
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
23
|
Computational simulation of liver fibrosis dynamics. Sci Rep 2022; 12:14112. [PMID: 35982187 PMCID: PMC9388486 DOI: 10.1038/s41598-022-18123-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/05/2022] [Indexed: 11/08/2022] Open
Abstract
Liver fibrosis is a result of homeostasis breakdown caused by repetitive injury. The accumulation of collagens disrupts liver structure and function, which causes serious consequences such as cirrhosis. Various mathematical simulation models have been developed to understand these complex processes. We employed the agent-based modelling (ABM) approach and implemented inflammatory processes in central venous regions. Collagens were individually modelled and visualised depending on their origin: myofibroblast and portal fibroblast. Our simulation showed that the administration of toxic compounds induced accumulation of myofibroblast-derived collagens in central venous regions and portal fibroblast-derived collagens in portal areas. Subsequently, these collagens were bridged between central-central areas and spread all over areas. We confirmed the consistent dynamic behaviour of collagen formulation in our simulation and from histological sections obtained via in vivo experiments. Sensitivity analyses identified dead hepatocytes caused by inflammation and the ratio of residential liver cells functioned as a cornerstone for the initiation and progression of liver fibrosis. The validated mathematical model demonstrated here shows virtual experiments that are complementary to biological experiments, which contribute to understanding a new mechanism of liver fibrosis.
Collapse
|
24
|
Fu H, Chu L, Yuan Y, Liao S, Wang G. circACTR2
activates
M2
polarization of macrophages through activating
YAP
signaling and contributes to renal fibrosis. Immunology 2022; 167:606-621. [DOI: 10.1111/imm.13558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/09/2022] [Indexed: 11/26/2022] Open
Affiliation(s)
- Hua Fu
- Department of Pathology, Third Xiangya Hospital Central South University Changsha Hunan Province P.R. China
| | - Ling Chu
- Department of Pathology, Third Xiangya Hospital Central South University Changsha Hunan Province P.R. China
| | - Yi‐Shu Yuan
- Department of Pathology, Third Xiangya Hospital Central South University Changsha Hunan Province P.R. China
| | - Shan Liao
- Department of Pathology, Third Xiangya Hospital Central South University Changsha Hunan Province P.R. China
| | - Guo‐Hui Wang
- Gastrointestinal surgery, Third Xiangya Hospital Central South University Changsha Hunan Province P.R. China
| |
Collapse
|
25
|
Pisklova M, Osmak G, Favorova O. Regulation of SMAD Signaling Pathway by miRNAs Associated with Myocardial Fibrosis: In silico Analysis of Target Gene Networks. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:832-838. [PMID: 36171647 DOI: 10.1134/s0006297922080144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 06/16/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a hereditary heart disease caused by mutations in the sarcomere genes, which is accompanied by myocardial fibrosis leading to progressive heart failure and arrhythmias. Recent studies suggest that the HCM development involves dysregulation of gene expression. Among the molecules involved in this process are microRNAs (miRNAs), which are short non-coding RNAs. Typically, one miRNA regulates several target genes post-transcriptionally, hence, it might be difficult to determine the role of a particular miRNA in the disease pathogenesis. In this study, using the PubMed database, we selected 15 miRNAs whose expression is associated with myocardial fibrosis, one of the critical pathological processes in HCM. We then used an earlier developed algorithm to search in silico for the signaling pathways regulated by these miRNAs and found that ten of them participate in the regulation of the TGF-β/SMAD signaling pathway. At the same time, among the SMAD signaling pathway genes, the target of the most identified miRNAs was the MYC gene, which is involved in the development of fibrosis in some tissues. In our earlier work, we found that the TGF-β/SMAD pathway is also regulated by a set of other miRNAs associated with the myocardial hypertrophy in HCM. The fact that two sets of miRNAs identified in two independent bioinformatic studies are involved in the regulation of the same signaling pathway indicates that the SMAD signaling cascade is indeed a key element in the regulation of pathological processes in HCM. The obtained data might contribute to understanding pathological processes underlying HCM development.
Collapse
Affiliation(s)
- Maria Pisklova
- Chazov National Medical Research Center of Cardiology, Moscow, 121552, Russia.
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - German Osmak
- Chazov National Medical Research Center of Cardiology, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - Olga Favorova
- Chazov National Medical Research Center of Cardiology, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
26
|
Defining the Profile: Characterizing Cytokines in Tendon Injury to Improve Clinical Therapy. JOURNAL OF IMMUNOLOGY AND REGENERATIVE MEDICINE 2022; 16. [PMID: 35309714 PMCID: PMC8932644 DOI: 10.1016/j.regen.2022.100059] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cytokine manipulation has been widely used to bolster innate healing mechanisms in an array of modern therapeutics. While other anatomical locations have a more definitive analysis of cytokine data, the tendon presents unique challenges to detection that make a complete portrayal of cytokine involvement during injury unattainable thus far. Without this knowledge, the advancement of tendon healing modalities is limited. In this review, we discuss what is known of the cytokine profile within the injured tendinous environment and the unique obstacles facing cytokine detection in the tendon while proposing possible solutions to these challenges. IL-1β, TNF-α, and IL-6 in particular have been identified as key cytokines for initiating tendon healing, but their function and temporal expression are still not well understood. Methods used for cytokine evaluation in the tendon including cell culture, tissue biopsy, and microdialysis have their strengths and limitations, but new methods and approaches are needed to further this research. We conclude that future study design for cytokine detection in the injured tendon should meet set criteria to achieve definitive characterization of cytokine expression to guide future therapeutics.
Collapse
|
27
|
Chen H, Agrawal DK, Thankam FG. Biomaterials-Driven Sterile Inflammation. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:22-34. [PMID: 33213285 PMCID: PMC8892963 DOI: 10.1089/ten.teb.2020.0253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Performance of the biomaterials used for regenerative medicine largely depends on biocompatibility; however, the biological mechanisms underlying biocompatibility of a biomaterial within the host system is poorly understood. In addition to the classical immune response against non-self-entities, the sterile inflammatory response could limit the compatibility of biological scaffolds. Whereas the immediate to short-term host response to a biomaterial implant have been characterized, the long-term progression of host-biomaterial relationship has not been described. This article explores the novel concept of biomaterials-driven sterile inflammation (BSI) in long-term biodegradable implants and throws light for possible explanation for the onset of BSI and the associated damage-associated molecular patterns. The understanding of BSI would advance the current strategies to improve biomaterial-host tissue integration and open novel translational avenues in biomaterials-based tissue regeneration. Impact statement Understanding the novel concept of biomaterials-driven sterile inflammation and associated damage-associated molecular patterns in long-term biodegradable implants would determine their success and improves the tissue engineering and regenerative strategies.
Collapse
Affiliation(s)
- Henry Chen
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| | - Devendra K. Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| | - Finosh G. Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
- Address correspondence to: Finosh G. Thankam, PhD, Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA 91766-1854, USA
| |
Collapse
|
28
|
Russo V, El Khatib M, Prencipe G, Citeroni MR, Faydaver M, Mauro A, Berardinelli P, Cerveró-Varona A, Haidar-Montes AA, Turriani M, Di Giacinto O, Raspa M, Scavizzi F, Bonaventura F, Stöckl J, Barboni B. Tendon Immune Regeneration: Insights on the Synergetic Role of Stem and Immune Cells during Tendon Regeneration. Cells 2022; 11:434. [PMID: 35159244 PMCID: PMC8834336 DOI: 10.3390/cells11030434] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
Tendon disorders represent a very common pathology in today's population, and tendinopathies that account 30% of tendon-related injuries, affect yearly millions of people which in turn cause huge socioeconomic and health repercussions worldwide. Inflammation plays a prominent role in the development of tendon pathologies, and advances in understanding the underlying mechanisms during the inflammatory state have provided additional insights into its potential role in tendon disorders. Different cell compartments, in combination with secreted immune modulators, have shown to control and modulate the inflammatory response during tendinopathies. Stromal compartment represented by tenocytes has shown to display an important role in orchestrating the inflammatory response during tendon injuries due to the interplay they exhibit with the immune-sensing and infiltrating compartments, which belong to resident and recruited immune cells. The use of stem cells or their derived secretomes within the regenerative medicine field might represent synergic new therapeutical approaches that can be used to tune the reaction of immune cells within the damaged tissues. To this end, promising opportunities are headed to the stimulation of macrophages polarization towards anti-inflammatory phenotype together with the recruitment of stem cells, that possess immunomodulatory properties, able to infiltrate within the damaged tissues and improve tendinopathies resolution. Indeed, the comprehension of the interactions between tenocytes or stem cells with the immune cells might considerably modulate the immune reaction solving hence the inflammatory response and preventing fibrotic tissue formation. The purpose of this review is to compare the roles of distinct cell compartments during tendon homeostasis and injury. Furthermore, the role of immune cells in this field, as well as their interactions with stem cells and tenocytes during tendon regeneration, will be discussed to gain insights into new ways for dealing with tendinopathies.
