101
|
Li L, Guan YC, Bai SY, Jin QW, Tao JP, Zhu GD, Huang SY. Mineralization Reduces the Toxicity and Improves Stability and Protective Immune Response Induced by Toxoplasma gondii. Vaccines (Basel) 2023; 12:35. [PMID: 38250848 PMCID: PMC10819335 DOI: 10.3390/vaccines12010035] [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: 11/25/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 01/23/2024] Open
Abstract
Vaccination is an ideal strategy for the control and prevention of toxoplasmosis. However, the thermostability and effectiveness of vaccines limit their application. Here, calcium mineralization was used to fabricate Toxoplasma gondii tachyzoites as immunogenic core-shell particles with improved immune response and thermostability. In the current study, T. gondii RH particles coated with mineralized shells were fabricated by calcium mineralization. The mineralized shells could maintain the T. gondii tachyzoites structural integrity for at least 12 months and weaken the virulence. Immunization of mice with mineralized tachyzoites induced high levels of T. gondii-specific antibodies and cytokines. The immunized mice were protected with a 100% survival rate in acute and chronic infection, and brain cyst burdens were significantly reduced. This study reported for the first time the strategy of calcium mineralization on T. gondii and proved that mineralized tachyzoites could play an immune protective role, thus expanding the application of biomineralization in T. gondii vaccine delivery.
Collapse
Affiliation(s)
- Ling Li
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China; (L.L.); (Y.-C.G.); (S.-Y.B.); (Q.-W.J.); (J.-P.T.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yong-Chao Guan
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China; (L.L.); (Y.-C.G.); (S.-Y.B.); (Q.-W.J.); (J.-P.T.)
| | - Shao-Yuan Bai
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China; (L.L.); (Y.-C.G.); (S.-Y.B.); (Q.-W.J.); (J.-P.T.)
| | - Qi-Wang Jin
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China; (L.L.); (Y.-C.G.); (S.-Y.B.); (Q.-W.J.); (J.-P.T.)
| | - Jian-Ping Tao
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China; (L.L.); (Y.-C.G.); (S.-Y.B.); (Q.-W.J.); (J.-P.T.)
| | - Guo-Ding Zhu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China;
| | - Si-Yang Huang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China; (L.L.); (Y.-C.G.); (S.-Y.B.); (Q.-W.J.); (J.-P.T.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
102
|
Malih S, Song YS, Sorenson CM, Sheibani N. Choroidal Mast Cells and Pathophysiology of Age-Related Macular Degeneration. Cells 2023; 13:50. [PMID: 38201254 PMCID: PMC10778483 DOI: 10.3390/cells13010050] [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: 10/20/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Age-related macular degeneration (AMD) remains a leading cause of vision loss in elderly patients. Its etiology and progression are, however, deeply intertwined with various cellular and molecular interactions within the retina and choroid. Among the key cellular players least studied are choroidal mast cells, with important roles in immune and allergic responses. Here, we will review what is known regarding the pathophysiology of AMD and expand on the recently proposed intricate roles of choroidal mast cells and their activation in outer retinal degeneration and AMD pathogenesis. We will focus on choroidal mast cell activation, the release of their bioactive mediators, and potential impact on ocular oxidative stress, inflammation, and overall retinal and choroidal health. We propose an important role for thrombospondin-1 (TSP1), a major ocular angioinflammatory factor, in regulation of choroidal mast cell homeostasis and activation in AMD pathogenesis. Drawing from limited studies, this review underscores the need for further comprehensive studies aimed at understanding the precise roles changes in TSP1 levels and choroidal mast cell activity play in pathophysiology of AMD. We will also propose potential therapeutic strategies targeting these regulatory pathways, and highlighting the promise they hold for curbing AMD progression through modulation of mast cell activity. In conclusion, the evolving understanding of the role of choroidal mast cells in AMD pathogenesis will not only offer deeper insights into the underlying mechanisms but will also offer opportunities for development of novel preventive strategies.
Collapse
Affiliation(s)
- Sara Malih
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.M.); (Y.-S.S.)
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 15614, Iran
| | - Yong-Seok Song
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.M.); (Y.-S.S.)
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Christine M. Sorenson
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.M.); (Y.-S.S.)
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
103
|
Visani de Luna LA, Loret T, He Y, Legnani M, Lin H, Galibert AM, Fordham A, Holme S, Del Rio Castillo AE, Bonaccorso F, Bianco A, Flahaut E, Kostarelos K, Bussy C. Pulmonary Toxicity of Boron Nitride Nanomaterials Is Aspect Ratio Dependent. ACS NANO 2023; 17:24919-24935. [PMID: 38051272 PMCID: PMC10753895 DOI: 10.1021/acsnano.3c06599] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/26/2023] [Accepted: 12/01/2023] [Indexed: 12/07/2023]
Abstract
Boron nitride (BN) nanomaterials have drawn a lot of interest in the material science community. However, extensive research is still needed to thoroughly analyze their safety profiles. Herein, we investigated the pulmonary impact and clearance of two-dimensional hexagonal boron nitride (h-BN) nanosheets and boron nitride nanotubes (BNNTs) in mice. Animals were exposed by single oropharyngeal aspiration to h-BN or BNNTs. On days 1, 7, and 28, bronchoalveolar lavage (BAL) fluids and lungs were collected. On one hand, adverse effects on lungs were evaluated using various approaches (e.g., immune response, histopathology, tissue remodeling, and genotoxicity). On the other hand, material deposition and clearance from the lungs were assessed. Two-dimensional h-BN did not cause any significant immune response or lung damage, although the presence of materials was confirmed by Raman spectroscopy. In addition, the low aspect ratio h-BN nanosheets were internalized rapidly by phagocytic cells present in alveoli, resulting in efficient clearance from the lungs. In contrast, high aspect ratio BNNTs caused a strong and long-lasting inflammatory response, characterized by sustained inflammation up to 28 days after exposure and the activation of both innate and adaptive immunity. Moreover, the presence of granulomatous structures and an indication of ongoing fibrosis as well as DNA damage in the lung parenchyma were evidenced with these materials. Concurrently, BNNTs were identified in lung sections for up to 28 days, suggesting long-term biopersistence, as previously demonstrated for other high aspect ratio nanomaterials with poor lung clearance such as multiwalled carbon nanotubes (MWCNTs). Overall, we reveal the safer toxicological profile of BN-based two-dimensional nanosheets in comparison to their nanotube counterparts. We also report strong similarities between BNNTs and MWCNTs in lung response, emphasizing their high aspect ratio as a major driver of their toxicity.
Collapse
Affiliation(s)
- Luis Augusto Visani de Luna
- Nanomedicine
Lab, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, U.K.
- National
Graphene Institute, The University of Manchester, Manchester, M13 9PL, U.K.
- Lydia
Becker Institute of Immunology and Inflammation, Faculty of Biology,
Medicine and Health, The University of Manchester,
Manchester Academic Health Science Centre, Manchester M13 9PT, U.K.
| | - Thomas Loret
- Nanomedicine
Lab, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, U.K.
- National
Graphene Institute, The University of Manchester, Manchester, M13 9PL, U.K.
- Lydia
Becker Institute of Immunology and Inflammation, Faculty of Biology,
Medicine and Health, The University of Manchester,
Manchester Academic Health Science Centre, Manchester M13 9PT, U.K.
| | - Yilin He
- CNRS,
Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University
of Strasbourg, ISIS, 67000 Strasbourg, France
| | - Morgan Legnani
- CIRIMAT,
Université Toulouse 3 Paul Sabatier, Toulouse INP, CNRS, Université
de Toulouse, 118 Route de Narbonne, 31062 Toulouse cedex 9, France
| | - Hazel Lin
- CNRS,
Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University
of Strasbourg, ISIS, 67000 Strasbourg, France
| | - Anne Marie Galibert
- CIRIMAT,
Université Toulouse 3 Paul Sabatier, Toulouse INP, CNRS, Université
de Toulouse, 118 Route de Narbonne, 31062 Toulouse cedex 9, France
| | - Alexander Fordham
- Nanomedicine
Lab, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, U.K.
- National
Graphene Institute, The University of Manchester, Manchester, M13 9PL, U.K.
- Lydia
Becker Institute of Immunology and Inflammation, Faculty of Biology,
Medicine and Health, The University of Manchester,
Manchester Academic Health Science Centre, Manchester M13 9PT, U.K.
| | - Sonja Holme
- Nanomedicine
Lab, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, U.K.
- National
Graphene Institute, The University of Manchester, Manchester, M13 9PL, U.K.
- Lydia
Becker Institute of Immunology and Inflammation, Faculty of Biology,
Medicine and Health, The University of Manchester,
Manchester Academic Health Science Centre, Manchester M13 9PT, U.K.
| | | | - Francesco Bonaccorso
- BeDimensional
S.p.A., Lungo Torrente
Secca 30r, 16163 Genoa, Italy
- Istituto
Italiano di Tecnologia, Graphene Laboratories, Via Morego 30, 16163 Genoa, Italy
| | - Alberto Bianco
- CNRS,
Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University
of Strasbourg, ISIS, 67000 Strasbourg, France
| | - Emmanuel Flahaut
- CIRIMAT,
Université Toulouse 3 Paul Sabatier, Toulouse INP, CNRS, Université
de Toulouse, 118 Route de Narbonne, 31062 Toulouse cedex 9, France
| | - Kostas Kostarelos
- Nanomedicine
Lab, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, U.K.
- National
Graphene Institute, The University of Manchester, Manchester, M13 9PL, U.K.
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST,, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Cyrill Bussy
- Nanomedicine
Lab, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, U.K.
- National
Graphene Institute, The University of Manchester, Manchester, M13 9PL, U.K.
- Lydia
Becker Institute of Immunology and Inflammation, Faculty of Biology,
Medicine and Health, The University of Manchester,
Manchester Academic Health Science Centre, Manchester M13 9PT, U.K.
| |
Collapse
|
104
|
Crocetto F, Imbimbo C, Barone B, Turchino D, Bracale UM, Peluso A, Panagrosso M, Falcone A, Mirto BF, De Luca L, Sicignano E, Del Giudice F, Busetto GM, Lucarelli G, Giampaglia G, Manfredi C, Ferro M, Tarantino G. Which inflammatory marker, between systemic immune-inflammation index and neutrophil to eosinophil ratio, is associated with Peyronie's disease and are there any implications for a better understanding of its mechanisms? Basic Clin Androl 2023; 33:38. [PMID: 38110896 PMCID: PMC10729439 DOI: 10.1186/s12610-023-00213-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/04/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Peyronie's disease affects up to 9% of men and is often accompanied by pain and/or erectile dysfunction. It is characterized by an inflammatory process that is the grassroots of the subsequent fibrosis stage. There is an unmet need to evaluate its onset and progression. Among the newly proposed biomarkers of inflammation, authors developed a novel systemic immune-inflammation index (SII) based on lymphocyte, neutrophil, and platelet counts. Similarly, a recent study reported that a neutrophil-to-eosinophil ratio (NER) represents systemic inflammation. RESULTS A 49-patient group with Peyronie's disease as confronted with 50 well-matched for age and BMI controls. As laboratory evaluation of inflammation, SII, NER and the eosinophil to neutrophil ratio (ENR) were studied. As a likely risk factor for the presence of Peyronie's disease, a higher prevalence of hypercholesterolemia, hyperglycemia and hypertension was discovered in the patients compared to controls. A significant difference was found in the median values of the NER between the two selected groups, i.e., 32.5 versus 17.3 (p = 0.0021). As expected, also ENR was significantly different. The receiver operating characteristic curves for SII, ENR and NER were 0.55, 0.32 and 0.67, respectively, highlighting the best performance of NER. The cut-off for NER was 12.1, according to the Youden test. CONCLUSIONS According to our results, any evaluation of circulating eosinophil, evaluated as NER, beyond being a signature of immuno-inflammatory response, help assess tissue homeostasis, since eosinophils are now considered multifunctional leukocytes and give a picture of the inflammatory process and repair process belonging to Peyronie's disease.
Collapse
Affiliation(s)
- Felice Crocetto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", 80131, Naples, Italy
| | - Ciro Imbimbo
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", 80131, Naples, Italy
| | - Biagio Barone
- Division of Urology, Department of Surgical Sciences, AORN Sant'Anna e San Sebastiano, 81100, Caserta, Italy.
| | - Davide Turchino
- Department of Public Health, Vascular Surgery Unit, University of Naples Federico II, 80131, Naples, Italy
| | - Umberto Marcello Bracale
- Department of Public Health, Vascular Surgery Unit, University of Naples Federico II, 80131, Naples, Italy
| | - Antonio Peluso
- Department of Public Health, Vascular Surgery Unit, University of Naples Federico II, 80131, Naples, Italy
| | - Marco Panagrosso
- Department of Public Health, Vascular Surgery Unit, University of Naples Federico II, 80131, Naples, Italy
| | - Alfonso Falcone
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", 80131, Naples, Italy
| | - Benito Fabio Mirto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", 80131, Naples, Italy
| | - Luigi De Luca
- Division of Urology, AORN "Antonio Cardarelli", Naples, Italy
| | - Enrico Sicignano
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", 80131, Naples, Italy
| | - Francesco Del Giudice
- Department of Maternal Infant and Urologic Sciences, Policlinico Umberto I Hospital, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Gian Maria Busetto
- Department of Urology and Organ Transplantation, University of Foggia, 71122, Foggia, Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Kidney Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124, Bari, Italy
| | - Gaetano Giampaglia
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", 80131, Naples, Italy
| | - Celeste Manfredi
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", 80131, Naples, Italy
| | - Matteo Ferro
- Department of Urology, IEO, European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II Medical School of Naples, Naples, Italy
| |
Collapse
|
105
|
Hu W, Meng L, Wang C, Lu W, Tong X, Lin R, Xu T, Chen L, Cui A, Xu X, Li A, Tang J, Gao H, Pei Z, Zhang R, Wang Y, Wang Y, Han W, Jiang N, Xiong C, Feng Y, Lee K, Chen M. Spatiotemporal observations of host-pathogen interactions in mucosa during SARS-CoV-2 infection indicate a protective role of ILC2s. Microbiol Spectr 2023; 11:e0087823. [PMID: 37937994 PMCID: PMC10714800 DOI: 10.1128/spectrum.00878-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/30/2023] [Indexed: 11/09/2023] Open
Abstract
IMPORTANCE Our study revealed the spatial interaction between humanized ACE2 and pseudovirus expressing Spike, emphasizing the role of type 2 innate lymphoid cells during the initial phase of viral infection. These findings provide a foundation for the development of mucosal vaccines and other treatment approaches for both pre- and post-infection management of coronavirus disease 2019.
