1
|
Lin B, Fan Y, Yang X, Pathak JL, Zhong M. MMP-12 and Periodontitis: Unraveling the Molecular Pathways of Periodontal Tissue Destruction. J Inflamm Res 2024; 17:7793-7806. [PMID: 39494211 PMCID: PMC11529342 DOI: 10.2147/jir.s480466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024] Open
Abstract
Periodontal disease is a common disorder affecting a wide range of people and has a high prevalence globally. Periodontitis comprises a series of inflammatory conditions affecting periodontal support tissue, which could ultimately lead to tooth loss and reduce life quality and add to the financial burden of society. Matrix metalloproteinase-12 (MMP-12) is an elastase that is produced mostly by macrophages and could degrade a wide spectrum of extracellular matrix (ECM) and also contribute to several systematic pathological conditions. Recently, researchers have reported higher expression of MMP-12 in chronic periodontitis patients. However, there are few reports on the role of MMP-12 in periodontitis pathogenicity, and the interaction between MMP-12, periodontal pathogens, and periodontal tissues remains unclear. In this review, we introduce the potentially unique role of MMP-12 in the context of periodontal inflammation earlier, summarize the possible effects of MMP-12 on the pathological process of periodontitis and the interaction of host response under the challenge of various inflammatory factors, and provide possible diagnostic and therapeutic strategies targeting MMP-12 for the management of periodontitis. Future research and policies should focus on and implement effective chairside testing methods to reduce the prevalence of periodontal diseases.
Collapse
Affiliation(s)
- Bingpeng Lin
- Department of Orthodontics, School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou, 510180, People’s Republic of China
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, People’s Republic of China
| | - Yufei Fan
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, People’s Republic of China
| | - Xuechao Yang
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, People’s Republic of China
| | - Janak L Pathak
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, People’s Republic of China
| | - Mei Zhong
- Department of Prosthodontics, School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou, 510180, People’s Republic of China
| |
Collapse
|
2
|
Buerger M, Amor M, Akhmetshina A, Bianco V, Perfler B, Zebisch A, Weichhart T, Kratky D. Limited Alleviation of Lysosomal Acid Lipase Deficiency by Deletion of Matrix Metalloproteinase 12. Int J Mol Sci 2024; 25:11001. [PMID: 39456786 PMCID: PMC11506919 DOI: 10.3390/ijms252011001] [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: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Lysosomal acid lipase (LAL) is the only known enzyme that degrades cholesteryl esters and triglycerides at an acidic pH. In LAL deficiency (LAL-D), dysregulated expression of matrix metalloproteinase 12 (MMP-12) has been described. The overexpression of MMP-12 in myeloid lineage cells causes an immune cell dysfunction resembling that of Lal knockout (Lal KO) mice. Both models develop progressive lymphocyte dysfunction and expansion of myeloid-derived suppressor (CD11b+ Gr-1+) cells. To study whether MMP-12 might be a detrimental contributor to the pathology of LAL-D, we have generated Lal/Mmp12 double knockout (DKO) mice. The phenotype of Lal/Mmp12 DKO mice closely resembled that of Lal KO mice, while the weight and morphology of the thymus were improved in Lal/Mmp12 DKO mice. Cytological examination of blood smears showed a mildly reversed lymphoid-to-myeloid shift in DKO mice. Despite significant decreases in CD11b+ Ly6G+ cells in the peripheral blood, bone marrow, and spleen of Lal/Mmp12 DKO mice, the hematopoietic bone marrow progenitor compartment and markers for neutrophil chemotaxis were unchanged. Since the overall severity of LAL-D remains unaffected by the deletion of Mmp12, we conclude that MMP-12 does not represent a viable target for treating the inflammatory pathology in LAL-D.
Collapse
Affiliation(s)
- Martin Buerger
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (M.B.); (M.A.); (A.A.); (V.B.)
| | - Melina Amor
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (M.B.); (M.A.); (A.A.); (V.B.)
| | - Alena Akhmetshina
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (M.B.); (M.A.); (A.A.); (V.B.)
| | - Valentina Bianco
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (M.B.); (M.A.); (A.A.); (V.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Bianca Perfler
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology, and Inflammation, Medical University of Graz, 8010 Graz, Austria; (B.P.); (A.Z.)