Collapse
Affiliation(s)
- Valentina Russo
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Mohammad El Khatib
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maria Rita Citeroni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Melisa Faydaver
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Paolo Berardinelli
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Arlette A. Haidar-Montes
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maura Turriani
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Oriana Di Giacinto
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Marcello Raspa
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Ferdinando Scavizzi
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Fabrizio Bonaventura
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Johannes Stöckl
- Centre for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| |
Collapse
|
29
|
Bijarchian F, Taghiyar L, Azhdari Z, Baghaban Eslaminejad M. M2c Macrophages enhance phalange regeneration of amputated mice digits in an organ co-culture system. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1602-1612. [PMID: 35317116 PMCID: PMC8917845 DOI: 10.22038/ijbms.2021.57887.12870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/26/2021] [Indexed: 11/17/2022]
Abstract
Objectives Delayed anti-inflammatory responses and scar-formation are the main causes for inability of injured body parts such as phalanges to regrow in mammals. Salamanders can regenerate fully scar-free body structures, followed by the appearance of anti-inflammatory responses at the injured site immediately after amputation. This study aimed to evaluate the local regenerative effects of direct amplified anti-inflammatory signals on regeneration of amputated mice digit tips using M2c-macrophages in a co-cultured organ system for the first time. Materials and Methods We used the amputated digits from the paws of 18.5E day old C57BL/6J mice. Monocytes were obtained from peripheral blood and co-cultured with amputated digits, which subsequently enhanced the M2c macrophage phenotype induced by IL-10. We also examined the regenerative effects of IL-10 and transcription growth factor-beta 1 (TGF-β1). Results The regrowth of new tissue occurred 10 days post-amputation in all groups. This regrowth was related to enhanced Msh homeobox-1 (Msx1), Msh homeobox-2 (Msx2), and bone morphogenic protein-4 (Bmp4) genes. Increased expression of fibroblast growth factor-8 (Fgf-8) also increased the proliferation rate. Histological analyses indicated that epidermal-closure occurred at 3-dpa in all groups. We observed full digit tip regeneration in the co-cultured group. Particularly, there was new tissue regrowth observed with 40 µg/ml of IL-10 and 120 µg/ml of TGF-β. In contrast, the control group had no remarkable digit elongation. Conclusion We propose that a direct amplified anti-inflammatory response at the digit injury site can regenerate epithelial and mesenchymal tissues, and might be useful for limb regeneration without scar formation in adult mammals.
Collapse
Affiliation(s)
- Fatemeh Bijarchian
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Leila Taghiyar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Zahra Azhdari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran,Corresponding author: Mohamadreza Baghaban Eslaminejad. Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem Street, Tehran, Iran. Tel: +98-21-23562524; Fax: +98-21-23562507;
| |
Collapse
|
30
|
Nikovics K, Favier AL. Macrophage Identification In Situ. Biomedicines 2021; 9:1393. [PMID: 34680510 PMCID: PMC8533306 DOI: 10.3390/biomedicines9101393] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022] Open
Abstract
Understanding the processes of inflammation and tissue regeneration after injury is of great importance. For a long time, macrophages have been known to play a central role during different stages of inflammation and tissue regeneration. However, the molecular and cellular mechanisms by which they exert their effects are as yet mostly unknown. While in vitro macrophages have been characterized, recent progress in macrophage biology studies revealed that macrophages in vivo exhibited distinctive features. Actually, the precise characterization of the macrophages in vivo is essential to develop new healing treatments and can be approached via in situ analyses. Nowadays, the characterization of macrophages in situ has improved significantly using antigen surface markers and cytokine secretion identification resulting in specific patterns. This review aims for a comprehensive overview of different tools used for in situ macrophage identification, reporter genes, immunolabeling and in situ hybridization, discussing their advantages and limitations.
Collapse
Affiliation(s)
- Krisztina Nikovics
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France;
| | | |
Collapse
|
31
|
Chen Y, Yuan S, Cao Y, Kong G, Jiang F, Li Y, Wang Q, Tang M, Zhang Q, Wang Q, Liu L. Gasotransmitters: Potential Therapeutic Molecules of Fibrotic Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3206982. [PMID: 34594474 PMCID: PMC8478550 DOI: 10.1155/2021/3206982] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/31/2021] [Indexed: 02/06/2023]
Abstract
Fibrosis is defined as the pathological progress of excessive extracellular matrix (ECM), such as collagen, fibronectin, and elastin deposition, as the regenerative capacity of cells cannot satisfy the dynamic repair of chronic damage. The well-known features of tissue fibrosis are characterized as the presence of excessive activated and proliferated fibroblasts and the differentiation of fibroblasts into myofibroblasts, and epithelial cells undergo the epithelial-mesenchymal transition (EMT) to expand the number of fibroblasts and myofibroblasts thereby driving fibrogenesis. In terms of mechanism, during the process of fibrosis, the activations of the TGF-β signaling pathway, oxidative stress, cellular senescence, and inflammatory response play crucial roles in the activation and proliferation of fibroblasts to generate ECM. The deaths due to severe fibrosis account for almost half of the total deaths from various diseases, and few treatment strategies are available for the prevention of fibrosis as yet. Recently, numerous studies demonstrated that three well-defined bioactive gasotransmitters, including nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), generally exhibited anti-inflammatory, antioxidative, antiapoptotic, and antiproliferative properties. Besides these effects, a number of studies have reported that low-dose exogenous and endogenous gasotransmitters can delay and interfere with the occurrence and development of fibrotic diseases, including myocardial fibrosis, idiopathic pulmonary fibrosis, liver fibrosis, renal fibrosis, diabetic diaphragm fibrosis, and peritoneal fibrosis. Furthermore, in animal and clinical experiments, the inhalation of low-dose exogenous gas and intraperitoneal injection of gaseous donors, such as SNAP, CINOD, CORM, SAC, and NaHS, showed a significant therapeutic effect on the inhibition of fibrosis through modulating the TGF-β signaling pathway, attenuating oxidative stress and inflammatory response, and delaying the cellular senescence, while promoting the process of autophagy. In this review, we first demonstrate and summarize the therapeutic effects of gasotransmitters on diverse fibrotic diseases and highlight their molecular mechanisms in the process and development of fibrosis.
