1
|
Bijai LK, Aboalela AA, Albalawi F, Khanagar SB, Iyer K. Role of mast cells in the pathogenesis of oral submucous fibrosis: a systematic review and meta-analysis. BMC Oral Health 2024; 24:1219. [PMID: 39402549 PMCID: PMC11476548 DOI: 10.1186/s12903-024-05025-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Oral Submucous Fibrosis (OSMF) is an oral potentially malignant disorder (OPMD) that commonly occurs in the South Asian population as there is high usage of areca nut. There has been extensive research on the pathogenesis and treatment of this condition. It is well-established in the scientific literature that mast cells (MC) have a definitive role in several inflammatory disorders. OSMF being a chronic inflammatory disorder, is also expected to have increased MCs. Hence, this review aims to evaluate the role of MCs in the pathogenesis of OSMF. METHODS A systematic search of articles was performed by two of the authors independently in PubMed, Scopus, Embase, Web of Science, and Google Scholar using the appropriate keywords and Boolean terms. The risk of bias was assessed using the Modified Newcastle-Ottawa Scale. The meta-analysis was performed with R studio software (Version: 4.4.0, Year: 2024, Company: The R foundation for statistical computing). RESULTS The search retrieved 36 studies, of which 16 were suitable for the review. There is evidence for a marked increase in the number of MCs in OSMF than the normal mucosa upon analyzing the retrieved articles. However, when comparing the grades of OSMF, there are variations in the reports. As all the retrieved articles were case-control studies, the risk of bias was analyzed using the Modified New Castle Ottawa Scale. All the studies scored in the good category (Score 6-9). The pooled effect size shows the Standard Mean Deviation (SMD) to be 0.09, 95% confidence interval (CI) [-0.18;0.37] to lie on either side of no effect. Hence the role of MCs in OSMF has not been established because of homogeneity and consistent sampling error. CONCLUSION Our systematic review does suggest a definitive role of mast cells in the progression of OSMF. However, there is a lack of methodological rigor in the included studies. This may contribute to diluting the results.
Collapse
Affiliation(s)
- Laliytha Kumar Bijai
- Maxillofacial Surgery and Diagnostic Sciences Department, College of Dentistry, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 11426, Saudi Arabia.
- King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh, 11481, Saudi Arabia.
| | - Ali A Aboalela
- Maxillofacial Surgery and Diagnostic Sciences Department, College of Dentistry, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 11426, Saudi Arabia
- King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh, 11481, Saudi Arabia
| | - Farraj Albalawi
- King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh, 11481, Saudi Arabia
- Preventive Dental Science Department, College of Dentistry, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 11426, Saudi Arabia
| | - Sanjeev B Khanagar
- King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh, 11481, Saudi Arabia
- Preventive Dental Science Department, College of Dentistry, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 11426, Saudi Arabia
| | - Kiran Iyer
- King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh, 11481, Saudi Arabia
- Preventive Dental Science Department, College of Dentistry, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 11426, Saudi Arabia
| |
Collapse
|
2
|
Atiakshin D, Kostin A, Alekhnovich A, Volodkin A, Ignatyuk M, Klabukov I, Baranovskii D, Buchwalow I, Tiemann M, Artemieva M, Medvedeva N, LeBaron TW, Noda M, Medvedev O. The Role of Mast Cells in the Remodeling Effects of Molecular Hydrogen on the Lung Local Tissue Microenvironment under Simulated Pulmonary Hypertension. Int J Mol Sci 2024; 25:11010. [PMID: 39456794 PMCID: PMC11507233 DOI: 10.3390/ijms252011010] [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: 09/06/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Molecular hydrogen (H2) has antioxidant, anti-inflammatory, and anti-fibrotic effects. In a rat model simulating pulmonary fibrotic changes induced by monocrotaline-induced pulmonary hypertension (MPH), we had previously explored the impact of inhaled H2 on lung inflammation and blood pressure. In this study, we further focused the biological effects of H2 on mast cells (MCs) and the parameters of the fibrotic phenotype of the local tissue microenvironment. MPH resulted in a significantly increased number of MCs in both the pneumatic and respiratory parts of the lungs, an increased number of tryptase-positive MCs with increased expression of TGF-β, activated interaction with immunocompetent cells (macrophages and plasma cells) and fibroblasts, and increased MC colocalization with a fibrous component of the extracellular matrix of connective tissue. The alteration in the properties of the MC population occurred together with intensified collagen fibrillogenesis and an increase in the integral volume of collagen and elastic fibers of the extracellular matrix of the pulmonary connective tissue. The exposure of H2 together with monocrotaline (MCT), despite individual differences between animals, tended to decrease the intrapulmonary MC population and the severity of the fibrotic phenotype of the local tissue microenvironment compared to changes in animals exposed to the MCT effect alone. In addition, the activity of collagen fibrillogenesis associated with MCs and the expression of TGF-β and tryptase in MCs decreased, accompanied by a reduction in the absolute and relative content of reticular and elastic fibers in the lung stroma. Thus, with MCT exposure, inhaled H2 has antifibrotic effects involving MCs in the lungs of rats. This reveals the unknown development mechanisms of the biological effects of H2 on the remodeling features of the extracellular matrix under inflammatory background conditions of the tissue microenvironment.
Collapse
Affiliation(s)
- Dmitrii Atiakshin
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Andrey Kostin
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
| | - Alexander Alekhnovich
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
| | - Artem Volodkin
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
| | - Michael Ignatyuk
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
| | - Ilya Klabukov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva st. 4, 249036 Obninsk, Russia (D.B.)
| | - Denis Baranovskii
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva st. 4, 249036 Obninsk, Russia (D.B.)
| | - Igor Buchwalow
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany;
| | - Markus Tiemann
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany;
| | - Marina Artemieva
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory 1-12, 119234 Moscow, Russia; (M.A.); (N.M.)
| | - Nataliya Medvedeva
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory 1-12, 119234 Moscow, Russia; (M.A.); (N.M.)
| | - Tyler W. LeBaron
- Department of Kinesiology and Outdoor Recreation, Southern Utah University, Cedar City, UT 84720, USA;
- Molecular Hydrogen Institute, Cedar City, UT 84720, USA
| | - Mami Noda
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science, Xi’an Jiaotong University, Xi’an 710049, China
| | - Oleg Medvedev
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Prospect 27-1, 119991 Moscow, Russia
| |
Collapse
|
3
|
Atiakshin D, Nikolaeva E, Semyachkina A, Kostin A, Volodkin A, Morozov S, Ignatyuk M, Mikhaleva L, Demyashkin G, Elieh-Ali-Komi D, Buchwalow I, Tiemann M. The Contribution of Mast Cells to the Regulation of Elastic Fiber Tensometry in the Skin Dermis of Children with Marfan Syndrome. Int J Mol Sci 2024; 25:9191. [PMID: 39273142 PMCID: PMC11394836 DOI: 10.3390/ijms25179191] [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: 07/12/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Marfan syndrome (MFS) is a hereditary condition accompanied by disorders in the structural and regulatory properties of connective tissue, including elastic fibers, due to a mutation in the gene encodes for fibrillin-1 protein (FBN1 gene) and the synthesis of abnormal fibrillin-1 glycoprotein. Despite the high potential of mast cells (MCs) to remodel the extracellular matrix (ECM), their pathogenetic significance in MFS has not been considered yet. The group of patients with Marfan syndrome included two mothers and five children (three girls aged 4, 11, and 11 and two boys aged 12 and 13). Normal skin was examined in two children aged 11 and 12. Histochemical, monoplex, and multiplex immunohistochemical techniques; combined protocols of simultaneous histochemical and immunohistochemical staining (the results of staining were assessed using light, epifluorescence, and confocal microscopy); and bioinformatics algorithms for the quantitative analysis of detected targets were used to evaluate mast cells and their relationship with other cells from extracellular structures in the skin dermis. Analysis of the skin MC population in children with Marfan syndrome revealed a considerably increased number of intra-organic populations with the preservation of the specific Tryptase+Chymase+CPA3+ protease profile typical of the skin. The features of the MC histotopography phenotype in MFS consisted of closer colocalization with elastic fibers, smooth muscle cells, and fibroblasts. MCs formed many intradermal clusters that synchronized the activity of cell functions in the stromal landscape of the tissue microenvironment with the help of spatial architectonics, including the formation of cell chains and the creation of fibrous niches. In MCs, the expression of specific proteases, TGF-β, and heparin increased, with targeted secretion of biologically active substances relative to the dermal elastic fibers, which had specific structural features in MFS, including abnormal variability in thickness along their entire length, alternating thickened and thinned areas, and uneven surface topography. This paper discusses the potential role of MCs in strain analysis (tensometry) of the tissue microenvironment in MFS. Thus, the quantitative and qualitative rearrangements of the skin MC population in MFS are aimed at altering the stromal landscape of the connective tissue. The results obtained should be taken into account when managing clinical signs of MFS manifested in other pathogenetically critical structures of internal organs, including the aorta, tendons, cartilage, and parenchymal organs.
Collapse
Affiliation(s)
- Dmitrii Atiakshin
- RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russian Federation
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Ekaterina Nikolaeva
- Veltischev Research and Clinical Institute for Pediatrics & Pediatric Surgery of the Pirogov Russian National Research Medical University, 2, Taldomskaya St., 125412 Moscow, Russia
| | - Alla Semyachkina
- Veltischev Research and Clinical Institute for Pediatrics & Pediatric Surgery of the Pirogov Russian National Research Medical University, 2, Taldomskaya St., 125412 Moscow, Russia
| | - Andrey Kostin
- RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russian Federation
| | - Artem Volodkin
- RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russian Federation
| | - Sergey Morozov
- Veltischev Research and Clinical Institute for Pediatrics & Pediatric Surgery of the Pirogov Russian National Research Medical University, 2, Taldomskaya St., 125412 Moscow, Russia
| | - Michael Ignatyuk
- RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russian Federation
| | - Liudmila Mikhaleva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| | - Grigory Demyashkin
- Laboratory of Histology and Immunohistochemistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya St., 8/2, 119048 Moscow, Russia
| | - Daniel Elieh-Ali-Komi
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 12203 Berlin, Germany
| | - Igor Buchwalow
- RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russian Federation
- Institute for Hematopathology, Fangdieckstr, 75a, 22547 Hamburg, Germany
| | - Markus Tiemann
- Institute for Hematopathology, Fangdieckstr, 75a, 22547 Hamburg, Germany
| |
Collapse
|
4
|
Abdelnabi MN, Hassan GS, Shoukry NH. Role of the type 3 cytokines IL-17 and IL-22 in modulating metabolic dysfunction-associated steatotic liver disease. Front Immunol 2024; 15:1437046. [PMID: 39156888 PMCID: PMC11327067 DOI: 10.3389/fimmu.2024.1437046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/12/2024] [Indexed: 08/20/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) comprises a spectrum of liver diseases that span simple steatosis, metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis and may progress to cirrhosis and cancer. The pathogenesis of MASLD is multifactorial and is driven by environmental, genetic, metabolic and immune factors. This review will focus on the role of the type 3 cytokines IL-17 and IL-22 in MASLD pathogenesis and progression. IL-17 and IL-22 are produced by similar adaptive and innate immune cells such as Th17 and innate lymphoid cells, respectively. IL-17-related signaling is upregulated during MASLD resulting in increased chemokines and proinflammatory cytokines in the liver microenvironment, enhanced recruitment of myeloid cells and T cells leading to exacerbation of inflammation and liver disease progression. IL-17 may also act directly by activating hepatic stellate cells resulting in increased fibrosis. In contrast, IL-22 is a pleiotropic cytokine with a dominantly protective signature in MASLD and is currently being tested as a therapeutic strategy. IL-22 also exhibits beneficial metabolic effects and abrogates MASH-related inflammation and fibrosis development via inducing the production of anti-oxidants and anti-apoptotic factors. A sex-dependent effect has been attributed to both cytokines, most importantly to IL-22 in MASLD or related conditions. Altogether, IL-17 and IL-22 are key effectors in MASLD pathogenesis and progression. We will review the role of these two cytokines and cells that produce them in the development of MASLD, their interaction with host factors driving MASLD including sexual dimorphism, and their potential therapeutic benefits.
Collapse
Affiliation(s)
- Mohamed N. Abdelnabi
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
5
|
Gill K, Yoo HS, Chakravarthy H, Granville DJ, Matsubara JA. Exploring the role of granzyme B in subretinal fibrosis of age-related macular degeneration. Front Immunol 2024; 15:1421175. [PMID: 39091492 PMCID: PMC11291352 DOI: 10.3389/fimmu.2024.1421175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Age-related macular degeneration (AMD), a prevalent and progressive degenerative disease of the macula, is the leading cause of blindness in elderly individuals in developed countries. The advanced stages include neovascular AMD (nAMD), characterized by choroidal neovascularization (CNV), leading to subretinal fibrosis and permanent vision loss. Despite the efficacy of anti-vascular endothelial growth factor (VEGF) therapy in stabilizing or improving vision in nAMD, the development of subretinal fibrosis following CNV remains a significant concern. In this review, we explore multifaceted aspects of subretinal fibrosis in nAMD, focusing on its clinical manifestations, risk factors, and underlying pathophysiology. We also outline the potential sources of myofibroblast precursors and inflammatory mechanisms underlying their recruitment and transdifferentiation. Special attention is given to the potential role of mast cells in CNV and subretinal fibrosis, with a focus on putative mast cell mediators, tryptase and granzyme B. We summarize our findings on the role of GzmB in CNV and speculate how GzmB may be involved in the pathological transition from CNV to subretinal fibrosis in nAMD. Finally, we discuss the advantages and drawbacks of animal models of subretinal fibrosis and pinpoint potential therapeutic targets for subretinal fibrosis.