- Division of Hematology, Medical University of Graz, 8010 Graz, Austria
| | - Armin Zebisch
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology, and Inflammation, Medical University of Graz, 8010 Graz, Austria; (B.P.); (A.Z.)
- Division of Hematology, Medical University of Graz, 8010 Graz, Austria
| | - Thomas Weichhart
- Center for Pathobiochemistry & Genetics, Medical University of Vienna, 1090 Vienna, Austria;
| | - Dagmar Kratky
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (M.B.); (M.A.); (A.A.); (V.B.)
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
3
|
Zhang X, Wang Y, Li H, Wang DW, Chen C. Insights into the post-translational modifications in heart failure. Ageing Res Rev 2024; 100:102467. [PMID: 39187021 DOI: 10.1016/j.arr.2024.102467] [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: 10/25/2023] [Revised: 08/01/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Heart failure (HF), as the terminal manifestation of multiple cardiovascular diseases, causes a huge socioeconomic burden worldwide. Despite the advances in drugs and medical-assisted devices, the prognosis of HF remains poor. HF is well-accepted as a myriad of subcellular dys-synchrony related to detrimental structural and functional remodelling of cardiac components, including cardiomyocytes, fibroblasts, endothelial cells and macrophages. Through the covalent chemical process, post-translational modifications (PTMs) can coordinate protein functions, such as re-localizing cellular proteins, marking proteins for degradation, inducing interactions with other proteins and tuning enzyme activities, to participate in the progress of HF. Phosphorylation, acetylation, and ubiquitination predominate in the currently reported PTMs. In addition, advanced HF is commonly accompanied by metabolic remodelling including enhanced glycolysis. Thus, glycosylation induced by disturbed energy supply is also important. In this review, firstly, we addressed the main types of HF. Then, considering that PTMs are associated with subcellular locations, we summarized the leading regulation mechanisms in organelles of distinctive cell types of different types of HF, respectively. Subsequently, we outlined the aforementioned four PTMs of key proteins and signaling sites in HF. Finally, we discussed the perspectives of PTMs for potential therapeutic targets in HF.
Collapse
Affiliation(s)
- Xudong Zhang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Yan Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Huaping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| |
Collapse
|
4
|
Wänman M, Betnér S, Esberg A, Holm CK, Isehed C, Holmlund A, Palmqvist P, Lövgren A, Lindquist S, Hänström L, Lerner UH, Kindstedt E, Lundberg P. The PerioGene North Study Uncovers Serum Proteins Related to Periodontitis. J Dent Res 2024; 103:999-1007. [PMID: 39101637 PMCID: PMC11402264 DOI: 10.1177/00220345241263320] [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] [Indexed: 08/06/2024] Open
Abstract
The sequalae of periodontitis include irreversible degradation of tooth-supporting structures and circulatory spread of inflammatory mediators. However, the serum protein profile in periodontitis is not well described, which is partly attributable to the limited number of studies based on large and well-characterized periodontitis cohorts. This study aims to identify novel, circulating inflammation-related proteins associated with periodontitis within the PerioGene North case-control study, which includes 478 cases with severe periodontitis and 509 periodontally healthy controls. The serum concentrations of high-sensitivity C-reactive protein (hs-CRP) and a panel of 45 inflammation-related proteins were analyzed using targeted proteomics. A distinguishable serum protein profile was evident in periodontitis cases. The protein pattern could separate cases from controls with a sensitivity of 0.81 and specificity of 0.81 (area under the curve = 0.87). Adjusted levels for hs-CRP and 24 of the 45 proteins were different between cases and controls. High levels of hs-CRP and matrix metalloproteinase-12, and low levels of epidermal growth factor (EGF) and oxidized low-density lipoprotein receptor 1 (OLR-1) were detected among the cases. Furthermore, the levels of C-C motif chemokine-19, granulocyte colony-stimulating factor-3 (CSF-3), interleukin-7 (IL-7), and hs-CRP were significantly higher in cases with a high degree of gingival inflammation. The levels of CSF-3 and tumor necrosis factor ligand superfamily member-10 TNFSF-10 were higher in cases with many deep periodontal pockets. The PerioGene North study includes detailed clinical periodontal data and uncovers a distinct serum protein profile in periodontitis. The findings of lower EGF and OLR-1 among the cases are highlighted, as this has not been presented before. The role of EGF and OLR-1 in periodontitis pathogenesis and as possible future biomarkers should be further explored.
