1
|
Christopoulou ME, Aletras AJ, Papakonstantinou E, Stolz D, Skandalis SS. WISP1 and Macrophage Migration Inhibitory Factor in Respiratory Inflammation: Novel Insights and Therapeutic Potentials for Asthma and COPD. Int J Mol Sci 2024; 25:10049. [PMID: 39337534 PMCID: PMC11432718 DOI: 10.3390/ijms251810049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Recent advancements highlight the intricate interplay between the extracellular matrix (ECM) and immune responses, notably in respiratory diseases such as asthma and Chronic Obstructive Pulmonary Disease (COPD). The ECM, a dynamic structural framework within tissues, orches-trates a plethora of cellular processes, including immune cell behavior and tissue repair mecha-nisms. WNT1-inducible-signaling pathway protein 1 (WISP1), a key ECM regulator, controls immune cell behavior, cytokine production, and tissue repair by modulating integrins, PI3K, Akt, β-catenin, and mTOR signaling pathways. WISP1 also induces macrophage migration inhibitory factor (MIF) expression via Src kinases and epidermal growth factor receptor (EGFR) activation. MIF, through its wide range of activities, enhances inflammation and tissue restructuring. Rec-ognized for its versatile roles in regulating the immune system, MIF interacts with multiple immune components, such as the NLRP3 inflammasome, thereby sustaining inflammatory pro-cesses. The WISP1-MIF axis potentially unveils complex molecular mechanisms governing im-mune responses and inflammation. Understanding the intricate roles of WISP1 and MIF in the pathogenesis of chronic respiratory diseases such as asthma and COPD could lead to the identi-fication of novel targets for therapeutic intervention to alleviate disease severity and enhance patient outcomes.
Collapse
Affiliation(s)
- Maria-Elpida Christopoulou
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexios J Aletras
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Eleni Papakonstantinou
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Daiana Stolz
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Spyros S Skandalis
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
2
|
de la Bastida-Casero L, García-León B, Tura-Ceide O, Oliver E. The Relevance of the Endothelium in Cardiopulmonary Disorders. Int J Mol Sci 2024; 25:9260. [PMID: 39273209 PMCID: PMC11395528 DOI: 10.3390/ijms25179260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/16/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
The endothelium is a cell monolayer that lines vessels and separates tissues from blood flow. Endothelial cells (ECs) have a multitude of functions, including regulating blood flow and systemic perfusion through changes in vessel diameter. When an injury occurs, the endothelium is affected by altering its functions and structure, which leads to endothelial dysfunction, a characteristic of many vascular diseases. Understanding the role that the endothelium plays in pulmonary vascular and cardiopulmonary diseases, and exploring new therapeutic strategies is of utmost importance to advance clinically. Currently, there are several treatments able to improve patients' quality of life, however, none are effective nor curative. This review examines the critical role of the endothelium in the pulmonary vasculature, investigating the alterations that occur in ECs and their consequences for blood vessels and potential molecular targets to regulate its alterations. Additionally, we delve into promising non-pharmacological therapeutic strategies, such as exercise and diet. The significance of the endothelium in cardiopulmonary disorders is increasingly being recognized, making ECs a relevant target for novel therapies aimed at preserving their functional and structural integrity.
Collapse
Affiliation(s)
| | - Bertha García-León
- Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, 28040 Madrid, Spain
| | - Olga Tura-Ceide
- Translational Research Group on Cardiovascular Respiratory Diseases (CAREs), Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and Institut d'Investigació Biomèdica de Girona (IDIBGI-CERCA), 17190 Girona, Spain
- Department of Pulmonary Medicine, Servei de Pneumologia, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Eduardo Oliver
- Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, 28040 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28039 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| |
Collapse
|
3
|
Othman B, Zeef L, Szestak T, Rchiad Z, Storm J, Askonas C, Satyam R, Madkhali A, Haley M, Wagstaff S, Couper K, Pain A, Craig A. Different PfEMP1-expressing Plasmodium falciparum variants induce divergent endothelial transcriptional responses during co-culture. PLoS One 2023; 18:e0295053. [PMID: 38033133 PMCID: PMC10688957 DOI: 10.1371/journal.pone.0295053] [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: 06/14/2023] [Accepted: 11/14/2023] [Indexed: 12/02/2023] Open
Abstract
The human malaria parasite Plasmodium falciparum is responsible for the majority of mortality and morbidity caused by malaria infection and differs from other human malaria species in the degree of accumulation of parasite-infected red blood cells in the microvasculature, known as cytoadherence or sequestration. In P. falciparum, cytoadherence is mediated by a protein called PfEMP1 which, due to its exposure to the host immune system, undergoes antigenic variation resulting in the expression of different PfEMP1 variants on the infected erythrocyte membrane. These PfEMP1s contain various combinations of adhesive domains, which allow for the differential engagement of a repertoire of endothelial receptors on the host microvasculature, with specific receptor usage associated with severe disease. We used a co-culture model of cytoadherence incubating human brain microvascular endothelial cells with erythrocytes infected with two parasite lines expressing different PfEMP1s that demonstrate different binding profiles to vascular endothelium. We determined the transcriptional profile of human brain microvascular endothelial cells (HBMEC) following different incubation periods with infected erythrocytes, identifying different transcriptional profiles of pathways previously found to be involved in the pathology of severe malaria, such as inflammation, apoptosis and barrier integrity, induced by the two PfEMP1 variants.
Collapse
Affiliation(s)
- Basim Othman
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Leo Zeef
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Tadge Szestak
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Zineb Rchiad
- Pathogen Genomics Laboratory, Bioscience Program, Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal, KSA
| | - Janet Storm
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Caroline Askonas
- Pathogen Genomics Laboratory, Bioscience Program, Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal, KSA
| | - Rohit Satyam
- Pathogen Genomics Laboratory, Bioscience Program, Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal, KSA
| | - Aymen Madkhali
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Michael Haley
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Simon Wagstaff
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Kevin Couper
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Arnab Pain
- Pathogen Genomics Laboratory, Bioscience Program, Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal, KSA
| | - Alister Craig
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| |
Collapse
|
4
|
Marottoli FM, Zhang H, Flores-Barrera E, Artur de la Villarmois E, Damen FC, Miguelez Fernández AM, Blesson HV, Chaudhary R, Nguyen AL, Nwokeji AE, Talati R, John AS, Madadakere K, Lutz SE, Cai K, Tseng KY, Tai LM. Endothelial Cell APOE3 Regulates Neurovascular, Neuronal, and Behavioral Function. Arterioscler Thromb Vasc Biol 2023; 43:1952-1966. [PMID: 37650329 PMCID: PMC10521805 DOI: 10.1161/atvbaha.123.319816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/17/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Specialized brain endothelial cells and human APOE3 are independently important for neurovascular function, yet whether APOE3 expression by endothelial cells contributes to brain function is currently unknown. In the present study, we determined whether the loss of endothelial cell APOE3 impacts brain vascular and neural function. METHODS We developed APOE3fl/fl/Cdh5(PAC)-CreERT2+/- (APOE3Cre+/-) and APOE3fl/fl/Cdh5(PAC)-CreERT2-/- (APOE3Cre-/-, control) mice and induced endothelial cell APOE3 knockdown with tamoxifen at ≈4 to 5 weeks of age. Neurovascular and neuronal function were evaluated by biochemistry, immunohistochemistry, behavioral testing, and electrophysiology at 9 months of age. RESULTS We found that the loss of endothelial APOE3 expression was sufficient to cause neurovascular dysfunction including higher permeability and lower vessel coverage in tandem with deficits in spatial memory and fear memory extinction and a disruption of cortical excitatory/inhibitory balance. CONCLUSIONS Our data collectively support the novel concept that endothelial APOE3 plays a critical role in the regulation of the neurovasculature, neural circuit function, and behavior.
Collapse
Affiliation(s)
- Felecia M. Marottoli
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Hui Zhang
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Eden Flores-Barrera
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Emilce Artur de la Villarmois
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | | | - Anabel M.M. Miguelez Fernández
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Hannah V. Blesson
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Rohan Chaudhary
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Anthony L. Nguyen
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Amanda E. Nwokeji
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Ruju Talati
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Ashwin S. John
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Kushi Madadakere
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Sarah E. Lutz
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Kejia Cai
- Radiology (F.C.D., K.C.), University of Illinois at Chicago
- Bioengineering (K.C.), University of Illinois at Chicago
| | - Kuei Y. Tseng
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Leon M. Tai
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| |
Collapse
|
5
|
Zhang H, Hu YM, Wang YJ, Zhou Y, Zhu ZJ, Chen MH, Wang YJ, Xu H, Wang YH. Macrophage migration inhibitory factor facilitates astrocytic production of the CCL2 chemokine following spinal cord injury. Neural Regen Res 2023; 18:1802-1808. [PMID: 36751809 PMCID: PMC10154479 DOI: 10.4103/1673-5374.363184] [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: 06/28/2022] [Revised: 10/08/2022] [Accepted: 10/20/2022] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury causes accumulation of a large number of leukocytes at the lesion site where they contribute to excessive inflammation. Overproduced chemokines are responsible for the migratory process of the leukocytes, but the regulatory mechanism underlying the production of chemokines from resident cells of the spinal cord has not been fully elucidated. We examined the protein levels of macrophage migration inhibitory factor and chemokine C-C motif chemokine ligand 2 in a spinal cord contusion model at different time points following spinal cord injury. The elevation of macrophage migration inhibitory factor at the lesion site coincided with the increase of chemokine C-C motif chemokine ligand 2 abundance in astrocytes. Stimulation of primary cultured astrocytes with different concentrations of macrophage migration inhibitory factor recombinant protein induced chemokine C-C motif chemokine ligand 2 production from the cells, and the macrophage migration inhibitory factor inhibitor 4-iodo-6-phenylpyrimidine attenuated the stimulatory effect. Further investigation into the underlying mechanism on macrophage migration inhibitory factor-mediated astrocytic production of chemokine C-C motif chemokine ligand 2 revealed that macrophage migration inhibitory factor activated intracellular JNK signaling through binding with CD74 receptor. Administration of the macrophage migration inhibitory factor inhibitor 4-iodo-6-phenylpyrimidine following spinal cord injury resulted in the reduction of chemokine C-C motif chemokine ligand 2-recruited microglia/macrophages at the lesion site and remarkably improved the hindlimb locomotor function of rats. Our results have provided insights into the functions of astrocyte-activated chemokines in the recruitment of leukocytes and may be beneficial to develop interventions targeting chemokine C-C motif chemokine ligand 2 for neuroinflammation after spinal cord injury.