Collapse
Affiliation(s)
- Karanvir Gill
- Department of Ophthalmology and Visual Sciences, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Hyung-Suk Yoo
- Department of Ophthalmology and Visual Sciences, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Harshini Chakravarthy
- Department of Ophthalmology and Visual Sciences, University of British Columbia (UBC), Vancouver, BC, Canada
| | - David J. Granville
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Joanne A. Matsubara
- Department of Ophthalmology and Visual Sciences, University of British Columbia (UBC), Vancouver, BC, Canada
| |
Collapse
|
6
|
Su H, Zou R, Su J, Chen X, Yang H, An N, Yang C, Tang J, Liu H, Yao C. Sterile inflammation of peritoneal membrane caused by peritoneal dialysis: focus on the communication between immune cells and peritoneal stroma. Front Immunol 2024; 15:1387292. [PMID: 38779674 PMCID: PMC11109381 DOI: 10.3389/fimmu.2024.1387292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024] Open
Abstract
Peritoneal dialysis is a widely used method for treating kidney failure. However, over time, the peritoneal structure and function can deteriorate, leading to the failure of this therapy. This deterioration is primarily caused by infectious and sterile inflammation. Sterile inflammation, which is inflammation without infection, is particularly concerning as it can be subtle and often goes unnoticed. The onset of sterile inflammation involves various pathological processes. Peritoneal cells detect signals that promote inflammation and release substances that attract immune cells from the bloodstream. These immune cells contribute to the initiation and escalation of the inflammatory response. The existing literature extensively covers the involvement of different cell types in the sterile inflammation, including mesothelial cells, fibroblasts, endothelial cells, and adipocytes, as well as immune cells such as macrophages, lymphocytes, and mast cells. These cells work together to promote the occurrence and progression of sterile inflammation, although the exact mechanisms are not fully understood. This review aims to provide a comprehensive overview of the signals from both stromal cells and components of immune system, as well as the reciprocal interactions between cellular components, during the initiation of sterile inflammation. By understanding the cellular and molecular mechanisms underlying sterile inflammation, we may potentially develop therapeutic interventions to counteract peritoneal membrane damage and restore normal function.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Huafeng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Cuiwei Yao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
7
|
Khoury P, Wechsler JB. Role of Mast Cells in Eosinophilic Gastrointestinal Diseases. Immunol Allergy Clin North Am 2024; 44:311-327. [PMID: 38575226 PMCID: PMC11220468 DOI: 10.1016/j.iac.2024.01.004] [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] [Indexed: 04/06/2024]
Abstract
Mast cells play a central role in the pathogenesis of eosinophilic gastrointestinal disorders (EGIDs), including eosinophilic esophagitis. Their interactions with immune and structural cells, involvement in tissue remodeling, and contribution to symptoms make them attractive targets for therapeutic intervention. More is being discovered regarding the intricate interplay of mast cells and eosinophils. Recent studies demonstrating that depletion of eosinophils is insufficient to improve symptoms of EGIDs have raised the question of whether other cells may play a role in symptomatology and pathogenesis of EGIDs.
Collapse
Affiliation(s)
- Paneez Khoury
- Human Eosinophil Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 12C103, Bethesda, MD 20892, USA.
| | - Joshua B Wechsler
- Simpson-Querrey 10-518, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E. Chicago Avenue, Box 65, Chicago, IL 60611, USA
| |
Collapse
|
8
|
Ning Y, Dou X, Wang Z, Shi K, Wang Z, Ding C, Sang X, Zhong X, Shao M, Han X, Cao G. SIRT3: A potential therapeutic target for liver fibrosis. Pharmacol Ther 2024; 257:108639. [PMID: 38561088 DOI: 10.1016/j.pharmthera.2024.108639] [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: 12/16/2023] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
Sirtuin3 (SIRT3) is a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase located in the mitochondria, which mainly regulates the acetylation of mitochondrial proteins. In addition, SIRT3 is involved in critical biological processes, including oxidative stress, inflammation, DNA damage, and apoptosis, all of which are closely related to the progression of liver disease. Liver fibrosis characterized by the deposition of extracellular matrix is a result of long termed or repeated liver damage, frequently accompanied by damaged hepatocytes, the recruitment of inflammatory cells, and the activation of hepatic stellate cells. Based on the functions and pharmacology of SIRT3, we will review its roles in liver fibrosis from three aspects: First, the main functions and pharmacological effects of SIRT3 were investigated based on its structure. Second, the roles of SIRT3 in major cells in the liver were summarized to reveal its mechanism in developing liver fibrosis. Last, drugs that regulate SIRT3 to prevent and treat liver fibrosis were discussed. In conclusion, exploring the pharmacological effects of SIRT3, especially in the liver, may be a potential strategy for treating liver fibrosis.
Collapse
Affiliation(s)
- Yan Ning
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinyue Dou
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhichao Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kao Shi
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zeping Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuan Ding
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiang Zhong
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meiyu Shao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Han
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China; The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Wang N, Zhang C. Oxidative Stress: A Culprit in the Progression of Diabetic Kidney Disease. Antioxidants (Basel) 2024; 13:455. [PMID: 38671903 PMCID: PMC11047699 DOI: 10.3390/antiox13040455] [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: 02/27/2024] [Revised: 04/01/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Diabetic kidney disease (DKD) is the principal culprit behind chronic kidney disease (CKD), ultimately developing end-stage renal disease (ESRD) and necessitating costly dialysis or kidney transplantation. The limited therapeutic efficiency among individuals with DKD is a result of our finite understanding of its pathogenesis. DKD is the result of complex interactions between various factors. Oxidative stress is a fundamental factor that can establish a link between hyperglycemia and the vascular complications frequently encountered in diabetes, particularly DKD. It is crucial to recognize the essential and integral role of oxidative stress in the development of diabetic vascular complications, particularly DKD. Hyperglycemia is the primary culprit that can trigger an upsurge in the production of reactive oxygen species (ROS), ultimately sparking oxidative stress. The main endogenous sources of ROS include mitochondrial ROS production, NADPH oxidases (Nox), uncoupled endothelial nitric oxide synthase (eNOS), xanthine oxidase (XO), cytochrome P450 (CYP450), and lipoxygenase. Under persistent high glucose levels, immune cells, the complement system, advanced glycation end products (AGEs), protein kinase C (PKC), polyol pathway, and the hexosamine pathway are activated. Consequently, the oxidant-antioxidant balance within the body is disrupted, which triggers a series of reactions in various downstream pathways, including phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), transforming growth factor beta/p38-mitogen-activated protein kinase (TGF-β/p38-MAPK), nuclear factor kappa B (NF-κB), adenosine monophosphate-activated protein kinase (AMPK), and the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling. The disease might persist even if strict glucose control is achieved, which can be attributed to epigenetic modifications. The treatment of DKD remains an unresolved issue. Therefore, reducing ROS is an intriguing therapeutic target. The clinical trials have shown that bardoxolone methyl, a nuclear factor erythroid 2-related factor 2 (Nrf2) activator, blood glucose-lowering drugs, such as sodium-glucose cotransporter 2 inhibitors, and glucagon-like peptide-1 receptor agonists can effectively slow down the progression of DKD by reducing oxidative stress. Other antioxidants, including vitamins, lipoic acid, Nox inhibitors, epigenetic regulators, and complement inhibitors, present a promising therapeutic option for the treatment of DKD. In this review, we conduct a thorough assessment of both preclinical studies and current findings from clinical studies that focus on targeted interventions aimed at manipulating these pathways. We aim to provide a comprehensive overview of the current state of research in this area and identify key areas for future exploration.
Collapse
Affiliation(s)
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
10
|
Ghosh K, Shome DK, Kulkarni B, Ghosh MK, Ghosh K. Fibrosis and bone marrow: understanding causation and pathobiology. J Transl Med 2023; 21:703. [PMID: 37814319 PMCID: PMC10561412 DOI: 10.1186/s12967-023-04393-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/28/2023] [Indexed: 10/11/2023] Open
Abstract
Bone marrow fibrosis represents an important structural change in the marrow that interferes with some of its normal functions. The aetiopathogenesis of fibrosis is not well established except in its primary form. The present review consolidates current understanding of marrow fibrosis. We searched PubMed without time restriction using key words: bone marrow and fibrosis as the main stem against the terms: growth factors, cytokines and chemokines, morphology, megakaryocytes and platelets, myeloproliferative disorders, myelodysplastic syndrome, collagen biosynthesis, mesenchymal stem cells, vitamins and minerals and hormones, and mechanism of tissue fibrosis. Tissue marrow fibrosis-related papers were short listed and analysed for the review. It emerged that bone marrow fibrosis is the outcome of complex interactions between growth factors, cytokines, chemokines and hormones together with their facilitators and inhibitors. Fibrogenesis is initiated by mobilisation of special immunophenotypic subsets of mesenchymal stem cells in the marrow that transform into fibroblasts. Fibrogenic stimuli may arise from neoplastic haemopoietic or non-hematopoietic cells, as well as immune cells involved in infections and inflammatory conditions. Autoimmunity is involved in a small subset of patients with marrow fibrosis. Megakaryocytes and platelets are either directly involved or are important intermediaries in stimulating mesenchymal stem cells. MMPs, TIMPs, TGF-β, PDGRF, and basic FGF and CRCXL4 chemokines are involved in these processes. Genetic and epigenetic changes underlie many of these conditions.
Collapse
Affiliation(s)
- Kanjaksha Ghosh
- National Institute of Immunohaematology, 13 Th Fl KEM Hospital, Parel, Mumbai, 400012, India.
| | - Durjoy K Shome
- Department of Pathophysiology, American University of Antigua College of Medicine, Coolidge, Antigua and Barbuda
| | - Bipin Kulkarni
- Department of Molecular Biology and Haemostasis, National Institute of Immunohaematology, 13Th Fl KEM Hospital, Parel, Mumbai, 400012, India
| | - Malay K Ghosh
- Department of Haematology, Nilratan Sarkar Medical College, Kolkata, 700014, West Bengal, India
| | - Kinjalka Ghosh
- Department of Clinical Biochemistry, Tata Medical Centre and Homi Bhaba National Institute, Parel, Mumbai, 400012, India
| |
Collapse
|
11
|
Mattoo H, Bangari DS, Cummings S, Humulock Z, Habiel D, Xu EY, Pate N, Resnick R, Savova V, Qian G, Beil C, Rao E, Nestle FO, Bryce PJ, Subramaniam A. Molecular Features and Stages of Pulmonary Fibrosis Driven by Type 2 Inflammation. Am J Respir Cell Mol Biol 2023; 69:404-421. [PMID: 37369139 DOI: 10.1165/rcmb.2022-0301oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 06/27/2023] [Indexed: 06/29/2023] Open
Abstract
Systemic sclerosis (SSc) is a progressive, multiorgan disease with limited treatment options. Although a recent proof-of-concept study using romilkimab or SAR156597, a bispecific IL-4/IL-13 antibody, suggests a direct role of these cytokines in the pathophysiology of SSc, their contributions to the balance between inflammation and fibrosis are unclear. Here, we determine the roles of type 2 inflammation in fibrogenesis using FRA2-Tg (Fos-related antigen 2-overexpressing transgenic) mice, which develop spontaneous, age-dependent progressive lung fibrosis. We defined the molecular signatures of inflammation and fibrosis at three key stages in disease progression, corresponding to preonset, inflammatory dominant, and fibrosis dominant biology, and revealed an early increase in cytokine-cytokine receptor interactions and antigen-processing and presentation pathways followed by enhanced Th2- and M2 macrophage-driven type 2 responses. This type 2 inflammation progressed to extensive fibrotic pathology by 14-18 weeks of age, with these gene signatures overlapping significantly with those seen in the lungs of patients with SSc with interstitial lung disease (ILD). These changes were also evident in the histopathology, which showed perivascular and peribronchiolar inflammation with prominent eosinophilia and accumulation of profibrotic M2-like macrophages followed by rapid progression to fibrosis with thickened alveolar walls with multifocal fibrotic bands and signs of interstitial pneumonia. Critically, treatment with a bispecific antibody targeting IL-4 and IL-13 during the inflammatory phase abrogated the Th2 and M2 responses and led to near-complete abrogation of lung fibrosis. These data recapitulate important features of fibrotic progression in the lungs of patients with SSc-ILD and enhance our understanding of the progressive pathobiology of SSc. This study also further establishes FRA2-Tg mice as a valuable tool for testing future therapeutic agents in SSc-ILD.
Collapse
Affiliation(s)
| | | | - Sheila Cummings
- Discovery Pathology, Translational In Vivo Models Platform, and
| | | | - David Habiel
- Immunology and Inflammation Research Therapeutic Area
| | - Ethan Y Xu
- Precision Medicine and Computational Biology
- Aspen Neuroscience, San Diego, California
| | - Nathan Pate
- Discovery Pathology, Translational In Vivo Models Platform, and
| | | | | | - George Qian
- Immunology and Inflammation Research Therapeutic Area
| | | | - Ercole Rao
- Biologics Research, Sanofi, Frankfurt, Germany; and
| | | | - Paul J Bryce
- Immunology and Inflammation Research Therapeutic Area
| | | |
Collapse
|
12
|
Xiong HD, Tang LL, Chen HJ, Wu Y, Li WY, Wen SJ, Lin YK. Identification of immune microenvironment changes, immune-related pathways and genes in male androgenetic alopecia. Medicine (Baltimore) 2023; 102:e35242. [PMID: 37746940 PMCID: PMC10519577 DOI: 10.1097/md.0000000000035242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/26/2023] Open
Abstract
BACKGROUND Although androgenetic alopecia (AGA) is classified as a non-inflammatory alopecia, histological evidence of microinflammation has long been recognized. However, changes in the immune microenvironment, immune-related pathways and the expression of immune-related genes (IRGs) involved in AGA remain unclear. METHODS The microarray gene expression data (GSE36169) from patients with male AGA were analyzed. gene set enrichment analysis (GSEA) among statistically changed genes was done. Kyoto Encyclopedia of Genes and Genomes and Gene Ontology analyses among differentially expressed genes were performed. differentially expressed genes were screened to identify IRGs based on the ImmPort database. The cytohubba-MCC plugin of Cytoscape was applied to screen hub immune genes. The infiltration levels of 28 immune cells were quantified adopting single-sample GSEA (ssGSEA) algorithm. The microarray gene expression data (GSE90594) of male AGA was analyzed to validate hub IRGs genes and differential infiltrated immune cells. RESULTS The ssGSEA revealed γδT cell, central memory CD8+ T cell, mast cell, immature B cell, activated CD8+ T cell, effector memory CD4+ T cell, eosinophil and neutrophil were significantly increased infiltration in the bald scalp. GSEA showed statistically changed genes were most enriched in immune related pathways, including innate immune system, adaptive immune system, cytokine signaling, interferon-γ signaling, interferon signaling and interleukins signaling. The 4 hub IRGs, including matrix metallopeptidase 9, protein tyrosine phosphatase receptor type C, bone morphogenetic protein 2, and thrombospondin 1, were enriched in the pathways of allograft rejection, coagulation and interferon-γ response. CONCLUSION In summary, we proposed that the increase in γδ T cells, central memory CD8+ T cells, activated CD8+ T cell as well as the infiltration of mast cells contributed to immune microenvironment changes in male AGA. The 4 hub IRGs may be involved in the development and progression of hair loss in male AGA through interferon-γ signal pathways.