Collapse
Affiliation(s)
- Yingqing Chen
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Shuo Yuan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002 Jilin Province, China
| | - Yuying Cao
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Guangyao Kong
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Feng Jiang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - You Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Qi Wang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Minli Tang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Qinggao Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002 Jilin Province, China
| | - Qianqian Wang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Liping Liu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| |
Collapse
|
32
|
Macrophage exosomes transfer angiotensin II type 1 receptor to lung fibroblasts mediating bleomycin-induced pulmonary fibrosis. Chin Med J (Engl) 2021; 134:2175-2185. [PMID: 34483252 PMCID: PMC8478379 DOI: 10.1097/cm9.0000000000001605] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Macrophages are involved in the pathogenesis of idiopathic pulmonary fibrosis, partially by activating lung fibroblasts. However, how macrophages communicate with lung fibroblasts is largely unexplored. Exosomes can mediate intercellular communication, whereas its role in lung fibrogenesis is unclear. Here we aim to investigate whether exosomes can mediate the crosstalk between macrophages and lung fibroblasts and subsequently induce fibrosis. METHODS In vivo, bleomycin (BLM)-induced lung fibrosis model was established and macrophages infiltration was examined. The effects of GW4869, an exosomes inhibitor, on lung fibrosis were assessed. Moreover, macrophage exosomes were injected into mice to observe its pro-fibrotic effects. In vitro, exosomes derived from angiotensin II (Ang II)-stimulated macrophages were collected. Then, lung fibroblasts were treated with the exosomes. Twenty-four hours later, protein levels of α-collagen I, angiotensin II type 1 receptor (AT1R), transforming growth factor-β (TGF-β), and phospho-Smad2/3 (p-Smad2/3) in lung fibroblasts were examined. The Student's t test or analysis of variance were used for statistical analysis. RESULTS In vivo, BLM-treated mice showed enhanced infiltration of macrophages, increased fibrotic alterations, and higher levels of Ang II and AT1R. GW4869 attenuated BLM-induced pulmonary fibrosis. Mice with exosomes injection showed fibrotic features with higher levels of Ang II and AT1R, which was reversed by irbesartan. In vitro, we found that macrophages secreted a great number of exosomes. The exosomes were taken by fibroblasts and resulted in higher levels of AT1R (0.22 ± 0.02 vs. 0.07 ± 0.02, t = 8.66, P = 0.001), TGF-β (0.54 ± 0.05 vs. 0.09 ± 0.06, t = 10.00, P < 0.001), p-Smad2/3 (0.58 ± 0.06 vs. 0.07 ± 0.03, t = 12.86, P < 0.001) and α-collagen I (0.27 ± 0.02 vs. 0.16 ± 0.01, t = 7.01, P = 0.002), and increased Ang II secretion (62.27 ± 7.32 vs. 9.56 ± 1.68, t = 12.16, P < 0.001). Interestingly, Ang II increased the number of macrophage exosomes, and the protein levels of Alix (1.45 ± 0.15 vs. 1.00 ± 0.10, t = 4.32, P = 0.012), AT1R (4.05 ± 0.64 vs. 1.00 ± 0.09, t = 8.17, P = 0.001), and glyceraldehyde-3-phosphate dehydrogenase (2.13 ± 0.36 vs. 1.00 ± 0.10, t = 5.28, P = 0.006) were increased in exosomes secreted by the same number of macrophages, indicating a positive loop between Ang II and exosomes production. CONCLUSIONS Exosomes mediate intercellular communication between macrophages and fibroblasts plays an important role in BLM-induced pulmonary fibrosis.
Collapse
|
33
|
Demichev V, Tober-Lau P, Lemke O, Nazarenko T, Thibeault C, Whitwell H, Röhl A, Freiwald A, Szyrwiel L, Ludwig D, Correia-Melo C, Aulakh SK, Helbig ET, Stubbemann P, Lippert LJ, Grüning NM, Blyuss O, Vernardis S, White M, Messner CB, Joannidis M, Sonnweber T, Klein SJ, Pizzini A, Wohlfarter Y, Sahanic S, Hilbe R, Schaefer B, Wagner S, Mittermaier M, Machleidt F, Garcia C, Ruwwe-Glösenkamp C, Lingscheid T, Bosquillon de Jarcy L, Stegemann MS, Pfeiffer M, Jürgens L, Denker S, Zickler D, Enghard P, Zelezniak A, Campbell A, Hayward C, Porteous DJ, Marioni RE, Uhrig A, Müller-Redetzky H, Zoller H, Löffler-Ragg J, Keller MA, Tancevski I, Timms JF, Zaikin A, Hippenstiel S, Ramharter M, Witzenrath M, Suttorp N, Lilley K, Mülleder M, Sander LE, Ralser M, Kurth F. A time-resolved proteomic and prognostic map of COVID-19. Cell Syst 2021; 12:780-794.e7. [PMID: 34139154 PMCID: PMC8201874 DOI: 10.1016/j.cels.2021.05.005] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/24/2021] [Accepted: 05/07/2021] [Indexed: 12/14/2022]
Abstract
COVID-19 is highly variable in its clinical presentation, ranging from asymptomatic infection to severe organ damage and death. We characterized the time-dependent progression of the disease in 139 COVID-19 inpatients by measuring 86 accredited diagnostic parameters, such as blood cell counts and enzyme activities, as well as untargeted plasma proteomes at 687 sampling points. We report an initial spike in a systemic inflammatory response, which is gradually alleviated and followed by a protein signature indicative of tissue repair, metabolic reconstitution, and immunomodulation. We identify prognostic marker signatures for devising risk-adapted treatment strategies and use machine learning to classify therapeutic needs. We show that the machine learning models based on the proteome are transferable to an independent cohort. Our study presents a map linking routinely used clinical diagnostic parameters to plasma proteomes and their dynamics in an infectious disease.
Collapse
Affiliation(s)
- Vadim Demichev
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany; The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK; The University of Cambridge, Department of Biochemistry and Cambridge Centre for Proteomics, Cambridge CB21GA, UK
| | - Pinkus Tober-Lau
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Oliver Lemke
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | - Tatiana Nazarenko
- University College London, Department of Mathematics, London WC1E 6BT, UK; University College London, Department of Women's Cancer, EGA Institute for Women'S Health, London WC1E 6BT, UK
| | - Charlotte Thibeault
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Harry Whitwell
- National Phenome Centre and Imperial Clinical Phenotyping Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW72AZ, UK; Lobachevsky University, Department of Applied Mathematics, Nizhny Novgorod 603105, Russia; Imperial College London, Section of Bioanalytical Chemistry, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, London SW7 2AZ, UK
| | - Annika Röhl
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | - Anja Freiwald
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | - Lukasz Szyrwiel
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK
| | - Daniela Ludwig
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | - Clara Correia-Melo
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK
| | - Simran Kaur Aulakh
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK
| | - Elisa T Helbig
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Paula Stubbemann
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Lena J Lippert
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Nana-Maria Grüning
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | - Oleg Blyuss
- Lobachevsky University, Department of Applied Mathematics, Nizhny Novgorod 603105, Russia; University of Hertfordshire, School of Physics, Astronomy and Mathematics, Hatfield AL10 9AB, UK; Sechenov First Moscow State Medical University, Department of Paediatrics and Paediatric Infectious Diseases, Moscow 119435, Russia
| | - Spyros Vernardis
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK
| | - Matthew White
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK
| | - Christoph B Messner
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany; The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK
| | - Michael Joannidis
- Medical University Innsbruck, Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, 6020 Innsbruck, Austria
| | - Thomas Sonnweber
- Medical University of Innsbruck, Department of Internal Medicine II, 6020 Innsbruck, Austria
| | - Sebastian J Klein
- Medical University Innsbruck, Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, 6020 Innsbruck, Austria
| | - Alex Pizzini
- Medical University of Innsbruck, Department of Internal Medicine II, 6020 Innsbruck, Austria
| | - Yvonne Wohlfarter
- Medical University of Innsbruck, Institute of Human Genetics, 6020 Innsbruck, Austria
| | - Sabina Sahanic
- Medical University of Innsbruck, Department of Internal Medicine II, 6020 Innsbruck, Austria
| | - Richard Hilbe
- Medical University of Innsbruck, Department of Internal Medicine II, 6020 Innsbruck, Austria
| | - Benedikt Schaefer
- Medical University of Innsbruck, Christian Doppler Laboratory for Iron and Phosphate Biology, Department of Internal Medicine I, 6020 Innsbruck, Austria
| | - Sonja Wagner
- Medical University of Innsbruck, Christian Doppler Laboratory for Iron and Phosphate Biology, Department of Internal Medicine I, 6020 Innsbruck, Austria