Collapse
Affiliation(s)
- Han Zhang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| | - Yu-Ming Hu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Ying-Jie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Yue Zhou
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Zhen-Jie Zhu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Min-Hao Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| | - Yong-Jun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Hua Xu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| | - You-Hua Wang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
6
|
Zheng R, Xu T, Lin Y, Huang X. Are endothelial cell proliferation and mesenchymal transition as distinguishing characteristics of 3-week Sugen5416/hypoxia mice model? Cardiovasc Res 2023; 119:e140-e141. [PMID: 37183500 DOI: 10.1093/cvr/cvad074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 03/30/2023] [Indexed: 05/16/2023] Open
Affiliation(s)
- Ruixuan Zheng
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, 1 Fanhai West Road, Ouhai District, Wenzhou City, Zhejiang Province 325000, China
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 1 Fanhai West Road, Ouhai District, Wenzhou City, Zhejiang Province 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou, 1 Fanhai West Road, Ouhai District, Wenzhou City, Zhejiang Province 325000, China
| | - Tingting Xu
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, 1 Fanhai West Road, Ouhai District, Wenzhou City, Zhejiang Province 325000, China
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 1 Fanhai West Road, Ouhai District, Wenzhou City, Zhejiang Province 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou, 1 Fanhai West Road, Ouhai District, Wenzhou City, Zhejiang Province 325000, China
| | - Ya Lin
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, 1 Fanhai West Road, Ouhai District, Wenzhou City, Zhejiang Province 325000, China
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 1 Fanhai West Road, Ouhai District, Wenzhou City, Zhejiang Province 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou, 1 Fanhai West Road, Ouhai District, Wenzhou City, Zhejiang Province 325000, China
| | - Xiaoying Huang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, 1 Fanhai West Road, Ouhai District, Wenzhou City, Zhejiang Province 325000, China
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 1 Fanhai West Road, Ouhai District, Wenzhou City, Zhejiang Province 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou, 1 Fanhai West Road, Ouhai District, Wenzhou City, Zhejiang Province 325000, China
| |
Collapse
|
7
|
Medina-Rodriguez EM, Han D, Lowell J, Beurel E. Stress promotes the infiltration of peripheral immune cells to the brain. Brain Behav Immun 2023; 111:412-423. [PMID: 37169132 PMCID: PMC10349920 DOI: 10.1016/j.bbi.2023.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 04/11/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023] Open
Abstract
Immune cells and the brain have a privileged interaction. Here, we report changes in the hippocampal immune microenvironment at the single cell level after stress, uncovering the tight orchestration of immune cell infiltration into the hippocampus after stress to maintain homeostasis. We show the distribution of several immune cell types in the hippocampus associated with their susceptibility or resilience to the learned helplessness paradigm in a sex- and microbiota-dependent manner using single-cell RNA sequencing and bioinformatic tools, flow cytometry, and immunofluorescence. We uncovered the presence of tissue-resident memory T cells that accumulate over time in the hippocampus of learned helpless mice, and the presence of CD74-expressing myeloid cells. These cells were found by a knockdown approach to be critical to induce resilience to learned helplessness. Altogether, these findings provide a novel overview of the neuro-immune repertoire and its impact on the landscape of the hippocampus after learned helplessness.
Collapse
Affiliation(s)
| | - Dongmei Han
- Department of Psychiatry and Behavioral Sciences, United States
| | - Jeffrey Lowell
- Department of Psychiatry and Behavioral Sciences, United States
| | - Eléonore Beurel
- Department of Psychiatry and Behavioral Sciences, United States; Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States.
| |
Collapse
|
8
|
Shu W, Wang Y, Li C, Zhang L, Zhuoma D, Yang P, Yan G, Chen C, Ba Y, Du P, Wang X. Single-cell Expression Atlas Reveals Cell Heterogeneity in the Creeping Fat of Crohn's Disease. Inflamm Bowel Dis 2023; 29:850-865. [PMID: 36715181 DOI: 10.1093/ibd/izac266] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Creeping fat (CrF) has been recognized to play a positive role in Crohn's disease (CD) progression, yet the cellular compositions within mesenteric adipose tissue (MAT) and their potential mechanism in CrF formation are poorly understood. METHODS Analysis of 10X single-cell RNA sequencing was performed on 67 064 cells from 3 pairs of surgically resected samples of CrF and their uninvolved MAT. The results were validated in another cohort with 6 paired MAT samples by immunofluorescence. RESULTS All samples manifested excellent consistency and repeatability in our study, and 10 cell types from the transcriptome atlas, including 20 clusters, were identified. In CrF, a specific vascular endothelial cell subpopulation highly expressing lipoprotein lipase was first identified, with a significantly increased proportion. This vascular endothelial cell subpopulation manifested robust peroxisome proliferator-activated receptor γ (PPARγ) transcription activity and an upregulated PPAR signaling pathway and was involved in lipid metabolism and the antibacterial response. A novel fibroblast subpopulation (FC3) with remarkable GREM1 and RFLNB expression was identified and validated to predominantly accumulate in the CrF. The FC3 was annotated as inflammation-associated fibroblasts, which are characterized by inflammatory responses and the regulation of Smad phosphorylation related to intestinal fibrosis. The trajectory of fibroblasts revealed their pro-inflammatory and profibrotic conversion tendency during CrF formation with corresponding gene dynamics. Additionally, we unprecedently dissected the different origins and functions of 6 macrophage subclusters within the myeloid compartment. CONCLUSIONS Our results uncover the cellular heterogeneity in the MAT of CD and the role of these various cellular compositions in CrF development. This comprehensive understanding of CrF provides future directions for in-depth research on and potential targets for MAT-based treatment.
Collapse
Affiliation(s)
- Weigang Shu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yongheng Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Chuanding Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Lei Zhang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Deji Zhuoma
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Pengyu Yang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Guorong Yan
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Chunqiu Chen
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yongbing Ba
- OE Biotech Co., Ltd., Shanghai 201114, China
| | - Peng Du
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200092, China
| | - Xiaolei Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
9
|
Geyer J, Krupa KA, Harris ZM, Sun Y, Sharma L, Würstle S, Hu B, Stanley G, Rajagopalan G, Pellot E, Koff JL, Robinson JB. A Novel Zinc (II) Porphyrin Is Synergistic with PEV2 Bacteriophage against Pseudomonas aeruginosa Infections. Antibiotics (Basel) 2023; 12:735. [PMID: 37107097 PMCID: PMC10135120 DOI: 10.3390/antibiotics12040735] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Pseudomonas aeruginosa (PsA) is an opportunistic bacterial pathogen that causes life-threatening infections in individuals with compromised immune systems and exacerbates health concerns for those with cystic fibrosis (CF). PsA rapidly develops antibiotic resistance; thus, novel therapeutics are urgently needed to effectively combat this pathogen. Previously, we have shown that a novel cationic Zinc (II) porphyrin (ZnPor) has potent bactericidal activity against planktonic and biofilm-associated PsA cells, and disassembles the biofilm matrix via interactions with eDNA In the present study, we report that ZnPor caused a significant decrease in PsA populations in mouse lungs within an in vivo model of PsA pulmonary infection. Additionally, when combined with an obligately lytic phage PEV2, ZnPor at its minimum inhibitory concentration (MIC) displayed synergy against PsA in an established in vitro lung model resulting in greater protection of H441 lung cells versus either treatment alone. Concentrations above the minimum bactericidal concentration (MBC) of ZnPor were not toxic to H441 cells; however, no synergy was observed. This dose-dependent response is likely due to ZnPor's antiviral activity, reported herein. Together, these findings show the utility of ZnPor alone, and its synergy with PEV2, which could be a tunable combination used in the treatment of antibiotic-resistant infections.
Collapse
Affiliation(s)
- Jessica Geyer
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
| | - Kristen A. Krupa
- Department of Chemical and Materials Engineering, University of Dayton, Dayton, OH 45469, USA
- Integrated Science and Engineering Center, University of Dayton, Dayton, OH 45469, USA
| | - Zachary M. Harris
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ying Sun
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Silvia Würstle
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
- School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Buqu Hu
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gail Stanley
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Govindarajan Rajagopalan
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Erin Pellot
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
| | - Jonathan L. Koff
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jayne B. Robinson
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
- Integrated Science and Engineering Center, University of Dayton, Dayton, OH 45469, USA
| |
Collapse
|
10
|
Cao L, Wang X, Liu X, Meng W, Guo W, Duan C, Liang X, Kang L, Lv P, Lin Q, Zhang R, Zhang X, Shen H. Tumor Necrosis Factor α-Dependent Lung Inflammation Promotes the Progression of Lung Adenocarcinoma Originating From Alveolar Type II Cells by Upregulating MIF-CD74. J Transl Med 2023; 103:100034. [PMID: 36925198 DOI: 10.1016/j.labinv.2022.100034] [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: 05/26/2022] [Revised: 08/28/2022] [Accepted: 10/25/2022] [Indexed: 01/11/2023] Open
Abstract
Lung adenocarcinoma is the most common type of lung cancer. We recently reported that inflammation-driven lung adenocarcinoma (IDLA) originates from alveolar type (AT)-II cells, which depend on major histocompatibility complex (MHC) class II to promote the expansion of regulatory T cells. The MHC class II-associated invariant chain (CD74) binds to the macrophage migration inhibitory factor (MIF), which is associated with promoting tumor growth and invasion. However, the role of MIF-CD74 in the progression of lung adenocarcinoma and the underlying mechanisms remain unclear. We aimed to explore the role of MIF-CD74 in the progression of lung adenocarcinoma and elucidate the mechanisms by which tumor necrosis (TNF)-α-mediated inflammation regulates CD74 and MIF expression in IDLA. In human lung adenocarcinoma, CD74 was upregulated on the surface of tumor cells originating from AT-II cells, which correlated positively with lymph node metastasis, tumor origin/nodal involvement/metastasis stage, and TNF-α expression. MIF interaction with CD74 promoted the proliferation and migration of A549 and H1299 cells in vitro. Using a urethane-induced IDLA mouse model, we observed that CD74 was upregulated in tumor cells and macrophages. MIF expression was upregulated in macrophages in IDLA. Blocking TNF-α-dependent inflammation downregulated CD74 expression in tumor cells and CD74 and MIF expression in macrophages in IDLA. Conditioned medium from A549 cells or activated mouse AT-II cells upregulated MIF in macrophages by secreting TNF-α. TNF-α-dependent lung inflammation contributes to the progression of lung adenocarcinoma by upregulating CD74 and MIF expression, and AT-II cells upregulate MIF expression in macrophages by secreting TNF-α. This study provides novel insights into the function of CD74 in the progression of IDLA.