Collapse
Affiliation(s)
- Hong-Di Xiong
- Department of Dermatology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lu-Lu Tang
- Department of Dermatology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hai-Ju Chen
- Department of Dermatology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yi Wu
- Department of Dermatology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wen-Yu Li
- Department of Dermatology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Si-Jian Wen
- Department of Dermatology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - You-Kun Lin
- Department of Dermatology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
13
|
Liu T, Zhuang XX, Gao JR. Identifying Aging-Related Biomarkers and Immune Infiltration Features in Diabetic Nephropathy Using Integrative Bioinformatics Approaches and Machine-Learning Strategies. Biomedicines 2023; 11:2454. [PMID: 37760894 PMCID: PMC10525809 DOI: 10.3390/biomedicines11092454] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Aging plays an essential role in the development of diabetic nephropathy (DN). This study aimed to identify and verify potential aging-related genes associated with DN using bioinformatics analysis. METHODS To begin with, we combined the datasets from GEO microarrays (GSE104954 and GSE30528) to find the genes that were differentially expressed (DEGs) across samples from DN and healthy patient populations. By overlapping DEGs, weighted co-expression network analysis (WGCNA), and 1357 aging-related genes (ARGs), differentially expressed ARGs (DEARGs) were discovered. We next performed functional analysis to determine DEARGs' possible roles. Moreover, protein-protein interactions were examined using STRING. The hub DEARGs were identified using the CytoHubba, MCODE, and LASSO algorithms. We next used two validation datasets and Receiver Operating Characteristic (ROC) curves to determine the diagnostic significance of the hub DEARGs. RT-qPCR, meanwhile, was used to confirm the hub DEARGs' expression levels in vitro. In addition, we investigated the relationships between immune cells and hub DEARGs. Next, Gene Set Enrichment Analysis (GSEA) was used to identify each biomarker's biological role. The hub DEARGs' subcellular location and cell subpopulations were both identified and predicted using the HPA and COMPARTMENTS databases, respectively. Finally, drug-protein interactions were predicted and validated using STITCH and AutoDock Vina. RESULTS A total of 57 DEARGs were identified, and functional analysis reveals that they play a major role in inflammatory processes and immunomodulation in DN. In particular, aging and the AGE-RAGE signaling pathway in diabetic complications are significantly enriched. Four hub DEARGs (CCR2, VCAM1, CSF1R, and ITGAM) were further screened using the interaction network, CytoHubba, MCODE, and LASSO algorithms. The results above were further supported by validation sets, ROC curves, and RT-qPCR. According to an evaluation of immune infiltration, DN had significantly more resting mast cells and delta gamma T cells but fewer regulatory T cells and active mast cells. Four DEARGs have statistical correlations with them as well. Further investigation revealed that four DEARGs were implicated in immune cell abnormalities and regulated a wide range of immunological and inflammatory responses. Furthermore, the drug-protein interactions included four possible therapeutic medicines that target four DEARGs, and molecular docking could make this association practical. CONCLUSIONS This study identified four DEARGs (CCR2, VCAM1, CSF1R, and ITGAM) associated with DN, which might play a key role in the development of DN and could be potential biomarkers in DN.
Collapse
Affiliation(s)
- Tao Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, China;
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, China
| | - Xing-Xing Zhuang
- Department of Pharmacy, Chaohu Hospital of Anhui Medical University, Chaohu 238000, China;
| | - Jia-Rong Gao
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, China;
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, China
| |
Collapse
|
14
|
Mihele DM, Nistor PA, Bruma G, Mitran CI, Mitran MI, Condrat CE, Tovaru M, Tampa M, Georgescu SR. Mast Cell Activation Syndrome Update-A Dermatological Perspective. J Pers Med 2023; 13:1116. [PMID: 37511729 PMCID: PMC10381535 DOI: 10.3390/jpm13071116] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Mast cells (MCs) are infamous for their role in potentially fatal anaphylaxis reactions. In the last two decades, a more complex picture has emerged, as it has become obvious that MCs are much more than just IgE effectors of anaphylaxis. MCs are defenders against a host of infectious and toxic aggressions (their interactions with other components of the immune system are not yet fully understood) and after the insult has ended, MCs continue to play a role in inflammation regulation and tissue repair. Unfortunately, MC involvement in pathology is also significant. Apart from their role in allergies, MCs can proliferate clonally to produce systemic mastocytosis. They have also been implicated in excessive fibrosis, keloid scaring, graft rejection and chronic inflammation, especially at the level of the skin and gut. In recent years, the term MC activation syndrome (MCAS) was proposed to account for symptoms caused by MC activation, and clear diagnostic criteria have been defined. However, not all authors agree with these criteria, as some find them too restrictive, potentially leaving much of the MC-related pathology unaccounted for. Here, we review the current knowledge on the physiological and pathological roles of MCs, with a dermatological emphasis, and discuss the MCAS classification.
Collapse
Affiliation(s)
- Dana Mihaela Mihele
- Dermatology Department, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania
- Dermatology Department, Victor Babes Clinical Hospital of Infectious and Tropical Diseases, 030303 Bucharest, Romania
| | - Paul Andrei Nistor
- Internal Medicine Department, Emergency University Hospital Bucharest, 169 Independence Blvd, 050098 Bucharest, Romania
| | - Gabriela Bruma
- Dermatology Department, Victor Babes Clinical Hospital of Infectious and Tropical Diseases, 030303 Bucharest, Romania
| | - Cristina Iulia Mitran
- Microbiology Department, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania
| | - Madalina Irina Mitran
- Microbiology Department, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania
| | - Carmen Elena Condrat
- Fetal Medicine Excellence Research Center, Alessandrescu-Rusescu National Institute for Mother and Child Health, 020395 Bucharest, Romania
- Department of Obstetrics and Gynecology, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania
| | - Mihaela Tovaru
- Dermatology Department, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania
- Dermatology Department, Victor Babes Clinical Hospital of Infectious and Tropical Diseases, 030303 Bucharest, Romania
| | - Mircea Tampa
- Dermatology Department, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania
- Dermatology Department, Victor Babes Clinical Hospital of Infectious and Tropical Diseases, 030303 Bucharest, Romania
| | - Simona Roxana Georgescu
- Dermatology Department, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania
- Dermatology Department, Victor Babes Clinical Hospital of Infectious and Tropical Diseases, 030303 Bucharest, Romania
| |
Collapse
|
15
|
Costanzo G, Costanzo GAML, Del Moro L, Nappi E, Pelaia C, Puggioni F, Canonica GW, Heffler E, Paoletti G. Mast Cells in Upper and Lower Airway Diseases: Sentinels in the Front Line. Int J Mol Sci 2023; 24:ijms24119771. [PMID: 37298721 DOI: 10.3390/ijms24119771] [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: 05/11/2023] [Revised: 06/01/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023] Open
Abstract
Mast cells (MCs) are fascinating cells of the innate immune system involved not only in allergic reaction but also in tissue homeostasis, response to infection, wound healing, protection against kidney injury, the effects of pollution and, in some circumstances, cancer. Indeed, exploring their role in respiratory allergic diseases would give us, perhaps, novel therapy targets. Based on this, there is currently a great demand for therapeutic regimens to enfeeble the damaging impact of MCs in these pathological conditions. Several strategies can accomplish this at different levels in response to MC activation, including targeting individual mediators released by MCs, blockade of receptors for MC-released compounds, inhibition of MC activation, limiting mast cell growth, or inducing mast cell apoptosis. The current work focuses on and summarizes the mast cells' role in pathogenesis and as a personalized treatment target in allergic rhinitis and asthma; even these supposed treatments are still at the preclinical stage.
Collapse
Affiliation(s)
- Giovanni Costanzo
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | | | - Lorenzo Del Moro
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy
| | - Emanuele Nappi
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Corrado Pelaia
- Department of Health Sciences, University 'Magna Græcia' of Catanzaro, 88100 Catanzaro, Italy
| | - Francesca Puggioni
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Giorgio Walter Canonica
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
| | - Enrico Heffler
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
| | - Giovanni Paoletti
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
| |
Collapse
|
16
|
Gupta S, Sharma M, Banerjee S, Holikatti K, Kamble P, Goyal JV. The Immunolocalization of Mast Cells in the Pathology of Oral Submucous Fibrosis. Cureus 2023; 15:e40069. [PMID: 37425595 PMCID: PMC10326457 DOI: 10.7759/cureus.40069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Background Oral submucous fibrosis (OSMF) is a prevalent precancerous condition of the oral cavity and an ambiguity to clinicians because of its indistinguishable etiopathogenesis. Previous studies could not establish a definite role of mast cells (MCs) in the fibrosis of stroma. The present study was done to study the histopathological changes in OSMF and to determine the association of mast cells (MCs) and their degranulated components with vascularity. Methods A retrospective case-control study involved 40 cases of various histopathological grades of OSMF and was compared with 10 cases of normal buccal mucosa by using a cluster of differentiation 117 (CD117) kit for the identification of MCs and Masson's trichrome stain to study the number of blood vessels (BVs). Results The present study indicated that advanced cases of OSMF had keratinized epithelium with atrophic changes and moderate to advanced fibrosis of stroma with the involvement of underlying muscles. The MC density and the number of blood vessels were progressively reduced in OSMF as the grade advanced compared to healthy controls. Conclusion An increase in the mast cell density in the initial stages of OSMF suggests their definite role in the initiation of fibrosis and secondary changes to the epithelium such as atrophy.
Collapse
Affiliation(s)
- Seema Gupta
- Orthodontics, Jawahar Medical Foundation's (JMF) Annasaheb Chudaman Patil Memorial (ACPM) Dental College, Dhule, IND
| | - Manish Sharma
- Oral Pathology and Microbiology, Jawahar Medical Foundation's (JMF) Annasaheb Chudaman Patil Memorial (ACPM) Dental College, Dhule, IND
| | - Satyabrat Banerjee
- Conservative Dentistry and Endodontics, Jawahar Medical Foundation's (JMF) Annasaheb Chudaman Patil Memorial (ACPM) Dental College, Dhule, IND
| | - Kiran Holikatti
- Oral Pathology and Microbiology, Jawahar Medical Foundation's (JMF) Annasaheb Chudaman Patil Memorial (ACPM) Dental College, Dhule, IND
| | - Priyanka Kamble
- Oral Pathology and Microbiology, Jawahar Medical Foundation's (JMF) Annasaheb Chudaman Patil Memorial (ACPM) Dental College, Dhule, IND
| | - Jay V Goyal
- Oral and Maxillofacial Surgery, Jawahar Medical Foundation's (JMF) Annasaheb Chudaman Patil Memorial (ACPM) Dental College, Dhule, IND
| |
Collapse
|
17
|
Rama TA, Henriques AF, Matito A, Jara-Acevedo M, Caldas C, Mayado A, Muñoz-González JI, Moreira A, Cavaleiro-Rufo J, García-Montero A, Órfão A, Sanchez-Muñoz L, Álvarez-Twose I. Bone and Cytokine Markers Associated With Bone Disease in Systemic Mastocytosis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:1536-1547. [PMID: 36801493 DOI: 10.1016/j.jaip.2023.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/05/2023] [Accepted: 02/03/2023] [Indexed: 02/19/2023]
Abstract
BACKGROUND Mastocytosis encompasses a heterogeneous group of diseases characterized by tissue accumulation of clonal mast cells, which frequently includes bone involvement. Several cytokines have been shown to play a role in the pathogenesis of bone mass loss in systemic mastocytosis (SM), but their role in SM-associated osteosclerosis remains unknown. OBJECTIVE To investigate the potential association between cytokine and bone remodeling markers with bone disease in SM, aiming at identifying biomarker profiles associated with bone loss and/or osteosclerosis. METHODS A total of 120 adult patients with SM, divided into 3 age and sex-matched groups according to their bone status were studied: (1) healthy bone (n = 46), (2) significant bone loss (n = 47), and (3) diffuse bone sclerosis (n = 27). Plasma levels of cytokines and serum baseline tryptase and bone turnover marker levels were measured at diagnosis. RESULTS Bone loss was associated with significantly higher levels of serum baseline tryptase (P = .01), IFN-γ (P = .05), IL-1β (P = .05), and IL-6 (P = .05) versus those found in patients with healthy bone. In contrast, patients with diffuse bone sclerosis showed significantly higher levels of serum baseline tryptase (P < .001), C-terminal telopeptide (P < .001), amino-terminal propeptide of type I procollagen (P < .001), osteocalcin (P < .001), bone alkaline phosphatase (P < .001), osteopontin (P < .01), and the C-C Motif Chemokine Ligand 5/RANTES chemokine (P = .01), together with lower IFN-γ (P = .03) and RANK-ligand (P = .04) plasma levels versus healthy bone cases. CONCLUSIONS SM with bone mass loss is associated with a proinflammatory cytokine profile in plasma, whereas diffuse bone sclerosis shows increased serum/plasma levels of biomarkers related to bone formation and turnover, in association with an immunosuppressive cytokine secretion profile.