| | - Mirja Mittermaier
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany; Berlin Institute of Health, 10178 Berlin, Germany
| | - Felix Machleidt
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Carmen Garcia
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Christoph Ruwwe-Glösenkamp
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Tilman Lingscheid
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Laure Bosquillon de Jarcy
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Miriam S Stegemann
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Moritz Pfeiffer
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Linda Jürgens
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Sophy Denker
- Charité Universitätsmedizin Berlin, Medical Department of Hematology, Oncology & Tumor Immunology, Virchow Campus & Molekulares Krebsforschungszentrum, 13353 Berlin, Germany; Berlin Institute of Health, 10178 Berlin, Germany
| | - Daniel Zickler
- Charité Universitätsmedizin Berlin, Department of Nephrology and Internal Intensive Care Medicine, 10117 Berlin, Germany
| | - Philipp Enghard
- Charité Universitätsmedizin Berlin, Department of Nephrology and Internal Intensive Care Medicine, 10117 Berlin, Germany
| | - Aleksej Zelezniak
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK; Chalmers Tekniska Högskola, Department of Biology and Biological Engineering, SE-412 96 Gothenburg, Sweden
| | - Archie Campbell
- University of Edinburgh, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, Edinburgh EH4 2XU, UK; University of Edinburgh, Usher Institute, Edinburgh EH16 4UX, UK
| | - Caroline Hayward
- University of Edinburgh, MRC Human Genetics Unit, Institute of Genetics and Cancer, Edinburgh EH4 2XU, UK
| | - David J Porteous
- University of Edinburgh, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, Edinburgh EH4 2XU, UK; University of Edinburgh, Usher Institute, Edinburgh EH16 4UX, UK
| | - Riccardo E Marioni
- University of Edinburgh, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, Edinburgh EH4 2XU, UK
| | - Alexander Uhrig
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Holger Müller-Redetzky
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Heinz Zoller
- Medical University of Innsbruck, Christian Doppler Laboratory for Iron and Phosphate Biology, Department of Internal Medicine I, 6020 Innsbruck, Austria
| | - Judith Löffler-Ragg
- Medical University of Innsbruck, Department of Internal Medicine II, 6020 Innsbruck, Austria
| | - Markus A Keller
- Medical University of Innsbruck, Institute of Human Genetics, 6020 Innsbruck, Austria
| | - Ivan Tancevski
- Medical University of Innsbruck, Department of Internal Medicine II, 6020 Innsbruck, Austria
| | - John F Timms
- University College London, Department of Women's Cancer, EGA Institute for Women'S Health, London WC1E 6BT, UK
| | - Alexey Zaikin
- University College London, Department of Mathematics, London WC1E 6BT, UK; University College London, Department of Women's Cancer, EGA Institute for Women'S Health, London WC1E 6BT, UK; Lobachevsky University, Laboratory of Systems Medicine of Healthy Ageing, Nizhny Novgorod 603105, Russia
| | - Stefan Hippenstiel
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany; German Centre for Lung Research, 35392 Gießen, Germany
| | - Michael Ramharter
- Bernhard Nocht Institute for Tropical Medicine, Department of Tropical Medicine, and University Medical Center Hamburg-Eppendorf, Department of Medicine, 20359 Hamburg, Germany
| | - Martin Witzenrath
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany; German Centre for Lung Research, 35392 Gießen, Germany
| | - Norbert Suttorp
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany; German Centre for Lung Research, 35392 Gießen, Germany
| | - Kathryn Lilley
- The University of Cambridge, Department of Biochemistry and Cambridge Centre for Proteomics, Cambridge CB21GA, UK
| | - Michael Mülleder
- Charité - Universitätsmedizin Berlin, Core Facility - High-Throughput Mass Spectrometry, 10117 Berlin, Germany
| | - Leif Erik Sander
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany; German Centre for Lung Research, 35392 Gießen, Germany
| | - Markus Ralser
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany; The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK.
| | - Florian Kurth
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany; Bernhard Nocht Institute for Tropical Medicine, Department of Tropical Medicine, and University Medical Center Hamburg-Eppendorf, Department of Medicine, 20359 Hamburg, Germany
| |
Collapse
|
34
|
Influence of Secretome of Different Functional Phenotypes of Macrophages on Proliferation, Differentiation, and Collagen-Producing Activity of Dermal Fibroblasts In Vitro. Bull Exp Biol Med 2021; 171:49-52. [PMID: 34057617 DOI: 10.1007/s10517-021-05170-y] [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: 09/04/2020] [Indexed: 10/21/2022]
Abstract
We studied the effect of conditioned media of GM-CSF-differentiated human macrophages polarized in M1(LPS), M2a(IL-4), M2c(dexamethasone), and M2(low serum) phenotypes on proliferation, differentiation, and collagen-producing activity of dermal fibroblasts. It was found that M1(LPS) and M2a(IL-4) were characterized by moderate influence on functional activity of fibroblasts. At the same time, soluble factors of M2c(dexamethasone) significantly enhanced the proliferative response of fibroblasts, but not their differentiation and type I collagen production. On the contrary, M2(low serum) generated under conditions of growth factors deficiency had a pronounced stimulating effect on the differentiation of fibroblasts and production of type I collagen by these cells, but moderately stimulated the fibroblast proliferation. Thus, the secretory activity of various functional phenotypes of macrophages is an important mechanism of fibrogenesis regulation.
Collapse
|
35
|
Pandey A, Kulshrestha R, Bansal SK. Dynamic role of LMW-hyaluronan fragments and Toll-like receptors 2,4 in progression of bleomycin induced lung parenchymal injury to fibrosis. THE EGYPTIAN JOURNAL OF BRONCHOLOGY 2021. [PMCID: PMC8138115 DOI: 10.1186/s43168-021-00073-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Pulmonary fibrosis (PF) is a progressive and lethal lung disease of elderly whose incidence has been increasing following the Covid-19 pandemic caused by severe acute respiratory syndrome corona virus 2 (SARS-CoV-2). PF immunopathogenesis involves progressive alveolar epithelial cell damage, release of damage-associated molecular patterns (DAMPs), and extracellular matrix (ECM) injury. We assessed the dynamic role of LMW-hyaluronan (LMW-HA) as DAMP in initiation of host immune TLR-2,4 responses and as determinant in progression of ECM injury to fibrosis. Male Wistar rats were divided into Group I (saline control, n = 24) and Group II (intratracheal bleomycin, 7 U/kg/animal, n = 24). Animals were euthanized on 0, 7, 14, and 28 days. The time course of release of LMW-HA, TLR-2,4 mRNA and protein levels, and NF-κB-p65 levels after bleomycin injury were correlated with the development of parenchymal inflammation, remodelling, and fibrosis. Results Acute lung injury caused by bleomycin significantly increases the pro-inflammatory LMW-HA levels and elevates TLR-2,4 levels on day 7. Subsequently, TLR-2 upregulation, TLR-4 downregulation, and NF-κB signalling follow on days 14 and 28. This results in progressive tissue inflammation, alveolar and interstitial macrophage accumulation, and fibrosis. Conclusions LMW-HA significantly increases in PF caused by non-infectious and infectious (Covid-19) etiologies. The accumulating HA fragments function as endogenous DAMPs and trigger inflammatory responses, through differential TLR2 and TLR4 signalling, thus promoting inflammation and macrophage influx. LMW-HA are reflective of the state of ongoing tissue inflammation and may be considered as a natural biosensor for fibrotic lung diseases and as potential therapeutic targets.
Collapse
|
36
|
Sicherre E, Favier AL, Riccobono D, Nikovics K. Non-Specific Binding, a Limitation of the Immunofluorescence Method to Study Macrophages In Situ. Genes (Basel) 2021; 12:649. [PMID: 33925331 PMCID: PMC8145419 DOI: 10.3390/genes12050649] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 12/24/2022] Open
Abstract
Advances in understanding tissue regenerative mechanisms require the characterization of in vivo macrophages as those play a fundamental role in this process. This characterization can be approached using the immuno-fluorescence method with widely studied and used pan-markers such as CD206 protein. This work investigated CD206 expression in an irradiated-muscle pig model using three different antibodies. Surprisingly, the expression pattern during immunodetection differed depending on the antibody origin and could give some false results. False results are rarely described in the literature, but this information is essential for scientists who need to characterize macrophages. In this context, we showed that in situ hybridization coupled with hybridization-chain-reaction detection (HCR) is an excellent alternative method to detect macrophages in situ.