Collapse
Affiliation(s)
- Lei Cao
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, Hebei Province, China; The Third Department of Geriatrics, Hebei General Hospital, Shijiazhuang, Hebei Province, China
| | - Xiuqing Wang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xiaoyi Liu
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Wei Meng
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Wenli Guo
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Chenyang Duan
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xiaoyan Liang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Lifei Kang
- Department of Pathology, Hebei Chest Hospital, Shijiazhuang, Hebei Province, China
| | - Ping Lv
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Qiang Lin
- Department of Oncology, North China Petroleum Bureau General Hospital of Hebei Medical University, Renqiu, Hebei Province, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Xianghong Zhang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, Hebei Province, China.
| | - Haitao Shen
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, Hebei Province, China.
| |
Collapse
|
11
|
Li QL, Tang J, Zhao L, Ruze A, Shan XF, Gao XM. The role of CD74 in cardiovascular disease. Front Cardiovasc Med 2023; 9:1049143. [PMID: 36712241 PMCID: PMC9877307 DOI: 10.3389/fcvm.2022.1049143] [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: 09/20/2022] [Accepted: 12/21/2022] [Indexed: 01/15/2023] Open
Abstract
Leukocyte differentiation antigen 74 (CD74), also known as invariant chain, is a molecular chaperone of major histocompatibility complex class II (MHC II) molecules involved in antigen presentation. CD74 has recently been shown to be a receptor for the macrophage migration inhibitory factor family proteins (MIF/MIF2). Many studies have revealed that CD74 plays an important role in cardiovascular disease. In this review, we summarize the structure and main functions of CD74 and then focus on the recent research progress on the role of CD74 in cardiovascular diseases. In addition, we also discuss potential treatment strategies that target CD74. Our systematic review of the role of CD74 in cardiovascular disease will fill some knowledge gaps in the field.
Collapse
Affiliation(s)
- Qiu-Lin Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China
| | - Jing Tang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China,Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Ling Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China
| | - Amanguli Ruze
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China
| | - Xue-Feng Shan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China,Clinical Medical Research Institute of Xinjiang Medical University, Ürümqi, China,*Correspondence: Xiao-Ming Gao,
| |
Collapse
|
12
|
Harris ZM, Sun Y, Joerns J, Clark B, Hu B, Korde A, Sharma L, Shin HJ, Manning EP, Placek L, Unutmaz D, Stanley G, Chun H, Sauler M, Rajagopalan G, Zhang X, Kang MJ, Koff JL. Epidermal Growth Factor Receptor Inhibition Is Protective in Hyperoxia-Induced Lung Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9518592. [PMID: 36193076 PMCID: PMC9526641 DOI: 10.1155/2022/9518592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 01/01/2023]
Abstract
Aims Studies have linked severe hyperoxia, or prolonged exposure to very high oxygen levels, with worse clinical outcomes. This study investigated the role of epidermal growth factor receptor (EGFR) in hyperoxia-induced lung injury at very high oxygen levels (>95%). Results Effects of severe hyperoxia (100% oxygen) were studied in mice with genetically inhibited EGFR and wild-type littermates. Despite the established role of EGFR in lung repair, EGFR inhibition led to improved survival and reduced acute lung injury, which prompted an investigation into this protective mechanism. Endothelial EGFR genetic knockout did not confer protection. EGFR inhibition led to decreased levels of cleaved caspase-3 and poly (ADP-ribosyl) polymerase (PARP) and decreased terminal dUTP nick end labeling- (TUNEL-) positive staining in alveolar epithelial cells and reduced ERK activation, which suggested reduced apoptosis in vivo. EGFR inhibition decreased hyperoxia (95%)-induced apoptosis and ERK in murine alveolar epithelial cells in vitro, and CRISPR-mediated EGFR deletion reduced hyperoxia-induced apoptosis and ERK in human alveolar epithelial cells in vitro. Innovation. This work defines a protective role of EGFR inhibition to decrease apoptosis in lung injury induced by 100% oxygen. This further characterizes the complex role of EGFR in acute lung injury and outlines a novel hyperoxia-induced cell death pathway that warrants further study. Conclusion In conditions of severe hyperoxia (>95% for >24 h), EGFR inhibition led to improved survival, decreased lung injury, and reduced cell death. These findings further elucidate the complex role of EGFR in acute lung injury.
Collapse
Affiliation(s)
- Zachary M. Harris
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Ying Sun
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - John Joerns
- Division of Pulmonary and Critical Care; Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA 75390
| | - Brian Clark
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Buqu Hu
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Asawari Korde
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Hyeon Jun Shin
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Edward P. Manning
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Lindsey Placek
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | - Derya Unutmaz
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | - Gail Stanley
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Hyung Chun
- Section of Cardiovascular Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Maor Sauler
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Govindarajan Rajagopalan
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Xuchen Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Min-Jong Kang
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Jonathan L. Koff
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| |
Collapse
|
13
|
Li Q, Jiang Y, Song N, Zhou B, Li Z, Lin L. An Immune-Related Genetic Feature Depicted the Heterogeneous Nature of Lung Adenocarcinoma and Squamous Cell Carcinoma and Their Distinctive Predicted Drug Responses. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8447083. [PMID: 36071867 PMCID: PMC9442502 DOI: 10.1155/2022/8447083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022]
Abstract
One of the primary causes of global cancer-associated mortality is lung cancer (LC). Current improvements in the management of LC rely mainly on the advancement of patient stratification, both molecularly and clinically, to achieve the maximal therapeutic benefit, while most LC screening protocols remain underdeveloped. In this research, we first employed two algorithms (ESTIMATE and xCell) to calculate the immune/stromal infiltration scores. This helped identify the altered immune infiltration landscapes in lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC). Afterward, based on their immune-related characteristics, we successfully stratified the LUAD and LUSC into 2 and 3 clusters, respectively. Different from the conventional bioinformatic approaches that start from the investigation of differential expression of single genes, differentially enriched curated gene sets identified through gene set variation analyses (GSVA) were curated, and gene names were reconstructed afterward. Furthermore, weighted gene correlation network analyses (WGCNA) were used to reveal hub genes highly connected with the clustering process. Actual expression levels of critical hub genes among different clusters were compared and so were the functional pathways these genes enriched into. Lastly, a computational method was applied to predict and compare the responses of each cluster to primary therapeutic agents. The heterogeneity presented in our study, along with the drug responses expected for identified clusters, may shed light on future exploration of combination immunochemotherapy that facilitates the optimization of individualized therapy.
Collapse
Affiliation(s)
- Qiuyuan Li
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, No. 507 Zhengmin Rd., Shanghai, China
| | - Yan Jiang
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, No. 507 Zhengmin Rd., Shanghai, China
| | - Nan Song
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, No. 507 Zhengmin Rd., Shanghai, China
| | - Bin Zhou
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, No. 507 Zhengmin Rd., Shanghai, China
| | - Zhao Li
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, No. 507 Zhengmin Rd., Shanghai, China
| | - Lei Lin
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, No. 507 Zhengmin Rd., Shanghai, China
| |
Collapse
|
14
|
Identification of Hub Genes and Immune Infiltration in Pediatric Biliary Atresia by Comprehensive Bioinformatics Analysis. CHILDREN 2022; 9:children9050697. [PMID: 35626874 PMCID: PMC9140130 DOI: 10.3390/children9050697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/21/2022] [Accepted: 05/06/2022] [Indexed: 12/04/2022]
Abstract
Background: Biliary atresia (BA) is the leading cause of pediatric liver failure and pediatric liver transplantation worldwide. Evidence suggests that the immune system plays a central role in the pathogenesis of BA. Methods: In this work, the novel immune-related genes between BA and normal samples were investigated based on weighted gene co-expression network analysis (WGCNA) and the deconvolution algorithm of CIBERSORT. Results: Specifically, 650 DEGs were identified between the BA and normal groups. The blue module was the most positively correlated with BA containing 3274 genes. Totally, 610 overlapping BA-related genes of DEGs and WGCNA were further used to identify IRGs. Three IRGs including VCAM1, HLA-DRA, and CD74 were finally identified as the candidate biomarkers. Particularly, the CD74 biomarker was discovered for the first as a potential immune biomarker for BA. Conclusions: Possibly, these 3 IRGs might serve as candidate biomarkers and guide the individualized treatment strategies for BA patients. Our results would provide great insights for a deeper understanding of both the occurrence and the treatment of BA.