Collapse
Affiliation(s)
- Wei Hu
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Lu Meng
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Chao Wang
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenhan Lu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Rui Lin
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Tao Xu
- Department of Infectious Diseases, National Medical Center for Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liang Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - An Cui
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoqing Xu
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Anni Li
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jia Tang
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongru Gao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhenle Pei
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Ruonan Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Yicong Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Yu Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Wendong Han
- Biosafety Level 3 Laboratory, Shanghai Medical College Fudan University, Shanghai, China
| | - Ning Jiang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Chenglong Xiong
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Kuinyu Lee
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Mingquan Chen
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
106
|
Pereira ENGDS, de Araujo BP, Rodrigues KL, Silvares RR, Guimarães FV, Martins CSM, Flores EEI, Silva PMRE, Daliry A. Cholesterol Exacerbates the Pathophysiology of Non-Alcoholic Steatohepatitis by Upregulating Hypoxia-Inducible Factor 1 and Modulating Microcirculatory Dysfunction. Nutrients 2023; 15:5034. [PMID: 38140293 PMCID: PMC10745917 DOI: 10.3390/nu15245034] [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: 11/04/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Cholesterol is a pivotal lipotoxic molecule that contributes to the progression of Non-Alcoholic Steatohepatitis NASH). Additionally, microcirculatory changes are critical components of Non-Alcoholic Fatty Liver Disease (NAFLD) pathogenesis. This study aimed to investigate the role of cholesterol as an insult that modulates microcirculatory damage in NAFLD and the underlying mechanisms. The experimental model was established in male C57BL/6 mice fed a high-fat high-carbohydrate (HFHC) diet for 39 weeks. Between weeks 31-39, 2% cholesterol was added to the HFHC diet in a subgroup of mice. Leukocyte recruitment and hepatic stellate cells (HSC) activation in microcirculation were assessed using intravital microscopy. The hepatic microvascular blood flow (HMBF) was measured using laser speckle flowmetry. High cholesterol levels exacerbated hepatomegaly, hepatic steatosis, inflammation, fibrosis, and leukocyte recruitment compared to the HFHC group. In addition, cholesterol decreased the HMBF-cholesterol-induced activation of HSC and increased HIF1A expression in the liver. Furthermore, cholesterol promoted a pro-inflammatory cytokine profile with a Th1-type immune response (IFN-γ/IL-4). These findings suggest cholesterol exacerbates NAFLD progression through microcirculatory dysfunction and HIF1A upregulation through hypoxia and inflammation. This study highlights the importance of cholesterol-induced lipotoxicity, which causes microcirculatory dysfunction associated with NAFLD pathology, thus reinforcing the potential of lipotoxicity and microcirculation as therapeutic targets for NAFLD.
Collapse
Affiliation(s)
- Evelyn Nunes Goulart da Silva Pereira
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| | - Beatriz Peres de Araujo
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| | - Karine Lino Rodrigues
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| | - Raquel Rangel Silvares
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| | - Fernanda Verdini Guimarães
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
| | - Carolina Souza Machado Martins
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| | - Edgar Eduardo Ilaquita Flores
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| | | | - Anissa Daliry
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| |
Collapse
|
107
|
Casari M, Siegl D, Deppermann C, Schuppan D. Macrophages and platelets in liver fibrosis and hepatocellular carcinoma. Front Immunol 2023; 14:1277808. [PMID: 38116017 PMCID: PMC10728659 DOI: 10.3389/fimmu.2023.1277808] [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: 08/15/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023] Open
Abstract
During fibrosis, (myo)fibroblasts deposit large amounts of extracellular matrix proteins, thereby replacing healthy functional tissue. In liver fibrosis, this leads to the loss of hepatocyte function, portal hypertension, variceal bleeding, and increased susceptibility to infection. At an early stage, liver fibrosis is a dynamic and reversible process, however, from the cirrhotic stage, there is significant progression to hepatocellular carcinoma. Both liver-resident macrophages (Kupffer cells) and monocyte-derived macrophages are important drivers of fibrosis progression, but can also induce its regression once triggers of chronic inflammation are eliminated. In liver cancer, they are attracted to the tumor site to become tumor-associated macrophages (TAMs) polarized towards a M2- anti-inflammatory/tumor-promoting phenotype. Besides their role in thrombosis and hemostasis, platelets can also stimulate fibrosis and tumor development by secreting profibrogenic factors and regulating the innate immune response, e.g., by interacting with monocytes and macrophages. Here, we review recent literature on the role of macrophages and platelets and their interplay in liver fibrosis and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Martina Casari
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Dominik Siegl
- Institute for Translational Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Carsten Deppermann
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immune Therapy Forschungszentrum für Immuntherapie (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Detlef Schuppan
- Institute for Translational Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immune Therapy Forschungszentrum für Immuntherapie (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
108
|
Chen X, Yang X, Ren Y, Xie B, Xie S, Zhao L, Wang S, Geng J, Jiang D, Luo S, He J, Shu S, Hu Y, Zhu L, Li Z, Zhang X, Liu M, Dai H. Clinical characteristics of hypersensitivity pneumonitis: non-fibrotic and fibrotic subtypes. Chin Med J (Engl) 2023; 136:2839-2846. [PMID: 37464421 PMCID: PMC10686610 DOI: 10.1097/cm9.0000000000002613] [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: 12/13/2022] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND The presence of fibrosis is a criterion for subtype classification in the newly updated hypersensitivity pneumonitis (HP) guidelines. The present study aimed to summarize differences in clinical characteristics and prognosis of non-fibrotic hypersensitivity pneumonitis (NFHP) and fibrotic hypersensitivity pneumonitis (FHP) and explore factors associated with the presence of fibrosis. METHODS In this prospective cohort study, patients diagnosed with HP through a multidisciplinary discussion were enrolled. Collected data included demographic and clinical characteristics, laboratory findings, and radiologic and histopathological features. Logistic regression analyses were performed to explore factors related to the presence of fibrosis. RESULTS A total of 202 patients with HP were enrolled, including 87 (43.1%) NFHP patients and 115 (56.9%) FHP patients. Patients with FHP were older and more frequently presented with dyspnea, crackles, and digital clubbing than patients with NFHP. Serum levels of carcinoembryonic antigen, carbohydrate antigen 125, carbohydrate antigen 153, gastrin-releasing peptide precursor, squamous cell carcinoma antigen, and antigen cytokeratin 21-1, and count of bronchoalveolar lavage (BAL) eosinophils were higher in the FHP group than in the NFHP group. BAL lymphocytosis was present in both groups, but less pronounced in the FHP group. Multivariable regression analyses revealed that older age, <20% of lymphocyte in BAL, and ≥1.75% of eosinophil in BAL were risk factors for the development of FHP. Twelve patients developed adverse outcomes, with a median survival time of 12.5 months, all of whom had FHP. CONCLUSIONS Older age, <20% of lymphocyte in BAL, and ≥1.75% of eosinophil in BAL were risk factors associated with the development of FHP. Prognosis of patients with NFHP was better than that of patients with FHP. These results may provide insights into the mechanisms of fibrosis in HP.
Collapse
Affiliation(s)
- Xueying Chen
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| | - Xiaoyan Yang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing 100029, China
| | - Yanhong Ren
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| | - Bingbing Xie
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| | - Sheng Xie
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Ling Zhao
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Shiyao Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| | - Jing Geng
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| | - Dingyuan Jiang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| | - Sa Luo
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| | - Jiarui He
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| | - Shi Shu
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| | - Yinan Hu
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| | - Lili Zhu
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| | - Zhen Li
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| | - Xinran Zhang
- Institute of Clinical Medical Sciences, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Min Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100029, China
| |
Collapse
|
109
|
Yang B, Rutkowski N, Elisseeff J. The foreign body response: emerging cell types and considerations for targeted therapeutics. Biomater Sci 2023; 11:7730-7747. [PMID: 37904536 DOI: 10.1039/d3bm00629h] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
The foreign body response (FBR) remains a clinical challenge in the field of biomaterials due to its ability to elicit a chronic and sustained immune response. Modulating the immune response to materials is a modern paradigm in tissue engineering to enhance repair while limiting fibrous encapsulation and implant isolation. Though the classical mediators of the FBR are well-characterized, recent studies highlight that our understanding of the cell types that shape the FBR may be incomplete. In this review, we discuss the emerging role of T cells, stromal-immune cell interactions, and senescent cells in the biomaterial response, particularly to synthetic materials. We emphasize future studies that will deepen the field's understanding of these cell types in the FBR, with the goal of identifying therapeutic targets that will improve implant integration. Finally, we briefly review several considerations that may influence our understanding of the FBR in humans, including rodent models, aging, gut microbiota, and sex differences. A better understanding of the heterogeneous host cell response during the FBR can enable the design and development of immunomodulatory materials that favor healing.
Collapse
Affiliation(s)
- Brenda Yang
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | - Natalie Rutkowski
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | - Jennifer Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
110
|
Zielinski CE. T helper cell subsets: diversification of the field. Eur J Immunol 2023; 53:e2250218. [PMID: 36792132 DOI: 10.1002/eji.202250218] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
Polarized T helper cell (Th cell) responses are important determinants of host protection. Th cell subsets tailor their functional repertoire of cytokines to their cognate antigens to efficiently contribute to their clearance. In contrast, in settings of immune abrogation, these polarized cytokine patterns of Th cells can mediate tissue damage and pathology resulting in allergy or autoimmunity. Recent technological developments in single-cell genomics and proteomics as well as advances in the high-dimensional bioinformatic analysis of complex datasets have challenged the prevailing Th cell subset classification into Th1, Th2, Th17, and other subsets. Additionally, systems immunology approaches have revealed that instructive input from the peripheral tissue microenvironment can have differential effects on the overall phenotype and molecular wiring of Th cells depending on their spatial distribution. Th cells from the blood or secondary lymphoid organs are therefore expected to follow distinct rules of regulation. In this review, the functional heterogeneity of Th cell subsets will be reviewed in the context of new technological developments and T-cell compartmentalization in tissue niches. This work will especially focus on challenges to the traditional boundaries of Th cell subsets and will discuss the underlying regulatory checkpoints, which could reveal new therapeutic strategies for various immune-mediated diseases.
Collapse
Affiliation(s)
- Christina E Zielinski
- Department of Infection Immunology, Leibniz Institute for Natural Products Research and Infection Biology, Jena, Germany
- Institute of Microbiology, Faculty of Biosciences, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
111
|
Xiao SQ, Wen TZ, Chen XY, Chen HY, Li Z, He ZC, Luo T, Tang R, Fu WJ, Cao MF, Chen L, Niu Q, Wang S, Lan Y, Ge J, Li QR, Guo HT, Wang YX, Ping YF, Shen H, Wang Y, Ding YQ, Bian XW, Yao XH. Autopsy analysis reveals increased macrophage infiltration and cell apoptosis in COVID-19 patients with severe pulmonary fibrosis. Pathol Res Pract 2023; 252:154920. [PMID: 37948998 DOI: 10.1016/j.prp.2023.154920] [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: 10/17/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023]
Abstract
Clinical data indicates that SARS-CoV-2 infection-induced respiratory failure is a fatal condition for severe COVID-19 patients. However, the pathological alterations of different types of respiratory failure remained unknown for severe COVID-19 patients. This study aims to evaluate whether there are differences in the performance of various types of respiratory failure in severe COVID-19 patients and investigate the pathological basis for these differences. The lung tissue sections of severe COVID-19 patients were assessed for the degree of injury and immune responses. Transcriptome data were used to analyze the molecular basis in severe COVID-19 patients. Severe COVID-19 patients with combined oxygenation and ventilatory failure presented more severe pulmonary fibrosis, airway obstruction, and prolonged disease course. The number of M2 macrophages increased with the degree of fibrosis in patients, suggesting that it may be closely related to the development of pulmonary fibrosis. The co-existence of pro-inflammatory and anti-inflammatory cytokines in the pulmonary environment could also participate in the progression of pulmonary fibrosis. Furthermore, the increased apoptosis in the lungs of COVID-19 patients with severe pulmonary fibrosis may represent a critical factor linking sustained inflammatory responses to fibrosis. Our findings indicate that during the extended phase of COVID-19, antifibrotic and antiapoptotic treatments should be considered in conjunction with the progression of the disease.