Collapse
Affiliation(s)
- Emma Sicherre
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France; (E.S.); (A.-L.F.)
| | - Anne-Laure Favier
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France; (E.S.); (A.-L.F.)
| | - Diane Riccobono
- Radiobiology Unit, Department of NRBC Defence, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France;
| | - Krisztina Nikovics
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France; (E.S.); (A.-L.F.)
| |
Collapse
|
37
|
Yang CY, Chang PY, Chen JY, Wu BS, Yang AH, Lee OKS. Adipose-derived mesenchymal stem cells attenuate dialysis-induced peritoneal fibrosis by modulating macrophage polarization via interleukin-6. Stem Cell Res Ther 2021; 12:193. [PMID: 33741073 PMCID: PMC7977319 DOI: 10.1186/s13287-021-02270-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/05/2021] [Indexed: 12/15/2022] Open
Abstract
Background Life-long peritoneal dialysis (PD) as a renal replacement therapy is limited by peritoneal fibrosis. Previous studies showed immunomodulatory and antifibrotic effects of adipose-derived mesenchymal stem cells (ADSCs) on peritoneal fibrosis. However, the role of the peritoneal macrophage in this process remains uninvestigated. Methods We examined the therapeutic effects of ADSC and bone marrow-derived mesenchymal stem cells (BM-MSC) in the rat model of dialysis-induced peritoneal fibrosis using methylglyoxal. In addition, treatment of macrophages with the conditioned medium of ADSC and BM-MSC was performed individually to identify the beneficial component of the stem cell secretome. Results In the in vivo experiments, we found dialysis-induced rat peritoneal fibrosis was attenuated by both ADSC and BM-MSC. Interestingly, ADSC possessed a more prominent therapeutic effect than BM-MSC in ameliorating peritoneal membrane thickening while also upregulating epithelial cell markers in rat peritoneal tissues. The therapeutic effects of ADSC were positively associated with M2 macrophage polarization. In the in vitro experiments, we confirmed that interleukin-6 (IL-6) secreted by MSCs upon transforming growth factor-β1 stimulation promotes M2 macrophage polarization. Conclusions In dialysis-induced peritoneal fibrosis, MSCs are situated in an inflammatory environment of TGF-β1 and secrete IL-6 to polarize macrophages into the M2 phenotype. Our findings reveal a previously unidentified role of tissue macrophage in this antifibrotic process. ADSC has the advantage of abundance and accessibility, making the application values extremely promising. Graphical abstract In dialysis-induced peritoneal fibrosis, peritoneal mesothelial cells secrete transforming growth factor-β1 (TGF-β1) when exposed to methylglyoxal (MGO)-containing peritoneal dialysate. When situated in TGF-β1, the inflammatory environment induces mesenchymal stem cells to secrete interleukin-6 (IL-6), IL-6 polarizes macrophages into the M2 phenotype. The dominant peritoneal tissue M2 macrophages, marked by upregulated Arg-1 expression, account for the attenuation of MGO-induced dedifferentiation of peritoneal mesothelial cells to maintain epithelial integrity.
![]() Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02270-4.
Collapse
Affiliation(s)
- Chih-Yu Yang
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, 2F, Shou-Ren Bldg., No.155, Sec.2, Li-Nong St., Beitou Dist, Taipei, 11221, Taiwan. .,Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan. .,Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, 11217, Taiwan. .,Stem Cell Research Center, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan. .,Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), Hsinchu, 30010, Taiwan.
| | - Pu-Yuan Chang
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, 2F, Shou-Ren Bldg., No.155, Sec.2, Li-Nong St., Beitou Dist, Taipei, 11221, Taiwan
| | - Jun-Yi Chen
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, 2F, Shou-Ren Bldg., No.155, Sec.2, Li-Nong St., Beitou Dist, Taipei, 11221, Taiwan
| | - Bo-Sheng Wu
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - An-Hang Yang
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, 2F, Shou-Ren Bldg., No.155, Sec.2, Li-Nong St., Beitou Dist, Taipei, 11221, Taiwan.,Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan.,Department of Pathology, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
| | - Oscar Kuang-Sheng Lee
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, 2F, Shou-Ren Bldg., No.155, Sec.2, Li-Nong St., Beitou Dist, Taipei, 11221, Taiwan.,Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan.,Stem Cell Research Center, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan.,Department of Orthopedics, China Medical University Hospital, Taichung, 40447, Taiwan
| |
Collapse
|
38
|
Afroj T, Mitsuhashi A, Ogino H, Saijo A, Otsuka K, Yoneda H, Tobiume M, Nguyen NT, Goto H, Koyama K, Sugimoto M, Kondoh O, Nokihara H, Nishioka Y. Blockade of PD-1/PD-L1 Pathway Enhances the Antigen-Presenting Capacity of Fibrocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:1204-1214. [PMID: 33504617 PMCID: PMC7939041 DOI: 10.4049/jimmunol.2000909] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023]
Abstract
Fibrocytes, a distinct population of collagen-producing, monocyte-derived cells, are involved in wound healing as well as fibrotic diseases. Recently, fibrocytes have been revealed to play a role in the tumor microenvironment, particularly under antiangiogenic therapy. In addition, combination cancer immunotherapy with immune checkpoint inhibitor and antiangiogenic agents have been developed for various cancers in the clinical setting, although the immunological background is not clear. In the current study, we aimed to determine the function of fibrocytes in tumor immunity induced by immune checkpoint inhibitor therapy. Human and murine fibrocytes were generated from PBMCs and lungs, respectively. The expression of costimulatory and inhibitory molecules on fibrocytes was examined by flow cytometry. The stimulation of CD8+ T cells by fibrocytes was examined in MLRs with a 3H-thymidine incorporation assay. Fibrocytes expressed CD80low and CD86high as a costimulatory molecule, and expressed PD-L1high, but not PD-L2, as a coinhibitory molecule. Without any stimulation, fibrocytes strongly enhanced the proliferation of CD8+ T cells in mice and humans. Treatment with anti-CD86 and -CD54 Abs inhibited the growth of CD8+ T cells induced by fibrocytes. Anti-PD-L1 Ab further enhanced the proliferation of CD8+ T cells, even in the OVA-specific MLR with OT-1Rag-/- mice. Importantly, fibrocytes derived from PBMCs of patients with lung adenocarcinoma or murine MC38 tumors augmented the proliferation of CD8+ T cells with PD-L1 blockade. These results suggest that fibrocytes infiltrating tumor sites may play a role in the antitumor immunity mediated by CD8+ T cells when the activity is further enhanced by PD-L1/PD-1 blockade.
Collapse
Affiliation(s)
- Tania Afroj
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Atsushi Mitsuhashi
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Hirokazu Ogino
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Atsuro Saijo
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Kenji Otsuka
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Hiroto Yoneda
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Makoto Tobiume
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Na Thi Nguyen
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Hisatsugu Goto
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Kazuya Koyama
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Masamichi Sugimoto
- Product Research Department, Kamakura Research Laboratories, Chugai Pharmaceutical, Co., Ltd., Kanagawa 247-8530, Japan
| | - Osamu Kondoh
- Product Research Department, Kamakura Research Laboratories, Chugai Pharmaceutical, Co., Ltd., Kanagawa 247-8530, Japan
| | - Hiroshi Nokihara
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| |
Collapse
|
39
|
Sok MCP, Baker N, McClain C, Lim HS, Turner T, Hymel L, Ogle M, Olingy C, Palacios JI, Garcia JR, Srithar K, García AJ, Qiu P, Botchwey EA. Dual delivery of IL-10 and AT-RvD1 from PEG hydrogels polarize immune cells towards pro-regenerative phenotypes. Biomaterials 2021; 268:120475. [PMID: 33321293 PMCID: PMC11129952 DOI: 10.1016/j.biomaterials.2020.120475] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 09/29/2020] [Accepted: 10/18/2020] [Indexed: 02/06/2023]
Abstract
Inflammation after traumatic injury or surgical intervention is both a protective tissue response leading to regeneration and a potential cause of wound complications. One potentially successful strategy to harness to pro-regenerative roles of host inflammation is the localized delivery of bioactive materials to induce immune suppressive cellular responses by cells responding to injury. In this study, we designed a fully synthetic poly (ethylene) glycol (PEG)-based hydrogel to release the specialized pro-resolving lipid mediator aspirin-triggered resolvin-D1 (AT-RvD1) and recombinant human interleukin 10 (IL-10). We utilized a unique side-by-side internally controlled implant design wherein bioactive hydrogels were implanted adjacent to control hydrogels devoid of immune modulatory factors in the dorsal skinfold window chamber. We also explored single-immune cell data with unsupervised approaches such as SPADE. First, we show that RGD-presenting hydrogel delivery results in enhanced immune cell recruitment to the site of injury. We then use intra-vital imaging to assess cellular recruitment and microvascular remodeling to show an increase in the caliber and density of local microvessels. Finally, we show that the recruitment and re-education of mononuclear phagocytes by combined delivery IL-10 and AT-RvD1 localizes immune suppressive subsets to the hydrogel, including CD206+ macrophages (M2a/c) and IL-10 expressing dendritic cells in the context of chronic inflammation following surgical tissue disruption. These data demonstrate the potential of combined delivery on the recruitment of regenerative cell subsets involved in wound healing complications.