Collapse
|
15
|
Sidramagowda Patil S, Soundararajan R, Fukumoto J, Breitzig M, Hernández-Cuervo H, Alleyn M, Lin M, Narala VR, Lockey R, Kolliputi N, Galam L. Mitochondrial Protein Akap1 Deletion Exacerbates Endoplasmic Reticulum Stress in Mice Exposed to Hyperoxia. Front Pharmacol 2022; 13:762840. [PMID: 35370705 PMCID: PMC8964370 DOI: 10.3389/fphar.2022.762840] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 01/27/2022] [Indexed: 01/02/2023] Open
Abstract
Acute lung injury (ALI) and its severe manifestation, acute respiratory distress syndrome (ARDS), are treated with high concentrations of supplementary oxygen. However, prolonged exposure to high oxygen concentrations stimulates the production of reactive oxygen species (ROS), which damages the mitochondria and accumulates misfolded proteins in the endoplasmic reticulum (ER). The mitochondrial protein A-kinase anchoring protein 1 (Akap1) is critical for mitochondrial homeostasis. It is known that Akap1 deficiency results in heart damage, neuronal development impairment, and mitochondrial malfunction in preclinical studies. Our laboratory recently revealed that deleting Akap1 increases the severity of hyperoxia-induced ALI in mice. To assess the role of Akap1 deletion in ER stress in lung injury, wild-type and Akap1−/− mice were exposed to hyperoxia for 48 h. This study indicates that Akap1−/− mice exposed to hyperoxia undergo ER stress, which is associated with an increased expression of BiP, JNK phosphorylation, eIF2α phosphorylation, ER stress-induced cell death, and autophagy. This work demonstrates that deleting Akap1 results in increased ER stress in the lungs of mice and that hyperoxia exacerbates ER stress-related consequences.
Collapse
Affiliation(s)
- Sahebgowda Sidramagowda Patil
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Ramani Soundararajan
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Jutaro Fukumoto
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Mason Breitzig
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States.,Washington University in St. Louis, Brown School, St. Louis, MO, United States
| | - Helena Hernández-Cuervo
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States.,University of South Florida, Department of Molecular Medicine, College of Medicine, Tampa, FL, United States
| | - Matthew Alleyn
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Muling Lin
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | | | - Richard Lockey
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Narasaiah Kolliputi
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States.,University of South Florida, Department of Molecular Medicine, College of Medicine, Tampa, FL, United States
| | - Lakshmi Galam
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| |
Collapse
|
16
|
Harrison DE, Strong R, Reifsnyder P, Kumar N, Fernandez E, Flurkey K, Javors MA, Lopez‐Cruzan M, Macchiarini F, Nelson JF, Markewych A, Bitto A, Sindler AL, Cortopassi G, Kavanagh K, Leng L, Bucala R, Rosenthal N, Salmon A, Stearns TM, Bogue M, Miller RA. 17-a-estradiol late in life extends lifespan in aging UM-HET3 male mice; nicotinamide riboside and three other drugs do not affect lifespan in either sex. Aging Cell 2021; 20:e13328. [PMID: 33788371 PMCID: PMC8135004 DOI: 10.1111/acel.13328] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/27/2020] [Accepted: 01/12/2021] [Indexed: 01/23/2023] Open
Abstract
In genetically heterogeneous mice produced by the CByB6F1 x C3D2F1 cross, the "non-feminizing" estrogen, 17-α-estradiol (17aE2), extended median male lifespan by 19% (p < 0.0001, log-rank test) and 11% (p = 0.007) when fed at 14.4 ppm starting at 16 and 20 months, respectively. 90th percentile lifespans were extended 7% (p = 0.004, Wang-Allison test) and 5% (p = 0.17). Body weights were reduced about 20% after starting the 17aE2 diets. Four other interventions were tested in males and females: nicotinamide riboside, candesartan cilexetil, geranylgeranylacetone, and MIF098. Despite some data suggesting that nicotinamide riboside would be effective, neither it nor the other three increased lifespans significantly at the doses tested. The 17aE2 results confirm and extend our original reports, with very similar results when started at 16 months compared with mice started at 10 months of age in a prior study. The consistently large lifespan benefit in males, even when treatment is started late in life, may provide information on sex-specific aspects of aging.
Collapse
Affiliation(s)
| | - Randy Strong
- Barshop Institute for Longevity and Aging StudiesThe University of Texas Health Science CenterSan AntonioTXUSA
- Geriatric ResearchEducation and Clinical CenterSan AntonioTXUSA
- Research ServiceSouth Texas Veterans Health Care SystemSan AntonioTXUSA
- Department of PharmacologyThe University of Texas Health Science CenterSan AntonioTXUSA
| | | | - Navasuja Kumar
- Department of Pathology and Geriatrics CenterUniversity of MichiganAnn ArborMIUSA
| | - Elizabeth Fernandez
- Barshop Institute for Longevity and Aging StudiesThe University of Texas Health Science CenterSan AntonioTXUSA
- Geriatric ResearchEducation and Clinical CenterSan AntonioTXUSA
- Department of PharmacologyThe University of Texas Health Science CenterSan AntonioTXUSA
| | | | - Martin A. Javors
- Department of PsychiatryThe University of Texas Health Science CenterSan AntonioTXUSA
| | - Marisa Lopez‐Cruzan
- Department of PsychiatryThe University of Texas Health Science CenterSan AntonioTXUSA
| | | | - James F. Nelson
- Barshop Institute for Longevity and Aging StudiesThe University of Texas Health Science CenterSan AntonioTXUSA
- Department of PhysiologyThe University of Texas Health Science CenterSan AntonioTXUSA
| | - Adrian Markewych
- Department of PathologyUniversity of Washington Medical CenterSeattleWAUSA
| | - Alessandro Bitto
- Department of PathologyUniversity of Washington Medical CenterSeattleWAUSA
| | - Amy L. Sindler
- Department of Health & Human Physiology and Department of BiochemistryUniversity of IowaIowa CityIAUSA
| | - Gino Cortopassi
- Department of Molecular BiosciencesUniversity of CaliforniaDavisCAUSA
| | - Kylie Kavanagh
- Department of Pathology and Internal MedicineWake Forest School of MedicineWinston‐SalemNCUSA
| | - Lin Leng
- Department of Internal MedicineYale UniversityNew HavenConnecticutUSA
| | - Richard Bucala
- Department of Internal MedicineYale UniversityNew HavenConnecticutUSA
| | | | - Adam Salmon
- Barshop Institute for Longevity and Aging StudiesThe University of Texas Health Science CenterSan AntonioTXUSA
- Geriatric ResearchEducation and Clinical CenterSan AntonioTXUSA
- Research ServiceSouth Texas Veterans Health Care SystemSan AntonioTXUSA
- Department of Molecular MedicineThe University of Texas Health Science CenterSan AntonioTXUSA
| | | | | | - Richard A. Miller
- Department of Pathology and Geriatrics CenterUniversity of MichiganAnn ArborMIUSA
| |
Collapse
|
17
|
Sidramagowda Patil S, Hernández-Cuervo H, Fukumoto J, Krishnamurthy S, Lin M, Alleyn M, Breitzig M, Narala VR, Soundararajan R, Lockey RF, Kolliputi N, Galam L. Alda-1 Attenuates Hyperoxia-Induced Acute Lung Injury in Mice. Front Pharmacol 2021; 11:597942. [PMID: 33597876 PMCID: PMC7883597 DOI: 10.3389/fphar.2020.597942] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 10/19/2020] [Indexed: 12/31/2022] Open
Abstract
Acute lung injury (ALI), a milder form of acute respiratory distress syndrome (ARDS), is a leading cause of mortality in older adults with an increasing prevalence. Oxygen therapy, is a common treatment for ALI, involving exposure to a high concentration of oxygen. Unfortunately, hyperoxia induces the formation of reactive oxygen species which can cause an increase in 4-HNE (4-hydroxy 2 nonenal), a toxic byproduct of lipid peroxidation. Mitochondrial aldehyde dehydrogenase 2 (ALDH2) serves as an endogenous shield against oxidative stress-mediated damage by clearing 4-HNE. Alda-1 [(N-(1, 3 benzodioxol-5-ylmethyl)-2, 6- dichloro-benzamide)], a small molecular activator of ALDH2, protects against reactive oxygen species-mediated oxidative stress by promoting ALDH2 activity. As a result, Alda-1 shields against ischemic reperfusion injury, heart failure, stroke, and myocardial infarction. However, the mechanisms of Alda-1 in hyperoxia-induced ALI remains unclear. C57BL/6 mice implanted with Alzet pumps received Alda-1 in a sustained fashion while being exposed to hyperoxia for 48 h. The mice displayed suppressed immune cell infiltration, decreased protein leakage and alveolar permeability compared to controls. Mechanistic analysis shows that mice pretreated with Alda-1 also experience decreased oxidative stress and enhanced levels of p-Akt and mTOR pathway associated proteins. These results show that continuous delivery of Alda-1 protects against hyperoxia-induced lung injury in mice.