Collapse
Affiliation(s)
- Shi-Qi Xiao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Tian-Zi Wen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xin-Yu Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - He-Yuan Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Zhuang Li
- Department of Neurology, Armed Corps Police Hospital of Chongqing, Chongqing, China
| | - Zhi-Cheng He
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Tao Luo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Rui Tang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Wen-Juan Fu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Mian-Fu Cao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Lu Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Qin Niu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Shuai Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Yang Lan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Jia Ge
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Qing-Rui Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Hai-Tao Guo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Yan-Xia Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Yi-Fang Ping
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Hong Shen
- Department of Pathology, Southern Medical University, Guangzhou, China
| | - Yan Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Yan-Qing Ding
- Department of Pathology, Southern Medical University, Guangzhou, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xiao-Hong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China.
| |
Collapse
|
112
|
Jiang S, Feng J, Jiang Y, Lu Z, Kong J, Li X, Lian H, Zhang F, Li Y, Li J. Triptolide attenuates CCL 4-induced liver fibrosis by regulating the differentiation of CD 4+ T cells in mice. Int Immunopharmacol 2023; 125:111206. [PMID: 37956491 DOI: 10.1016/j.intimp.2023.111206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/26/2023] [Accepted: 11/07/2023] [Indexed: 11/15/2023]
Abstract
Liver fibrosis is a major global health issue, and immune dysregulation is a main contributor. Triptolide is a natural immunosuppressive agent with demonstrated effectiveness in ameliorating liver fibrosis, but whether it exerts anti-liver fibrotic effects via immunoregulation remains obscure. In this study, first, by employing a CCL4-induced liver fibrosis mouse model, we demonstrated that triptolide could alleviate pathological damage to liver tissue and attenuate liver function damaged by CCL4. In addition, triptolide inhibited the expression of liver fibrotic markers such as hydroxyproline, collagen type IV, hyaluronidase, laminin, and procollagen type III, and the protein expression of α-SMA in CCL4-induced liver fibrosis. Second, with the help of network pharmacology, we predicted that triptolide's anti-liver fibrotic effects might occur through the regulation of Th17, Th1, and Th2 cell differentiation, which indicated that triptolide might mitigate liver fibrosis via immunoregulation. Finally, multiplex immunoassays and flow cytometry were adopted to verify this prediction. The results suggested that triptolide could reverse the aberrant expression of inflammatory cytokines caused by CCL4 and regulate the differentiation of Th1, Th2, Th17, and Treg cells. In conclusion, triptolide could attenuate CCL4-induced liver fibrosis by regulating the differentiation of CD4+ T cells. The results obtained in this study extended the application of triptolide and introduced a new mechanism of triptolide's anti-liver fibrotic effects.
Collapse
Affiliation(s)
- Shiyuan Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jing Feng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yanling Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhihao Lu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jingwei Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xueming Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hui Lian
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Fang Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yu Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
113
|
Satzinger S, Willenborg S, Ding X, Klose CSN, Radtke D, Voehringer D, Eming SA. Type 2 Immunity Regulates Dermal White Adipose Tissue Function. J Invest Dermatol 2023; 143:2456-2467.e5. [PMID: 37295491 DOI: 10.1016/j.jid.2023.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 04/19/2023] [Accepted: 05/09/2023] [Indexed: 06/12/2023]
Abstract
Type 2 immune responses have been increasingly linked with tissue maintenance, regeneration, and metabolic homeostasis. The molecular basis of regulator and effector mechanisms of type 2 immunity in skin regeneration and homeostasis is still lacking. In this study, we analyzed the role of IL-4Rα signaling in the regeneration of diverse cellular compartments in the skin. Mutants with global IL-4Rα deficiency showed two major phenotypes: first, a pronounced atrophy of the interfollicular epidermis, and second, a significant increase in dermal white adipose tissue thickness in mice aged 3 weeks (postnatal day 21) compared with littermate controls. Notably, IL-4Rα deficiency decreased the activation of hormone-sensitive lipase, a rate-limiting step in lipolysis. Immunohistochemical and FACS analysis in IL-4/enhanced GFP reporter mice showed that IL-4 expression peaked on postnatal day 21 and that eosinophils are the predominant IL-4-expressing cells. Eosinophil-deficient mice recapitulated the lipolytic-defective dermal white adipose tissue phenotype of Il4ra-deficient mice, showing that eosinophils are necessary for dermal white adipose tissue lipolysis. Collectively, we provide mechanistic insights into the regulation of interfollicular epidermis and hormone-sensitive lipase-mediated lipolysis in dermal white adipose tissue in early life by IL-4Rα, and our findings show that eosinophils play a critical role in this process.
Collapse
Affiliation(s)
| | | | - Xiaolei Ding
- Department of Dermatology, University of Cologne, Cologne, Germany; Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China; Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
| | - Christoph S N Klose
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Daniel Radtke
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sabine A Eming
- Department of Dermatology, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany.
| |
Collapse
|
114
|
Hattori K, Sakaguchi Y, Oka T, Asahina Y, Kawaoka T, Yamamoto R, Matsui I, Mizui M, Kaimori JY, Isaka Y. Interstitial Eosinophilic Aggregates and Kidney Outcome in Patients with CKD. Clin J Am Soc Nephrol 2023; 18:1563-1572. [PMID: 37639279 PMCID: PMC10723926 DOI: 10.2215/cjn.0000000000000277] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/18/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Interstitial eosinophilic aggregates are observed in various kidney diseases, but their clinical implications remain unknown. We assessed the association between interstitial eosinophilic aggregates and kidney outcomes and further analyzed the association between blood eosinophil count, as a surrogate for interstitial eosinophilic aggregates, and the risk of kidney failure in patients with advanced CKD. METHODS We analyzed datasets from two retrospective cohort studies: ( 1 ) the kidney biopsy cohort including 563 patients who underwent native kidney biopsy at Osaka University Hospital between 2009 and 2021 and ( 2 ) the retrospective CKD cohort including 2877 patients with an eGFR of 10-60 ml/min per 1.73 m 2 referred to the nephrology outpatient center at Osaka University Hospital between 2005 and 2018. Interstitial eosinophilic aggregates were defined as ≥5 interstitial eosinophils in the high-power field on hematoxylin and eosin staining. This study outcome was initiation of KRT or ≥40% decline in eGFR. RESULTS In the kidney biopsy cohort, interstitial eosinophilic aggregates were found in 17% of patients, most frequently in those with diabetic nephropathy (50%). Interstitial eosinophilic aggregates were associated with a higher rate of the composite kidney outcome after adjustment for clinical and histological variables (hazard ratio, 3.61; 95% confidence interval, 2.47 to 5.29; P < 0.001). LASSO revealed that blood eosinophil count was the strongest predictor of interstitial eosinophilic aggregates. In the retrospective CKD cohort, higher baseline and time-updated blood eosinophil counts were significantly associated with a higher rate of KRT initiation in Cox proportional hazards models and marginal structural models. CONCLUSIONS Interstitial eosinophilic aggregates were associated with a higher risk of a composite of KRT initiation or ≥40% decline in eGFR. PODCAST This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/CJASN/2023_12_08_CJN0000000000000277.mp3.
Collapse
Affiliation(s)
- Koki Hattori
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yusuke Sakaguchi
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tatsufumi Oka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuta Asahina
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takayuki Kawaoka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ryohei Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
- Health and Counseling Center, Osaka University, Toyonaka, Japan
| | - Isao Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jun-Ya Kaimori
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
115
|
Zhang M, Jing Y, Xu W, Shi X, Zhang W, Chen P, Cao X, Han X, Duan X, Ren J. The C-type lectin COLEC10 is predominantly produced by hepatic stellate cells and involved in the pathogenesis of liver fibrosis. Cell Death Dis 2023; 14:785. [PMID: 38036508 PMCID: PMC10689734 DOI: 10.1038/s41419-023-06324-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/05/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023]
Abstract
Hepatic stellate cell is one of the major nonparenchymal cell types in liver. It has been proved the hepatic stellate cells are activated upon liver injury and produce excessive extracellular matrix to induce liver fibrosis. Single-cell RNA sequencing has been introduced to identify the subpopulations and function of hepatic stellate cells for its remarkable resolution of representation of single-cell transcriptome. According to the re-analysis of single-cell RNA sequencing data and pseudotime trajectory inference, we have found the C-type lectins including Colec10 and Colec11 are not produced by hepatocytes but predominantly produced by hepatic stellate cells, especially quiescent ones in the mice livers. In addition, the expression of Colec10 is decreased in the fibrotic livers of CCl4-challenged mice. COLEC10 is also mainly expressed in the hepatic stellate cells of human livers and the expression of COLEC10 is decreased with the progression of liver fibrosis. The bulk RNA sequencing data of the lentivirus transfected LX-2 cells indicates the function of COLEC10 is associated with inflammation, angiogenesis and extracellular matrix alteration. Surprisingly, the in vitro overexpression of COLEC10 in LX-2 cells promotes the mRNA expression of extracellular matrix components including COL1A1, COL1A2 and COL3A1 and the extracellular matrix degradation enzyme MMP2. To further investigate the role of COLEC10 in the pathogenesis of liver fibrosis, the serum concentration of COLEC10 in patients with chronic liver disease and healthy donors is measured. The serum concentration of COLEC10 is elevated in the patients with chronic liver disease compared to the healthy donors and positively correlated with serum concentration of the D-dimer but not the most of liver function markers. Altogether, we conclude that the C-type lectin COLEC10 is predominantly produced by the hepatic stellate cells and involved in the pathogenesis of liver fibrosis.
Collapse
Affiliation(s)
- Mengfan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, China
| | - Yang Jing
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300070, China
| | - Wenze Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, China
| | - Xiaojing Shi
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300070, China
| | - Wenguang Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, China
| | - Pengfei Chen
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, China
| | - Xiaocang Cao
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300070, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, China.
| | - Xuhua Duan
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, China.
| | - Jianzhuang Ren
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
116
|
Singh G, Warang P, García-Bernalt Diego J, Chang L, Bykov Y, Singh S, Pache L, Cuadrado-Castano S, Webb B, Garcia-Sastre A, Schotsaert M. Host immune responses associated with SARS-CoV-2 Omicron infection result in protection or pathology during reinfection depending on mouse genetic background. RESEARCH SQUARE 2023:rs.3.rs-3637405. [PMID: 38077015 PMCID: PMC10705603 DOI: 10.21203/rs.3.rs-3637405/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Rapid emergence of antigenic distinct SARS-CoV-2 variants implies a greater risk of reinfection as viruses can escape neutralizing antibodies induced by vaccination or previous viral exposure. Disease severity during COVID-19 depends on many variables such as age-related comorbidities, host immune status and genetic variation. The host immune response during infection with SARS-CoV-2 may contribute to disease severity, which can range from asymptomatic to severe with fatal outcome. Furthermore, the extent of host immune response activation may rely on underlying genetic predisposition for disease or protection. To address these questions, we performed immune profiling studies in mice with different genetic backgrounds - transgenic K18-hACE2 and wild-type 129S1 mice - subjected to reinfection with the severe disease-causing SARS-CoV-2 B.1.351 variant, 30 days after experimental milder BA.1 infection. BA.1 preinfection conferred protection against B.1.351-induced morbidity in K18-hACE2 mice but aggravated disease in 129S1 mice. We found that he cytokine/chemokine profile in B.1.351 re-infected 129S1mice is similar to that during severe SARS-CoV-2 infection in humans and is characterized by a much higher level of IL-10, IL-1β, IL-18 and IFN-γ, whereas in B.1.351 re-infected K18-hACE2 mice, the cytokine profile echoes the signature of naïve mice undergoing viral infection for the first time. Interestingly, the enhanced pathology observed in 129S1 mice upon reinfection cannot be attributed to a less efficient induction of adaptive immune responses to the initial BA.1 infection, as both K18-hACE2 and 129S1 mice exhibited similar B and T cell responses at 30 DPI against BA.1, with similar anti-BA.1 or B.1.351 spike-specific ELISA binding titers, levels of germinal center B-cells, and SARS-CoV-2-Spike specific tissue-resident T-cells. Long-term effects of BA.1 infection are associated with differential transcriptional changes in bronchoalveolar lavage-derived CD11c + immune cells from K18-hACE2 and 129S1, with K18-hACE2 CD11c + cells showing a strong antiviral defense gene expression profile whereas 129S1 CD11c + cells showed a more pro-inflammatory response. In conclusion, initial infection with BA.1 induces cross-reactive adaptive immune responses in both K18-hACE2 and 129S1 mice, however the different disease outcome of reinfection seems to be driven by differential responses of CD11c + cells in the alveolar space.
Collapse
Affiliation(s)
| | | | | | | | | | - Sarabjot Singh
- RT-PCR COVID-19 Laboratory, Civil Hospital, Moga, Punjab, India
| | - Lars Pache
- Sanford Burnham Prebys Medical Discovery Institute
| | | | - Brett Webb
- Department of Veterinary Sciences, University of Wyoming
| | | | | |
Collapse
|
117
|
Maseda D, Manfredo-Vieira S, Payne AS. T cell and bacterial microbiota interaction at intestinal and skin epithelial interfaces. DISCOVERY IMMUNOLOGY 2023; 2:kyad024. [PMID: 38567051 PMCID: PMC10917213 DOI: 10.1093/discim/kyad024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/28/2023] [Accepted: 11/24/2023] [Indexed: 04/04/2024]
Abstract
Graphical Abstract.
Collapse
Affiliation(s)
- Damian Maseda
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Silvio Manfredo-Vieira
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aimee S Payne
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
118
|
Zang Y, Liu S, Rao Z, Wang Y, Zhang B, Li H, Cao Y, Zhou J, Shen Z, Duan S, He D, Xu H. Retinoid X receptor gamma dictates the activation threshold of group 2 innate lymphoid cells and limits type 2 inflammation in the small intestine. Immunity 2023; 56:2542-2554.e7. [PMID: 37714152 DOI: 10.1016/j.immuni.2023.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 06/18/2023] [Accepted: 08/22/2023] [Indexed: 09/17/2023]
Abstract
Group 2 innate lymphoid cells (ILC2s) are crucial in promoting type 2 inflammation that contributes to both anti-parasite immunity and allergic diseases. However, the molecular checkpoints in ILC2s that determine whether to immediately launch a proinflammatory response are unknown. Here, we found that retinoid X receptor gamma (Rxrg) was highly expressed in small intestinal ILC2s and rapidly suppressed by alarmin cytokines. Genetic deletion of Rxrg did not impact ILC2 development but facilitated ILC2 responses and the tissue inflammation induced by alarmins. Mechanistically, RXRγ maintained the expression of its target genes that support intracellular cholesterol efflux, which in turn reduce ILC2 proliferation. Furthermore, RXRγ expression prevented ILC2 response to mild stimulations, including low doses of alarmin cytokine and mechanical skin injury. Together, we propose that RXRγ expression and its mediated lipid metabolic states function as a cell-intrinsic checkpoint that confers the threshold of ILC2 activation in the small intestine.