Collapse
Affiliation(s)
- Mary Caitlin P Sok
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Emory University Medical Scientist Training Program, USA
| | - Nusaiba Baker
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Emory University Medical Scientist Training Program, USA
| | - Claire McClain
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Hong Seo Lim
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Thomas Turner
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Lauren Hymel
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Molly Ogle
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Claire Olingy
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Joshua I Palacios
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - José R Garcia
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Krithik Srithar
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Andrés J García
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Peng Qiu
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Edward A Botchwey
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
40
|
Nikovics K, Morin H, Riccobono D, Bendahmane A, Favier AL. Hybridization-chain-reaction is a relevant method for in situ detection of M2d-like macrophages in a mini-pig model. FASEB J 2020; 34:15675-15686. [PMID: 33078886 DOI: 10.1096/fj.202001496r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/20/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022]
Abstract
Macrophages are a heterogeneous population of cells with an important role in innate immunity and tissue regeneration. Based on in vitro experiments, macrophages have been subdivided into five distinct subtypes named M1, M2a, M2b, M2c, and M2d, depending on the means of their activation and the cell surface markers they display. Whether all subtypes can be detected in vivo is still unclear. The identification of macrophages in vivo in the regenerating muscle could be used as a new diagnostic tool to monitor therapeutic strategies for tissue repair. The use of classical immunolabeling techniques is unable to discriminate between different M2 macrophages and a functional characterization of these macrophages is lacking. Using in situ hybridization coupled with hybridization-chain-reaction detection (HCR), we achieved the identification of M2d-like macrophages within regenerating muscle and applied this technique to understand the role of M2 macrophages in the regeneration of irradiated pig-muscle after adipose tissue stem cell treatment. Our work highlights the limits of immunolabeling and the usefulness of HCR analysis to provide valuable information for macrophage characterization.
Collapse
Affiliation(s)
- Krisztina Nikovics
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, Brétigny-sur-Orge Cedex, France
| | - Halima Morin
- Institute of Plant Sciences Paris-Saclay (IPS2), University Paris-Saclay, INRAE, CNRS, Univ Evry, Orsay, France
| | - Diane Riccobono
- Radiobiology Unit, Department of NRBC Defens, French Armed Forces Biomedical Research Institute, Brétigny-sur-Orge Cedex, France
| | - Abdelhafid Bendahmane
- Institute of Plant Sciences Paris-Saclay (IPS2), University Paris-Saclay, INRAE, CNRS, Univ Evry, Orsay, France
| | - Anne-Laure Favier
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, Brétigny-sur-Orge Cedex, France
| |
Collapse
|
41
|
Characterization of macrophages, giant cells and granulomas during muscle regeneration after irradiation. Cytokine 2020; 137:155318. [PMID: 33045525 DOI: 10.1016/j.cyto.2020.155318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/12/2020] [Accepted: 09/22/2020] [Indexed: 12/26/2022]
Abstract
Macrophages play a fundamental role in the different stages of muscle regeneration although the precise mechanisms involved are not entirely understood. Here we investigated the types of macrophages and cytokines that appeared in muscles after local gamma irradiation of mini-pigs that underwent no subsequent treatment or received three successive adipose tissue-derived stem cell (ASC) injections. Although some variability was observed among the three animals included in each study group, a general picture emerged. No macrophages appeared in control muscles from regions that had not been irradiated nor in muscles from irradiated regions derived from two animals. A third irradiated, but untreated animal, with characteristic muscle fibrosis and necrosis due to irradiation, showed invasion of M2 macrophages within small muscle lesions. In contrast, among the three ASC-treated and irradiated animals, one of them had completely recovered normal muscle architecture at the time of sampling with no invading macrophages, muscle from a second one contained mostly M1 macrophages and some M2-like macrophages whereas muscle from a third one displayed granulomas and giant cells. ASC treatment was associated with the presence of similar levels of pro-inflammatory cytokines within the two animals in the process of muscle regeneration whereas the levels of IL-4 and IL-10 expression were distinct from one animal to another. Microspectrofluorimetry and in situ hybridization revealed strong expression of TGF-β1 and TNFα in regenerating muscle. Overall, the data confirm the critical role of macrophages in muscle regeneration and suggest the involvement of a complex network of cytokine expression for successful recovery.
Collapse
|
42
|
Anisiewicz A, Pawlik A, Filip-Psurska B, Wietrzyk J. Differential Impact of Calcitriol and Its Analogs on Tumor Stroma in Young and Aged Ovariectomized Mice Bearing 4T1 Mammary Gland Cancer. Int J Mol Sci 2020; 21:E6359. [PMID: 32887237 PMCID: PMC7503326 DOI: 10.3390/ijms21176359] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022] Open
Abstract
(1) Background: Vitamin D compounds (VDC) are extensively studied in the field of anticancer properties, including breast cancer. Previously, we showed that calcitriol and its analogs (PRI-2191 and PRI-2205) stimulate metastasis in 4T1 murine mammary gland cancer models in young mice, whereas the reverse effect was observed in aged ovariectomized (OVX) mice; (2) Methods: We determined the phenotype of monocytes/macrophages using FACS and examined the expression of selected genes and proteins by Real-Time PCR and ELISA; (3) Results: Activities of VDC are accompanied by an increase in the percentage of Ly6Clow anti-inflammatory monocytes in the spleen of young and a decrease in aged OVX mice. Treatment of young mice with VDC resulted in an increase of CCL2 plasma and tumor concentration and Arg1 in tumor. In later stage of tumor progression the expression of genes related to metastasis in lung tissue was decreased or increased, in old OVX or young mice, respectively; (4) Conclusions: Pro- or anti-metastatic effects of calcitriol and its analogs in young or aged OVX mice, respectively, can be attributed to the differences in the effects of VDC on the tumor microenvironment, as a consequence of differences in the immunity status of young and aged mice.
Collapse
Affiliation(s)
- Artur Anisiewicz
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.P.); (B.F.-P.); (J.W.)
| | | | | | | |
Collapse
|
43
|
Trichostatin A Alleviates Renal Interstitial Fibrosis Through Modulation of the M2 Macrophage Subpopulation. Int J Mol Sci 2020; 21:ijms21175966. [PMID: 32825118 PMCID: PMC7503910 DOI: 10.3390/ijms21175966] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 08/16/2020] [Accepted: 08/16/2020] [Indexed: 12/16/2022] Open
Abstract
Mounting evidence indicates that an increase in histone deacetylation contributes to renal fibrosis. Although inhibition of histone deacetylase (HDAC) can reduce the extent of fibrosis, whether HDAC inhibitors exert the antifibrotic effect through modulating the phenotypes of macrophages, the key regulator of renal fibrosis, remains unknown. Moreover, the functional roles of the M2 macrophage subpopulation in fibrotic kidney diseases remain incompletely understood. Herein, we investigated the role of HDAC inhibitors on renal fibrogenesis and macrophage plasticity. We found that HDAC inhibition by trichostatin A (TSA) reduced the accumulation of interstitial macrophages, suppressed the activation of myofibroblasts and attenuated the extent of fibrosis in obstructive nephropathy. Moreover, TSA inhibited M1 macrophages and augmented M2 macrophage infiltration in fibrotic kidney tissue. Interestingly, TSA preferentially upregulated M2c macrophages and suppressed M2a macrophages in the obstructed kidneys, which was correlated with a reduction of interstitial fibrosis. TSA also repressed the expression of proinflammatory and profibrotic molecules in cultured M2a macrophages and inhibited the activation of renal myofibroblasts. In conclusion, our study was the first to show that HDAC inhibition by TSA alleviates renal fibrosis in obstructed kidneys through facilitating an M1 to M2c macrophage transition.
Collapse
|
44
|
Masaki S, Hashimoto Y, Kunisho S, Kimoto A, Kitadai Y. Fatty change of the liver microenvironment influences the metastatic potential of colorectal cancer. Int J Exp Pathol 2020; 101:162-170. [PMID: 32783302 PMCID: PMC7495750 DOI: 10.1111/iep.12371] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
Fatty liver is the most common cause of liver disease, and its prevalence has been increasing globally. Colorectal cancer (CRC) accounts for approximately 10% of all cancers and metastasizes most commonly to the liver. Paget's ‘Seed and Soil’ theory of metastasis proposed that the secondary growth of cancer cells is dependent on the distal organ microenvironment. This implies that the risk of metastasis may change due to changes in the microenvironment of target organs. However, the association between steatosis, fatty change in the liver microenvironment, and liver metastasis has not been clarified. Here, we induced fatty liver conditions in BALB/c mice using a choline‐deficient high‐fat diet with 0.1% methionine (CDAHFD) and then injected the CT26 cells to produce experimental metastasis. The number of metastatic tumours was significantly increased in mice with severe fatty liver as compared to control mice. The average size of metastatic tumours was smaller in mice with moderate fatty liver than in control mice. The stromal components, including cancer‐associated fibroblasts, tumour‐associated macrophages and tumour‐infiltrating lymphocytes, were also examined. Metastatic tumours in fatty liver showed invasive growth patterns without a fibrotic capsule. Compared to control groups, the polarization of macrophages and subtypes of tumour‐infiltrating lymphocytes differed depending on the extent of fatty liver progression. These results indicated that fatty changes in the liver influenced liver metastasis of CRC. Although moderate fatty changes suppress the growth of metastatic tumours in the liver, a severe fatty microenvironment may promote invasion and metastasis through alteration of the tumour microenvironment (TME).