Collapse
Affiliation(s)
- M Wänman
- Department of Odontology, Umeå University, Section for Molecular Periodontology, Umeå, Sweden
| | - S Betnér
- Northern Registry Centre, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - A Esberg
- Department of Odontology, Umeå University, Umeå, Sweden
| | - C K Holm
- Department of Odontology, Umeå University, Section for Molecular Periodontology, Umeå, Sweden
| | - C Isehed
- Gävle County Hospital, Department of Periodontology, Public Dental Health County Council of Gävleborg, Gävle, Sweden
- Center for Research and Development Uppsala University/Region Gävleborg, Gävle, Sweden
| | - A Holmlund
- Gävle County Hospital, Department of Periodontology, Public Dental Health County Council of Gävleborg, Gävle, Sweden
- Center for Research and Development Uppsala University/Region Gävleborg, Gävle, Sweden
| | - P Palmqvist
- Department of Periodontology, County Council of Västerbotten, Umeå, Sweden
| | - A Lövgren
- Department of Odontology, Umeå University, Section for Clinical Oral Physiology, Umeå, Sweden
| | - S Lindquist
- Department of Odontology, Umeå University, Section for Molecular Periodontology, Umeå, Sweden
- Lipum AB, Umeå, Sweden
| | - L Hänström
- Department of Odontology, Umeå University, Section for Molecular Periodontology, Umeå, Sweden
| | - U H Lerner
- Department of Odontology, Umeå University, Section for Molecular Periodontology, Umeå, Sweden
- Sahlgrenska Ostoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - E Kindstedt
- Department of Odontology, Umeå University, Section for Molecular Periodontology, Umeå, Sweden
| | - P Lundberg
- Department of Odontology, Umeå University, Section for Molecular Periodontology, Umeå, Sweden
| |
Collapse
|
5
|
Herreros-Cabello A, Del Moral-Salmoral J, Morato E, Marina A, Barrocal B, Fresno M, Gironès N. Quantitative Proteomic Analysis of Macrophages Infected with Trypanosoma cruzi Reveals Different Responses Dependent on the SLAMF1 Receptor and the Parasite Strain. Int J Mol Sci 2024; 25:7493. [PMID: 39000601 PMCID: PMC11242706 DOI: 10.3390/ijms25137493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
Chagas disease is caused by the intracellular protozoan parasite Trypanosoma cruzi. This disease affects mainly rural areas in Central and South America, where the insect vector is endemic. However, this disease has become a world health problem since migration has spread it to other continents. It is a complex disease with many reservoirs and vectors and high genetic variability. One of the host proteins involved in the pathogenesis is SLAMF1. This immune receptor acts during the infection of macrophages controlling parasite replication and thus affecting survival in mice but in a parasite strain-dependent manner. Therefore, we studied the role of SLAMF1 by quantitative proteomics in a macrophage in vitro infection and the different responses between Y and VFRA strains of Trypanosoma cruzi. We detected different significant up- or downregulated proteins involved in immune regulation processes, which are SLAMF1 and/or strain-dependent. Furthermore, independently of SLAMF1, this parasite induces different responses in macrophages to counteract the infection and kill the parasite, such as type I and II IFN responses, NLRP3 inflammasome activation, IL-18 production, TLR7 and TLR9 activation specifically with the Y strain, and IL-11 signaling specifically with the VFRA strain. These results have opened new research fields to elucidate the concrete role of SLAMF1 and discover new potential therapeutic approaches for Chagas disease.