Collapse
Affiliation(s)
- Sahebgowda Sidramagowda Patil
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Helena Hernández-Cuervo
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jutaro Fukumoto
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sudarshan Krishnamurthy
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Muling Lin
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Matthew Alleyn
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Mason Breitzig
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Brown School, Washington University, St. Louis, MO, United States
| | | | - Ramani Soundararajan
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Richard F Lockey
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Lakshmi Galam
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
18
|
Li R, Wang F, Wei J, Lin Y, Tang G, Rao L, Ma L, Xu Q, Wu J, Lv Q, Zhou R, Lei H, Zhao X, Yao D, Xiao B, Huang H, Zhang J, Mo B. The Role of Macrophage Migration Inhibitory Factor (MIF) in Asthmatic Airway Remodeling. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2021; 13:88-105. [PMID: 33191679 PMCID: PMC7680835 DOI: 10.4168/aair.2021.13.1.88] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/10/2020] [Accepted: 05/17/2020] [Indexed: 12/23/2022]
Abstract
Purpose Recent studies have demonstrated that macrophage migration inhibitory factor (MIF) is of importance in asthmatic inflammation. The role of MIF in modulating airway remodeling has not yet been thoroughly elucidated to date. In the present study, we hypothesized that MIF promoted airway remodeling by intensifying airway smooth muscle cell (ASMC) autophagy and explored the specific mechanisms. Methods MIF knockdown in the lung tissues of C57BL/6 mice was conducted by instilling intratracheally adeno-associated virus (AAV) vectors (MIF-mutant AAV9) into mouse lung tissues. Mice genetically deficient in the autophagy marker ATG5 (ATG5+/−) was used to detect the role of autophagy in ovalbumin (OVA)-asthmatic murine models. Moreover, to block the expression of MIF and CD74 in vitro models, inhibitors, antibodies and lentivirus transfection techniques were employed. Results First, MIF knockdown in the lung tissues of mice showed markedly reduced airway remodeling in OVA murine mice models. Secondly, ASMC autophagy was increased in the OVA-challenged models. Mice genetically deficient in the autophagy marker ATG5 (ATG5+/−) that were primed and challenged with OVA showed lower airway remodeling than genetically wild-type asthmatic mice. Thirdly, MIF can induce ASMC autophagy in vitro. Moreover, the cellular source of MIF which promoted ASMC autophagy was macrophages. Finally, MIF promoted ASMC autophagy in a CD74-dependent manner. Conclusions MIF can increase asthmatic airway remodeling by enhancing ASMC autophagy. Macrophage-derived MIF can promote ASMC autophagy by targeting CD74.
Collapse
Affiliation(s)
- Ruyi Li
- Department of Respiratory and Critical Care Medicine, Key Laboratory of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Feiyun Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianghong Wei
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yun Lin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Guofang Tang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Lizong Rao
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Libing Ma
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Qing Xu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jingjie Wu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Qian Lv
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Rui Zhou
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Changsha, China
| | - Huiren Lei
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Xueqiang Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Dong Yao
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Bo Xiao
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Haiming Huang
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Jiange Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Biwen Mo
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China.
| |
Collapse
|
19
|
Huidobro C, Martín-Vicente P, López-Martínez C, Alonso-López I, Amado-Rodríguez L, Crespo I, M Albaiceta G. Cellular and molecular features of senescence in acute lung injury. Mech Ageing Dev 2020; 193:111410. [PMID: 33249191 DOI: 10.1016/j.mad.2020.111410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 11/20/2020] [Indexed: 12/14/2022]
Abstract
A wide range of insults can trigger acute injury in the lungs, which eventually may lead to respiratory failure and death of patients. Current treatment relies mainly on supportive measures and mechanical ventilation. Even so, survivors frequently develop important sequels that compromise quality of life. In the search for new approaches to prevent and treat acute lung injury, many investigations have focused on molecular and cellular pathways which could exert a pathogenic role in this disease. Herein, we review recent findings in the literature suggesting that cellular senescence could be involved in lung injury and discuss the potential use of senotherapies to prevent disease progression.
Collapse
Affiliation(s)
- Covadonga Huidobro
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain.
| | - Paula Martín-Vicente
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Instituto Universitario de Oncología del Principado de Asturias, Calle Fernando Bongera s/n, 33006, Oviedo, Spain
| | - Cecilia López-Martínez
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Instituto Universitario de Oncología del Principado de Asturias, Calle Fernando Bongera s/n, 33006, Oviedo, Spain
| | - Inés Alonso-López
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Instituto Universitario de Oncología del Principado de Asturias, Calle Fernando Bongera s/n, 33006, Oviedo, Spain
| | - Laura Amado-Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Avenida de Roma s/n, 33011, Oviedo, Spain
| | - Irene Crespo
- Departamento de Biología Funcional. Universidad de Oviedo, C/ Julián Clavería s/n, 33006, Oviedo, Spain
| | - Guillermo M Albaiceta
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Instituto Universitario de Oncología del Principado de Asturias, Calle Fernando Bongera s/n, 33006, Oviedo, Spain; Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Avenida de Roma s/n, 33011, Oviedo, Spain; Departamento de Biología Funcional. Universidad de Oviedo, C/ Julián Clavería s/n, 33006, Oviedo, Spain
| |
Collapse
|
20
|
Farr L, Ghosh S, Moonah S. Role of MIF Cytokine/CD74 Receptor Pathway in Protecting Against Injury and Promoting Repair. Front Immunol 2020; 11:1273. [PMID: 32655566 PMCID: PMC7325688 DOI: 10.3389/fimmu.2020.01273] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
Wound healing after an injury is essential for life. An in-depth understanding of the healing process is necessary to ultimately improve the currently limited treatment options for patients suffering as a result of damage to various organs and tissues. Injuries, even the most minor, trigger an inflammatory response that protects the host and activates repair pathways. In recent years, substantial progress has been made in delineating the mechanisms by which inflammatory cytokines and their receptors facilitate tissue repair and regeneration. This mini review focuses on emerging literature on the role of the cytokine macrophage migration inhibitory factor (MIF) and its cell membrane receptor CD74, in protecting against injury and promoting healing in different parts of the body.
Collapse
Affiliation(s)
- Laura Farr
- Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Swagata Ghosh
- Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Shannon Moonah
- Department of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
21
|
Kim BS, Breuer B, Arnke K, Ruhl T, Hofer T, Simons D, Knobe M, Ganse B, Guidi M, Beier JP, Fuchs PC, Pallua N, Bernhagen J, Grieb G. The effect of the macrophage migration inhibitory factor (MIF) on excisional wound healing in vivo. J Plast Surg Hand Surg 2020; 54:137-144. [PMID: 32281469 DOI: 10.1080/2000656x.2019.1710710] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Background: The macrophage migration inhibitory factor (MIF) has been determined as a cytokine exerting a multitude of effects in inflammation and angiogenesis. Earlier studies have indicated that MIF may also be involved in wound healing and flap surgery. Methods: We investigated the effect of MIF in an excisional wound model in wildtype, Mif-/- and recombinant MIF treated mice. Wound closure rates as well as the macrophage marker Mac-3, the pro-inflammatory cytokine tumor necrosis factor α (TNFα) and the pro-angiogenic factor von Willebrand factor (vWF) were measured. Finally, we used a flap model in Mif-/- and WT mice with an established perfusion gradient to identify MIF's contribution in flap perfusion. Results: In the excision wound model, we found reduced wound healing after MIF injection, whereas Mif deletion improved wound healing. Furthermore, a reduced expression of Mac-3, TNFα and vWF in Mif-/- mice was seen when compared to WT mice. In the flap model, Mif-/- knockout mice showed mitigated flap perfusion with lower hemoglobin content and oxygen saturation as measured by O2C measurements when compared to WT mice. Conclusions: Our data suggest an inhibiting effect of MIF in wound healing with increased inflammation and perfusion. In flaps, by contrast, MIF may contribute to flap vascularization.
Collapse
Affiliation(s)
- Bong-Sung Kim
- Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, RWTH Aachen University Hospital, Aachen, Germany.,Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Benjamin Breuer
- Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, RWTH Aachen University Hospital, Aachen, Germany
| | - Kevin Arnke
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Tim Ruhl
- Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, RWTH Aachen University Hospital, Aachen, Germany
| | - Tanja Hofer
- Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, RWTH Aachen University Hospital, Aachen, Germany
| | - David Simons
- Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, RWTH Aachen University Hospital, Aachen, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Knobe
- Department of Orthopaedic Trauma, RWTH Aachen University Hospital, Aachen, Germany.,Division of Trauma Surgery, Kantonsspital Luzern, Luzern, Switzerland
| | - Bergita Ganse
- Department of Orthopaedic Trauma, RWTH Aachen University Hospital, Aachen, Germany
| | - Marco Guidi
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Justus P Beier
- Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, RWTH Aachen University Hospital, Aachen, Germany
| | - Paul C Fuchs
- Department of Plastic Surgery, Hand Surgery - Burn Center, Cologne-Merheim Medical Center, Witten/Herdecke University, Cologne, Germany
| | - Norbert Pallua
- Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, RWTH Aachen University Hospital, Aachen, Germany.,Aesthetic Elite International - Private Clinic, Düsseldorf, Germany
| | - Jürgen Bernhagen
- Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Gerrit Grieb
- Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, RWTH Aachen University Hospital, Aachen, Germany.,Department of Plastic Surgery and Hand Surgery, Gemeinschaftskrankenhaus Havelhoehe, Teaching Hospital of the Charité University, Berlin, Germany
| |
Collapse
|
22
|
Huang H, Chen D, Pu J, Yuan A, Fu Q, Li J, Leng L, Bucala R, Ye S, Lu L. The small molecule macrophage migration inhibitory factor antagonist MIF098, inhibits pulmonary hypertension associated with murine SLE. Int Immunopharmacol 2019; 76:105874. [PMID: 31499270 DOI: 10.1016/j.intimp.2019.105874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/31/2019] [Accepted: 09/01/2019] [Indexed: 01/11/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a severe complication of systemic lupus erythematosus (SLE), with unclear etiopathogenesis. We evaluated the role of macrophage migration inhibitory factor (MIF), which has been implicated in idiopathic pulmonary hypertension (PH), in SLE-associated PAH. Circulating MIF was measured in SLE patients, SLE-PAH patients, and healthy donors. In situ pulmonary artery MIF protein expression was determined in spontaneous SLE mice (MRL/lpr) and hypoxia-induced C57BL/6J mice. Daily MIF098 was administered to C57BL/6J mice, and these mice were maintained in a hypoxic chamber for 4 weeks. The right ventricular systolic pressure (RVSP) and pathological characteristics of the pulmonary artery (PA), such as hyperproliferation, muscularization, and fibrosis were then measured in each group of mice. Data were also obtained in vitro using pulmonary smooth muscle cells (PASMC) challenged with platelet-derived growth factor (PDGF)-BB or 1% O2 hypoxia. As a result, circulating MIF was elevated in SLE-PAH patients compared with SLE patients or healthy donors. Higher RVSP SLE mice produced more MIF protein than lower RVSP SLE mice in the pulmonary artery. MIF098 decreased RVSP and inhibited distal pulmonary artery hyperproliferation, muscularization, and collagen deposition in hypoxia challenged mice. In addition, MIF098 inhibited PASMC proliferation and migration by regulating mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 (MAPK/ERK1/2) signal- and cell-cycle-related proteins. MIF098 also reduced collagen synthesis by inhibiting the TGFβ1/Smad2/Smad3 pathway in cell-based experiments. In conclusion, MIF may serve as a biomarker and a therapeutic target of SLE-associated PAH. Pharmacologic MIF antagonism may be an effective means to ameliorate SLE-PAH.