Collapse
Affiliation(s)
- Satoshi Masaki
- Department of Health and Science, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Yoshimi Hashimoto
- Department of Health and Science, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Shoma Kunisho
- Department of Health and Science, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Akiko Kimoto
- Department of Health and Science, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Yasuhiko Kitadai
- Department of Health and Science, Prefectural University of Hiroshima, Hiroshima, Japan
| |
Collapse
|
45
|
Metabolic immunomodulation of macrophage functional plasticity in nonhealing wounds. Curr Opin Infect Dis 2020; 32:204-209. [PMID: 30950855 DOI: 10.1097/qco.0000000000000550] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Despite modern advances in medicine, nonhealing wounds are the number one cause of nontraumatic, lower-limb amputation. Nonhealing wounds are characterized by a healing process stalled between inflammation and tissue remodel/repair, a stage characterized by a shift in macrophage functional phenotype. Characterization of diversity in macrophage functional phenotype in wounds and metabolic contributions to macrophage polarization are discussed. RECENT FINDINGS Macrophage functional diversity in phenotype has recently evolved from duality (classically activated, pro-inflammatory M1 and alternatively activated, anti-inflammatory M2) to include an additional four alternately activated subphenotypes (M2a, M2b, M2c and M2d). Metabolic pathway utilization shifts characterize macrophage polarization with resulting metabolic and immune outcomes impacting host-pathogen interactions during wound healing. SUMMARY Recognition of the key role macrophage diversity plays in wound healing, along with better characterization of diverse macrophage phenotypes, will inform our understanding of pathogenicity in wound healing. Comprehensive profiling of the metabolism regulating macrophage polarization and host-pathogen interaction creates opportunity of discovery for innovative new diagnostics and therapeutics for treating nonhealing wounds.
Collapse
|
46
|
Pupkaite J, Sedlakova V, Eren Cimenci C, Bak M, McLaughlin S, Ruel M, Alarcon EI, Suuronen EJ. Delivering More of an Injectable Human Recombinant Collagen III Hydrogel Does Not Improve Its Therapeutic Efficacy for Treating Myocardial Infarction. ACS Biomater Sci Eng 2020; 6:4256-4265. [PMID: 33463355 DOI: 10.1021/acsbiomaterials.0c00418] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Injectable hydrogels are a promising method to enhance repair in the heart after myocardial infarction (MI). However, few studies have compared different strategies for the application of biomaterial treatments. In this study, we use a clinically relevant mouse MI model to assess the therapeutic efficacy of different treatment protocols for intramyocardial injection of a recombinant human collagen III (rHCIII) thermoresponsive hydrogel. Comparing a single hydrogel injection at an early time point (3 h) versus injections at multiple time points (3 h, 1 week, and 2 weeks) post-MI revealed that the single injection group led to superior cardiac function, reduced scar size and inflammation, and increased vascularization. Omitting the 3 h time point and delivering the hydrogel at 1 and 2 weeks post-MI led to poorer cardiac function. The positive effects of the single time point injection (3 h) on scar size and vascular density were lost when the hydrogel's collagen concentration was increased from 1% to 2%, and it did not confer any additional functional improvement. This study shows that early treatment with a rHCIII hydrogel can improve cardiac function post-MI but that injecting more rHCIII (by increased concentration or more over time) can reduce its efficacy, thus highlighting the importance of investigating optimal treatment strategies of biomaterial therapy for MI.
Collapse
Affiliation(s)
- Justina Pupkaite
- BEaTS Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, K1Y 4W7 Ontario, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, K1H 8M5 Ontario, Canada.,Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, Linköping 582 25, Sweden
| | - Veronika Sedlakova
- BEaTS Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, K1Y 4W7 Ontario, Canada
| | - Cagla Eren Cimenci
- BEaTS Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, K1Y 4W7 Ontario, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, K1H 8M5 Ontario, Canada
| | - Madison Bak
- BEaTS Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, K1Y 4W7 Ontario, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, K1H 8M5 Ontario, Canada
| | - Sarah McLaughlin
- BEaTS Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, K1Y 4W7 Ontario, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, K1H 8M5 Ontario, Canada
| | - Marc Ruel
- BEaTS Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, K1Y 4W7 Ontario, Canada
| | - Emilio I Alarcon
- BEaTS Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, K1Y 4W7 Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, K1H 8M5 Ontario, Canada
| | - Erik J Suuronen
- BEaTS Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, K1Y 4W7 Ontario, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, K1H 8M5 Ontario, Canada
| |
Collapse
|
47
|
Effendi WI, Nagano T, Hasan H, Yudhawati R. Immunoregulatory Property of C-Type Lectin-Like Receptors in Fibrosing Interstitial Lung Diseases. Int J Mol Sci 2020; 21:E3665. [PMID: 32455964 PMCID: PMC7279300 DOI: 10.3390/ijms21103665] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 11/16/2022] Open
Abstract
The innate immune system identifies exogenous threats or endogenous stress through germline-encoded receptors called pattern recognition receptors (PRRs) that initiate consecutive downstream signaling pathways to control immune responses. However, the contribution of the immune system and inflammation to fibrosing interstitial lung diseases (ILD) remains poorly understood. Immunoreceptor tyrosine-based motif-bearing C-type lectin-like receptors (CTLRs) may interact with various immune cells during tissue injury and wound repair processes. Dectin-1 is a CTLR with dominant mechanisms manifested through its intracellular signaling cascades, which regulate fibrosis-promoting properties through gene transcription and cytokine activation. Additionally, immune impairment in ILD facilitates microbiome colonization; hence, Dectin-1 is the master protector in host pulmonary defense against fungal invasion. Recent progress in determining the signaling pathways that control the balance of fibrosis has implicated immunoreceptor tyrosine-based motif-bearing CTLRs as being involved, either directly or indirectly, in the pathogenesis of fibrosing ILD.
Collapse
Affiliation(s)
- Wiwin Is Effendi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan;
- Department of Pulmonology and Respiratory Medicine, Medical Faculty of Airlangga University, Surabaya 60131, Indonesia; (H.H.); (R.Y.)
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan;
| | - Helmia Hasan
- Department of Pulmonology and Respiratory Medicine, Medical Faculty of Airlangga University, Surabaya 60131, Indonesia; (H.H.); (R.Y.)
| | - Resti Yudhawati
- Department of Pulmonology and Respiratory Medicine, Medical Faculty of Airlangga University, Surabaya 60131, Indonesia; (H.H.); (R.Y.)