Collapse
Affiliation(s)
- Tiago Azenha Rama
- Serviço de Imunoalergologia, Centro Hospitalar Universitário São João, Porto, Portugal; Serviço de Imunologia Básica e Clínica, Departamento de Patologia, Faculdade de Medicina da Universidade do Porto, Porto, Portugal; EPIUnit - Institute of Public Health, University of Porto, Porto, Portugal; Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal.
| | - Ana Filipa Henriques
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast) - Reference Center (CSUR) for Mastocytosis, Hospital Virgen del Valle, Complejo Hospitalario Universitario de Toledo, Toledo, Spain; Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Almudena Matito
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast) - Reference Center (CSUR) for Mastocytosis, Hospital Virgen del Valle, Complejo Hospitalario Universitario de Toledo, Toledo, Spain; Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Maria Jara-Acevedo
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; DNA Sequencing Service (NUCLEUS), Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Caldas
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; DNA Sequencing Service (NUCLEUS), Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Andrea Mayado
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain; Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine, Cytometry Service (NUCLEUS) Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Javier I Muñoz-González
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain; Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine, Cytometry Service (NUCLEUS) Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - André Moreira
- Serviço de Imunoalergologia, Centro Hospitalar Universitário São João, Porto, Portugal; Serviço de Imunologia Básica e Clínica, Departamento de Patologia, Faculdade de Medicina da Universidade do Porto, Porto, Portugal; EPIUnit - Institute of Public Health, University of Porto, Porto, Portugal; Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
| | - João Cavaleiro-Rufo
- EPIUnit - Institute of Public Health, University of Porto, Porto, Portugal; Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
| | - Andrés García-Montero
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain; Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine, Cytometry Service (NUCLEUS) Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Alberto Órfão
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain; Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine, Cytometry Service (NUCLEUS) Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Laura Sanchez-Muñoz
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast) - Reference Center (CSUR) for Mastocytosis, Hospital Virgen del Valle, Complejo Hospitalario Universitario de Toledo, Toledo, Spain; Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Iván Álvarez-Twose
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast) - Reference Center (CSUR) for Mastocytosis, Hospital Virgen del Valle, Complejo Hospitalario Universitario de Toledo, Toledo, Spain; Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
18
|
Anggraini N, Siregar NC, Sitorus RS. Recurrence of Idiopathic Orbital Inflammation: An 11-year Retrospective Study. Middle East Afr J Ophthalmol 2023; 30:89-97. [PMID: 39006932 PMCID: PMC11238927 DOI: 10.4103/meajo.meajo_225_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 07/16/2024] Open
Abstract
PURPOSE The high recurrence rate of idiopathic orbital inflammation (IOI) has been reported. This study aims to determine existing predictive factors for the recurrence of IOI. METHODS This was an 11-year retrospective study with at least a 12-month follow-up. Fifty patients with biopsy-proven IOI admitted between 2006 and 2017 at our tertiary hospital were observed. We compared the clinical characteristics, histopathological profile, and biomarker expressions (mast cell, immunoglobulin G4, tumor necrosis factor-alpha, and transforming growth factor-beta) of 16 patients with recurrence (Group I) and 34 patients with no recurrence (Group II). Statistical comparison and multivariate analysis were performed to establish the predictive factors. RESULTS We discovered five recurrence predictive factors: presentation of proptosis (odds ratio [OR] 4.96, 95% confidence interval [CI] 1.36-18.03), visual impairment (OR 15, 95% CI 1.58-142.72), extraocular muscle (EOM) restriction (OR 3.86, 95% CI 1.07-13.94), nonanterior involvement (OR 7.94, 95% CI 1.88-33.5), and corticosteroid (CS) alone treatment (OR 7.20, 95% CI 1.87-27.8). On multivariate analysis, nonanterior involvement and CS alone treatment were validated as predictive factors (area under the curve = 0.807 [95% CI 0.69-0.92]). Histopathological profile and biomarker expressions were not associated with recurrence. However, there was a 22-fold higher recurrence risk for granulomatous-type patients given CS alone treatment. CONCLUSION Unlike the five clinical characteristics mentioned, both histopathology and biomarker variables were not associated with recurrence. CS alone treatment for patients with nonanterior involvement or granulomatous type is proven to increase the risk of recurrence. Therefore, we suggest not giving CS without any combination treatment with other modalities for this group of patients.
Collapse
Affiliation(s)
- Neni Anggraini
- Department of Ophthalmology, Universitas Indonesia / Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Nurjati C Siregar
- Department of Anatomical Pathology, Universitas Indonesia / Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Rita S Sitorus
- Department of Ophthalmology, Universitas Indonesia / Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| |
Collapse
|
19
|
Su S, Ma Z, Wu H, Xu Z, Yi H. Oxidative stress as a culprit in diabetic kidney disease. Life Sci 2023; 322:121661. [PMID: 37028547 DOI: 10.1016/j.lfs.2023.121661] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/26/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023]
Abstract
Diabetic kidney disease (DKD) has become the leading cause of end-stage renal disease (ESRD), and the prevalence of DKD has increased worldwide during recent years. DKD is associated with poor therapeutic outcomes in most patients, but there is limited understanding of its pathogenesis. This review suggests that oxidative stress interacts with many other factors in causing DKD. Highly active mitochondria and NAD(P)H oxidase are major sources of oxidants, and they significantly affect the risk for DKD. Oxidative stress and inflammation may be considered reciprocal causes of DKD, in that each is a cause and an effect of DKD. Reactive oxygen species (ROS) can act as second messengers in various signaling pathways and as regulators of metabolism, activation, proliferation, differentiation, and apoptosis of immune cells. Epigenetic modifications, such as DNA methylation, histone modifications, and non-coding RNAs can modulate oxidative stress. The development of new technologies and identification of new epigenetic mechanisms may provide novel opportunities for the diagnosis and treatment of DKD. Clinical trials demonstrated that novel therapies which reduce oxidative stress can slow the progression of DKD. These therapies include the NRF2 activator bardoxolone methyl, new blood glucose-lowering drugs such as sodium-glucose cotransporter 2 inhibitors, and glucagon-like peptide-1 receptor agonists. Future studies should focus on improving early diagnosis and the development of more effective combination treatments for this multifactorial disease.
Collapse
|
20
|
van der Elst G, Varol H, Hermans M, Baan CC, Duong-van Huyen JP, Hesselink DA, Kramann R, Rabant M, Reinders MEJ, von der Thüsen JH, van den Bosch TPP, Clahsen-van Groningen MC. The mast cell: A Janus in kidney transplants. Front Immunol 2023; 14:1122409. [PMID: 36891297 PMCID: PMC9986315 DOI: 10.3389/fimmu.2023.1122409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
Mast cells (MCs) are innate immune cells with a versatile set of functionalities, enabling them to orchestrate immune responses in various ways. Aside from their known role in allergy, they also partake in both allograft tolerance and rejection through interaction with regulatory T cells, effector T cells, B cells and degranulation of cytokines and other mediators. MC mediators have both pro- and anti-inflammatory actions, but overall lean towards pro-fibrotic pathways. Paradoxically, they are also seen as having potential protective effects in tissue remodeling post-injury. This manuscript elaborates on current knowledge of the functional diversity of mast cells in kidney transplants, combining theory and practice into a MC model stipulating both protective and harmful capabilities in the kidney transplant setting.
Collapse
Affiliation(s)
- G van der Elst
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - H Varol
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - M Hermans
- Department of Internal Medicine, Division of Allergy and Clinical Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - C C Baan
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | - D A Hesselink
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - R Kramann
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany.,Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - M Rabant
- Department of Pathology, Necker Hospital, APHP, Paris, France
| | - M E J Reinders
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - J H von der Thüsen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - T P P van den Bosch
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - M C Clahsen-van Groningen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
21
|
St John AL, Rathore APS, Ginhoux F. New perspectives on the origins and heterogeneity of mast cells. Nat Rev Immunol 2023; 23:55-68. [PMID: 35610312 DOI: 10.1038/s41577-022-00731-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 01/06/2023]
Abstract
Mast cells are immune cells of the haematopoietic lineage that are now thought to have multifaceted functions during homeostasis and in various disease states. Furthermore, while mast cells have been known for a long time to contribute to allergic disease in adults, recent studies, mainly in mice, have highlighted their early origins during fetal development and potential for immune functions, including allergic responses, in early life. Our understanding of the imprinting of mast cells by particular tissues of residence and their potential for regulatory interactions with organ systems such as the peripheral immune, nervous and vascular systems is also rapidly evolving. Here, we discuss the origins of mast cells and their diverse and plastic phenotypes that are influenced by tissue residence. We explore how divergent phenotypes and functions might result from both their hard-wired 'nature' defined by their ontogeny and the 'nurture' they receive within specialized tissue microenvironments.
Collapse
Affiliation(s)
- Ashley L St John
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
- Department of Pathology, Duke University Medical Center, Durham, NC, USA.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- SingHealth Duke-NUS Global Health Institute, Singapore, Singapore.
| | - Abhay P S Rathore
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Florent Ginhoux
- Singapore Immunology Network, A*STAR, Singapore, Singapore.
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.
| |
Collapse
|
22
|
Tziastoudi M, Theoharides TC, Nikolaou E, Efthymiadi M, Eleftheriadis T, Stefanidis I. Key Genetic Components of Fibrosis in Diabetic Nephropathy: An Updated Systematic Review and Meta-Analysis. Int J Mol Sci 2022; 23:15331. [PMID: 36499658 PMCID: PMC9736240 DOI: 10.3390/ijms232315331] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/27/2022] [Accepted: 11/29/2022] [Indexed: 12/09/2022] Open
Abstract
Renal fibrosis (RF) constitutes the common end-point of all kinds of chronic kidney disease (CKD), regardless of the initial cause of disease. The aim of the present study was to identify the key players of fibrosis in the context of diabetic nephropathy (DN). A systematic review and meta-analysis of all available genetic association studies regarding the genes that are included in signaling pathways related to RF were performed. The evaluated studies were published in English and they were included in PubMed and the GWAS Catalog. After an extensive literature review and search of the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, eight signaling pathways related to RF were selected and all available genetic association studies of these genes were meta-analyzed. ACE, AGT, EDN1, EPO, FLT4, GREM1, IL1B, IL6, IL10, IL12RB1, NOS3, TGFB1, IGF2/INS/TH cluster, and VEGFA were highlighted as the key genetic components driving the fibrosis process in DN. The present systematic review and meta-analysis indicate, as key players of fibrosis in DN, sixteen genes. However, the results should be interpreted with caution because the number of studies was relatively small.
Collapse
Affiliation(s)
- Maria Tziastoudi
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece
| | - Theoharis C. Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA 02155, USA
- School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02155, USA
- Departments of Internal Medicine and Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, MA 02155, USA
- Institute of Neuro-Immune Medicine, Nova Southeastern University, Clearwater, FL 33314, USA
| | - Evdokia Nikolaou
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece
| | - Maria Efthymiadi
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece
| | - Theodoros Eleftheriadis
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece
| | - Ioannis Stefanidis
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece
| |
Collapse
|
23
|
Jia Q, Huang Z, Wang G, Sun X, Wu Y, Yang B, Yang T, Liu J, Li P, Li J. Osteopontin: An important protein in the formation of kidney stones. Front Pharmacol 2022; 13:1036423. [PMID: 36452224 PMCID: PMC9703462 DOI: 10.3389/fphar.2022.1036423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/26/2022] [Indexed: 09/10/2024] Open
Abstract
The incidence of kidney stones averages 10%, and the recurrence rate of kidney stones is approximately 10% at 1 year, 35% at 5 years, 50% at 10 years, and 75% at 20 years. However, there is currently a lack of good medicines for the prevention and treatment of kidney stones. Osteopontin (OPN) is an important protein in kidney stone formation, but its role is controversial, with some studies suggesting that it inhibits stone formation, while other studies suggest that it can promote stone formation. OPN is a highly phosphorylated protein, and with the deepening of research, there is growing evidence that it promotes stone formation, and the phosphorylated protein is believed to have adhesion effect, promote stone aggregation and nucleation. In addition, OPN is closely related to immune cell infiltration, such as OPN as a pro-inflammatory factor, which can activate mast cells (degranulate to release various inflammatory factors), macrophages (differentiated into M1 macrophages), and T cells (differentiated into T1 cells) etc., and these inflammatory cells play a role in kidney damage and stone formation. In short, OPN mainly exists in the phosphorylated form in kidney stones, plays an important role in the formation of stones, and may be an important target for drug therapy of kidney stones.
Collapse
Affiliation(s)
- Qingxia Jia
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Ziye Huang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Guang Wang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xia Sun
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yuyun Wu
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bowei Yang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Tongxin Yang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jianhe Liu
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Pei Li
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jiongming Li
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
24
|
Feng TY, Azar FN, Dreger SA, Buchta Rosean C, McGinty MT, Putelo AM, Kolli SH, Carey MA, Greenfield S, Fowler WJ, Robinson SD, Rutkowski MR. Reciprocal Interactions Between the Gut Microbiome and Mammary Tissue Mast Cells Promote Metastatic Dissemination of HR+ Breast Tumors. Cancer Immunol Res 2022; 10:1309-1325. [PMID: 36040846 PMCID: PMC9633553 DOI: 10.1158/2326-6066.cir-21-1120] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/02/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022]
Abstract
Establishing commensal dysbiosis, defined as an inflammatory gut microbiome with low biodiversity, before breast tumor initiation, enhances early dissemination of hormone receptor-positive (HR+) mammary tumor cells. Here, we sought to determine whether cellular changes occurring in normal mammary tissues, before tumor initiation and in response to dysbiosis, enhanced dissemination of HR+ tumors. Commensal dysbiosis increased both the frequency and profibrogenicity of mast cells in normal, non-tumor-bearing mammary tissues, a phenotypic change that persisted after tumor implantation. Pharmacological and adoptive transfer approaches demonstrated that profibrogenic mammary tissue mast cells from dysbiotic animals were sufficient to enhance dissemination of HR+ tumor cells. Using archival HR+ patient samples, we determined that enhanced collagen levels in tumor-adjacent mammary tissue positively correlated with mast cell abundance and HR+ breast cancer recurrence. Together, these data demonstrate that mast cells programmed by commensal dysbiosis activate mammary tissue fibroblasts and orchestrate early dissemination of HR+ breast tumors.