Collapse
Affiliation(s)
- Yang Zang
- School of Basic Medical Sciences, Fudan University, Shanghai 200433, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Shaorui Liu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Zebing Rao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Yinsheng Wang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Boya Zhang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Hui Li
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Yingjiao Cao
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jie Zhou
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zhuxia Shen
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Shengzhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Danyang He
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Laboratory of Neuroimmunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Heping Xu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China.
| |
Collapse
|
119
|
Pattnaik B, Negi V, Chaudhuri R, Desiraju K, Faizan MI, Akhtar A, Ansari MS, Shakir M, Gheware A, Prakash YS, Guleria R, Ghosh B, Agrawal A, Ahmad T. MiR-326-mediated overexpression of NFIB offsets TGF-β induced epithelial to mesenchymal transition and reverses lung fibrosis. Cell Mol Life Sci 2023; 80:357. [PMID: 37950757 PMCID: PMC11072886 DOI: 10.1007/s00018-023-05005-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 11/13/2023]
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a progressively fatal and incurable disease characterized by the loss of alveolar structures, increased epithelial-mesenchymal transition (EMT), and aberrant tissue repair. In this study, we investigated the role of Nuclear Factor I-B (NFIB), a transcription factor critical for lung development and maturation, in IPF. Using both human lung tissue samples from patients with IPF, and a mouse model of lung fibrosis induced by bleomycin, we showed that there was a significant reduction of NFIB both in the lungs of patients and mice with IPF. Furthermore, our in vitro experiments using cultured human lung cells demonstrated that the loss of NFIB was associated with the induction of EMT by transforming growth factor beta (TGF-β). Knockdown of NFIB promoted EMT, while overexpression of NFIB suppressed EMT and attenuated the severity of bleomycin-induced lung fibrosis in mice. Mechanistically, we identified post-translational regulation of NFIB by miR-326, a miRNA with anti-fibrotic effects that is diminished in IPF. Specifically, we showed that miR-326 stabilized and increased the expression of NFIB through its 3'UTR target sites for Human antigen R (HuR). Moreover, treatment of mice with either NFIB plasmid or miR-326 reversed airway collagen deposition and fibrosis. In conclusion, our study emphasizes the critical role of NFIB in lung development and maturation, and its reduction in IPF leading to EMT and loss of alveolar structures. Our study highlights the potential of miR-326 as a therapeutic intervention for IPF. The schema shows the role of NFIB in maintaining the normal epithelial cell characteristics in the lungs and how its reduction leads to a shift towards mesenchymal cell-like features and pulmonary fibrosis. A In normal lungs, NFIB is expressed abundantly in the epithelial cells, which helps in maintaining their shape, cell polarity and adhesion molecules. However, when the lungs are exposed to factors that induce pulmonary fibrosis, such as bleomycin, or TGF-β, the epithelial cells undergo epithelial to mesenchymal transition (EMT), which leads to a decrease in NFIB. B The mesenchymal cells that arise from EMT appear as spindle-shaped with loss of cell junctions, increased cell migration, loss of polarity and expression of markers associated with mesenchymal cells/fibroblasts. C We designed a therapeutic approach that involves exogenous administration of NFIB in the form of overexpression plasmid or microRNA-326. This therapeutic approach decreases the mesenchymal cell phenotype and restores the epithelial cell phenotype, thus preventing the development or progression of pulmonary fibrosis.
Collapse
Affiliation(s)
- Bijay Pattnaik
- Molecular Immunogenetics Laboratory and Centre of Excellence for Translational Research in Asthma & Lung Disease, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India
- Department of Pulmonary, Critical Care & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Vinny Negi
- Molecular Immunogenetics Laboratory and Centre of Excellence for Translational Research in Asthma & Lung Disease, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India
| | - Rituparna Chaudhuri
- Molecular Immunogenetics Laboratory and Centre of Excellence for Translational Research in Asthma & Lung Disease, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India
| | - Koundinya Desiraju
- Molecular Immunogenetics Laboratory and Centre of Excellence for Translational Research in Asthma & Lung Disease, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India
| | - Md Imam Faizan
- Multidisciplinary Centre for Advanced Research & Studies, Jamia Millia Islamia, New Delhi, 110025, India
| | - Areej Akhtar
- Multidisciplinary Centre for Advanced Research & Studies, Jamia Millia Islamia, New Delhi, 110025, India
| | - Md Sufyan Ansari
- Multidisciplinary Centre for Advanced Research & Studies, Jamia Millia Islamia, New Delhi, 110025, India
| | - Md Shakir
- Multidisciplinary Centre for Advanced Research & Studies, Jamia Millia Islamia, New Delhi, 110025, India
| | - Atish Gheware
- Molecular Immunogenetics Laboratory and Centre of Excellence for Translational Research in Asthma & Lung Disease, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India
| | - Y S Prakash
- Departments of Anesthesiology, Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Randeep Guleria
- Department of Pulmonary, Critical Care & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Balaram Ghosh
- Molecular Immunogenetics Laboratory and Centre of Excellence for Translational Research in Asthma & Lung Disease, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India
| | - Anurag Agrawal
- Molecular Immunogenetics Laboratory and Centre of Excellence for Translational Research in Asthma & Lung Disease, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India.
- Trivedi School of Biosciences, Ashoka University, NH 44, Rajiv Gandhi Education City, Sonipat, Haryana, 131029, India.
| | - Tanveer Ahmad
- Molecular Immunogenetics Laboratory and Centre of Excellence for Translational Research in Asthma & Lung Disease, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India.
- Multidisciplinary Centre for Advanced Research & Studies, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
120
|
Yuan W, Ferreira LDAQ, Yu B, Ansari S, Moshaverinia A. Dental-derived stem cells in tissue engineering: the role of biomaterials and host response. Regen Biomater 2023; 11:rbad100. [PMID: 38223292 PMCID: PMC10786679 DOI: 10.1093/rb/rbad100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 01/16/2024] Open
Abstract
Dental-derived stem cells (DSCs) are attractive cell sources due to their easy access, superior growth capacity and low immunogenicity. They can respond to multiple extracellular matrix signals, which provide biophysical and biochemical cues to regulate the fate of residing cells. However, the direct transplantation of DSCs suffers from poor proliferation and differentiation toward functional cells and low survival rates due to local inflammation. Recently, elegant advances in the design of novel biomaterials have been made to give promise to the use of biomimetic biomaterials to regulate various cell behaviors, including proliferation, differentiation and migration. Biomaterials could be tailored with multiple functionalities, e.g., stimuli-responsiveness. There is an emerging need to summarize recent advances in engineered biomaterials-mediated delivery and therapy of DSCs and their potential applications. Herein, we outlined the design of biomaterials for supporting DSCs and the host response to the transplantation.
Collapse
Affiliation(s)
- Weihao Yuan
- Weintraub Center for Reconstructive Biotechnology, Section of Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Luiza de Almeida Queiroz Ferreira
- Weintraub Center for Reconstructive Biotechnology, Section of Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bo Yu
- Section of Restorative Dentistry, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sahar Ansari
- Weintraub Center for Reconstructive Biotechnology, Section of Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Section of Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
121
|
Sawada K, Chung H, Softic S, Moreno-Fernandez ME, Divanovic S. The bidirectional immune crosstalk in metabolic dysfunction-associated steatotic liver disease. Cell Metab 2023; 35:1852-1871. [PMID: 37939656 PMCID: PMC10680147 DOI: 10.1016/j.cmet.2023.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is an unabated risk factor for end-stage liver diseases with no available therapies. Dysregulated immune responses are critical culprits of MASLD pathogenesis. Independent contributions from either the innate or adaptive arms of the immune system or their unidirectional interplay are commonly studied in MASLD. However, the bidirectional communication between innate and adaptive immune systems and its impact on MASLD remain insufficiently understood. Given that both innate and adaptive immune cells are indispensable for the development and progression of inflammation in MASLD, elucidating pathogenic contributions stemming from the bidirectional interplay between these two arms holds potential for development of novel therapeutics for MASLD. Here, we review the immune cell types and bidirectional pathways that influence the pathogenesis of MASLD and highlight potential pharmacologic approaches to combat MASLD based on current knowledge of this bidirectional crosstalk.
Collapse
Affiliation(s)
- Keisuke Sawada
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Hak Chung
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Samir Softic
- Department of Pediatrics and Gastroenterology, University of Kentucky, Lexington, KY 40536, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
122
|
Wang X, Wen S, Du X, Zhang Y, Yang X, Zou R, Feng B, Fu X, Jiang F, Zhou G, Liu Z, Zhu W, Ma R, Feng J, Shen B. SAA suppresses α-PD-1 induced anti-tumor immunity by driving T H2 polarization in lung adenocarcinoma. Cell Death Dis 2023; 14:718. [PMID: 37925492 PMCID: PMC10625560 DOI: 10.1038/s41419-023-06198-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 11/06/2023]
Abstract
Cancer stem cells (CSCs) are believed to be crucial in the initiation, progression, and recurrence of cancer. CSCs are also known to be more resistant to cancer treatments. However, the interaction between CSCs and the immune microenvironment is complex and not fully understood. In current study we used single cell RNA sequence (scRNA-Seq, public dataset) technology to identify the characteristic of CSCs. We found that the lung adenocarcinoma cancer stem population is highly inflammatory and remodels the tumor microenvironment by secreting inflammatory factors, specifically the acute phase protein serum amyloid A (SAA). Next, we developed an ex-vivo autologous patient-derived organoids (PDOs) and peripheral blood mononuclear cells (PBMCs) co-culture model to evaluate the immune biological impact of SAA. We found that SAA not only promotes chemoresistance by inducing cancer stem transformation, but also restricts anti-tumor immunity and promotes tumor fibrosis by driving type 2 immunity, and α-SAA neutralization antibody could restrict treatment resistant and tumor fibrosis. Mechanically, we found that the malignant phenotype induced by SAA is dependent on P2X7 receptor. Our data indicate that cancer stem cells secreted SAA have significant biological impact to promote treatment resistant and tumor fibrosis by driving cancer stemness transformation and type 2 immunity polarization via P2X7 receptor. Notably, α-SAA neutralization antibody shows therapeutic potential by restricting these malignant phenotypes.
Collapse
Affiliation(s)
- Xin Wang
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Shaodi Wen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xiaoyue Du
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yihan Zhang
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xiao Yang
- Department of Clinical Laboratory, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Renrui Zou
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Bing Feng
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xiao Fu
- Department of General Surgery, Nanjing Drum Tower Hospital, Nanjing, China
- Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Jiang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Guoren Zhou
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Zi Liu
- Nanjing Advanced Analysis Tech. (NAAT) Co., LTD, Nanjing, China
| | - Wei Zhu
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Rong Ma
- Research Center for Clinical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Jifeng Feng
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Bo Shen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
| |
Collapse
|
123
|
Yan L, Hou C, Liu J, Wang Y, Zeng C, Yu J, Zhou T, Zhou Q, Duan S, Xiong W. Local administration of liposomal-based Plekhf1 gene therapy attenuates pulmonary fibrosis by modulating macrophage polarization. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2571-2586. [PMID: 37340175 DOI: 10.1007/s11427-022-2314-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/01/2023] [Indexed: 06/22/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal interstitial lung disease with limited therapeutic options. Macrophages, particularly alternatively activated macrophages (M2), have been recognized to contribute to the pathogenesis of pulmonary fibrosis. Therefore, targeting macrophages might be a viable therapeutic strategy for IPF. Herein, we report a potential nanomedicine-based gene therapy for IPF by modulating macrophage M2 activation. In this study, we illustrated that the levels of pleckstrin homology and FYVE domain containing 1 (Plekhf1) were increased in the lungs originating from IPF patients and PF mice. Further functionality studies identified the pivotal role of Plekhf1 in macrophage M2 activation. Mechanistically, Plekhf1 was upregulated by IL-4/IL-13 stimulation, after which Plekhf1 enhanced PI3K/Akt signaling to promote the macrophage M2 program and exacerbate pulmonary fibrosis. Therefore, intratracheal administration of Plekhf1 siRNA-loaded liposomes could effectively suppress the expression of Plekhf1 in the lungs and notably protect mice against BLM-induced lung injury and fibrosis, concomitant with a significant reduction in M2 macrophage accumulation in the lungs. In conclusion, Plekhf1 may play a crucial role in the pathogenesis of pulmonary fibrosis, and Plekhf1 siRNA-loaded liposomes might be a promising therapeutic approach against pulmonary fibrosis.
Collapse
Affiliation(s)
- Lifeng Yan
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chenchen Hou
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Juan Liu
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yi Wang
- Department of Pulmonary and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chenxi Zeng
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, 430030, China
| | - Jun Yu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, 430030, China
| | - Tianyu Zhou
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Pulmonary and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qing Zhou
- Department of Pulmonary and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Shengzhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China.
| | - Weining Xiong
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
124
|
Bernstein ZJ, Shenoy A, Chen A, Heller NM, Spangler JB. Engineering the IL-4/IL-13 axis for targeted immune modulation. Immunol Rev 2023; 320:29-57. [PMID: 37283511 DOI: 10.1111/imr.13230] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 05/19/2023] [Indexed: 06/08/2023]
Abstract
The structurally and functionally related interleukin-4 (IL-4) and IL-13 cytokines play pivotal roles in shaping immune activity. The IL-4/IL-13 axis is best known for its critical role in T helper 2 (Th2) cell-mediated Type 2 inflammation, which protects the host from large multicellular pathogens, such as parasitic helminth worms, and regulates immune responses to allergens. In addition, IL-4 and IL-13 stimulate a wide range of innate and adaptive immune cells, as well as non-hematopoietic cells, to coordinate various functions, including immune regulation, antibody production, and fibrosis. Due to its importance for a broad spectrum of physiological activities, the IL-4/IL-13 network has been targeted through a variety of molecular engineering and synthetic biology approaches to modulate immune behavior and develop novel therapeutics. Here, we review ongoing efforts to manipulate the IL-4/IL-13 axis, including cytokine engineering strategies, formulation of fusion proteins, antagonist development, cell engineering approaches, and biosensor design. We discuss how these strategies have been employed to dissect IL-4 and IL-13 pathways, as well as to discover new immunotherapies targeting allergy, autoimmune diseases, and cancer. Looking ahead, emerging bioengineering tools promise to continue advancing fundamental understanding of IL-4/IL-13 biology and enabling researchers to exploit these insights to develop effective interventions.