| |
Collapse
|
48
|
Qadri M, Jay GD, Zhang LX, Richendrfer H, Schmidt TA, Elsaid KA. Proteoglycan-4 regulates fibroblast to myofibroblast transition and expression of fibrotic genes in the synovium. Arthritis Res Ther 2020; 22:113. [PMID: 32404156 PMCID: PMC7222325 DOI: 10.1186/s13075-020-02207-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/30/2020] [Indexed: 12/19/2022] Open
Abstract
Background Synovial tissue fibrosis is common in advanced OA with features including the presence of stress fiber-positive myofibroblasts and deposition of cross-linked collagen type-I. Proteoglycan-4 (PRG4) is a mucinous glycoprotein secreted by synovial fibroblasts and is a major component of synovial fluid. PRG4 is a ligand of the CD44 receptor. Our objective was to examine the role of PRG4-CD44 interaction in regulating synovial tissue fibrosis in vitro and in vivo. Methods OA synoviocytes were treated with TGF-β ± PRG4 for 24 h and α-SMA content was determined using immunofluorescence. Rhodamine-labeled rhPRG4 was incubated with OA synoviocytes ± anti-CD44 or isotype control antibodies and cellular uptake of rhPRG4 was determined following a 30-min incubation and α-SMA expression following a 24-h incubation. HEK-TGF-β cells were treated with TGF-β ± rhPRG4 and Smad3 phosphorylation was determined using immunofluorescence and TGF-β/Smad pathway activation was determined colorimetrically. We probed for stress fibers and focal adhesions (FAs) in TGF-β-treated murine fibroblasts and fibroblast migration was quantified ± rhPRG4. Synovial expression of fibrotic markers: α-SMA, collagen type-I, and PLOD2 in Prg4 gene-trap (Prg4GT) and recombined Prg4GTR animals were studied at 2 and 9 months of age. Synovial expression of α-SMA and PLOD2 was determined in 2-month-old Prg4GT/GT&Cd44−/− and Prg4GTR/GTR&Cd44−/− animals. Results PRG4 reduced α-SMA content in OA synoviocytes (p < 0.001). rhPRG4 was internalized by OA synoviocytes via CD44 and CD44 neutralization attenuated rhPRG4’s antifibrotic effect (p < 0.05). rhPRG4 reduced pSmad3 signal in HEK-TGF-β cells (p < 0.001) and TGF-β/Smad pathway activation (p < 0.001). rhPRG4 reduced the number of stress fiber-positive myofibroblasts, FAs mean size, and cell migration in TGF-β-treated NIH3T3 fibroblasts (p < 0.05). rhPRG4 inhibited fibroblast migration in a macrophage and fibroblast co-culture model without altering active or total TGF-β levels. Synovial tissues of 9-month-old Prg4GT/GT animals had higher α-SMA, collagen type-I, and PLOD2 (p < 0.001) content and Prg4 re-expression reduced these markers (p < 0.01). Prg4 re-expression also reduced α-SMA and PLOD2 staining in CD44-deficient mice. Conclusion PRG4 is an endogenous antifibrotic modulator in the joint and its effect on myofibroblast formation is partially mediated by CD44, but CD44 is not required to demonstrate an antifibrotic effect in vivo.
Collapse
Affiliation(s)
- Marwa Qadri
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Sciences Campus, 9401 Jeronimo Road, Irvine, CA, 92618, USA.,Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, 82826, Saudi Arabia
| | - Gregory D Jay
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Ling X Zhang
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Holly Richendrfer
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Tannin A Schmidt
- Biomedical Engineering Department, School of Dental Medicine, University of Connecticut, Farmington, CT, USA
| | - Khaled A Elsaid
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Sciences Campus, 9401 Jeronimo Road, Irvine, CA, 92618, USA.
| |
Collapse
|
49
|
Renaud L, da Silveira WA, Takamura N, Hardiman G, Feghali-Bostwick C. Prominence of IL6, IGF, TLR, and Bioenergetics Pathway Perturbation in Lung Tissues of Scleroderma Patients With Pulmonary Fibrosis. Front Immunol 2020; 11:383. [PMID: 32210969 PMCID: PMC7075854 DOI: 10.3389/fimmu.2020.00383] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/18/2020] [Indexed: 12/21/2022] Open
Abstract
Scleroderma-associated pulmonary fibrosis (SSc-PF) and idiopathic pulmonary fibrosis (IPF) are two of many chronic fibroproliferative diseases that are responsible for nearly 45% of all deaths in developed countries. While sharing several pathobiological characteristics, they also have very distinct features. Currently no effective anti-fibrotic treatments exist that can halt the progression of PF or reverse it. Our goal is to uncover potential gene targets for the development of anti-fibrotic therapies efficacious in both diseases, and those specific to SSc-PF, by identifying universal pathways and molecules driving fibrosis in SSc-PF and IPF tissues as well as those unique to SSc-PF. Using DNA microarray data, a meta-analysis of the differentially expressed (DE) genes in SSc-PF and IPF lung tissues (diseased vs. normal) was performed followed by a full systems level analysis of the common and unique transcriptomic signatures obtained. Protein-protein interaction networks were generated to identify hub proteins and explore the data using the centrality principle. Our results suggest that therapeutic strategies targeting IL6 trans-signaling, IGFBP2, IGFL2, and the coagulation cascade may be efficacious in both SSc-PF and IPF. Further, our data suggest that the expression of matrikine-producing collagens is also perturbed in PF. Lastly, an overall perturbation of bioenergetics, specifically between glycolysis and fatty acid metabolism, was uncovered in SSc-PF. Our findings provide insights into potential targets for the development of anti-fibrotic therapies that could be effective in both IPF and SSc-PF.
Collapse
Affiliation(s)
- Ludivine Renaud
- Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Willian A. da Silveira
- School of Biological Sciences, Institute for Global Food Security, Queens University Belfast, Belfast, United Kingdom
| | - Naoko Takamura
- Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Gary Hardiman
- Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
- School of Biological Sciences, Institute for Global Food Security, Queens University Belfast, Belfast, United Kingdom
| | - Carol Feghali-Bostwick
- Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
50
|
Saldarriaga OA, Freiberg B, Krishnan S, Rao A, Burks J, Booth AL, Dye B, Utay N, Ferguson M, Akil A, Yi M, Beretta L, Stevenson HL. Multispectral Imaging Enables Characterization of Intrahepatic Macrophages in Patients With Chronic Liver Disease. Hepatol Commun 2020; 4:708-723. [PMID: 32363321 PMCID: PMC7193134 DOI: 10.1002/hep4.1494] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/07/2020] [Indexed: 12/24/2022] Open
Abstract
Intrahepatic macrophages influence the composition of the microenvironment, host immune response to liver injury, and development of fibrosis. Compared with stellate cells, the role of macrophages in the development of fibrosis remains unclear. Multispectral imaging allows detection of multiple markers in situ in human formalin‐fixed, paraffin‐embedded tissue. This cutting‐edge technology is ideal for analyzing human liver tissues, as it allows spectral unmixing of fluorophore signals, subtraction of auto‐fluorescence, and preservation of hepatic architecture. We analyzed five different antibodies commonly observed on macrophage populations (CD68, MAC387, CD163, CD14, and CD16). After optimization of the monoplex stains and development of a Spectral Library, we combined all of the antibodies into a multiplex protocol and used them to stain biopsies collected from representative patients with chronic liver diseases, including chronic hepatitis C, nonalcoholic steatohepatitis, and autoimmune hepatitis. Various imaging modalities were tested, including cell phenotyping, tissue segmentation, t‐distributed stochastic neighbor embedding plots, and phenotype matrices that facilitated comparison and visualization of the identified macrophage and other cellular profiles. We then tested the feasibility of this platform to analyze numerous regions of interest from liver biopsies with multiple patients per group, using batch analysis algorithms. Five populations showed significant differences between patients positive for hepatitis C virus with advanced fibrosis when compared with controls. Three of these were significantly increased in patients with advanced fibrosis when compared to controls, and these included CD163+CD16+, CD68+, and CD68+MAC387+. Conclusion: Spectral imaging microscopy is a powerful tool that enables in situ analysis of macrophages and other cells in human liver biopsies and may lead to more personalized therapeutic approaches in the future.
Collapse
Affiliation(s)
| | | | - Santhoshi Krishnan
- Department of Electrical and Computer Engineering Rice University Houston TX.,Department of Computational Medicine and Bioinformatics University of Michigan Ann Arbor MI
| | - Arvind Rao
- Department of Electrical and Computer Engineering Rice University Houston TX.,Department of Computational Medicine and Bioinformatics University of Michigan Ann Arbor MI.,Department of Radiation Oncology University of Michigan Ann Arbor MI
| | - Jared Burks
- Department of Leukemia University of Texas MD Anderson Cancer Center Houston TX
| | - Adam L Booth
- Department of Pathology University of Texas Medical Branch Galveston TX
| | - Bradley Dye
- Department of Pathology University of Texas Medical Branch Galveston TX
| | - Netanya Utay
- Department of Internal Medicine University of Texas Health Science Center at Houston Houston TX
| | - Monique Ferguson
- Department of Internal Medicine University of Texas Medical Branch Galveston TX
| | - Abdellah Akil
- Department of Microbiology and Immunology University of Texas Medical Branch Galveston TX
| | - Minkyung Yi
- Department of Microbiology and Immunology University of Texas Medical Branch Galveston TX
| | - Laura Beretta
- Department of Molecular and Cellular Oncology University of Texas MD Anderson Cancer Center Houston TX
| | | |
Collapse
|