Collapse
Affiliation(s)
- Tzu-Yu Feng
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Francesca N. Azar
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Sally A. Dreger
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Claire Buchta Rosean
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Mitchell T. McGinty
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Audrey M. Putelo
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Sree H. Kolli
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Maureen A. Carey
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, VA, USA
| | - Stephanie Greenfield
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Wesley J. Fowler
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Stephen D. Robinson
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Melanie R. Rutkowski
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| |
Collapse
|
25
|
Menkü Özdemir FD, Üstün GG, Kősemehmetoğlu K, İspirli M, Boynuyoğun E, Uzun H. Comparison of Cromolyn Sodium, Montelukast, and Zafirlukast Prophylaxis for Capsular Contracture. Plast Reconstr Surg 2022; 150:1005e-1014e. [PMID: 35994348 DOI: 10.1097/prs.0000000000009653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Capsular contracture is the most common complication following breast augmentation. Recently, prophylaxis studies aiming to inhibit the release of profibrotic substances to prevent capsular contracture have gained in importance. This study investigated the effects of cromolyn sodium, montelukast, and zafirlukast on capsular contracture in a rat model. METHODS Thirty female Wistar albino rats were randomly divided into five groups: control, sham, cromolyn sodium, montelukast, and zafirlukast. Intraperitoneal injections were administered daily to the sham (1 ml per day), cromolyn sodium (10 mg/kg per day), montelukast (10 mg/kg per day), and zafirlukast (1.25 mg/kg per day) groups 1 month before surgery. Miniature breast implants were then placed on the backs of the rats in each group. Injections were continued for the next 3 months. The rats were subsequently killed, and the capsules were harvested and assessed histopathologically. The histopathologic outcomes were acute inflammation status, inflammation severity, synovial metaplasia, foreign body reaction, mast cell count, and capsular thickness. RESULTS The cromolyn sodium, montelukast, and zafirlukast groups had less acute inflammation and lower mean inflammation severity scores, foreign body reaction occurrence, mast cell counts, and capsular thickness than the control and sham groups ( p < 0.05). These parameters were better in the cromolyn sodium group than in the montelukast and zafirlukast groups ( p < 0.05). CONCLUSIONS Cromolyn sodium appears to inhibit capsular contracture more efficiently than montelukast and zafirlukast. This report may be a pioneer study for the prophylactic use of cromolyn sodium in capsular contracture. CLINICAL RELEVANCE STATEMENT The prophylactic administration of cromolyn sodium appears to reduce capsular contracture more efficiently than that of montelukast and zafirlukast. This report might constitute a pioneer study for the prophylactic use of cromolyn sodium in capsular contracture.
Collapse
Affiliation(s)
- Fethiye Damla Menkü Özdemir
- From the Departments of Plastic Reconstructive and Aesthetic Surgery, Pathology, and Pharmacology, Hacettepe University Faculty of Medicine
| | - Galip Gencay Üstün
- From the Departments of Plastic Reconstructive and Aesthetic Surgery, Pathology, and Pharmacology, Hacettepe University Faculty of Medicine
| | - Kemal Kősemehmetoğlu
- From the Departments of Plastic Reconstructive and Aesthetic Surgery, Pathology, and Pharmacology, Hacettepe University Faculty of Medicine
| | - Mukaddes İspirli
- From the Departments of Plastic Reconstructive and Aesthetic Surgery, Pathology, and Pharmacology, Hacettepe University Faculty of Medicine
| | - Etkin Boynuyoğun
- From the Departments of Plastic Reconstructive and Aesthetic Surgery, Pathology, and Pharmacology, Hacettepe University Faculty of Medicine
| | - Hakan Uzun
- From the Departments of Plastic Reconstructive and Aesthetic Surgery, Pathology, and Pharmacology, Hacettepe University Faculty of Medicine
| |
Collapse
|
26
|
Mast cells as a therapeutic target in myeloproliferative neoplasms. Trends Mol Med 2022; 28:902-905. [PMID: 36064534 DOI: 10.1016/j.molmed.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 08/16/2022] [Indexed: 11/23/2022]
Abstract
Mast cells have been implicated as mediators of bone marrow fibrosis and pruritus in myeloproliferative neoplasms (MPNs) with JAK2V617F or calreticulin mutations. We hypothesize that potent KIT inhibitors, already in clinical use for systemic mastocytosis, have therapeutic potential for the treatment of MPNs by directly targeting mast cells.
Collapse
|
27
|
Focal adhesion kinase priming in pancreatic cancer, altering biomechanics to improve chemotherapy. Biochem Soc Trans 2022; 50:1129-1141. [PMID: 35929603 PMCID: PMC9444069 DOI: 10.1042/bst20220162] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022]
Abstract
The dense desmoplastic and fibrotic stroma is a characteristic feature of pancreatic ductal adenocarcinoma (PDAC), regulating disease progression, metastasis and response to treatment. Reciprocal interactions between the tumour and stroma are mediated by bidirectional integrin-mediated signalling, in particular by Focal Adhesion Kinase (FAK). FAK is often hyperactivated and overexpressed in aggressive cancers, promoting stromal remodelling and inducing tissue stiffness which can accelerate cancer cell proliferation, survival and chemoresistance. Therapeutic targeting of the PDAC stroma is an evolving area of interest for pre-clinical and clinical research, where a subtle reshaping of the stromal architecture prior to chemotherapy may prove promising in the clinical management of disease and overall patient survival. Here, we describe how transient stromal manipulation (or ‘priming’) via short-term FAK inhibition, rather than chronic treatment, can render PDAC cells exquisitely vulnerable to subsequent standard-of-care chemotherapy. We assess how our priming publication fits with the recent literature and describe in this perspective how this could impact future cancer treatment. This highlights the significance of treatment timing and warrants further consideration of anti-fibrotic therapies in the clinical management of PDAC and other fibrotic diseases.
Collapse
|
28
|
Lind T, Melo FR, Gustafson AM, Sundqvist A, Zhao XO, Moustakas A, Melhus H, Pejler G. Mast Cell Chymase Has a Negative Impact on Human Osteoblasts. Matrix Biol 2022; 112:1-19. [PMID: 35908613 DOI: 10.1016/j.matbio.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 07/07/2022] [Accepted: 07/26/2022] [Indexed: 10/16/2022]
Abstract
Mast cells have been linked to osteoporosis and bone fractures, and in a previous study we found that mice lacking a major mast cell protease, chymase, develop increased diaphyseal bone mass. These findings introduce the possibility that mast cell chymase can regulate bone formation, but the underlying mechanism(s) has not previously been investigated. Here we hypothesized that chymase might exert such effects through a direct negative impact on osteoblasts, i.e., the main bone-building cells. Indeed, we show that chymase has a distinct impact on human primary osteoblasts. Firstly, chymase was shown to have pronounced effects on the morphological features of osteoblasts, including extensive cell contraction and actin reorganization. Chymase also caused a profound reduction in the output of collagen from the osteoblasts, and was shown to degrade osteoblast-secreted fibronectin and to activate pro-matrix metallopeptidase-2 released by the osteoblasts. Further, chymase was shown to have a preferential impact on the gene expression, protein output and phosphorylation status of TGFβ-associated signaling molecules. A transcriptomic analysis was conducted and revealed a significant effect of chymase on several genes of importance for bone metabolism, including a reduction in the expression of osteoprotegerin, which was confirmed at the protein level. Finally, we show that chymase interacts with human osteoblasts and is taken up by the cells. Altogether, the present findings provide a functional link between mast cell chymase and osteoblast function, and can form the basis for a further evaluation of chymase as a potential target for intervention in metabolic bone diseases.
Collapse
Affiliation(s)
- Thomas Lind
- Uppsala University Hospital, Department of Medical Sciences, Section of Clinical Pharmacology, Uppsala, Sweden.
| | - Fabio Rabelo Melo
- Uppsala University, Department of Medical Biochemistry and Microbiology, Uppsala, Sweden
| | - Ann-Marie Gustafson
- Uppsala University Hospital, Department of Medical Sciences, Section of Clinical Pharmacology, Uppsala, Sweden; Uppsala University, Department of Medical Biochemistry and Microbiology, Uppsala, Sweden
| | - Anders Sundqvist
- Uppsala University, Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala, Sweden
| | - Xinran O Zhao
- Uppsala University, Department of Medical Biochemistry and Microbiology, Uppsala, Sweden
| | - Aristidis Moustakas
- Uppsala University, Department of Medical Biochemistry and Microbiology, Uppsala, Sweden
| | - Håkan Melhus
- Uppsala University Hospital, Department of Medical Sciences, Section of Clinical Pharmacology, Uppsala, Sweden
| | - Gunnar Pejler
- Uppsala University, Department of Medical Biochemistry and Microbiology, Uppsala, Sweden
| |
Collapse
|
29
|
Jin J, Jiang Y, Chakrabarti S, Su Z. Cardiac Mast Cells: A Two-Head Regulator in Cardiac Homeostasis and Pathogenesis Following Injury. Front Immunol 2022; 13:963444. [PMID: 35911776 PMCID: PMC9334794 DOI: 10.3389/fimmu.2022.963444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiac mast cells (CMCs) are multifarious immune cells with complex roles both in cardiac physiological and pathological conditions, especially in cardiac fibrosis. Little is known about the physiological importance of CMCs in cardiac homeostasis and inflammatory process. Therefore, the present review will summarize the recent progress of CMCs on origin, development and replenishment in the heart, including their effects on cardiac development, function and ageing under physiological conditions as well as the roles of CMCs in inflammatory progression and resolution. The present review will shed a light on scientists to understand cardioimmunology and to develop immune treatments targeting on CMCs following cardiac injury.
Collapse
Affiliation(s)
- Jing Jin
- International Genome Center, Jiangsu University, Zhenjiang, China
- Institute of Immunology, Jiangsu University, Zhenjiang, China
| | - Yuanyuan Jiang
- Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang, China
- Institute of Immunology, Jiangsu University, Zhenjiang, China
- *Correspondence: Zhaoliang Su,
| |
Collapse
|
30
|
Wang EHC, Monga I, Sallee BN, Chen JC, Abdelaziz AR, Perez-Lorenzo R, Bordone LA, Christiano AM. Primary cicatricial alopecias are characterized by dysregulation of shared gene expression pathways. PNAS NEXUS 2022; 1:pgac111. [PMID: 35899069 PMCID: PMC9308563 DOI: 10.1093/pnasnexus/pgac111] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/07/2022] [Indexed: 02/06/2023]
Abstract
The primary forms of cicatricial (scarring) alopecia (PCA) are a group of inflammatory, irreversible hair loss disorders characterized by immune cell infiltrates targeting hair follicles (HFs). Lichen planopilaris (LPP), frontal fibrosing alopecia (FFA), and centrifugal cicatricial alopecia (CCCA) are among the main subtypes of PCAs. The pathogenesis of the different types of PCAs are poorly understood, and current treatment regimens yield inconsistent and unsatisfactory results. We performed high-throughput RNA-sequencing on scalp biopsies of a large cohort PCA patients to develop gene expression-based signatures, trained into machine-learning-based predictive models and pathways associated with dysregulated gene expression. We performed morphological and cytokine analysis to define the immune cell populations found in PCA subtypes. We identified a common PCA gene signature that was shared between LPP, FFA, and CCCA, which revealed a significant over-representation of mast cell (MC) genes, as well as downregulation of cholesterogenic pathways and upregulation of fibrosis and immune signaling genes. Immunohistological analyses revealed an increased presence of MCs in PCAs lesions. Our gene expression analyses revealed common pathways associated with PCAs, with a strong association with MCs. The indistinguishable differences in gene expression profiles and immune cell signatures between LPP, FFA, and CCCA suggest that similar treatment regimens may be effective in treating these irreversible forms of hair loss.
Collapse
Affiliation(s)
- Eddy H C Wang
- Department of Dermatology, Columbia University Irving Medical Center, 1150 St. Nicholas Ave, New York, NY 10032, USA
| | - Isha Monga
- Department of Dermatology, Columbia University Irving Medical Center, 1150 St. Nicholas Ave, New York, NY 10032, USA
| | - Brigitte N Sallee
- Department of Dermatology, Columbia University Irving Medical Center, 1150 St. Nicholas Ave, New York, NY 10032, USA
| | - James C Chen
- Department of Dermatology, Columbia University Irving Medical Center, 1150 St. Nicholas Ave, New York, NY 10032, USA
| | - Alexa R Abdelaziz
- Department of Dermatology, Columbia University Irving Medical Center, 1150 St. Nicholas Ave, New York, NY 10032, USA
| | - Rolando Perez-Lorenzo
- Department of Dermatology, Columbia University Irving Medical Center, 1150 St. Nicholas Ave, New York, NY 10032, USA
| | - Lindsey A Bordone
- Department of Dermatology, Columbia University Irving Medical Center, 1150 St. Nicholas Ave, New York, NY 10032, USA
| | | |
Collapse
|
31
|
Lin X, Wang Y, Liu Z, Lin S, Tan J, He J, Hu F, Wu X, Ghosh S, Chen M, Liu F, Mao R. Intestinal strictures in Crohn's disease: a 2021 update. Therap Adv Gastroenterol 2022; 15:17562848221104951. [PMID: 35757383 PMCID: PMC9218441 DOI: 10.1177/17562848221104951] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Intestinal strictures remain one of the most intractable and common complications of Crohn's disease (CD). Approximately 70% of CD patients will develop fibrotic strictures after 10 years of CD diagnosis. Since specific antifibrotic therapies are unavailable, endoscopic balloon dilation and surgery remain the mainstay treatments despite a high recurrence rate. Besides, there are no reliable methods for accurately evaluating intestinal fibrosis. This is largely due to the fact that the mechanisms of initiation and propagation of intestinal fibrosis are poorly understood. There is growing evidence implying that the pathogenesis of stricturing CD involves the intricate interplay of factors including aberrant immune and nonimmune responses, host-microbiome dysbiosis, and genetic susceptibility. Currently, the progress on intestinal strictures has been fueled by the advent of novel techniques, such as single-cell sequencing, multi-omics, and artificial intelligence. Here, we perform a timely and comprehensive review of the substantial advances in intestinal strictures in 2021, aiming to provide prompt information regarding fibrosis and set the stage for the improvement of diagnosis, treatment, and prognosis of intestinal strictures.