Collapse
Affiliation(s)
- Alfonso Herreros-Cabello
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Javier Del Moral-Salmoral
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Esperanza Morato
- Unidad de Proteómica, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain
| | - Anabel Marina
- Unidad de Proteómica, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain
- Unidad de Técnicas Bioanalíticas (BAT), Instituto de Investigación de Ciencias de la Alimentación (CIAL), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Beatriz Barrocal
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Manuel Fresno
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
- Instituto Universitario de Biología Molecular, Universidad Autónoma de Madrid (IUBM-UAM), 28049 Madrid, Spain
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Núria Gironès
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
- Instituto Universitario de Biología Molecular, Universidad Autónoma de Madrid (IUBM-UAM), 28049 Madrid, Spain
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, 28006 Madrid, Spain
| |
Collapse
|
6
|
Silva SV, Lima MA, Hodgson L, Freitas VM, Rodríguez-Manzaneque JC. ADAMTS-1 has nuclear localization in cells with epithelial origin and leads to decreased cell migration. Exp Cell Res 2023; 433:113852. [PMID: 37951335 PMCID: PMC10841765 DOI: 10.1016/j.yexcr.2023.113852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/13/2023]
Abstract
In the study of tumorigenesis, the involvement of molecules within the extracellular matrix (ECM) is crucial. ADAMTSs (A Disintegrin and Metalloproteinase with Thrombospondin motifs), a group of secreted proteases known for their role in ECM remodeling, were primarily considered to be extracellular proteases. However, our research specifically detected ADAMTS-1, a member of this family, predominantly within the nucleus of mammary cells. Our main objective was to understand the mechanism of ADAMTS-1 translocation to the nucleus and its functional significance in this cellular compartment. Our investigation uncovered that nuclear ADAMTS-1 was present in cells exhibiting an epithelial phenotype, while cells of mesenchymal origin contained the protease in the cytoplasm. Moreover, disruption of ADAMTS-1 secretion, induced by Monensin treatment, resulted in its accumulation in the cytoplasm. Notably, our research indicated that alterations in the secretory pathways could influence the protease's compartmentalization. Additionally, experiments with conditioned medium from cells containing nuclear ADAMTS-1 demonstrated its internalization into the nucleus by HT-1080 cells and fibroblasts. Furthermore, heightened levels of ADAMTS-1 within the ECM reduced the migratory potential of mesenchymal cells. This highlights the potential significance of nuclear ADAMTS-1 as a critical component within the tumor microenvironment due to its functional activity in this specific cellular compartment.
Collapse
Affiliation(s)
- Suély V Silva
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil.
| | - Maíra A Lima
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Louis Hodgson
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Vanessa M Freitas
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil.
| | | |
Collapse
|
7
|
Kuntschar S, Cardamone G, Klann K, Bauer R, Meyer SP, Raue R, Rappl P, Münch C, Brüne B, Schmid T. Mmp12 Is Translationally Regulated in Macrophages during the Course of Inflammation. Int J Mol Sci 2023; 24:16981. [PMID: 38069304 PMCID: PMC10707645 DOI: 10.3390/ijms242316981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Despite the importance of rapid adaptive responses in the course of inflammation and the notion that post-transcriptional regulation plays an important role herein, relevant translational alterations, especially during the resolution phase, remain largely elusive. In the present study, we analyzed translational changes in inflammatory bone marrow-derived macrophages upon resolution-promoting efferocytosis. Total RNA-sequencing confirmed that apoptotic cell phagocytosis induced a pro-resolution signature in LPS/IFNγ-stimulated macrophages (Mϕ). While inflammation-dependent transcriptional changes were relatively small between efferocytic and non-efferocytic Mϕ; considerable differences were observed at the level of de novo synthesized proteins. Interestingly, translationally regulated targets in response to inflammatory stimuli were mostly downregulated, with only minimal impact of efferocytosis. Amongst these targets, pro-resolving matrix metallopeptidase 12 (Mmp12) was identified as a translationally repressed candidate during early inflammation that recovered during the resolution phase. Functionally, reduced MMP12 production enhanced matrix-dependent migration of Mϕ. Conclusively, translational control of MMP12 emerged as an efficient strategy to alter the migratory properties of Mϕ throughout the inflammatory response, enabling Mϕ migration within the early inflammatory phase while restricting migration during the resolution phase.