Collapse
Affiliation(s)
- Huijing Huang
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dandan Chen
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ancai Yuan
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiong Fu
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jia Li
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lin Leng
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Richard Bucala
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Shuang Ye
- Department of Rheumatology, Ren Ji Hospital South Campus, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Liangjing Lu
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
23
|
Tilstam PV, Pantouris G, Corman M, Andreoli M, Mahboubi K, Davis G, Du X, Leng L, Lolis E, Bucala R. A selective small-molecule inhibitor of macrophage migration inhibitory factor-2 (MIF-2), a MIF cytokine superfamily member, inhibits MIF-2 biological activity. J Biol Chem 2019; 294:18522-18531. [PMID: 31578280 DOI: 10.1074/jbc.ra119.009860] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/06/2019] [Indexed: 12/13/2022] Open
Abstract
Cytokine macrophage migration inhibitory factor-2 (MIF-2 or D-dopachrome tautomerase) is a recently characterized second member of the MIF cytokine superfamily in mammalian genomes. MIF-2 shares pro-inflammatory and tumorigenic properties with the clinical target MIF (MIF-1), but the precise contribution of MIF-2 to immune physiology or pathology is unclear. Like MIF-1, MIF-2 has intrinsic keto-enol tautomerase activity and mediates biological functions by engaging the cognate, common MIF family receptor CD74. Evidence that the catalytic site of MIF family cytokines has a structural role in receptor binding has prompted exploration of tautomerase inhibitors as potential biological antagonists and therapeutic agents, although few catalytic inhibitors inhibit receptor activation. Here we describe the discovery and biochemical characterization of a selective small-molecule inhibitor of MIF-2. An in silico screen of 1.6 million compounds targeting the MIF-2 tautomerase site yielded several hits for potential catalytic inhibitors of MIF-2 and identified 4-(3-carboxyphenyl)-2,5-pyridinedicarboxylic acid (4-CPPC) as the most functionally potent compound. We found that 4-CPPC has an enzymatic IC50 of 27 μm and 17-fold selectivity for MIF-2 versus MIF-1. An in vitro binding assay for MIF-1/MIF-2 to the CD74 ectodomain (sCD74) indicated that 4-CPPC inhibits MIF-2-CD74 binding in a dose-dependent manner (0.01-10 μm) without influencing MIF-1-CD74 binding. Notably, 4-CPPC inhibited MIF-2-mediated activation of CD74 and reduced CD74-dependent signal transduction. These results open opportunities for development of more potent and pharmacologically auspicious MIF-2 inhibitors to investigate the distinct functions of this MIF family member in vivo.
Collapse
Affiliation(s)
| | - Georgios Pantouris
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut 06510
| | - Michael Corman
- The Institutes for Pharmaceutical Discovery, LLC, Branford, Connecticut 06405
| | - Monica Andreoli
- The Institutes for Pharmaceutical Discovery, LLC, Branford, Connecticut 06405
| | - Keyvan Mahboubi
- The Institutes for Pharmaceutical Discovery, LLC, Branford, Connecticut 06405
| | - Gary Davis
- The Institutes for Pharmaceutical Discovery, LLC, Branford, Connecticut 06405
| | - Xin Du
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut 06510
| | - Lin Leng
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut 06510
| | - Elias Lolis
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut 06510; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut 06510
| | - Richard Bucala
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut 06510; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut 06510.
| |
Collapse
|
24
|
Stoppe C, Averdunk L, Goetzenich A, Soppert J, Marlier A, Kraemer S, Vieten J, Coburn M, Kowark A, Kim BS, Marx G, Rex S, Ochi A, Leng L, Moeckel G, Linkermann A, El Bounkari O, Zarbock A, Bernhagen J, Djudjaj S, Bucala R, Boor P. The protective role of macrophage migration inhibitory factor in acute kidney injury after cardiac surgery. Sci Transl Med 2019; 10:10/441/eaan4886. [PMID: 29769287 DOI: 10.1126/scitranslmed.aan4886] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 12/22/2017] [Accepted: 04/13/2018] [Indexed: 12/20/2022]
Abstract
Acute kidney injury (AKI) represents the most frequent complication after cardiac surgery. Macrophage migration inhibitory factor (MIF) is a stress-regulating cytokine that was shown to protect the heart from myocardial ischemia-reperfusion injury, but its role in the pathogenesis of AKI remains unknown. In an observational study, serum and urinary MIF was quantified in 60 patients scheduled for elective conventional cardiac surgery with the use of cardiopulmonary bypass. Cardiac surgery triggered an increase in MIF serum concentrations, and patients with high circulating MIF (>median) 12 hours after surgery had a significantly reduced risk of developing AKI (relative risk reduction, 72.7%; 95% confidence interval, 12 to 91.5%; P = 0.03). Experimental AKI was induced in wild-type and Mif-/- mice by 30 min of ischemia followed by 6 or 24 hours of reperfusion, or by rhabdomyolysis. Mif-deficient mice exhibited increased tubular cell injury, increased regulated cell death (necroptosis and ferroptosis), and enhanced oxidative stress. Therapeutic administration of recombinant MIF after ischemia-reperfusion in mice ameliorated AKI. In vitro treatment of tubular epithelial cells with recombinant MIF reduced cell death and oxidative stress as measured by glutathione and thiobarbituric acid reactive substances in the setting of hypoxia. Our data provide evidence of a renoprotective role of MIF in experimental ischemia-reperfusion injury by protecting renal tubular epithelial cells, consistent with our observation that high MIF in cardiac surgery patients is associated with a reduced incidence of AKI.
Collapse
Affiliation(s)
- Christian Stoppe
- Department of Intensive Care Medicine, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany.
| | - Luisa Averdunk
- Department of Intensive Care Medicine, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany
| | - Andreas Goetzenich
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Josefin Soppert
- Department of Intensive Care Medicine, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany.,Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Arnaud Marlier
- Department of Nephrology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Sandra Kraemer
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Jil Vieten
- Department of Intensive Care Medicine, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany
| | - Mark Coburn
- Department of Anesthesiology, University Hospital, RWTH Aachen, Aachen, Germany
| | - Ana Kowark
- Department of Anesthesiology, University Hospital, RWTH Aachen, Aachen, Germany
| | - Bong-Song Kim
- Department of Plastic and Reconstructive Surgery, Hand Surgery, Burn Center, RWTH Aachen, Aachen, Germany
| | - Gernot Marx
- Department of Intensive Care Medicine, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany
| | - Steffen Rex
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| | - Akinobu Ochi
- Department of Nephropathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gilbert Moeckel
- Department of Nephropathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Omar El Bounkari
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Critical Care Medicine and Pain Therapy, University Hospital Münster, Münster, Germany
| | - Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany. .,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Munich Cluster for Systems Neurology (EXC 1010 SyNergy), Munich, Germany
| | - Sonja Djudjaj
- Institute of Pathology and Department of Nephrology, University Hospital, RWTH Aachen, Aachen, Germany
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Peter Boor
- Institute of Pathology and Department of Nephrology, University Hospital, RWTH Aachen, Aachen, Germany. .,Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
25
|
Poulsen KL, McMullen MR, Huang E, Kibler CD, Sheehan MM, Leng L, Bucala R, Nagy LE. Novel Role of Macrophage Migration Inhibitory Factor in Upstream Control of the Unfolded Protein Response After Ethanol Feeding in Mice. Alcohol Clin Exp Res 2019; 43:1439-1451. [PMID: 31009094 DOI: 10.1111/acer.14065] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/12/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF), a pluripotent immune regulator, is an emerging mediator in alcohol-related liver disease (ALD). MIF is associated with ALD progression through its chemokine- and cytokine-like activities. METHODS Mechanistic studies into the role of MIF in ethanol (EtOH)-induced liver injury were performed in Mif-/- mice and in C57BL/6J mice treated with a small-molecule MIF antagonist, MIF098, after Gao-Binge (acute-on-chronic) EtOH feeding, an EtOH feeding protocol associated with hepatic neutrophilia and induction of the unfolded protein response (UPR). RESULTS The MIF axis, for example, MIF and MIF receptors invariant polypeptide of major histocompatibility complex, class II antigen-associated (CD74), CXCR2, CXCR4, and CXCR7, was enhanced in the livers of alcoholic hepatitis (AH) patients as compared to healthy controls. Mif-/- mice were protected from hepatocellular injury after Gao-Binge feeding, independent of neutrophilia and inflammation, but were associated with the UPR. Interestingly, the UPR signature in AH patients and in mice following Gao-Binge feeding was biased toward cell death with increased expression of pro-cell death CCAAT-enhancer-binding protein homologous protein (CHOP) and decreased prosurvival GRP78. The UPR and liver injury 6 hours after binge were prevented both in Mif-/- mice and in MIF098-treated mice. However, both MIF interventions led to increased liver injury and exacerbated the hepatic UPR 9 hours after binge. Induction of upstream UPR signaling and expression of CHOP protein by thapsigargin in alpha mouse liver 12 hepatocytes were blunted by coexposure to MIF098, directly connecting MIF to UPR in hepatocytes. CONCLUSIONS The current study revealed that, in addition to its cytokine/chemokine functions, MIF is an upstream regulator of UPR in response to EtOH feeding in mice. Importantly, both MIF and UPR can either protect or contribute to liver injury, dependent upon the stage or severity of EtOH-induced liver injury.