Collapse
Affiliation(s)
- Xiaoxuan Lin
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zishan Liu
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sinan Lin
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jinyu Tan
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jinshen He
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fan Hu
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaomin Wu
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Subrata Ghosh
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fen Liu
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road 2nd, Guangzhou 510080, People’s Republic of China
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road 2nd, Guangzhou 510080, People’s Republic of China
- Department of Gastroenterology, Huidong People’s Hospital, Huizhou 516399, China
| |
Collapse
|
32
|
da Luz MJ, da Costa VAA, Balbi APC, Bispo-da-Silva LB. Effects of Disodium Cromoglycate Treatment in the Early Stage of Diabetic Nephropathy: Focus on Collagen Deposition. Biol Pharm Bull 2022; 45:245-249. [PMID: 35228391 DOI: 10.1248/bpb.b21-00662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inflammation is part of the pathophysiology of diabetic nephropathy (DN), and mast cells (MCs) appear to increase in number within the kidney of humans and animals with diabetes. Disodium cromoglycate (CG) not only inhibits the degranulation of MCs but also has several secondary effects that may improve inflammation. However, little is known about the effects of CG treatment on kidney collagen deposition and myofibroblast population in animals with type I diabetes (DM1). Data presented here suggest that the increases in the density and activity of MCs within the kidney in the early stages of DN contribute to tubulointerstitial collagen deposition, even in the absence of alterations in the renal myofibroblast population. Moreover, CG treatment showed renoprotective effects in rats with DM1, which appear to be linked to its mast cell stabilizing property and its ability to avoid some detrimental morphofunctional alterations.
Collapse
Affiliation(s)
- Mateus Jacinto da Luz
- Department of Pharmacology, Institute of Biomedical Sciences, Federal University of Uberlândia, ICBIM-UFU
| | | | - Ana Paula Coelho Balbi
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlândia, ICBIM-UFU
| | - Luiz Borges Bispo-da-Silva
- Department of Pharmacology, Institute of Biomedical Sciences, Federal University of Uberlândia, ICBIM-UFU
| |
Collapse
|
33
|
Wan J, Wu T, Liu Y, Yang M, Fichna J, Guo Y, Yin L, Chen C. Mast Cells Tryptase Promotes Intestinal Fibrosis in Natural Decellularized Intestinal Scaffolds. Tissue Eng Regen Med 2022; 19:717-726. [PMID: 35218507 PMCID: PMC9294124 DOI: 10.1007/s13770-022-00433-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/18/2021] [Accepted: 01/08/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Standard two-dimensional (2D) culture has confirmed the mechanism of mast cells (MCs) in the pathogenesis of inflammatory bowel disease (IBD), but the regulation of signaling responses of MCs may well differ in three-dimensional (3D) microenvironments. The aim of the study was to develop a 3D culture model based on decellularized intestinal scaffolds (DIS) and verify how MCs influenced fibroblasts phenotype in the 3D model. METHODS DIS were achieved using the detergent technique and extracellular matrix (ECM) components were verified by histologic analysis, quantification and scanning electron microscope. After human colon fibroblasts recellularized into the scaffolds and activated by MCs tryptase and TGFβ1, the changes in genes and signaling pathways during fibroblasts activation in 3D were studied and compared with the changes in 2D cell culture on plastic plates. RESULTS Decellularization process effectively removed native cell debris while retaining natural ECM components and structure. The engrafted fibroblasts could penetrate into the scaffolds and maintain its phenotype. No matter whether fibroblasts were cultured in 2D or 3D, MCs tryptase and transforming growth factor β1 (TGF-β1) could promote the differentiation of fibroblasts into fibrotic-phenotype myofibroblasts through Akt and Smad2/3 signaling pathways. Furthermore, the pro-collagen1α1 and fibronectin synthesis of myofibroblasts in 3D was higher than in 2D culture. CONCLUSION Our results demonstrated that the DIS can be used as a bioactive microenvironment for the study of intestinal fibrosis, providing an innovative platform for future intestinal disease modeling and screening of genes and signaling pathways.
Collapse
Affiliation(s)
- Jian Wan
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Tianqi Wu
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Ying Liu
- Department of General Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Muqing Yang
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Yibing Guo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226000 China
| | - Lu Yin
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Chunqiu Chen
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
34
|
The Alleviating Effect of Lagerstroemia indica Flower Extract on Stretch Marks through Regulation of Mast Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041274. [PMID: 35209065 PMCID: PMC8877584 DOI: 10.3390/molecules27041274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 11/17/2022]
Abstract
Striae distensae (SD) or stretch marks are common linear scars of atrophic skin with disintegrating extracellular matrix (ECM) structures. Although fibroblasts contribute to the construction of ECM structure in SD, some studies have reported that mast cell degranulation causes the disruption of ECM in early SD lesions. Lagerstroemia indica flower (LIF) has traditionally been used in India as a diuretic. However, little is known about the effect and molecular action of Lagerstroemia indica flower extract (LIFE) on alleviating SD. This study evaluated the effects of LIFE on mast cell degranulation and the synthesis of ECM components in fibroblasts. LIFE inhibits the adhesion of rat basophilic leukemia (RBL) cells, RBL-2H3 on fibronectin (FN) and the expression of integrin, a receptor for FN, thereby reducing focal adhesion kinase (FAK) phosphorylation. In addition, LIFE attenuated the allergen-induced granules and cytokine interleukin 3 (IL-3) through the adhesion with FN. Moreover, the conditioned medium (CM) of activated mast cells decreases the synthesis of ECM components, and LIFE restores the abnormal expressions induced by activated mast cells. These results demonstrate that LIFE suppresses FN-induced mast cell activation and promotes the synthesis of ECM components in fibroblast, which indicates that LIFE may be a useful cosmetic agent for SD treatment.
Collapse
|
35
|
Stamenov N, Kotov G, Iliev A, Landzhov B, Kirkov V, Stanchev S. Mast cells and basic fibroblast growth factor in physiological aging of rat heart and kidney. Biotech Histochem 2022; 97:504-518. [DOI: 10.1080/10520295.2021.2024251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Affiliation(s)
- Nikola Stamenov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Sofia, Bulgaria
| | - Georgi Kotov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Sofia, Bulgaria
| | - Alexandar Iliev
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Sofia, Bulgaria
| | - Boycho Landzhov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Sofia, Bulgaria
| | - Vidin Kirkov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Sofia, Bulgaria
| | - Stancho Stanchev
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
36
|
Le Joncour A, Desbois AC, Leroyer AS, Tellier E, Régnier P, Maciejewski-Duval A, Comarmond C, Barete S, Arock M, Bruneval P, Launay JM, Fouret P, Blank U, Rosenzwajg M, Klatzmann D, Jarraya M, Chiche L, Koskas F, Cacoub P, Kaplanski G, Saadoun D. Mast cells drive pathologic vascular lesions in Takayasu arteritis. J Allergy Clin Immunol 2022; 149:292-301.e3. [PMID: 33992671 DOI: 10.1016/j.jaci.2021.05.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Takayasu arteritis (TAK) is a large vessel vasculitis resulting in artery wall remodeling with segmental stenosis and/or aneurysm formation. Mast cells (MCs) are instrumental in bridging cell injury and inflammatory response. OBJECTIVES This study sought to investigate the contribution of MCs on vessel permeability, angiogenesis, and fibrosis in patients with TAK. METHODS MC activation and their tissue expression were assessed in sera and in aorta from patients with TAK and from healthy donors (HDs). In vivo permeability was assessed using a modified Miles assay. Subconfluent cultured human umbilic vein endothelial cells and fibroblasts were used in vitro to investigate the effects of MC mediators on angiogenesis and fibrogenesis. RESULTS This study found increased levels of MC activation markers (histamine and indoleamine 2,3-dioxygenase) in sera of patients with TAK compared with in sera of HDs. Marked expression of MCs was shown in aortic lesions of patients with TAK compared with in those of noninflammatory aorta controls. Using Miles assay, this study showed that sera of patients with TAK significantly increased vascular permeability in vivo as compared with that of HDs. Vessel permeability was abrogated in MC-deficient mice. MCs stimulated by sera of patients with TAK supported neoangiogenesis (increased human umbilic vein endothelial cell proliferation and branches) and fibrosis by inducing increased production of fibronectin, type 1 collagen, and α-smooth muscle actin by fibroblasts as compared to MCs stimulated by sera of HD. CONCLUSIONS MCs are a key regulator of vascular lesions in patients with TAK and may represent a new therapeutic target in large vessel vasculitis.
Collapse
Affiliation(s)
- Alexandre Le Joncour
- Department of Immunology-Immunopathology-Immunotherapy, Université Pierre-et-Marie-Curie Université de Paris 06, Unite Mixte de Recherche (UMR)S959, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Universités, Paris, France; Department of Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France; Department of Internal Medicine and Clinical Immunology, Centre National de Références Maladies Autoimmunes et Systémiques Rares, Centre National de Références Maladies Autoinflammatoires Rares et Amylose Inflammatoire, Paris, France
| | - Anne-Claire Desbois
- Department of Immunology-Immunopathology-Immunotherapy, Université Pierre-et-Marie-Curie Université de Paris 06, Unite Mixte de Recherche (UMR)S959, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Universités, Paris, France; Department of Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France; Department of Internal Medicine and Clinical Immunology, Centre National de Références Maladies Autoimmunes et Systémiques Rares, Centre National de Références Maladies Autoinflammatoires Rares et Amylose Inflammatoire, Paris, France
| | - Aurélie S Leroyer
- Centre de Recherche en CardioVasculaire et Nutrition, INSERM U1263, Inrae 1260, Aix-Marseille Université, Marseille, France
| | - Edwige Tellier
- Centre de Recherche en CardioVasculaire et Nutrition, INSERM U1263, Inrae 1260, Aix-Marseille Université, Marseille, France
| | - Paul Régnier
- Department of Immunology-Immunopathology-Immunotherapy, Université Pierre-et-Marie-Curie Université de Paris 06, Unite Mixte de Recherche (UMR)S959, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Universités, Paris, France; Department of Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France
| | - Anna Maciejewski-Duval
- Department of Immunology-Immunopathology-Immunotherapy, Université Pierre-et-Marie-Curie Université de Paris 06, Unite Mixte de Recherche (UMR)S959, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Universités, Paris, France; Department of Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France
| | - Cloé Comarmond
- Department of Immunology-Immunopathology-Immunotherapy, Université Pierre-et-Marie-Curie Université de Paris 06, Unite Mixte de Recherche (UMR)S959, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Universités, Paris, France; Department of Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France; Department of Internal Medicine and Clinical Immunology, Centre National de Références Maladies Autoimmunes et Systémiques Rares, Centre National de Références Maladies Autoinflammatoires Rares et Amylose Inflammatoire, Paris, France
| | - Stéphane Barete
- Department of Internal Medicine and Clinical Immunology, Centre National de Références Maladies Autoimmunes et Systémiques Rares, Centre National de Références Maladies Autoinflammatoires Rares et Amylose Inflammatoire, Paris, France; Department of Dermatology DMU3ID, Unité Fonctionnelle de Dermatologie, Groupe Hospitalier Pitié-Salpêtrière-C. Foix, Paris, France
| | - Michel Arock
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France; Laboratoire d'Hématologie Biologique, Hôpital Pitié-Salpêtrière, Paris, France
| | - Patrick Bruneval
- Laboratoire d'anatomopathologie, Hôpital Européen Georges Pompidou, Paris, France
| | | | - Pierre Fouret
- Laboratoire d'Anatomopathologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Ulrich Blank
- Center of Research on Inflammation, INSERM UMR S1149 and Centre National de la Recherche Scientifique Experimental Research Laboratory 8252, Universite de Paris, Sorbonne Paris Cite, Laboratoire d'Excellence INFLAMEX, Paris, France
| | - Michelle Rosenzwajg
- Department of Immunology-Immunopathology-Immunotherapy, Université Pierre-et-Marie-Curie Université de Paris 06, Unite Mixte de Recherche (UMR)S959, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Universités, Paris, France; Department of Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France; Department of Internal Medicine and Clinical Immunology, Centre National de Références Maladies Autoimmunes et Systémiques Rares, Centre National de Références Maladies Autoinflammatoires Rares et Amylose Inflammatoire, Paris, France
| | - David Klatzmann
- Department of Immunology-Immunopathology-Immunotherapy, Université Pierre-et-Marie-Curie Université de Paris 06, Unite Mixte de Recherche (UMR)S959, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Universités, Paris, France; Department of Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France; Department of Internal Medicine and Clinical Immunology, Centre National de Références Maladies Autoimmunes et Systémiques Rares, Centre National de Références Maladies Autoinflammatoires Rares et Amylose Inflammatoire, Paris, France
| | - Mohamed Jarraya
- Banque des Tissus Humains, Hôpital Saint Louis, Paris, France
| | - Laurent Chiche
- Service de Chirurgie Vasculaire, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Fabien Koskas
- Service de Chirurgie Vasculaire, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Patrice Cacoub
- Department of Immunology-Immunopathology-Immunotherapy, Université Pierre-et-Marie-Curie Université de Paris 06, Unite Mixte de Recherche (UMR)S959, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Universités, Paris, France; Department of Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France; Department of Internal Medicine and Clinical Immunology, Centre National de Références Maladies Autoimmunes et Systémiques Rares, Centre National de Références Maladies Autoinflammatoires Rares et Amylose Inflammatoire, Paris, France
| | - Gilles Kaplanski
- Centre de Recherche en CardioVasculaire et Nutrition, INSERM U1263, Inrae 1260, Aix-Marseille Université, Marseille, France; Service de Médecine Interne, Centre Hospitalier Universitaire Conception, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - David Saadoun
- Department of Immunology-Immunopathology-Immunotherapy, Université Pierre-et-Marie-Curie Université de Paris 06, Unite Mixte de Recherche (UMR)S959, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Universités, Paris, France; Department of Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France; Department of Internal Medicine and Clinical Immunology, Centre National de Références Maladies Autoimmunes et Systémiques Rares, Centre National de Références Maladies Autoinflammatoires Rares et Amylose Inflammatoire, Paris, France.