Collapse
Affiliation(s)
- Zachary J Bernstein
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anjali Shenoy
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amy Chen
- Department of Molecular and Cellular Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nicola M Heller
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
- Division of Allergy and Clinical Immunology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jamie B Spangler
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Sidney Kimmel Cancer Center, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
125
|
Wang Z, Xu Z, Yang X, Li M, Yip RCS, Li Y, Chen H. Current application and modification strategy of marine polysaccharides in tissue regeneration: A review. BIOMATERIALS ADVANCES 2023; 154:213580. [PMID: 37634336 DOI: 10.1016/j.bioadv.2023.213580] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/24/2023] [Accepted: 08/04/2023] [Indexed: 08/29/2023]
Abstract
Marine polysaccharides (MPs) are exceptional bioactive materials that possess unique biochemical mechanisms and pharmacological stability, making them ideal for various tissue engineering applications. Certain MPs, including agarose, alginate, carrageenan, chitosan, and glucan have been successfully employed as biological scaffolds in animal studies. As carriers of signaling molecules, scaffolds can enhance the adhesion, growth, and differentiation of somatic cells, thereby significantly improving the tissue regeneration process. However, the biological benefits of pure MPs composite scaffold are limited. Therefore, physical, chemical, enzyme modification and other methods are employed to expand its efficacy. Chemically, the structural properties of MPs scaffolds can be altered through modifications to functional groups or molecular weight reduction, thereby enhancing their biological activities. Physically, MPs hydrogels and sponges emulate the natural extracellular matrix, creating a more conducive environment for tissue repair. The porosity and high permeability of MPs membranes and nanomaterials expedite wound healing. This review explores the distinctive properties and applications of select MPs in tissue regeneration, highlighting their structural versatility and biological applicability. Additionally, we provide a brief overview of common modification strategies employed for MP scaffolds. In conclusion, MPs have significant potential and are expected to be a novel regenerative material for tissue engineering.
Collapse
Affiliation(s)
- Zhaokun Wang
- Marine College, Shandong University, NO. 180 Wenhua West Road, Gao Strict, Weihai 264209, China.
| | - Zhiwen Xu
- Marine College, Shandong University, NO. 180 Wenhua West Road, Gao Strict, Weihai 264209, China.
| | - Xuan Yang
- Marine College, Shandong University, NO. 180 Wenhua West Road, Gao Strict, Weihai 264209, China.
| | - Man Li
- Marine College, Shandong University, NO. 180 Wenhua West Road, Gao Strict, Weihai 264209, China.
| | - Ryan Chak Sang Yip
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Yuanyuan Li
- Department of Food Science, Cornell University, Stocking Hall, Ithaca, NY 14853, USA.
| | - Hao Chen
- Marine College, Shandong University, NO. 180 Wenhua West Road, Gao Strict, Weihai 264209, China; The Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Jiangnan University, NO. 1800 Lihu Road, Wuxi 214122, China.
| |
Collapse
|
126
|
Kumari D, Gautam J, Sharma V, Gupta SK, Sarkar S, Jana P, Singhal V, Babele P, Kamboj P, Bajpai S, Tandon R, Kumar Y, Dikshit M. Effect of herbal extracts and Saroglitazar on high-fat diet-induced obesity, insulin resistance, dyslipidemia, and hepatic lipidome in C57BL/6J mice. Heliyon 2023; 9:e22051. [PMID: 38027691 PMCID: PMC10663915 DOI: 10.1016/j.heliyon.2023.e22051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
We evaluated the effects of select herbal extracts (Tinospora cordifolia [TC], Tinospora cordifolia with Piper longum [TC + PL], Withania somnifera [WS], Glycyrrhiza glabra [GG], AYUSH-64 [AY-64], and Saroglitazar [S]) on various parameters in a diet-induced obesity mouse model. After 12 weeks of oral administration of the herbal extracts in high-fat diet (HFD)-fed C57BL/6J mice, we analyzed plasma biochemical parameters, insulin resistance (IR), liver histology, and the expression of inflammatory and fibrosis markers, along with hepatic lipidome. We also used a 3D hepatic spheroid model to assess their impact on profibrotic gene expression. Among the extracts, TC + PL showed a significant reduction in IR, liver weight, TNF-α, IL4, IL10 expression, and hepatic lipid levels (saturated triglycerides, ceramides, lysophosphocholines, acylcarnitines, diglycerides, and phosphatidylinositol levels). Saroglitazar reversed changes in body weight, IR, plasma triglycerides, glucose, insulin, and various hepatic lipid species (fatty acids, phospholipids, glycerophospholipids, sphingolipids, and triglycerides). With the exception of GG, Saroglitazar, and other extracts protected against palmitic acid-induced fibrosis marker gene expression in the 3D spheroids. TC + PL and Saroglitazar also effectively prevented HFD-induced insulin resistance, inflammation, and specific harmful lipid species in the liver.
Collapse
Affiliation(s)
- Deepika Kumari
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Jyoti Gautam
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Vipin Sharma
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Sonu Kumar Gupta
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Soumalya Sarkar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Pradipta Jana
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Vikas Singhal
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Prabhakar Babele
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Parul Kamboj
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Sneh Bajpai
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | | | - Yashwant Kumar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Madhu Dikshit
- Central Drug Research Institute, Sitapur Rd, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India.
| |
Collapse
|
127
|
Taylor J, Uhl L, Moll I, Hasan SS, Wiedmann L, Morgenstern J, Giaimo BD, Friedrich T, Alsina-Sanchis E, De Angelis Rigotti F, Mülfarth R, Kaltenbach S, Schenk D, Nickel F, Fleming T, Sprinzak D, Mogler C, Korff T, Billeter AT, Müller-Stich BP, Berriel Diaz M, Borggrefe T, Herzig S, Rohm M, Rodriguez-Vita J, Fischer A. Endothelial Notch1 signaling in white adipose tissue promotes cancer cachexia. NATURE CANCER 2023; 4:1544-1560. [PMID: 37749321 PMCID: PMC10663158 DOI: 10.1038/s43018-023-00622-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/20/2023] [Indexed: 09/27/2023]
Abstract
Cachexia is a major cause of morbidity and mortality in individuals with cancer and is characterized by weight loss due to adipose and muscle tissue wasting. Hallmarks of white adipose tissue (WAT) remodeling, which often precedes weight loss, are impaired lipid storage, inflammation and eventually fibrosis. Tissue wasting occurs in response to tumor-secreted factors. Considering that the continuous endothelium in WAT is the first line of contact with circulating factors, we postulated whether the endothelium itself may orchestrate tissue remodeling. Here, we show using human and mouse cancer models that during precachexia, tumors overactivate Notch1 signaling in distant WAT endothelium. Sustained endothelial Notch1 signaling induces a WAT wasting phenotype in male mice through excessive retinoic acid production. Pharmacological blockade of retinoic acid signaling was sufficient to inhibit WAT wasting in a mouse cancer cachexia model. This demonstrates that cancer manipulates the endothelium at distant sites to mediate WAT wasting by altering angiocrine signals.
Collapse
Affiliation(s)
- Jacqueline Taylor
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Leonie Uhl
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Iris Moll
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sana Safatul Hasan
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Lena Wiedmann
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob Morgenstern
- Department of Internal Medicine Endocrinology and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | | | - Tobias Friedrich
- Institute of Biochemistry, University of Giessen, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Science Unit for Basic and Clinical Medicine, Giessen, Germany
| | - Elisenda Alsina-Sanchis
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Francesca De Angelis Rigotti
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Tumor-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Ronja Mülfarth
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sarah Kaltenbach
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Darius Schenk
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Nickel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thomas Fleming
- Department of Internal Medicine Endocrinology and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
- German Center of Diabetes Research (DZD), Neuherberg, Germany
| | - David Sprinzak
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Carolin Mogler
- Institute of Pathology, Technical University of Munich School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas Korff
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, University of Heidelberg, Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Adrian T Billeter
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Beat P Müller-Stich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Unit, Department of Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Tilman Borggrefe
- Institute of Biochemistry, University of Giessen, Giessen, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Unit, Department of Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
- Chair Molecular Metabolic Control, Technical University of Munich, Munich, Germany
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Unit, Department of Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Juan Rodriguez-Vita
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Tumor-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Andreas Fischer
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
128
|
Sikking MA, Stroeks SL, Marelli-Berg F, Heymans SR, Ludewig B, Verdonschot JA. Immunomodulation of Myocardial Fibrosis. JACC Basic Transl Sci 2023; 8:1477-1488. [PMID: 38093747 PMCID: PMC10714184 DOI: 10.1016/j.jacbts.2023.03.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 06/27/2024]
Abstract
Immunotherapy is a potential cornerstone in the treatment of myocardial fibrosis. During a myocardial insult or heart failure, danger signals stimulate innate immune cells to produce chemokines and profibrotic cytokines, which initiate self-escalating inflammatory processes by attracting and stimulating adaptive immune cells. Stimulation of fibroblasts by inflammatory processes and the need to replace damaged cardiomyocytes fosters reshaping of the cardiac fibroblast landscape. In this review, we discuss new immunomodulatory strategies that manipulate and direct cardiac fibroblast activation and differentiation. In particular, we highlight immunomodulatory strategies that target fibroblasts such as chimeric antigen receptor T cells, interleukin-11, and invariant natural killer T-cells. Moreover, we discuss the potential of manipulating both innate and adaptive immune system components for the translation into clinical validation. Clearly, multiple pathways should be considered to develop innovative approaches to ameliorate myocardial fibrosis and hence to reduce the risk of heart failure.
Collapse
Affiliation(s)
- Maurits A. Sikking
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Sophie L.V.M. Stroeks
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Federica Marelli-Berg
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Stephane R.B. Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
- Department of Cardiovascular Research, University of Leuven, Leuven, Belgium
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Job A.J. Verdonschot
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
- Department of Clinical Genetics, Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| |
Collapse
|
129
|
Guo X, Wang Z, Qu M, Guo Y, Yu M, Hong W, Zhang C, Fan X, Song J, Xu R, Zhang J, Huang H, Linghu E, Wang FS, Sun L, Jiao YM. Abnormal blood microbiota profiles are associated with inflammation and immune restoration in HIV/AIDS individuals. mSystems 2023; 8:e0046723. [PMID: 37698407 PMCID: PMC10654078 DOI: 10.1128/msystems.00467-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/22/2023] [Indexed: 09/13/2023] Open
Abstract
IMPORTANCE The characteristics of blood microbiota in HIV-infected individuals and their relevance to disease progression are still unknown, despite alterations in gut microbiota diversity and composition in HIV-infected individuals. Here, we present evidence of increased blood microbiota diversity in HIV-infected individuals, which may result from gut microbiota translocation. Also, we identify a group of microbes, Porphyromonas gingivalis, Prevotella sp. CAG:5226, Eubacterium sp. CAG:251, Phascolarctobacterium succinatutens, Anaerobutyricum hallii, Prevotella sp. AM34-19LB, and Phocaeicola plebeius, which are linked to poor immunological recovery. This work provides a scientific foundation toward therapeutic strategies targeting blood microbiota for immune recovery of HIV infection.
Collapse
Affiliation(s)
- Xiaoyan Guo
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Zerui Wang
- Department of Gastroenterology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Mengmeng Qu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yuntian Guo
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Minrui Yu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Weiguo Hong
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Chao Zhang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Xing Fan
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jinwen Song
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ruonan Xu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jiyuan Zhang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Huihuang Huang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Enqiang Linghu
- Department of Gastroenterology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Lijun Sun
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yan-Mei Jiao
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| |
Collapse
|
130
|
Diny NL, Wood MK, Won T, Talor MV, Lukban C, Bedja D, Wang N, Kalinoski H, Daoud A, Talbot CC, Leei Lin B, Čiháková D. Hypereosinophilia causes progressive cardiac pathologies in mice. iScience 2023; 26:107990. [PMID: 37829205 PMCID: PMC10565781 DOI: 10.1016/j.isci.2023.107990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 08/02/2023] [Accepted: 09/16/2023] [Indexed: 10/14/2023] Open
Abstract
Hypereosinophilic syndrome is a progressive disease with extensive eosinophilia that results in organ damage. Cardiac pathologies are the main reason for its high mortality rate. A better understanding of the mechanisms of eosinophil-mediated tissue damage would benefit therapeutic development. Here, we describe the cardiac pathologies that developed in a mouse model of hypereosinophilic syndrome. These IL-5 transgenic mice exhibited decreased left ventricular function at a young age which worsened with age. Mechanistically, we demonstrated infiltration of activated eosinophils into the heart tissue that led to an inflammatory environment. Gene expression signatures showed tissue damage as well as repair and remodeling processes. Cardiomyocytes from IL-5Tg mice exhibited significantly reduced contractility relative to wild type (WT) controls. This impairment may result from the inflammatory stress experienced by the cardiomyocytes and suggest that dysregulation of contractility and Ca2+ reuptake in cardiomyocytes contributes to cardiac dysfunction at the whole organ level in hypereosinophilic mice.