Collapse
Affiliation(s)
- Silvia Kuntschar
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Giulia Cardamone
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Kevin Klann
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Rebekka Bauer
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Sofie Patrizia Meyer
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Rebecca Raue
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Peter Rappl
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Christian Münch
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, 60596 Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, 60596 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, 60596 Frankfurt, Germany
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
| |
Collapse
|
8
|
Chalise U, Becirovic‐Agic M, Lindsey ML. The cardiac wound healing response to myocardial infarction. WIREs Mech Dis 2023; 15:e1584. [PMID: 36634913 PMCID: PMC10077990 DOI: 10.1002/wsbm.1584] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/31/2022] [Accepted: 05/18/2022] [Indexed: 01/14/2023]
Abstract
Myocardial infarction (MI) is defined as evidence of myocardial necrosis consistent with prolonged ischemia. In response to MI, the myocardium undergoes a series of wound healing events that initiate inflammation and shift to anti-inflammation before transitioning to tissue repair that culminates in scar formation to replace the region of the necrotic myocardium. The overall response to MI is determined by two major steps, the first of which is the secretion of proteases by infiltrating leukocytes to breakdown extracellular matrix (ECM) components, a necessary step to remove necrotic cardiomyocytes. The second step is the generation of new ECM that comprises the scar; and this step is governed by the cardiac fibroblasts as the major source of new ECM synthesis. The leukocyte component resides in the middle of the two-step process, contributing to both sides as the leukocytes transition from pro-inflammatory to anti-inflammatory and reparative cell phenotypes. The balance between the two steps determines the final quantity and quality of scar formed, which in turn contributes to chronic outcomes following MI, including the progression to heart failure. This review will summarize our current knowledge regarding the cardiac wound healing response to MI, primarily focused on experimental models of MI in mice. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Immune System Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| | - Mediha Becirovic‐Agic
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| | - Merry L. Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| |
Collapse
|
9
|
Kubat Oktem E, Aydin B, Yazar M, Arga KY. Integrative Analysis of Motor Neuron and Microglial Transcriptomes from SOD1 G93A Mice Models Uncover Potential Drug Treatments for ALS. J Mol Neurosci 2022; 72:2360-2376. [PMID: 36178612 DOI: 10.1007/s12031-022-02071-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 09/19/2022] [Indexed: 02/07/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disease of motor neurons that mainly affects the motor cortex, brainstem, and spinal cord. Under disease conditions, microglia could possess two distinct profiles, M1 (toxic) and M2 (protective), with the M2 profile observed at disease onset. SOD1 (superoxide dismutase 1) gene mutations account for up to 20% of familial ALS cases. Comparative gene expression differences in M2-protective (early) stage SOD1G93A microglia and age-matched SOD1G93A motor neurons are poorly understood. We evaluated the differential gene expression profiles in SOD1G93A microglia and SOD1G93A motor neurons utilizing publicly available transcriptomics data and bioinformatics analyses, constructed biomolecular networks around them, and identified gene clusters as potential drug targets. Following a drug repositioning strategy, 5 small compounds (belinostat, auranofin, BRD-K78930611, AZD-8055, and COT-10b) were repositioned as potential ALS therapeutic candidates that mimic the protective state of microglia and reverse the toxic state of motor neurons. We anticipate that this study will provide new insights into the ALS pathophysiology linking the M2 state of microglia and drug repositioning.
Collapse
Affiliation(s)
- Elif Kubat Oktem
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Istanbul Medeniyet University, Kuzey Yerleşkesi H Blok, Ünalan Sk. D100 Karayolu Yanyol 34700, Istanbul, Turkey.