| |
Collapse
|
37
|
Kamel AH, Hassanin Sherif EA, Khaled. El Zawawy W, El-shinawy NA. Therapeutic potential of dexamethasone Nano chitosan synthesized from chitosan as a novel treatment of pulmonary fibrosis in C57BL/6 mice. ALEXANDRIA JOURNAL OF MEDICINE 2021. [DOI: 10.1080/20905068.2021.1987795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Afaf Hendawy Kamel
- Department of Zoology, Faculty of Women for Arts, Science and Education, Ain Shams University, Heliopolis, Cairo, Egypt
| | - Eman Adel Hassanin Sherif
- Department of Zoology, Faculty of Women for Arts, Science and Education, Ain Shams University, Heliopolis, Cairo, Egypt
| | - Waleed Khaled. El Zawawy
- Department of Chemical Industries Research Division, National Research Centre, Dokki, Giza, Egypt
| | - Nashwa Ahmed El-shinawy
- Department of Physiology, Zoology Department, Department of Women for Arts, Science and Education, Ain Shams University, Heliopolis, Cairo, Egypt
| |
Collapse
|
38
|
Pejler G, Alanazi S, Grujic M, Adler J, Olsson AK, Sommerhoff CP, Rabelo Melo F. Mast Cell Tryptase Potentiates Neutrophil Extracellular Trap Formation. J Innate Immun 2021; 14:433-446. [PMID: 34937018 PMCID: PMC9485958 DOI: 10.1159/000520972] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/26/2021] [Indexed: 11/26/2022] Open
Abstract
Previous research has indicated an intimate functional communication between mast cells (MCs) and neutrophils during inflammatory conditions, but the nature of such communication is not fully understood. Activated neutrophils are known to release DNA-containing extracellular traps (neutrophil extracellular traps [NETs]) and, based on the known ability of tryptase to interact with negatively charged polymers, we here hypothesized that tryptase might interact with NET-contained DNA and thereby regulate NET formation. In support of this, we showed that tryptase markedly enhances NET formation in phorbol myristate acetate-activated human neutrophils. Moreover, tryptase was found to bind vividly to the NETs, to cause proteolysis of core histones and to cause a reduction in the levels of citrullinated histone-3. Secretome analysis revealed that tryptase caused increased release of numerous neutrophil granule compounds, including gelatinase, lactoferrin, and myeloperoxidase. We also show that DNA can induce the tetrameric, active organization of tryptase, suggesting that NET-contained DNA can maintain tryptase activity in the extracellular milieu. In line with such a scenario, DNA-stabilized tryptase was shown to efficiently degrade numerous pro-inflammatory compounds. Finally, we showed that tryptase is associated with NET formation in vivo in a melanoma setting and that NET formation in vivo is attenuated in mice lacking tryptase expression. Altogether, these findings reveal that NET formation can be regulated by MC tryptase, thus introducing a novel mechanism of communication between MCs and neutrophils.
Collapse
Affiliation(s)
- Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- *Gunnar Pejler,
| | - Sultan Alanazi
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Mirjana Grujic
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jeremy Adler
- Department of Immunology, Genetics and Pathology − BioVis, Uppsala University, Uppsala, Sweden
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | | | - Fabio Rabelo Melo
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- **Fabio Rabelo Melo,
| |
Collapse
|
39
|
Kochetkova M, Samuel MS. Differentiation of the tumor microenvironment: are CAFs the Organizer? Trends Cell Biol 2021; 32:285-294. [PMID: 34895986 DOI: 10.1016/j.tcb.2021.11.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022]
Abstract
Cancers contain a suite of genetically stable cells within an extracellular matrix, collectively termed the tumor microenvironment (TME). The TME strongly influences disease outcome for patients. Gleaning clues from the literature, we propose that the TME should be viewed not as disparate populations of cells constituting a pathological lesion, but as a cohesive tissue constituting a novel pathological organ, arising from the coordinated differentiation of its constituent cell types - a process we have termed tumor-associated neodifferentiation (TAND). We also discuss why cancer-associated fibroblasts (CAFs) may assume the role of Organizer of this organ, directing the recruitment and differentiation of cells within the TME. Viewing the microenvironment in this way will reveal new cancer vulnerabilities that may be exploited for therapy.
Collapse
Affiliation(s)
- Marina Kochetkova
- Centre for Cancer Biology, an alliance between SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia
| | - Michael Susithiran Samuel
- Centre for Cancer Biology, an alliance between SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia.
| |
Collapse
|
40
|
Burgener SS, Brügger M, Leborgne NGF, Sollberger S, Basilico P, Kaufmann T, Bird PI, Benarafa C. Granule Leakage Induces Cell-Intrinsic, Granzyme B-Mediated Apoptosis in Mast Cells. Front Cell Dev Biol 2021; 9:630166. [PMID: 34858967 PMCID: PMC8630627 DOI: 10.3389/fcell.2021.630166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 10/14/2021] [Indexed: 11/29/2022] Open
Abstract
Mast cells are multifunctional immune cells scattered in tissues near blood vessels and mucosal surfaces where they mediate important reactions against parasites and contribute to the pathogenesis of allergic reactions. Serine proteases released from secretory granules upon mast cell activation contribute to these functions by modulating cytokine activity, platelet activation and proteolytic neutralization of toxins. The forced release of granule proteases into the cytosol of mast cells to induce cell suicide has recently been proposed as a therapeutic approach to reduce mast cell numbers in allergic diseases, but the molecular pathways involved in granule-mediated mast cell suicide are incompletely defined. To identify intrinsic granule proteases that can cause mast cell death, we used mice deficient in cytosolic serine protease inhibitors and their respective target proteases. We found that deficiency in Serpinb1a, Serpinb6a, and Serpinb9a or in their target proteases did not alter the kinetics of apoptosis induced by growth factor deprivation in vitro or the number of peritoneal mast cells in vivo. The serine protease cathepsin G induced marginal cell death upon mast cell granule permeabilization only when its inhibitors Serpinb1a or Serpinb6a were deleted. In contrast, the serine protease granzyme B was essential for driving apoptosis in mast cells. On granule permeabilization, granzyme B was required for caspase-3 processing and cell death. Moreover, cytosolic granzyme B inhibitor Serpinb9a prevented caspase-3 processing and mast cell death in a granzyme B-dependent manner. Together, our findings demonstrate that cytosolic serpins provide an inhibitory shield preventing granule protease-induced mast cell apoptosis, and that the granzyme B-Serpinb9a-caspase-3 axis is critical in mast cell survival and could be targeted in the context of allergic diseases.
Collapse
Affiliation(s)
- Sabrina Sofia Burgener
- Institute of Virology and Immunology (IVI), Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Melanie Brügger
- Institute of Virology and Immunology (IVI), Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Science, University of Bern, Bern, Switzerland
| | - Nathan Georges François Leborgne
- Institute of Virology and Immunology (IVI), Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Sophia Sollberger
- Institute of Virology and Immunology (IVI), Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Paola Basilico
- Graduate School for Cellular and Biomedical Science, University of Bern, Bern, Switzerland.,Theodor Kocher Institute, Department of Preclinical Medicine, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Thomas Kaufmann
- Institute of Pharmacology, Department of Preclinical Medicine, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Phillip Ian Bird
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Charaf Benarafa
- Institute of Virology and Immunology (IVI), Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
41
|
Bradding P. Mechanisms of Mast Cell Activation in Severe Asthma - Beyond IgE. Am J Respir Crit Care Med 2021; 205:375-377. [PMID: 34856107 PMCID: PMC8886944 DOI: 10.1164/rccm.202110-2322ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Peter Bradding
- University of Leicester, 4488, Respiratory Sciences, Leicester, United Kingdom of Great Britain and Northern Ireland;
| |
Collapse
|
42
|
Ozpinar EW, Frey AL, Cruse G, Freytes DO. Mast Cell-Biomaterial Interactions and Tissue Repair. TISSUE ENGINEERING. PART B, REVIEWS 2021; 27:590-603. [PMID: 33164714 PMCID: PMC8739845 DOI: 10.1089/ten.teb.2020.0275] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Tissue engineers often use biomaterials to provide structural support along with mechanical and chemical signals to modulate the wound healing process. Biomaterials that are implanted into the body interact with a heterogeneous and dynamic inflammatory environment that is present at the site of injury. Whether synthetically derived, naturally derived, or a combination of both, it is important to assess biomaterials for their ability to modulate inflammation to understand their potential clinical use. One important, but underexplored cell in the context of biomaterials is the mast cell (MC). MCs are granulocytic leukocytes that engage in a variety of events in both the innate and adaptive immune systems. Although highly recognized for their roles in allergic reactions, MCs play an important role in wound healing by recognizing antigens through pattern recognition receptors and the high-affinity immunoglobulin E receptor (FceRI) and releasing granules that affect cell recruitment, fibrosis, extracellular matrix deposition, angiogenesis, and vasculogenesis. MCs also mediate the foreign body response, contributing to the incorporation or rejection of implants. Studies of MC-biomaterial interactions can aid in the elucidation of MC roles during the host tissue response and tissue repair. This review is designed for those in the tissue engineering and biomaterial fields who are interested in exploring the role MCs may play in wound-biomaterial interactions and wound healing. With this review, we hope to inspire more research in the MC-biomaterial space to accelerate the design and construction of optimized implants. Impact statement Mast cells (MCs) are highly specialized inflammatory cells that have crucial, but not fully understood, roles in wound healing and tissue repair. Upon stimulation, they recognize foreign antigens and release granules that help orchestrate the inflammatory response after tissue damage or biomaterial implantation. This review summarizes the current use of MCs in biomaterial research along with literature from the past decade focusing on MC interactions with materials used for tissue repair and regeneration. Studying MC-biomaterial interactions will help (i) further understand the process of inflammation and (ii) design biomaterials and tissue-engineered constructs for optimal repair and regeneration.
Collapse
Affiliation(s)
- Emily W Ozpinar
- The Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Ariana L Frey
- The Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
| | - Glenn Cruse
- The Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Donald O Freytes
- The Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
43
|
Mace EL, Zhao S, Lipscomb B, Wootten CT, Belcher RH. Clinical Significance of Mast Cells in the Supraglottic Larynx of Children With Aerodigestive Disease. Otolaryngol Head Neck Surg 2021; 167:375-381. [PMID: 34699295 DOI: 10.1177/01945998211055139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To analyze the association of mast cells found on supraglottic biopsy of pediatric patients with common aerodigestive diseases. STUDY DESIGN Cross-sectional study. SETTING Tertiary care children's hospital. METHODS A total of 461 pediatric patients undergoing otolaryngology aerodigestive procedures provided consent between 2014 and 2019, and biopsies of the supraglottic larynx were collected at the time of their surgery. Pathologists reviewed biopsies for the presence and number of mast cells per high-power field. The patients' electronic health records were reviewed for relevant demographic data and clinical diagnoses present at the time of biopsy. Multivariate logistic regression was used to assess the relationship of mast cells with odds of aerodigestive disease. RESULTS Patients with mast cells in their biopsy had significantly higher odds of asthma (odds ratio [OR], 2.02; 95% CI, 1.17-3.46), gastroesophageal reflux disease (OR, 2.36; 95% CI, 1.47-3.77), laryngomalacia (OR, 2.98; 95% CI, 1.80-4.94), laryngeal anomalies (OR, 2.32; 95% CI, 1.52-3.55), and obstructive sleep apnea (OR, 2.16; 95% CI, 1.35-3.45). When mast cells were evaluated as a continuous variable, there was a nonlinear relationship between increasing mast cell count and odds of disease. CONCLUSIONS Mast cells are known to be associated with inflammatory conditions, though little is known about their presence in laryngeal inflammation. Results from our study demonstrate an association between mast cells in the pediatric larynx and asthma, gastroesophageal reflux disease, laryngomalacia, laryngeal anomalies, and obstructive sleep apnea. Our study also showed a nonlinear relationship between number of mast cells and odds of disease diagnosis.
Collapse
Affiliation(s)
- Emily L Mace
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Brittany Lipscomb
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Christopher T Wootten
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ryan H Belcher
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
44
|
Glutamate triggers the expression of functional ionotropic and metabotropic glutamate receptors in mast cells. Cell Mol Immunol 2021; 18:2383-2392. [PMID: 32313211 PMCID: PMC8484602 DOI: 10.1038/s41423-020-0421-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 03/16/2020] [Indexed: 02/07/2023] Open
Abstract
Mast cells are emerging as players in the communication between peripheral nerve endings and cells of the immune system. However, it is not clear the mechanism by which mast cells communicate with peripheral nerves. We previously found that mast cells located within healing tendons can express glutamate receptors, raising the possibility that mast cells may be sensitive to glutamate signaling. To evaluate this hypothesis, we stimulated primary mast cells with glutamate and showed that glutamate induced the profound upregulation of a panel of glutamate receptors of both the ionotropic type (NMDAR1, NMDAR2A, and NMDAR2B) and the metabotropic type (mGluR2 and mGluR7) at both the mRNA and protein levels. The binding of glutamate to glutamate receptors on the mast cell surface was confirmed. Further, glutamate had extensive effects on gene expression in the mast cells, including the upregulation of pro-inflammatory components such as IL-6 and CCL2. Glutamate also induced the upregulation of transcription factors, including Egr2, Egr3 and, in particular, FosB. The extensive induction of FosB was confirmed by immunofluorescence assessment. Glutamate receptor antagonists abrogated the responses of the mast cells to glutamate, supporting the supposition of a functional glutamate-glutamate receptor axis in mast cells. Finally, we provide in vivo evidence supporting a functional glutamate-glutamate receptor axis in the mast cells of injured tendons. Together, these findings establish glutamate as an effector of mast cell function, thereby introducing a novel principle for how cells in the immune system can communicate with nerve cells.