Collapse
Affiliation(s)
- Kyle Lauren Poulsen
- Department of Inflammation and Immunity, Center for Liver Disease Research, Cleveland Clinic, Cleveland, Ohio
| | - Megan R McMullen
- Department of Inflammation and Immunity, Center for Liver Disease Research, Cleveland Clinic, Cleveland, Ohio
| | - Emily Huang
- Department of Inflammation and Immunity, Center for Liver Disease Research, Cleveland Clinic, Cleveland, Ohio
| | - Christopher D Kibler
- Department of Inflammation and Immunity, Center for Liver Disease Research, Cleveland Clinic, Cleveland, Ohio
| | - Megan M Sheehan
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Laura E Nagy
- Department of Inflammation and Immunity, Center for Liver Disease Research, Cleveland Clinic, Cleveland, Ohio.,Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
26
|
Roy CJ, Ehrbar DJ, Bohorova N, Bohorov O, Kim D, Pauly M, Whaley K, Rong Y, Torres-Velez FJ, Vitetta ES, Didier PJ, Doyle-Meyers L, Zeitlin L, Mantis NJ. Rescue of rhesus macaques from the lethality of aerosolized ricin toxin. JCI Insight 2019; 4:124771. [PMID: 30626745 DOI: 10.1172/jci.insight.124771] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/27/2018] [Indexed: 12/12/2022] Open
Abstract
Ricin toxin (RT) ranks at the top of the list of bioweapons of concern to civilian and military personnel alike, due to its high potential for morbidity and mortality after inhalation. In nonhuman primates, aerosolized ricin triggers severe acute respiratory distress characterized by perivascular and alveolar edema, neutrophilic infiltration, and severe necrotizing bronchiolitis and alveolitis. There are currently no approved countermeasures for ricin intoxication. Here, we report the therapeutic potential of a humanized mAb against an immunodominant epitope on ricin's enzymatic A chain (RTA). Rhesus macaques that received i.v. huPB10 4 hours after a lethal dose of ricin aerosol exposure survived toxin challenge, whereas control animals succumbed to ricin intoxication within 30 hours. Antibody intervention at 12 hours resulted in the survival of 1 of 5 monkeys. Changes in proinflammatory cytokine, chemokine, and growth factor profiles in bronchial alveolar lavage fluids before and after toxin challenge successfully clustered animals by treatment group and survival, indicating a relationship between local tissue damage and experimental outcome. This study represents the first demonstration, to our knowledge, in nonhuman primates that the lethal effects of inhalational ricin exposure can be negated by a drug candidate, and it opens up a path forward for product development.
Collapse
Affiliation(s)
- Chad J Roy
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Dylan J Ehrbar
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | | | | | - Do Kim
- Mapp Biopharmaceutical Inc., San Diego, California, USA
| | - Michael Pauly
- Mapp Biopharmaceutical Inc., San Diego, California, USA
| | - Kevin Whaley
- Mapp Biopharmaceutical Inc., San Diego, California, USA
| | - Yinghui Rong
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Fernando J Torres-Velez
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Ellen S Vitetta
- Departments of Immunology and Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Peter J Didier
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Lara Doyle-Meyers
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Larry Zeitlin
- Mapp Biopharmaceutical Inc., San Diego, California, USA
| | - Nicholas J Mantis
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| |
Collapse
|
27
|
Zhang C, Ramsey C, Berical A, Yu L, Leng L, McGinnis KA, Song Y, Michael H, McCormack MC, Allore H, Morris A, Crothers K, Bucala R, Lee PJ, Sauler M. A functional macrophage migration inhibitory factor promoter polymorphism is associated with reduced diffusing capacity. Am J Physiol Lung Cell Mol Physiol 2018; 316:L400-L405. [PMID: 30520689 DOI: 10.1152/ajplung.00439.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cigarette smoke exposure is the leading modifiable risk factor for chronic obstructive pulmonary disease (COPD); however, the clinical and pathologic consequences of chronic cigarette smoke exposure are variable among smokers. Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine implicated in the pathogenesis of COPD. Within the promoter of the MIF gene is a functional polymorphism that regulates MIF expression (-794 CATT5-8 microsatellite repeat) ( rs5844572 ). The role of this polymorphim in mediating disease susceptibility to COPD-related traits remains unknown. We performed a cross-sectional analysis of DNA samples from 641 subjects to analyze MIF-794 CATT5-8 ( rs5844572 ) polymorphism by standard methods. We generated multivariable logistic regression models to determine the risk of low expressing MIF alleles for airflow obstruction [defined by forced expiratory volume in 1 s (FEV1)/forced vital capacity ratio <0.70] and an abnormal diffusion capacity [defined by a diffusion capacity for carbon monoxide (DLCO) percent predicted <80%]. We then used generalized linear models to determine the association of MIF genotypes with FEV1 percent predicted and DLCO percent predicted. The MIF-794 CATT5 allele was associated with an abnormal diffusion capacity in two cohorts [odds ratio (OR): 9.31, 95% confidence interval (CI): 1.97-4.06; and OR: 2.21, 95% CI: 1.03-4.75]. Similarly, the MIF-794 CATT5 allele was associated with a reduced DLCO percentage predicted in these two cohorts: 63.5 vs. 70.0 ( P = 0.0023) and 60.1 vs. 65.4 ( P = 0.059). This study suggests an association between a common genetic polymorphism of an endogenous innate immune gene, MIF, with reduced DLCO, an important measurement of COPD severity.
Collapse
Affiliation(s)
- C Zhang
- Department of Medicine Saint Louis University Hospital , Saint Louis, Missouri
| | - C Ramsey
- Yale Center for Medical Informatics, Yale School of Medicine , New Haven, Connecticut
| | - A Berical
- Department of Medicine, Boston University School of Medicine , Boston, Massachusetts
| | - L Yu
- Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts
| | - L Leng
- Department of Medicine, Yale School of Medicine , New Haven, Connecticut
| | - K A McGinnis
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Y Song
- Department of Medicine, Yale School of Medicine , New Haven, Connecticut
| | - H Michael
- Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - M C McCormack
- Department of Medicine, Johns Hopkins University , Baltimore, Maryland
| | - H Allore
- Department of Medicine, Yale School of Medicine , New Haven, Connecticut
| | - A Morris
- Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - K Crothers
- Department of Medicine, University of Washington School of Medicine , Seattle, Washington
| | - R Bucala
- Department of Medicine, Yale School of Medicine , New Haven, Connecticut
| | - P J Lee
- Department of Medicine, Yale School of Medicine , New Haven, Connecticut
| | - M Sauler
- Department of Medicine, Yale School of Medicine , New Haven, Connecticut
| |
Collapse
|
28
|
Shin MS, Kang Y, Wahl ER, Park HJ, Lazova R, Leng L, Mamula M, Krishnaswamy S, Bucala R, Kang I. Macrophage Migration Inhibitory Factor Regulates U1 Small Nuclear RNP Immune Complex-Mediated Activation of the NLRP3 Inflammasome. Arthritis Rheumatol 2018; 71:109-120. [PMID: 30009530 DOI: 10.1002/art.40672] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 07/12/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE High-expression alleles of macrophage migration inhibitory factor (MIF) are linked genetically to the severity of systemic lupus erythematosus (SLE). The U1 small nuclear RNP (snRNP) immune complex containing U1 snRNP and anti-U1 snRNP antibodies, which are found in patients with SLE, activates the NLRP3 inflammasome, comprising NLRP3, ASC, and procaspase 1, in human monocytes, leading to the production of interleukin-1β (IL-1β). This study was undertaken to investigate the role of the snRNP immune complex in up-regulating the expression of MIF and its interface with the NLRP3 inflammasome. METHODS MIF, IL-1β, NLRP3, caspase 1, ASC, and MIF receptors were analyzed by enzyme-linked immunosorbent assay, Western blotting, quantitative polymerase chain reaction, and cytometry by time-of-flight mass spectrometry (CytoF) in human monocytes incubated with or without the snRNP immune complex. MIF pathway responses were probed with the novel small molecule antagonist MIF098. RESULTS The snRNP immune complex induced the production of MIF and IL-1β from human monocytes. High-dimensional, single-cell CytoF analysis established that MIF regulates activation of the NLRP3 inflammasome, including findings of a quantitative relationship between MIF and its receptors and IL-1β levels in the monocytes. MIF098, which blocks MIF binding to its cognate receptor, suppressed the production of IL-1β, the up-regulation of NLRP3, which is a rate-limiting step in NLRP3 inflammasome activation, and the activation of caspase 1 in snRNP immune complex-stimulated human monocytes. CONCLUSION The U1 snRNP immune complex is a specific stimulus of MIF production in human monocytes, with MIF having an upstream role in defining the inflammatory characteristics of activated monocytes by regulating NLRP3 inflammasome activation and downstream IL-1β production. These findings provide mechanistic insight and a therapeutic rationale for targeting MIF in subgroups of lupus patients, such as those classified as high genotypic MIF expressers or those with anti-snRNP antibodies.