| | - Busra Aydin
- Department of Bioengineering, Faculty of Engineering and Architecture, Konya Food and Agriculture University, Konya, Turkey
| | - Metin Yazar
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul, Turkey.,Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul, Turkey
| | - Kazim Yalcin Arga
- Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul, Turkey.,Genetic and Metabolic Diseases Research and Investigation Center, Marmara University, Istanbul, Turkey
| |
Collapse
|
10
|
Pro-oxidative priming but maintained cardiac function in a broad spectrum of murine models of chronic kidney disease. Redox Biol 2022; 56:102459. [PMID: 36099852 PMCID: PMC9482130 DOI: 10.1016/j.redox.2022.102459] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
Aims Patients with chronic kidney disease (CKD) have an increased risk of cardiovascular events and exhibit myocardial changes including left ventricular (LV) hypertrophy and fibrosis, overall referred to as ‘uremic cardiomyopathy’. Although different CKD animal models have been studied for cardiac effects, lack of consistent reporting on cardiac function and pathology complicates clear comparison of these models. Therefore, this study aimed at a systematic and comprehensive comparison of cardiac function and cardiac pathophysiological characteristics in eight different CKD models and mouse strains, with a main focus on adenine-induced CKD. Methods and results CKD of different severity and duration was induced by subtotal nephrectomy or adenine-rich diet in various strains (C57BL/6J, C57BL/6 N, hyperlipidemic C57BL/6J ApoE−/−, 129/Sv), followed by the analysis of kidney function and morphology, blood pressure, cardiac function, cardiac hypertrophy, fibrosis, myocardial calcification and inflammation using functional, histological and molecular techniques, including cardiac gene expression profiling supplemented by oxidative stress analysis. Intriguingly, despite uremia of variable degree, neither cardiac dysfunction, hypertrophy nor interstitial fibrosis were observed. However, already moderate CKD altered cardiac oxidative stress responses and enhanced oxidative stress markers in each mouse strain, with cardiac RNA sequencing revealing activation of oxidative stress signaling as well as anti-inflammatory feedback responses. Conclusion This study considerably expands the knowledge on strain- and protocol-specific differences in the field of cardiorenal research and reveals that several weeks of at least moderate experimental CKD increase oxidative stress responses in the heart in a broad spectrum of mouse models. However, this was insufficient to induce relevant systolic or diastolic dysfunction, suggesting that additional “hits” are required to induce uremic cardiomyopathy. Translational perspective Patients with chronic kidney disease (CKD) have an increased risk of cardiovascular adverse events and exhibit myocardial changes, overall referred to as ‘uremic cardiomyopathy’. We revealed that CKD increases cardiac oxidative stress responses in the heart. Nonetheless, several weeks of at least moderate experimental CKD do not necessarily trigger cardiac dysfunction and remodeling, suggesting that additional “hits” are required to induce uremic cardiomyopathy in the clinical setting. Whether the altered cardiac oxidative stress balance in CKD may increase the risk and extent of cardiovascular damage upon additional cardiovascular risk factors and/or events will be addressed in future studies. Development of a CKD mouse model with a clear cardiac functional or morphological phenotype is challenging. Cardiac oxidative stress response as well as oxidative stress markers are increased in a broad spectrum of CKD mouse models. Our findings suggest need of additional cardiovascular hits to clearly induce uremic cardiomyopathy as observed in patients.