Collapse
Affiliation(s)
- Nicola Laura Diny
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Megan Kay Wood
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Taejoon Won
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Monica Vladut Talor
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Clarisse Lukban
- Department of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Djahida Bedja
- Department of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nadan Wang
- Department of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hannah Kalinoski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Abdel Daoud
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - C. Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Brian Leei Lin
- Department of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daniela Čiháková
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
131
|
Pu S, Yang Z, Zhang X, Li M, Han N, Yang X, He J, Yu G, Meng X, Jia Q, Shao H. Fermented cordyceps powder alleviates silica-induced pulmonary inflammation and fibrosis in rats by regulating the Th immune response. Chin Med 2023; 18:131. [PMID: 37828528 PMCID: PMC10571334 DOI: 10.1186/s13020-023-00823-8] [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: 05/09/2023] [Accepted: 08/20/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND Silicosis is an important occupational disease caused by inhalation of free silica and is characterized by persistent pulmonary inflammation, subsequent fibrosis and lung dysfunction. Until now, there has been no effective treatment for the disease due to the complexity of pathogenesis. Fermented cordyceps powder (FCP) has a similar effect to natural cordyceps in tonifying the lung and kidney. It has started to be used in the adjuvant treatment of silicosis. This work aimed to verify the protective effects of FCP against silicosis, and to explore the related mechanism. METHODS Wistar rats were randomly divided into four groups including the saline-instilled group, the silica-exposed group, the silica + FCP (300 mg/kg) group and the silica + FCP (600 mg/kg) group. Silicosis rat models were constructed by intratracheal instillation of silica (50 mg). Rats in the FCP intervention groups received the corresponding dose of FCP daily by intragastric gavage. Rats were sacrificed on days 7, 28 and 56 after treatment, then samples were collected for further analysis. RESULTS FCP intervention reduced the infiltration of inflammatory cells and the concentration of interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and transforming growth factor-β1 (TGF-β1) at days 7, 28, 56, and decreased the expression of collagen, α-smooth muscle actin (α-SMA) and fibronectin (FN) at days 28 and 56 in the lung of silicosis rats. FCP also decreased the immune response of Th1 and Th17 at days 7, 28, 56 and inhibited the enhancement of the Th2 response at day 56. CONCLUSIONS FCP intervention could alleviate silica-induced pulmonary inflammation and fibrosis, the protective effect may be achieved by reducing Th1 and Th17 immune responses and inhibiting the enhancement of the Th2 response.
Collapse
Affiliation(s)
- Shuangshuang Pu
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Changqing District, Jinan, 250355, Shandong, China
- Hospital Affiliated to Shandong University of Traditional Chinese Medicine, 16369 Jingshi Road, Lixia District, Jinan, 250014, Shandong, China
| | - Zhifeng Yang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China
| | - Xiaofeng Zhang
- Linyi County Center for Disease Control and Prevention, Linyi County, 91 Yongxing Street, Dezhou, 251500, Shandong, China
| | - Ming Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China
| | - Na Han
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Changqing District, Jinan, 250355, Shandong, China
| | - Xiaohan Yang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China
| | - Jin He
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China
| | - Gongchang Yu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China
| | - Xiangjing Meng
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China.
| | - Hua Shao
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Changqing District, Jinan, 250355, Shandong, China.
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China.
| |
Collapse
|
132
|
Maizels RM, Gause WC. Targeting helminths: The expanding world of type 2 immune effector mechanisms. J Exp Med 2023; 220:e20221381. [PMID: 37638887 PMCID: PMC10460967 DOI: 10.1084/jem.20221381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/24/2023] [Accepted: 08/09/2023] [Indexed: 08/29/2023] Open
Abstract
In this new review, Rick Maizels and Bill Gause summarize how type 2 immune responses combat helminth parasites through novel mechanisms, coordinating multiple innate and adaptive cell and molecular players that can eliminate infection and repair-resultant tissue damage.
Collapse
Affiliation(s)
- Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - William C. Gause
- Center for Immunity and Inflammation, Rutgers Biomedical Health Sciences Institute for Infectious and Inflammatory Diseases, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, USA
| |
Collapse
|
133
|
Russi AE, Shivakumar P, Luo Z, Bezerra J. Plasticity between type 2 innate lymphoid cell subsets and amphiregulin expression regulates epithelial repair in biliary atresia. Hepatology 2023; 78:1035-1049. [PMID: 37078450 PMCID: PMC10524120 DOI: 10.1097/hep.0000000000000418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/22/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND AND AIMS Although a dysregulated type 1 immune response is integral to the pathogenesis of biliary atresia, studies in both humans and mice have uncovered a type 2 response, primarily driven by type 2 innate lymphoid cells. In nonhepatic tissues, natural type 2 innate lymphoid cell (nILC2s) regulate epithelial proliferation and tissue repair, whereas inflammatory ILC2s (iIlC2s) drive tissue inflammation and injury. The aim of this study is to determine the mechanisms used by type 2 innate lymphoid cell (ILC2) subpopulations to regulate biliary epithelial response to an injury. APPROACH AND RESULTS Using Spearman correlation analysis, nILC2 transcripts, but not those of iILC2s, are positively associated with cholangiocyte abundance in biliary atresia patients at the time of diagnosis. nILC2s are identified in the mouse liver through flow cytometry. They undergo expansion and increase amphiregulin production after IL-33 administration. This drives epithelial proliferation dependent on the IL-13/IL-4Rα/STAT6 pathway as determined by decreased nILC2s and reduced epithelial proliferation in knockout strains. The addition of IL-2 promotes inter-lineage plasticity towards a nILC2 phenotype. In experimental biliary atresia induced by rotavirus, this pathway promotes epithelial repair and tissue regeneration. The genetic loss or molecular inhibition of any part of this circuit switches nILC2s to inflammatory type 2 innate lymphoid cell-like, resulting in decreased amphiregulin production, decreased epithelial proliferation, and the full phenotype of experimental biliary atresia. CONCLUSIONS These findings identify a key function of the IL-13/IL-4Rα/STAT6 pathway in ILC2 plasticity and an alternate circuit driven by IL-2 to promote nILC2 stability and amphiregulin expression. This pathway induces epithelial homeostasis and repair in experimental biliary atresia.
Collapse
Affiliation(s)
- Abigail E Russi
- Division of Gastroenterology, Hepatology and Nutrition at Cincinnati Children’s Hospital Medical Center; Cincinnati OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine; Cincinnati OH, USA
| | - Pranavkumar Shivakumar
- Division of Gastroenterology, Hepatology and Nutrition at Cincinnati Children’s Hospital Medical Center; Cincinnati OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine; Cincinnati OH, USA
| | - Zhenhua Luo
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China, 510080
| | - Jorge Bezerra
- Department of Pediatrics, University of Texas Southwestern Medical Center and Children’s Health of Dallas, TX, USA
| |
Collapse
|
134
|
Liu T, Liu J, Chen H, Zhou X, Fu W, Cao Y, Yang J. Cannabinoid receptor 2 signal promotes type 2 immunity in the lung. CELL INSIGHT 2023; 2:100124. [PMID: 37868095 PMCID: PMC10585230 DOI: 10.1016/j.cellin.2023.100124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/04/2023] [Accepted: 10/04/2023] [Indexed: 10/24/2023]
Abstract
Type 2 immunity in the lung protects against pathogenic infection and facilitates tissue repair, but its dysregulation may lead to severe human diseases. Notably, cannabis usage for medical or recreational purposes has increased globally. However, the potential impact of the cannabinoid signal on lung immunity is incompletely understood. Here, we report that cannabinoid receptor 2 (CB2) is highly expressed in group 2 innate lymphoid cells (ILC2s) of mouse and human lung tissues. Of importance, the CB2 signal enhances the IL-33-elicited immune response of ILC2s. In addition, the chemogenetic manipulation of inhibitory G proteins (Gi) downstream of CB2 produces a similarly promotive effect. Conversely, the genetic deletion of CB2 mitigates the IL-33-elicited type 2 immunity in the lung. Also, such ablation of the CB2 signal ameliorates papain-induced tissue inflammation. Together, these results have elucidated a critical aspect of the CB2 signal in lung immunity, implicating its potential involvement in pulmonary diseases.
Collapse
Affiliation(s)
- Tingting Liu
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Jiaqi Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Hongjie Chen
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xin Zhou
- Department of General Surgery, Peking University Third Hospital, Beijing, 100191, China
- Peking University Third Hospital Cancer Center, Beijing, 100191, China
| | - Wei Fu
- Department of General Surgery, Peking University Third Hospital, Beijing, 100191, China
- Peking University Third Hospital Cancer Center, Beijing, 100191, China
| | - Ying Cao
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Jing Yang
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
- Peking University Third Hospital Cancer Center, Beijing, 100191, China
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, 518055, China
| |
Collapse
|
135
|
Wang J, Liu X, Sun R, Mao H, Liu M, Jin X. Akkermansia muciniphila participates in the host protection against helminth-induced cardiac fibrosis via TLR2. PLoS Pathog 2023; 19:e1011683. [PMID: 37788279 PMCID: PMC10547169 DOI: 10.1371/journal.ppat.1011683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/12/2023] [Indexed: 10/05/2023] Open
Abstract
Helminth Trichinella spiralis (Ts) is one of the major pathogens of human infective myocarditis that can lead to cardiac fibrosis (CF). The gut microbiota involved in this pathology are of interest. Here, we use mice infected with Ts as a model to examine the interactions between gut microbes and host protection to CF. Infected mice show enhanced CF severity. We find that antibiotics treatment to deplete the microbiota aggravates the disease phenotype. Attempts to restore microbiota using fecal microbiota transplantation ameliorates helminth-induced CF. 16S rRNA gene sequencing and metagenomics sequencing reveal a higher abundance of Akkermansia muciniphila in gut microbiomes of Ts-infected mice. Oral supplementation with alive or pasteurized A. muciniphila improves CF via TLR2. This work represents a substantial advance toward our understanding of causative rather than correlative relationships between the gut microbiota and CF.
Collapse
Affiliation(s)
- Jiaqi Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Animal Sciences, Jilin University, Changchun, China
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaolei Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ruohang Sun
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hanhai Mao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Mingyuan Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xuemin Jin
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
136
|
Ezzo M, Hinz B. Novel approaches to target fibroblast mechanotransduction in fibroproliferative diseases. Pharmacol Ther 2023; 250:108528. [PMID: 37708995 DOI: 10.1016/j.pharmthera.2023.108528] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/09/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
The ability of cells to sense and respond to changes in mechanical environment is vital in conditions of organ injury when the architecture of normal tissues is disturbed or lost. Among the various cellular players that respond to injury, fibroblasts take center stage in re-establishing tissue integrity by secreting and organizing extracellular matrix into stabilizing scar tissue. Activation, activity, survival, and death of scar-forming fibroblasts are tightly controlled by mechanical environment and proper mechanotransduction ensures that fibroblast activities cease after completion of the tissue repair process. Conversely, dysregulated mechanotransduction often results in fibroblast over-activation or persistence beyond the state of normal repair. The resulting pathological accumulation of extracellular matrix is called fibrosis, a condition that has been associated with over 40% of all deaths in the industrialized countries. Consequently, elements in fibroblast mechanotransduction are scrutinized for their suitability as anti-fibrotic therapeutic targets. We review the current knowledge on mechanically relevant factors in the fibroblast extracellular environment, cell-matrix and cell-cell adhesion structures, stretch-activated membrane channels, stress-regulated cytoskeletal structures, and co-transcription factors. We critically discuss the targetability of these elements in therapeutic approaches and their progress in pre-clinical and/or clinical trials to treat organ fibrosis.
Collapse
Affiliation(s)
- Maya Ezzo
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, and Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, and Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
137
|
Kumar V, Hertz M, Agro A, Byrne AJ. Type 1 invariant natural killer T cells in chronic inflammation and tissue fibrosis. Front Immunol 2023; 14:1260503. [PMID: 37818376 PMCID: PMC10561218 DOI: 10.3389/fimmu.2023.1260503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/06/2023] [Indexed: 10/12/2023] Open
Abstract
Chronic tissue inflammation often results in fibrosis characterized by the accumulation of extracellular matrix components remodeling normal tissue architecture and function. Recent studies have suggested common immune mechanisms despite the complexity of the interactions between tissue-specific fibroblasts, macrophages, and distinct immune cell populations that mediate fibrosis in various tissues. Natural killer T (NKT) cells recognizing lipid antigens bound to CD1d molecules have been shown to play an important role in chronic inflammation and fibrosis. Here we review recent data in both experimental models and in humans that suggest a key role of type 1 invariant NKT (iNKT) cell activation in the progression of inflammatory cascades leading to recruitment of neutrophils and activation of the inflammasome, macrophages, fibroblasts, and, ultimately, fibrosis. Emerging evidence suggests that iNKT-associated mechanisms contribute to type 1, type 2 and type 3 immune pathways mediating tissue fibrosis, including idiopathic pulmonary fibrosis (IPF). Thus, targeting a pathway upstream of these immune mechanisms, such as the inhibition of iNKT activation, may be important in modulating various fibrotic conditions.
Collapse
Affiliation(s)
- Vipin Kumar
- Laboratory of Immune Regulation, Department of Medicine, University of California San Diego, La Jolla, CA, United States
- GRI Bio, La Jolla, CA, United States
| | | | | | - Adam J. Byrne
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- School of Medicine and Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
138
|
Lopez K, Deng JJ, Xu Y, Sharkey FE, Wang P, Liu J. Exploring the Role of YAP1 and TAZ in Pancreatic Acinar Cells and the Therapeutic Potential of VT-104 in Pancreatic Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.558321. [PMID: 37781601 PMCID: PMC10541090 DOI: 10.1101/2023.09.18.558321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Increasing evidences have linked the hippo pathway with the fibroinflammatory diseases. We generated a series of genetic knockout mice for targeting the key components of Hippo pathway to examine the individual effects of YAP1 and TAZ on pancreatic inflammation and evaluated the therapeutic potential of the YAP1/TAZ inhibitor VT-104. Mice with acinar-specific knockout of YAP1/TAZ did not exhibit any histological abnormalities in the pancreas. LATS1/2 deficiency induced acinar-to-ductal metaplasia, immune cell infiltration and fibroblast activation, which were rescued by the homozygous knockout YAP1, but not TAZ. Additionally, treatment with VT-104 also decreased pathological alterations induced by deletions of LATS1 and LATS2 in acinar cells. Our findings highlight the critical role of YAP1 in modulating pancreatic inflammation and demonstrate that VT-104 holds therapeutic potential to mitigate pancreatitis-associated pathological manifestations. Further exploration is necessary to unravel the underlying mechanisms and translate these insights into clinical applications.