Collapse
Affiliation(s)
- Min Sun Shin
- Yale University School of Medicine, New Haven, Connecticut
| | - Youna Kang
- Yale University School of Medicine, New Haven, Connecticut
| | - Elizabeth R Wahl
- Yale University School of Medicine, New Haven, Connecticut, and University of Washington, Seattle
| | - Hong-Jai Park
- Yale University School of Medicine, New Haven, Connecticut
| | - Rossitza Lazova
- Yale University School of Medicine, New Haven, Connecticut, and California Skin Institute, San Jose
| | - Lin Leng
- Yale University School of Medicine, New Haven, Connecticut
| | - Mark Mamula
- Yale University School of Medicine, New Haven, Connecticut
| | | | - Richard Bucala
- Yale University School of Medicine, New Haven, Connecticut
| | - Insoo Kang
- Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
29
|
Ma F, Hunt DE, Leng L, Bucala R, Meyer-Siegler KL, Vera PL. Protease activated-receptor 4 activation as a model of persistent bladder pain: Essential role of macrophage migration inhibitory factor and high mobility group box 1. Int J Urol 2018; 25:887-893. [PMID: 30112848 DOI: 10.1111/iju.13778] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/12/2018] [Indexed: 12/28/2022]
Abstract
OBJECTIVES To develop a rodent model of persistent non-inflammatory bladder pain and to test macrophage migration inhibitory factor and high mobility box group 1 as mediators of bladder pain. METHODS Female C57BL/6 mice received intravesical instillations of protease activated receptor 4 (100 μmol/L, for 1 h) three times every other day and abdominal mechanical hypersensitivity (50% mechanical threshold) was tested on day 0 (baseline), and at days 1, 2, 3, 4, 7 and 9 after the first protease-activated receptor 4 injection. At the end of the experiment, micturition changes were measured and bladders were examined for histological changes. Macrophage migration inhibitory factor antagonist (MIF098; 40 mg/kg i.p. b.i.d.) or high mobility group box 1 inhibitor (glycyrrhizin; 50 mg/kg i.p. daily) was administered from day 2 until day 8. RESULTS There was a significant and persistent decrease in abdominal mechanical threshold starting from day 3 in the protease-activated receptor 4-treated group that persisted until day 9 (5 days post-last instillation), but not in the control group. Glycyrrhizin fully reversed while MIF098 partially reversed abdominal mechanical hypersensitivity in protease-activated receptor 4-treated mice. The changes started on day 3 after the first protease-activated receptor 4 instillation, and analgesic effects lasted throughout the rest of the testing period. None of the groups had significant micturition changes or overt bladder histological changes. CONCLUSIONS Repeated intravesical protease activated receptor 4 instillations produce persistent bladder pain without inflammation. Macrophage migration inhibitory factor and high mobility group box 1 are possible effective target molecules for bladder pain alleviation.
Collapse
Affiliation(s)
- Fei Ma
- Research and Development, Lexington Veterans Affairs Medical Center, Lexington, Kentucky, USA.,Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - David E Hunt
- Research and Development, Lexington Veterans Affairs Medical Center, Lexington, Kentucky, USA
| | - Lin Leng
- Department of Internal Medicine, Yale University, New Haven, Connecticut, USA
| | - Richard Bucala
- Department of Internal Medicine, Yale University, New Haven, Connecticut, USA
| | | | - Pedro L Vera
- Research and Development, Lexington Veterans Affairs Medical Center, Lexington, Kentucky, USA.,Department of Physiology, University of Kentucky, Lexington, Kentucky, USA.,Department of Surgery, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
30
|
The biological function and significance of CD74 in immune diseases. Inflamm Res 2016; 66:209-216. [DOI: 10.1007/s00011-016-0995-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 09/30/2016] [Indexed: 12/25/2022] Open
|
31
|
Relationship between elevated soluble CD74 and severity of experimental and clinical ALI/ARDS. Sci Rep 2016; 6:30067. [PMID: 27444250 PMCID: PMC4957083 DOI: 10.1038/srep30067] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/28/2016] [Indexed: 01/11/2023] Open
Abstract
CD74 is expressed on the cell surface of pulmonary macrophages and contributes to macrophage migration inhibitory factor (MIF)-induced inflammatory response in acute lung injury (ALI). A circulating form of CD74 (soluble CD74, sCD74) was recently discovered in autoimmune liver disease. Using two murine ALI models and cells culture, we examined the presence of sCD74 in circulation and alveolar space and preliminarily assessed the biological function of sCD74. The concentrations of sCD74 were increased in serum and bronchoalveolar lavage fluids (BALF) of murine ALI models. The elevated levels of sCD74 in BALF positively correlated with lung permeability and inflammation. In addition, sCD74 is secreted by macrophages in response to MIF stimulation and itself can stimulate the production of inflammatory cytokines. Our clinical study confirmed some findings of basic research. Moreover, we also found Day 3 serum sCD74 levels were associated with worse clinical outcomes. In conclusion, higher serum sCD74 levels may reflect more severe lung injury and may be used to help physicians determine prognosis of acute respiratory distress syndrome (ARDS).
Collapse
|
32
|
Husebø GR, Bakke PS, Grønseth R, Hardie JA, Ueland T, Aukrust P, Eagan TML. Macrophage migration inhibitory factor, a role in COPD. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1-7. [PMID: 27190066 DOI: 10.1152/ajplung.00461.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 05/17/2016] [Indexed: 01/08/2023] Open
Abstract
Macrophage migration inhibitor factor (MIF) is a pluripotent cytokine associated with several different inflammatory conditions, but its role within lung inflammation and chronic obstructive pulmonary disease (COPD) is unclear. This study aimed to examine MIF in both stable COPD and during acute exacerbations (AECOPD). The study included 433 patients with COPD aged 41-76 and 325 individuals from the Bergen COPD cohort study who served as controls. All patients had an FEV1 of <80% predicted, FEV1/FVC ratio of <0.7, and a smoking history >10 pack-years. Serum levels of MIF were compared between the two groups at baseline, and for 149 patients, measurements were also carried out during AECOPD. Linear regression models were fitted with MIF as the outcome variable and adjusted for sex, age, body composition, smoking, and Charlson Comorbidity Score (CCS). Median MIF (interquartile range) in patients with COPD was 20.1 ng/ml (13.5-30.9) compared with 14.9 ng/ml (11.1-21.6) in controls (P < 0.01). MIF was bivariately associated with sex, body composition, and CCS (P < 0.05 for all). In the regression analyses, MIF was significantly higher in patients with COPD, coefficient 1.32 (P < 0.01) and 1.30 (P < 0.01) unadjusted and adjusted, respectively. In addition, in 149 patients during episodes of AECOPD, MIF was significantly elevated, with a median of 23.2 ng/ml (14.1-42.3) compared with measurements at stable disease of 19.3 ng/ml (12.4-31.3, P < 0.01). Serum levels of MIF were significantly higher in patients with COPD compared with controls. We also identified an additional increase in MIF levels during episodes of AECOPD.
Collapse
Affiliation(s)
- Gunnar R Husebø
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen Norway; Department of Clinical Science, University of Bergen, Bergen, Norway;
| | - Per S Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Rune Grønseth
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jon A Hardie
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen Inflammatory Research Centre, University of Oslo, Oslo, Norway; and
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen Inflammatory Research Centre, University of Oslo, Oslo, Norway; and Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Tomas M L Eagan
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
33
|
Valiño-Rivas L, Baeza-Bermejillo C, Gonzalez-Lafuente L, Sanz AB, Ortiz A, Sanchez-Niño MD. CD74 in Kidney Disease. Front Immunol 2015; 6:483. [PMID: 26441987 PMCID: PMC4585214 DOI: 10.3389/fimmu.2015.00483] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/05/2015] [Indexed: 12/17/2022] Open
Abstract
CD74 (invariant MHC class II) regulates protein trafficking and is a receptor for macrophage migration inhibitory factor (MIF) and d-dopachrome tautomerase (d-DT/MIF-2). CD74 expression is increased in tubular cells and/or glomerular podocytes and parietal cells in human metabolic nephropathies, polycystic kidney disease, graft rejection and kidney cancer and in experimental diabetic nephropathy and glomerulonephritis. Stressors like abnormal metabolite (glucose, lyso-Gb3) levels and inflammatory cytokines increase kidney cell CD74. MIF activates CD74 to increase inflammatory cytokines in podocytes and tubular cells and proliferation in glomerular parietal epithelial cells and cyst cells. MIF overexpression promotes while MIF targeting protects from experimental glomerular injury and kidney cysts, and interference with MIF/CD74 signaling or CD74 deficiency protected from crescentic glomerulonephritis. However, CD74 may protect from interstitial kidney fibrosis. Furthermore, CD74 expression by stressed kidney cells raises questions about the kidney safety of cancer therapy strategies delivering lethal immunoconjugates to CD74-expressing cells. Thus, understanding CD74 biology in kidney cells is relevant for kidney therapeutics.
Collapse
Affiliation(s)
- Lara Valiño-Rivas
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Madrid , Spain
| | - Ciro Baeza-Bermejillo
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Madrid , Spain ; Red de Investigación Renal (REDINREN) , Madrid , Spain
| | - Laura Gonzalez-Lafuente
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Madrid , Spain
| | - Ana Belen Sanz
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Madrid , Spain ; Red de Investigación Renal (REDINREN) , Madrid , Spain
| | - Alberto Ortiz
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Madrid , Spain ; Red de Investigación Renal (REDINREN) , Madrid , Spain ; School of Medicine, Universidad Autónoma de Madrid , Madrid , Spain ; Fundacion Renal Iñigo Alvarez de Toledo-IRSIN , Madrid , Spain
| | - Maria Dolores Sanchez-Niño
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Madrid , Spain ; Red de Investigación Renal (REDINREN) , Madrid , Spain
| |
Collapse
|
34
|
Sauler M, Bucala R, Lee PJ. Role of macrophage migration inhibitory factor in age-related lung disease. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1-10. [PMID: 25957294 DOI: 10.1152/ajplung.00339.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 05/05/2015] [Indexed: 12/25/2022] Open
Abstract
The prevalence of many common respiratory disorders, including pneumonia, chronic obstructive lung disease, pulmonary fibrosis, and lung cancer, increases with age. Little is known of the host factors that may predispose individuals to such diseases. Macrophage migration inhibitory factor (MIF) is a potent upstream regulator of the immune system. MIF is encoded by variant alleles that occur commonly in the population. In addition to its role as a proinflammatory cytokine, a growing body of literature demonstrates that MIF influences diverse molecular processes important for the maintenance of cellular homeostasis and may influence the incidence or clinical manifestations of a variety of chronic lung diseases. This review highlights the biological properties of MIF and its implication in age-related lung disease.
Collapse
Affiliation(s)
- Maor Sauler
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Richard Bucala
- Section of Rheumatology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Patty J Lee
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| |
Collapse
|