Collapse
|
11
|
Chalise U, Becirovic-Agic M, Konfrst SR, Rodriguez-Paar JR, Cook LM, Lindsey ML. MMP-12 polarizes neutrophil signalome towards an apoptotic signature. J Proteomics 2022; 264:104636. [PMID: 35661763 DOI: 10.1016/j.jprot.2022.104636] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 11/18/2022]
Abstract
While macrophages are well-known to polarize across the inflammatory spectrum, neutrophils have only recently been found to activate in a similar fashion in response to pro- or anti-inflammatory stimuli. Matrix metalloproteinase (MMP)-12 mediates neutrophil physiology with direct signaling mechanisms yet to be investigated. We hypothesized MMP-12 may modify neutrophil signaling. Bone marrow neutrophils were stimulated with interleukin (IL-1β; pro-inflammatory), IL-4 (anti-inflammatory), or MMP-12. The secretome was mapped by multi-analyte profiling and intracellular signaling evaluated by array. IL-1β induced a cytokine-mediated inflammatory LPS-like signalome, with upregulation of pro-inflammatory cytokines such as interferon gamma (IFNγ,15.2-fold,p = 0.001), chemokine (C-X-C motif) ligand 1 (CXCL1,8.4-fold,p = 0.005), and tumor necrosis factor alpha (TNFα,11.2-fold,p = 0.004). IL-4 induced strong intracellular signaling with upregulation of mitogen-activated protein kinase kinase (MEK1;1.9-fold,p = 0.0005) and downregulation of signal transducer and activator of transcription 4 (STAT4;0.77-fold,0.001). MMP-12 increased IL-4 secretion 20-fold and induced a robust apoptotic neutrophil signalome with upregulation of forkhead box O1 (FOXO1;1.4-fold,p < 0.0001) and downregulation of WNT signaling with MMP-12 cleavage of the adherens junction components β-catenin, cahderin-3, and catenin-α2. In conclusion, neutrophils shifted phenotype by stimuli, with MMP-12 inducing a unique apoptotic signalome with higher resemblance to the anti-inflammatory signalome. SIGNIFICANCE: This study revealed that neutrophils demonstrate unique polarization signaling responses to specific stimuli, with the matrix metalloproteinase (MMP)-12 signalome showing similarity to the IL-4 signalome. MMP-12 polarized neutrophils towards a strong apoptotic signature by upregulating FOXO1 and downregulating WNT signaling. Our results highlight that neutrophils display more plasticity than previously appreciated.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198, United States of America; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States of America
| | - Mediha Becirovic-Agic
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198, United States of America; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States of America
| | - Shelby R Konfrst
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198, United States of America; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States of America
| | - Jocelyn R Rodriguez-Paar
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198, United States of America; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States of America
| | - Leah M Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States of America
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198, United States of America; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States of America.
| |
Collapse
|
12
|
Simões G, Pereira T, Caseiro A. Matrix metaloproteinases in vascular pathology. Microvasc Res 2022; 143:104398. [PMID: 35671836 DOI: 10.1016/j.mvr.2022.104398] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 05/01/2022] [Accepted: 06/01/2022] [Indexed: 12/07/2022]
Abstract
Vascular diseases are the main cause of morbidity and mortality. The vascular extracellular matrix (ECM) is essential in mechanical support, also regulating the cellular behavior fundamental to vascular function and homeostasis. Vascular remodeling is an adaptive response to various physiological and pathological changes and is associated with aging and vascular diseases. The aim of this review is provide a general overview of the involvement of MMPs in the pathogenesis of vascular diseases, namely, arterial hypertension, atherosclerosis, aortic aneurysms and myocardial infarction. The change in the composition of the ECM by matrix metalloproteinases (MMPs) generates a pro-inflammatory microenvironment that modifies the phenotypes of endothelial cells and vascular smooth muscle cells. They play a central role in morphogenesis, tissue repair and remodeling in response to injury, e.g., after myocardial infarction, and in progression of diseases such as atherosclerosis. Alterations in specific MMPs could influence arterial remodeling and lead to various pathological disorders such as hypertension and aneurysm formation. MMPs are regulated by endogenous tissue inhibitors of metalloproteinases (TIMPs), and the MMP/TIMP ratio generally determines the extent of ECM protein degradation and tissue remodeling. Studies are currently focused on improving the diagnostic and prognostic value of MMPs involved in the pathogenic process, increasing their therapeutic potential, and monitoring the disease. New selective MMP inhibitors may improve the specificity of these inhibitors, target specific MMPs in relevant pathological conditions and mitigate some of the side effects.
Collapse
Affiliation(s)
- Gonçalo Simões
- Politécnico de Coimbra, ESTeSC, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal.
| | - Telmo Pereira
- LABINSAÚDE - Laboratório de Investigação em Ciências Aplicadas à Saúde, Instituto Politécnico de Coimbra, ESTeSC, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; Politécnico de Coimbra, ESTeSC, Fisiologia Clínica, Rua 5 de Outubro, 3046-854 Coimbra, Portugal.