Collapse
|
139
|
Yuan X, Jiang H, Fu D, Rech JC, Robida A, Rajanayake K, Yuan H, He M, Wen B, Sun D, Liu C, Chinnaswamy K, Stuckey JA, Paczesny S, Yang CY. Prophylactic Mitigation of Acute Graft versus Host Disease by Novel 2-(Pyrrolidin-1-ylmethyl)pyrrole-Based Stimulation-2 (ST2) Inhibitors. ACS Pharmacol Transl Sci 2023; 6:1275-1287. [PMID: 37705593 PMCID: PMC10496145 DOI: 10.1021/acsptsci.3c00122] [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: 06/16/2023] [Indexed: 09/15/2023]
Abstract
Hematopoietic cell transplantation (HCT) is a proven and potentially curable therapy for hematological malignancies and inherited hematological disease. The main risk of HCT is the development of graft versus host disease (GVHD) acquired in up to 50% of patients. Upregulation of soluble ST2 (sST2) is a key clinical biomarker for GVHD prognosis and was shown to be a potential therapeutic target for GVHD. Agents targeting sST2 to reduce the sST2 level after HCT have the potential to mitigate GVHD progression. Here, we report 32 (or XY52) as the lead ST2 inhibitor from our optimization campaign. XY52 had improved inhibitory activity and metabolic stability in vitro and in vivo. XY52 suppressed proinflammatory T-cell proliferation while increasing regulatory T cells in vitro. In a clinically relevant GVHD model, a 21-day prophylactic regimen of XY52 reduced plasma sST2 and IFN-γ levels and GVHD score and extended survival in mice. XY52 represented a significant improvement over our previous compound, iST2-1, and further optimization of XY52 is warranted. The small-molecule ST2 inhibitors can potentially be used as a biomarker-guided therapy for mitigating GVHD in future clinical applications.
Collapse
Affiliation(s)
- Xinrui Yuan
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Hua Jiang
- Department
of Microbiology & Immunology, Medical
University of South Carolina, Charleston, South Carolina 29425-2503, United States
| | - Denggang Fu
- Department
of Microbiology & Immunology, Medical
University of South Carolina, Charleston, South Carolina 29425-2503, United States
| | - Jason C. Rech
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Aaron Robida
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Krishani Rajanayake
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hebao Yuan
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Miao He
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bo Wen
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Chen Liu
- Department
of Pathology, Yale University, New Haven, Connecticut 06520, United States
| | - Krishnapriya Chinnaswamy
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jeanne A. Stuckey
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Sophie Paczesny
- Department
of Microbiology & Immunology, Medical
University of South Carolina, Charleston, South Carolina 29425-2503, United States
| | - Chao-Yie Yang
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
140
|
Barone L, Gallazzi M, Rossi F, Papait R, Raspanti M, Zecca PA, Buonarrivo L, Bassani B, Bernardini G, Bruno A, Gornati R. Human Dental Pulp Mesenchymal Stem Cell-Derived Soluble Factors Combined with a Nanostructured Scaffold Support the Generation of a Vascular Network In Vivo. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2479. [PMID: 37686988 PMCID: PMC10489993 DOI: 10.3390/nano13172479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023]
Abstract
Among all strategies directed at developing new tools to support re-vascularization of damaged tissues, the use of pro-angiogenic soluble factors, derived from mesenchymal stem cells (MSCs), appears a promising approach for regenerative medicine. Here, we compared the feasibility of two devices, generated by coupling soluble factors of human dental pulp mesenchymal stem cells (DPSCs), with a nanostructured scaffold, to support angiogenesis once transplanted in mice. DPSCs were obtained from impacted wisdom tooth removal, usually considered surgical waste material. After 28 days, we verified the presence of active blood vessels inside the scaffold through optical and scansion electron microscopy. The mRNA expression of surface antigens related to macrophage polarization (CD68, CD80, CD86, CD163, CD206), as well as pro-angiogenic markers (CD31, CD34, CD105, Angpt1, Angpt2, CDH5) was evaluated by real-time PCR. Our results demonstrate the capability of DPSC-scaffold and DPSC soluble factors-scaffold to support angiogenesis, similarly to adipose stem cells, whereas the absence of blood vessels was found in the scaffold grafted alone. Our results provide evidence that DPSC-conditioned medium can be proposed as a cell-free preparation able to support angiogenesis, thus, providing a relevant tool to overcome the issues and restrictions associated with the use of cells.
Collapse
Affiliation(s)
- Ludovica Barone
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
| | - Matteo Gallazzi
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (M.G.); (B.B.)
| | - Federica Rossi
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
| | - Roberto Papait
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
| | - Mario Raspanti
- Department of Medicine and Innovative Technology, University of Insubria, 21100 Varese, Italy; (M.R.); (P.A.Z.)
| | - Piero Antonio Zecca
- Department of Medicine and Innovative Technology, University of Insubria, 21100 Varese, Italy; (M.R.); (P.A.Z.)
| | - Luca Buonarrivo
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (M.G.); (B.B.)
| | - Giovanni Bernardini
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
| | - Antonino Bruno
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (M.G.); (B.B.)
| | - Rosalba Gornati
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
| |
Collapse
|
141
|
Wang L, Li D, Zhu Z, Liao Y, Wu J, Liu Y, Yang R, Dai H, Wu Z, Sun X. Knockout of Sema4D alleviates liver fibrosis by suppressing AOX1 expression. Pharmacol Res 2023; 195:106886. [PMID: 37591326 DOI: 10.1016/j.phrs.2023.106886] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/30/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023]
Abstract
Liver fibrosis can occur in many chronic liver diseases, and no effective treatments are available due to the poorly characterized molecular pathogenesis. Semaphorin 4D (Sema4D) has immune functions and serves important roles in T cell priming. Here, we found that Sema4D was highly expressed in fibrotic liver, and the expression of Sema4D increased with hepatic stellate cells (HSCs) activation. Knockout of Sema4D alleviated liver fibrosis. Mechanistically, knockout of Sema4D alleviated liver fibrosis by suppressing the expression of AOX1 in retinol metabolism. Further investigation demonstrated that retinoic acid receptor α (RARA), an important nuclear receptor of retinoic acid, was reduced by Sema4D knockout during liver fibrogenesis. Sema4D knockout-mediated suppression of liver fibrosis was partly mediated by regulating the balance of Th1, Th2, Th17, and T-bet+Treg cells via inhibiting AOX1/RARA. Thus, targeting Sema4D may hold promise as a potential therapeutic approach for treating liver fibrosis.
Collapse
Affiliation(s)
- Lifu Wang
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 511436, China
| | - Dinghao Li
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou 510080, China
| | - Zifeng Zhu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou 510080, China
| | - Yao Liao
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 511436, China
| | - Ji Wu
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 511436, China
| | - Yuheng Liu
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 511436, China
| | - Ruibing Yang
- Guangzhou KingMed Diagnostic Laboratory Group Co Ltd, Guangzhou 510310, China
| | - Hanqiao Dai
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou 510080, China.
| | - Xi Sun
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou 510080, China.
| |
Collapse
|
142
|
Ghonim MA, Boyd DF, Flerlage T, Thomas PG. Pulmonary inflammation and fibroblast immunoregulation: from bench to bedside. J Clin Invest 2023; 133:e170499. [PMID: 37655660 PMCID: PMC10471178 DOI: 10.1172/jci170499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
In recent years, there has been an explosion of interest in how fibroblasts initiate, sustain, and resolve inflammation across disease states. Fibroblasts contain heterogeneous subsets with diverse functionality. The phenotypes of these populations vary depending on their spatial distribution within the tissue and the immunopathologic cues contributing to disease progression. In addition to their roles in structurally supporting organs and remodeling tissue, fibroblasts mediate critical interactions with diverse immune cells. These interactions have important implications for defining mechanisms of disease and identifying potential therapeutic targets. Fibroblasts in the respiratory tract, in particular, determine the severity and outcome of numerous acute and chronic lung diseases, including asthma, chronic obstructive pulmonary disease, acute respiratory distress syndrome, and idiopathic pulmonary fibrosis. Here, we review recent studies defining the spatiotemporal identity of the lung-derived fibroblasts and the mechanisms by which these subsets regulate immune responses to insult exposures and highlight past, current, and future therapeutic targets with relevance to fibroblast biology in the context of acute and chronic human respiratory diseases. This perspective highlights the importance of tissue context in defining fibroblast-immune crosstalk and paves the way for identifying therapeutic approaches to benefit patients with acute and chronic pulmonary disorders.
Collapse
Affiliation(s)
- Mohamed A. Ghonim
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al Azhar University, Cairo, Egypt
| | - David F. Boyd
- Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, USA
| | - Tim Flerlage
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
143
|
Ospino R, Berges A, Mafla L, Collins S, Li YC, Lina I, Gelbard A, Hillel AT, Motz K. Characterizing the Macrophage Population in Patients With Idiopathic Subglottic Stenosis. Laryngoscope 2023; 133:2308-2316. [PMID: 36524603 PMCID: PMC10272290 DOI: 10.1002/lary.30524] [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: 04/07/2022] [Revised: 09/19/2022] [Accepted: 11/20/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Idiopathic subglottic stenosis (iSGS) is characterized by progressive fibrosis and subglottic luminal narrowing. Currently, immune characterization has focused on T-cells; however, macrophages remain largely unexplored. The goals of this study are to characterize the transcriptome of iSGS macrophages and the fibrogenic nature of identifed biomarkers. STUDY DESIGN Bioinformatics and in vitro. METHODS Human tracheal biopsies from iSGS scar (n = 4), and matched non-scar (n = 4) regions were analyzed using single-cell RNA-seq (scRNA-seq). Immunofluorescence (IF) was performed on rapidly processed autopsies (RPA) and iSGS tracheal resections (n = 4) to co-localize S100A8/9 and CD11b. Collagen gene/protein expression was assessed in iSGS fibroblasts (n = 4) treated with protein S100A8/9 (1000 ng/ml). Macrophages were subclustered to identify distinct subpopulations. RESULTS scRNA-seq analysis revealed S100A8/S100A9 (fold change (FC) = 4.1/1.88, p < 0.001) as top differentially expressed genes in iSGS macrophages. IF exhibited increased CD11b+/S100A8/9+ cells in tracheal samples of iSGS versus RPA (26.75% ± 7.08 vs. 0.594% ± 0.974, n = 4, p = 0.029). iSGS fibroblasts treated with S100A8/9 demonstrated increased gene expression of COL1A1 (FC = 2.30 ± 0.45, p = 0.03, n = 4) and COL3A1 (FC = 2.44 ± 0.40, p = 0.03, n = 4). COL1A1 protein assays revealed an increase in the experimental group, albeit not significant, (p = 0.12, n = 4). Finally, macrophage sub clustering revealed one subpopulation as a predominant source of S100A8/S100A9 expression (FC = 7.94/5.47, p < 0.001). CONCLUSIONS S100A8/9 is a key biomarker in iSGS macrophages. Although S100A8/9 demonstrates profibrotic nature in vitro, the role of S100A8/9+ macrophages in vivo warrants further investigation. LEVEL OF EVIDENCE NA Laryngoscope, 133:2308-2316, 2023.
Collapse
Affiliation(s)
- Rafael Ospino
- Johns Hopkins University School of Medicine, 1800 Orleans Street, Baltimore, MD, 21287
| | - Alexandra Berges
- Johns Hopkins Department of Otolaryngology Head and Neck Surgery, 1800 Orleans Street, Baltimore, MD, 21287
| | - Laura Mafla
- Johns Hopkins University School of Medicine, 1800 Orleans Street, Baltimore, MD, 21287
| | - Samuel Collins
- Johns Hopkins Department of Otolaryngology Head and Neck Surgery, 1800 Orleans Street, Baltimore, MD, 21287
| | - Yee Chan Li
- Johns Hopkins Department of Otolaryngology Head and Neck Surgery, 1800 Orleans Street, Baltimore, MD, 21287
| | - Ioan Lina
- Johns Hopkins Department of Otolaryngology Head and Neck Surgery, 1800 Orleans Street, Baltimore, MD, 21287
| | - Alexander Gelbard
- Department of Otolaryngology - Head & Neck Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Alexander T. Hillel
- Johns Hopkins Department of Otolaryngology Head and Neck Surgery, 1800 Orleans Street, Baltimore, MD, 21287
| | - Kevin Motz
- Johns Hopkins Department of Otolaryngology Head and Neck Surgery, 1800 Orleans Street, Baltimore, MD, 21287
| |
Collapse
|
144
|
Sung EA, Park MH, Henegariu O, Sime PJ, Chae WJ. Dickkopf1 Promotes Pulmonary Fibrosis upon Bleomycin-Induced Lung Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1130-1142. [PMID: 37263344 PMCID: PMC10477954 DOI: 10.1016/j.ajpath.2023.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/21/2023] [Accepted: 05/19/2023] [Indexed: 06/03/2023]
Abstract
Orchestration of inflammation and tissue repair processes is critical to maintaining homeostasis upon tissue injury. Tissue fibrosis is a pathological process characterized by aberrant accumulation of extracellular matrix proteins, such as collagen, upon injury. Dickkopf1 (DKK1) is a quintessential Wnt antagonist. The role of DKK1 in bleomycin (BLM)-induced lung injury and fibrosis model remains elusive. This study shows that BLM-induced lung injury markedly elevated DKK1 protein expressions in the lungs in mice, consistent with human pulmonary fibrosis patient lung tissues. The elevated DKK1 levels coincided with immune cell infiltration and collagen deposition. Notably, the reduced expression of DKK1 in Dkk1 hypomorphic doubleridge (Dkk1d/d) mice abrogated BLM-induced lung inflammation and fibrosis. Immune cell infiltration, collagen deposition, expression of profibrotic cytokine transforming growth factor β1 (TGF-β1), and extracellular matrix protein-producing myofibroblast marker α-smooth muscle actin (α-SMA) were reduced in Dkk1d/d mice. Consistent with these results, local DKK1 antibody administration after BLM-induced lung injury substantially decreased lung inflammation and fibrosis phenotypes. Taken together, these results demonstrate that DKK1 is a proinflammatory and profibrotic ligand that promotes inflammation and fibrosis upon BLM-induced lung injury, placing it as an attractive molecular target for dysregulated pulmonary inflammation and tissue repair.