Collapse
|
45
|
Song J, He Z, Yang M, Yu T, Wang X, Liu B, Li J. HepaticIschemia/Reperfusion Injuryinvolves functional tryptase/PAR-2 signaling in liver sinusoidal endothelial cell population. Int Immunopharmacol 2021; 100:108052. [PMID: 34454294 DOI: 10.1016/j.intimp.2021.108052] [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: 04/04/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023]
Abstract
Mast cells (MCs) are tissue-resident effector cells that could be the earliest responder to release a unique, stimulus-specific set of mediators in hepatic ischemia-reperfusion (IR) injury However, how MCs function in the hepatic IR has remained a formidable challenge due to the substantial redundancy and functional diverse of these mediators. Tryptase is the main protease for degranulation of MCs and its receptor-protease-activated receptor 2 (PAR-2) is widely expressed in endothelial cells. It is unclear whether and how tryptase/PAR-2 axis participates in hepatic IR. We employed an experimental warm 70% liver IR model in mice and found that tryptase was accumulated in the circulation during hepatic IR and positively correlated with liver injury. Tryptase inhibition by protamine can significantly down-regulate the expression of adhesion molecules and reduce neutrophil infiltration within the liver. The level of inflammatory factors and chemokines were also consistent with the pathological change of the liver. In addition, the treatment with exogeneous tryptase in MC-deficient mice can induce the damage observed in wild type mice in the context of liver IR. In vitro, neutrophil infiltration and inflammatory factor secretion were regulated by Tryptase/PAR-2, involving the adhesion molecule expression to regulate neutrophil adhesion dependent on NF-κB pathway. Conclusion: tryptase/PAR-2 participates in liver injury through the activation of LSECs in the early phase of liver IR.
Collapse
Affiliation(s)
- Jian Song
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China; Department of General Surgery, Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, 68 Zhongshan Road, Wuxi 214002, Jiangsu, China
| | - Zhigang He
- Department of Plastic and Constructive Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Muqing Yang
- Department of General Surgery, Shanghai Tenth People's Hospital School of Medicine, Tongji University, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Tianyu Yu
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China
| | - Xiaodong Wang
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China
| | - Bin Liu
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China
| | - Jiyu Li
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China.
| |
Collapse
|
46
|
Babaei-Jadidi R, Dongre A, Miller S, Castellanos Uribe M, Stewart ID, Thompson ZM, Nateri AS, Bradding P, May ST, Clements D, Johnson SR. Mast-Cell Tryptase Release Contributes to Disease Progression in Lymphangioleiomyomatosis. Am J Respir Crit Care Med 2021; 204:431-444. [PMID: 33882264 DOI: 10.1164/rccm.202007-2854oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: Lymphangioleiomyomatosis (LAM) is a multisystem disease that causes lung cysts and respiratory failure. Loss of TSC (tuberous sclerosis complex) gene function results in a clone of "LAM cells" with dysregulated mTOR (mechanistic target of rapamycin) activity. LAM cells and fibroblasts form lung nodules that also contain mast cells, although their significance is unknown. Objectives: To understand the mechanism of mast-cell accumulation and the role of mast cells in the pathogenesis of LAM. Methods: Gene expression was examined using transcriptional profiling and qRT-PCR. Mast cell/LAM nodule interactions were examined in vitro using spheroid TSC2-null cell/fibroblast cocultures and in vivo using an immunocompetent Tsc2-null murine homograft model. Measurements and Main Results: LAM-derived cell/fibroblast cocultures induced multiple CXC chemokines in fibroblasts. LAM lungs had increased tryptase-positive mast cells expressing CXCRs (CXC chemokine receptors) (P < 0.05). Mast cells located around the periphery of LAM nodules were positively associated with the rate of lung function loss (P = 0.016). LAM spheroids attracted mast cells, and this process was inhibited by pharmacologic and CRISPR/cas9 inhibition of CXCR1 and CXCR2. LAM spheroids caused mast-cell tryptase release, which induced fibroblast proliferation and increased LAM-spheroid size (1.36 ± 0.24-fold; P = 0.0019). The tryptase inhibitor APC366 and sodium cromoglycate (SCG) inhibited mast cell-induced spheroid growth. In vivo, SCG reduced mast-cell activation and Tsc2-null lung tumor burden (vehicle: 32.5.3% ± 23.6%; SCG: 5.5% ± 4.3%; P = 0.0035). Conclusions: LAM-cell/fibroblast interactions attract mast cells where tryptase release contributes to disease progression. Repurposing SCG for use in LAM should be studied as an alternative or adjunct to mTOR inhibitor therapy.
Collapse
Affiliation(s)
- Roya Babaei-Jadidi
- Division of Respiratory Medicine, National Institute for Health Research Biomedical Research Centre and Biodiscovery Institute
| | - Arundhati Dongre
- Division of Respiratory Medicine, National Institute for Health Research Biomedical Research Centre and Biodiscovery Institute
| | - Suzanne Miller
- Division of Respiratory Medicine, National Institute for Health Research Biomedical Research Centre and Biodiscovery Institute
| | | | - Ian D Stewart
- Division of Respiratory Medicine, National Institute for Health Research Biomedical Research Centre and Biodiscovery Institute
| | - Zoe M Thompson
- Division of Respiratory Medicine, National Institute for Health Research Biomedical Research Centre and Biodiscovery Institute
| | - Abdolrahman S Nateri
- Cancer Genetics & Stem Cell Group, Division of Cancer and Stem Cells, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Peter Bradding
- Department of Respiratory Sciences, Institute for Lung Health, University of Leicester, Leicester, United Kingdom.,Respiratory Theme, National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom; and
| | - Sean T May
- Nottingham Arabidopsis Stock Centre, and
| | - Debbie Clements
- Division of Respiratory Medicine, National Institute for Health Research Biomedical Research Centre and Biodiscovery Institute
| | - Simon R Johnson
- Division of Respiratory Medicine, National Institute for Health Research Biomedical Research Centre and Biodiscovery Institute.,National Centre for Lymphangioleiomyomatosis, Nottingham University Hospitals National Health Service Trust, Nottingham, United Kingdom
| |
Collapse
|
47
|
Durant F, Whited JL. Finding Solutions for Fibrosis: Understanding the Innate Mechanisms Used by Super-Regenerator Vertebrates to Combat Scarring. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100407. [PMID: 34032013 PMCID: PMC8336523 DOI: 10.1002/advs.202100407] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/12/2021] [Indexed: 05/08/2023]
Abstract
Soft tissue fibrosis and cutaneous scarring represent massive clinical burdens to millions of patients per year and the therapeutic options available are currently quite limited. Despite what is known about the process of fibrosis in mammals, novel approaches for combating fibrosis and scarring are necessary. It is hypothesized that scarring has evolved as a solution to maximize healing speed to reduce fluid loss and infection. This hypothesis, however, is complicated by regenerative animals, which have arguably the most remarkable healing abilities and are capable of scar-free healing. This review explores the differences observed between adult mammalian healing that typically results in fibrosis versus healing in regenerative animals that heal scarlessly. Each stage of wound healing is surveyed in depth from the perspective of many regenerative and fibrotic healers so as to identify the most important molecular and physiological variances along the way to disparate injury repair outcomes. Understanding how these powerful model systems accomplish the feat of scar-free healing may provide critical therapeutic approaches to the treatment or prevention of fibrosis.
Collapse
Affiliation(s)
- Fallon Durant
- Department of Stem Cell and Regenerative BiologyHarvard UniversityCambridgeMA02138USA
| | - Jessica L. Whited
- Department of Stem Cell and Regenerative BiologyHarvard UniversityCambridgeMA02138USA
- The Harvard Stem Cell InstituteCambridgeMA02138USA
| |
Collapse
|
48
|
Liu X, Shi GP, Guo J. Innate Immune Cells in Pressure Overload-Induced Cardiac Hypertrophy and Remodeling. Front Cell Dev Biol 2021; 9:659666. [PMID: 34368120 PMCID: PMC8343105 DOI: 10.3389/fcell.2021.659666] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/28/2021] [Indexed: 12/23/2022] Open
Abstract
Pressure overload and heart failure are among the leading causes of cardiovascular morbidity and mortality. Accumulating evidence suggests that inflammatory cell activation and release of inflammatory mediators are of vital importance during the pathogenesis of these cardiac diseases. Yet, the roles of innate immune cells and subsequent inflammatory events in these processes remain poorly understood. Here, we outline the possible underlying mechanisms of innate immune cell participation, including mast cells, macrophages, monocytes, neutrophils, dendritic cells, eosinophils, and natural killer T cells in these pathological processes. Although these cells accumulate in the atrium or ventricles at different time points after pressure overload, their cardioprotective or cardiodestructive activities differ from each other. Among them, mast cells, neutrophils, and dendritic cells exert detrimental function in experimental models, whereas eosinophils and natural killer T cells display cardioprotective activities. Depending on their subsets, macrophages and monocytes may exacerbate cardiodysfunction or negatively regulate cardiac hypertrophy and remodeling. Pressure overload stimulates the secretion of cytokines, chemokines, and growth factors from innate immune cells and even resident cardiomyocytes that together assist innate immune cell infiltration into injured heart. These infiltrates are involved in pro-hypertrophic events and cardiac fibroblast activation. Immune regulation of cardiac innate immune cells becomes a promising therapeutic approach in experimental cardiac disease treatment, highlighting the significance of their clinical evaluation in humans.
Collapse
Affiliation(s)
- Xin Liu
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Junli Guo
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research & Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|
49
|
Michailidou D, Schwartz DM, Mustelin T, Hughes GC. Allergic Aspects of IgG4-Related Disease: Implications for Pathogenesis and Therapy. Front Immunol 2021; 12:693192. [PMID: 34305927 PMCID: PMC8292787 DOI: 10.3389/fimmu.2021.693192] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/03/2021] [Indexed: 01/05/2023] Open
Abstract
IgG4-related disease (IgG4-RD) is a rare systemic fibroinflammatory disease frequently associated with allergy. The pathogenesis of IgG4-RD is poorly understood, and effective therapies are limited. However, IgG4-RD appears to involve some of the same pathogenic mechanisms observed in allergic disease, such as T helper 2 (Th2) and regulatory T cell (Treg) activation, IgG4 and IgE hypersecretion, and blood/tissue eosinophilia. In addition, IgG4-RD tissue fibrosis appears to involve activation of basophils and mast cells and their release of alarmins and cytokines. In this article, we review allergy-like features of IgG4-RD and highlight targeted therapies for allergy that have potential in treating patients with IgG4-RD.
Collapse
Affiliation(s)
- Despina Michailidou
- Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Daniella Muallem Schwartz
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Tomas Mustelin
- Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Grant C. Hughes
- Division of Rheumatology, University of Washington, Seattle, WA, United States
| |
Collapse
|
50
|
Garcia-Rodriguez KM, Bini EI, Gamboa-Domínguez A, Espitia-Pinzón CI, Huerta-Yepez S, Bulfone-Paus S, Hernández-Pando R. Differential mast cell numbers and characteristics in human tuberculosis pulmonary lesions. Sci Rep 2021; 11:10687. [PMID: 34021178 PMCID: PMC8140073 DOI: 10.1038/s41598-021-89659-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/20/2021] [Indexed: 01/31/2023] Open
Abstract
Tuberculosis (TB) is still a major worldwide health threat and primarily a lung disease. The innate immune response against Mycobacterium tuberculosis (Mtb) is orchestrated by dendritic cells, macrophages, neutrophils, natural killer cells and apparently mast cells (MCs). MCs are located at mucosal sites including the lungs and contribute in host-defence against pathogens, but little is known about their role during Mtb infection. This study investigates the location and characteristics of MCs in TB lesions to assess their contribution to TB pathology. To this purpose, number, location and phenotype of MCs was studied in 11 necropsies of pulmonary TB and 3 necropsies of non-TB infected lungs that were used as controls. MCs were localised at pneumonic areas, in the granuloma periphery and particularly abundant in fibrotic tissue. Furthermore, MCs displayed intracellular Mtb and IL-17A and TGF-β immunostaining. These findings were validated by analysing, post-mortem lung tissue microarrays from 44 individuals with pulmonary TB and 25 control subjects. In affected lungs, increased numbers of MCs expressing intracellularly both tryptase and chymase were found at fibrotic sites. Altogether, our data suggest that MCs are recruited at the inflammatory site and that actively produce immune mediators such as proteases and TGF-β that may be contributing to late fibrosis in TB lesions.
Collapse
Affiliation(s)
- Karen Magdalena Garcia-Rodriguez
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester , Manchester, UK
| | - Estela Isabel Bini
- Seccion de Patologia Experimental, Instituto Nacional de Ciencias Medicas y Nutricion "Salvador Zubiran", Mexico City, Mexico
| | - Armando Gamboa-Domínguez
- Seccion de Patologia Experimental, Instituto Nacional de Ciencias Medicas y Nutricion "Salvador Zubiran", Mexico City, Mexico
| | - Clara Inés Espitia-Pinzón
- Departamento de Inmunologia, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Sara Huerta-Yepez
- Unidad de Investigacion en Enfermedades Oncologicas, Hospital Infantil de Mexico, Federico Gomez, Mexico City, Mexico
| | - Silvia Bulfone-Paus
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester , Manchester, UK.,Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Rogelio Hernández-Pando
- Seccion de Patologia Experimental, Instituto Nacional de Ciencias Medicas y Nutricion "Salvador Zubiran", Mexico City, Mexico.
| |
Collapse
|