Collapse
Affiliation(s)
- Eun-Ah Sung
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia; Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Min Hee Park
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia; Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Octavian Henegariu
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Patricia J Sime
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia; Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Wook-Jin Chae
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia; Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia.
| |
Collapse
|
145
|
Shi M, Lu Y, Mohyeddin A, Qi F, Pan Y. Preservation of Eschar Prevents Excessive Wound Healing by Reducing M2 Macrophages Polarization. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2023; 11:e5238. [PMID: 37731728 PMCID: PMC10508428 DOI: 10.1097/gox.0000000000005238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 07/11/2023] [Indexed: 09/22/2023]
Abstract
Background Removal of the eschar has gradually become a consensus on treatments of deep dermal necrosis after skin trauma in recent years, whereas exaggerated scar contracture and tissue proliferation developed during healing have received little attention. Here, the authors investigated the effects of eschar on excessive wound healing of small dermal damage and focused on the role M2 macrophages played, hoping to offer a theoretical basis to improve patients' cosmetic satisfaction. Methods A mouse dorsal wound model (n = 12) was established by electric heating pads heating for 20 seconds on each side of the spine, and the left side was the preserved group. Macrophage numbers, expression of wound-healing-associated proteins, and inflammatory cytokine levels were assessed at different time points by immunohistochemistry and quantitative real-time polymerase chain reaction. A co-culture system of M2 macrophages and myofibroblasts was created in vitro. Immunohistochemistry, real-time polymerase chain reaction, and Western blot were performed to evaluate the proliferation, migration, and protein expression of myofibroblasts. Results Preserving eschar inhibited contraction-associated proteins (α-smooth muscle actin and vimentin) and collagen expression, inflammatory cytokine (IL-1β, IL-10, TFN-α, and IL-4) expression, and M2 macrophage infiltration. Mechanistically, M2 macrophages potentially contributed to excessive wound healing by promoting myofibroblasts proliferation, migration, and production of contraction-associated proteins. Conclusion Eschar preservation in wounds could reduce inflammation and negatively modulate myofibroblasts by inhibiting M2 macrophage polarization and infiltration, preventing excessive wound contraction and collagen deposition.
Collapse
Affiliation(s)
- Mingyue Shi
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yao Lu
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ali Mohyeddin
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fazhi Qi
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuyan Pan
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
146
|
van der Ploeg EK, Krabbendam L, Vroman H, van Nimwegen M, de Bruijn MJW, de Boer GM, Bergen IM, Kool M, Tramper-Standers GA, Braunstahl GJ, Huylebroeck D, Hendriks RW, Stadhouders R. Type-2 CD8 + T-cell formation relies on interleukin-33 and is linked to asthma exacerbations. Nat Commun 2023; 14:5137. [PMID: 37612281 PMCID: PMC10447424 DOI: 10.1038/s41467-023-40820-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 08/11/2023] [Indexed: 08/25/2023] Open
Abstract
CD4+ T helper 2 (Th2) cells and group 2 innate lymphoid cells are considered the main producers of type-2 cytokines that fuel chronic airway inflammation in allergic asthma. However, CD8+ cytotoxic T (Tc) cells - critical for anti-viral defense - can also produce type-2 cytokines (referred to as 'Tc2' cells). The role of Tc cells in asthma and virus-induced disease exacerbations remains poorly understood, including which micro-environmental signals and cell types promote Tc2 cell formation. Here we show increased circulating Tc2 cell abundance in severe asthma patients, reaching peak levels during exacerbations and likely emerging from canonical IFNγ+ Tc cells through plasticity. Tc2 cell abundance is associated with increased disease burden, higher exacerbations rates and steroid insensitivity. Mouse models of asthma recapitulate the human disease by showing extensive type-2 skewing of lung Tc cells, which is controlled by conventional type-1 dendritic cells and IFNγ. Importantly, we demonstrate that the alarmin interleukin-33 (IL-33) critically promotes type-2 cytokine production by lung Tc cells in experimental allergic airway inflammation. Our data identify Tc cells as major producers of type-2 cytokines in severe asthma and during exacerbations that are remarkably sensitive to alterations in their inflammatory tissue micro-environment, with IL-33 emerging as an important regulator of Tc2 formation.
Collapse
Affiliation(s)
- Esmee K van der Ploeg
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Lisette Krabbendam
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Heleen Vroman
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Menno van Nimwegen
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Marjolein J W de Bruijn
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Geertje M de Boer
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, The Netherlands
| | - Ingrid M Bergen
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Mirjam Kool
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Gerdien A Tramper-Standers
- Department of Pediatric Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, The Netherlands
- Department of Neonatology, Sophia Children's Hospital, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Gert-Jan Braunstahl
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, The Netherlands
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
- Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
147
|
Qin S, Tan P, Xie J, Zhou Y, Zhao J. A systematic review of the research progress of traditional Chinese medicine against pulmonary fibrosis: from a pharmacological perspective. Chin Med 2023; 18:96. [PMID: 37537605 PMCID: PMC10398979 DOI: 10.1186/s13020-023-00797-7] [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: 04/27/2023] [Accepted: 07/06/2023] [Indexed: 08/05/2023] Open
Abstract
Pulmonary fibrosis is a chronic progressive interstitial lung disease caused by a variety of etiologies. The disease can eventually lead to irreversible damage to the lung tissue structure, severely affecting respiratory function and posing a serious threat to human health. Currently, glucocorticoids and immunosuppressants are the main drugs used in the clinical treatment of pulmonary fibrosis, but their efficacy is limited and they can cause serious adverse effects. Traditional Chinese medicines have important research value and potential for clinical application in anti-pulmonary fibrosis. In recent years, more and more scientific researches have been conducted on the use of traditional Chinese medicine to improve or reduce pulmonary fibrosis, and some important breakthroughs have been made. This review paper systematically summarized the research progress of pharmacological mechanism of traditional Chinese medicines and their active compounds in improving or reducing pulmonary fibrosis. We conducted a systematic search in several main scientific databases, including PubMed, Web of Science, and Google Scholar, using keywords such as idiopathic pulmonary fibrosis, pulmonary fibrosis, interstitial pneumonia, natural products, herbal medicine, and therapeutic methods. Ultimately, 252 articles were included and systematically evaluated in this analysis. The anti-fibrotic mechanisms of these traditional Chinese medicine studies can be roughly categorized into 5 main aspects, including inhibition of epithelial-mesenchymal transition, anti-inflammatory and antioxidant effects, improvement of extracellular matrix deposition, mediation of apoptosis and autophagy, and inhibition of endoplasmic reticulum stress. The purpose of this article is to provide pharmaceutical researchers with information on the progress of scientific research on improving or reducing Pulmonary fibrosis with traditional Chinese medicine, and to provide reference for further pharmacological research.
Collapse
Affiliation(s)
- Shanbo Qin
- Key Laboratory of Biological Evaluation of TCM Quality of State Administration of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Peng Tan
- Key Laboratory of Biological Evaluation of TCM Quality of State Administration of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China.
| | - Junjie Xie
- Key Laboratory of Biological Evaluation of TCM Quality of State Administration of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Yongfeng Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Junning Zhao
- Key Laboratory of Biological Evaluation of TCM Quality of State Administration of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China.
| |
Collapse
|
148
|
Lin Z, Chen Z, Chen Y, Yang N, Shi J, Tang Z, Zhang C, Lin H, Yin J. Hydrogenated silicene nanosheet functionalized scaffold enables immuno-bone remodeling. EXPLORATION (BEIJING, CHINA) 2023; 3:20220149. [PMID: 37933236 PMCID: PMC10624372 DOI: 10.1002/exp.20220149] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/03/2023] [Indexed: 11/08/2023]
Abstract
An ideal implant needs to have the ability to coordinate the foreign body response and tissue regeneration. Here, Hydrogenated-silicon nanosheets (H-Si NSs) with favorable biodegradability are integrated and functionalized into a β-tricalcium phosphate scaffold (H-Si TCP) for bone defect healing. H-Si TCP can greatly improve bone regeneration through osteoimmunomodulation-guided biodegradation in vivo. The spatiotemporal regulation of degradation products replenishes sufficient nutrients step by step for the entire process of bone repair. Extracellular and intracellular reactive oxygen species (ROS) are first downregulated by reaction with H-Si NSs, followed by marked M2 polarization, remodeling the micro-environment timely for immune-bone regeneration. The release of primary reaction products awakened bone marrow mesenchymal stem cells (BMSCs), which are converted into osteoblasts anchored on scaffolds. Subsequently, biomineralization is promoted by the final degradation products. The intrinsic ROS-responsive, immunoregulatory, and osteo-promotive capability of 2D H-Si NSs makes such composite H-Si TCP scaffold a highly potential alternative for the treatment of critical bone defect.
Collapse
Affiliation(s)
- Zixuan Lin
- Institute of Microsurgery on ExtremitiesDepartment of Orthopaedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Zhixin Chen
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of Ceramics Chinese Academy of SciencesShanghaiP. R. China
| | - Yiwei Chen
- Institute of Microsurgery on ExtremitiesDepartment of Orthopaedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Nan Yang
- Institute of Microsurgery on ExtremitiesDepartment of Orthopaedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of Ceramics Chinese Academy of SciencesShanghaiP. R. China
- Shanghai Tenth People's HospitalShanghai Frontiers Science Center of Nanocatalytic MedicineSchool of MedicineTongji UniversityShanghaiP. R. China
| | - Zhongmin Tang
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Changqing Zhang
- Institute of Microsurgery on ExtremitiesDepartment of Orthopaedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Han Lin
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of Ceramics Chinese Academy of SciencesShanghaiP. R. China
- Shanghai Tenth People's HospitalShanghai Frontiers Science Center of Nanocatalytic MedicineSchool of MedicineTongji UniversityShanghaiP. R. China
| | - Junhui Yin
- Institute of Microsurgery on ExtremitiesDepartment of Orthopaedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| |
Collapse
|
149
|
Chen M, Zhong W, Xu W. Alcohol and the mechanisms of liver disease. J Gastroenterol Hepatol 2023; 38:1233-1240. [PMID: 37423758 DOI: 10.1111/jgh.16282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023]
Abstract
Alcoholic liver disease (ALD), which is a leading cause of morbidity and mortality worldwide, covers a large spectrum of liver injuries ranging from simple steatosis to steatohepatitis, advanced fibrosis, cirrhosis, and hepatocellular carcinoma. The pathogenesis of ALD includes genetic and epigenetic alterations, oxidative stress, acetaldehyde-mediated toxicity and cytokine and chemokine-induced inflammation, metabolic reprogramming, immune damage, and dysbiosis of the gut microbiota. This review discusses the progress in the pathogenesis and molecular mechanism of ALD, which could provide evidence for further research on the potential therapeutic strategies targeting these pathways.
Collapse
Affiliation(s)
- Mo Chen
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wanglei Zhong
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Weiqi Xu
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
150
|
Feng T, Chen Y, Wei J, Tan S, Guangnan L. Distribution and chemotactic mechanism of CD4 + T cells in traumatic tracheal stenosis. Immun Inflamm Dis 2023; 11:e916. [PMID: 37647429 PMCID: PMC10411395 DOI: 10.1002/iid3.916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 09/01/2023] Open
Abstract
A systemic and local inflammatory immune imbalance is thought to be the cause of traumatic tracheal stenosis (TS). However, with CD4+ T lymphocytes being the predominant immune cells in TS, the mechanism of action and recruitment has not been described. In our research, using flow cytometry, ELISA, immunofluorescence, and Transwell chamber assays, the expression, distribution, and potential chemotactic function of CD4+ T cells in TS patients were examined before and after treatment. The results showed that the untreated group had significantly more CD4+ T cells and their secreted TGF-β1 than the treated group. Additionally, the untreated group's CD4+ T cells showed a significant rise in CCL22 and CCL1, as well as a larger proportion of CCR4 and CCR8. CD4+ T cells and CD68+ macrophages located in TS also expressed CCL1 and CCL22. In vitro, anti-CCL1 and anti-CCL22 can partially block the chemoattractant effect of TS bronchoalveolar lavage (BAL) on purified CD4+ T cells. The findings of this study indicated that TS contained unbalanced CD4 immune cells that were actively recruited locally by CCR4/CCL22 and CCR8/CCL1. As a result, it is anticipated that CD4 immune rebalancing can serve as a novel treatment for TS.
Collapse
Affiliation(s)
- Tingmei Feng
- Guangxi Medical UniversityNanningChina
- Department of Respiratory MedicineThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Yan Chen
- Department of Respiratory MedicineThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Jinmei Wei
- Department of Respiratory MedicineThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Sen Tan
- Department of Respiratory MedicineThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Liu Guangnan
- Department of Respiratory MedicineThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|