| | - Armando Caseiro
- Politécnico de Coimbra, ESTeSC, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; LABINSAÚDE - Laboratório de Investigação em Ciências Aplicadas à Saúde, Instituto Politécnico de Coimbra, ESTeSC, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; Unidade I&D Química-Física Molecular, Faculdade de Ciências e Tecnologia, Universidade de Coimbra, Coimbra, Portugal.
| |
Collapse
|
13
|
Kologrivova I, Shtatolkina M, Suslova T, Ryabov V. Cells of the Immune System in Cardiac Remodeling: Main Players in Resolution of Inflammation and Repair After Myocardial Infarction. Front Immunol 2021; 12:664457. [PMID: 33868315 PMCID: PMC8050340 DOI: 10.3389/fimmu.2021.664457] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
The burden of heart failure (HF), developing after myocardial infarction MI, still represents a major issue in clinical practice. Failure of appropriate resolution of inflammation during post-myocardial injury is associated with unsuccessful left ventricular remodeling and underlies HF pathogenesis. Cells of the immune system have been shown to mediate both protective and damaging effects in heart remodeling. This ambiguity of the role of the immune system and inconsistent results of the recent clinical trials question the benefits of anti-inflammatory therapies during acute MI. The present review will summarize knowledge of the roles that different cells of the immune system play in the process of post-infarct cardiac healing. Data on the phenotype, active molecules and functions of the immune cells, based on the results of both experimental and clinical studies, will be provided. For some cellular subsets, such as macrophages, neutrophils, dendritic cells and lymphocytes, an anti-inflammatory activity has been attributed to the specific subpopulations. Activity of other cells, such as eosinophils, mast cells, natural killer (NK) cells and NKT cells has been shown to be highly dependent of the signals created by micro-environment. Also, new approaches for classification of cellular phenotypes based on the single-cell RNA sequencing allow better understanding of the phenotype of the cells involved in resolution of inflammation. Possible perspectives of immune-mediated therapy for AMI patients are discussed in the conclusion. We also outline unresolved questions that need to be solved in order to implement the current knowledge on the role of the immune cells in post-MI tissue repair into practice.
Collapse
Affiliation(s)
- Irina Kologrivova
- Department of Clinical Laboratory Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Marina Shtatolkina
- Department of Emergency Cardiology, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Tatiana Suslova
- Department of Clinical Laboratory Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Vyacheslav Ryabov
- Department of Emergency Cardiology, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia.,Division of Cardiology, Department of Professional Development and Retraining, Siberian State Medical University, Tomsk, Russia
| |
Collapse
|
14
|
Liu T, Balzano-Nogueira L, Lleo A, Conesa A. Transcriptional Differences for COVID-19 Disease Map Genes between Males and Females Indicate a Different Basal Immunophenotype Relevant to the Disease. Genes (Basel) 2020; 11:genes11121447. [PMID: 33271804 PMCID: PMC7761414 DOI: 10.3390/genes11121447] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/28/2020] [Accepted: 11/29/2020] [Indexed: 12/13/2022] Open
Abstract
Worldwide COVID-19 epidemiology data indicate differences in disease incidence amongst sex and gender demographic groups. Specifically, male patients are at a higher death risk than female patients, and the older population is significantly more affected than young individuals. Whether this difference is a consequence of a pre-existing differential response to the virus, has not been studied in detail. We created DeCovid, an R shiny app that combines gene expression (GE) data of different human tissue from the Genotype-Tissue Expression (GTEx) project along with the COVID-19 Disease Map and COVID-19 related pathways gene collections to explore basal GE differences across healthy demographic groups. We used this app to study differential gene expression of COVID-19 associated genes in different age and sex groups. We identified that healthy women show higher expression-levels of interferon genes. Conversely, healthy men exhibit higher levels of proinflammatory cytokines. Additionally, young people present a stronger complement system and maintain a high level of matrix metalloproteases than older adults. Our data suggest the existence of different basal immunophenotypes amongst different demographic groups, which are relevant to COVID-19 progression and may contribute to explaining sex and age biases in disease severity. The DeCovid app is an effective and easy to use tool for exploring the GE levels relevant to COVID-19 across demographic groups and tissues.
Collapse
Affiliation(s)
- Tianyuan Liu
- Microbiology and Cell Science, Institute for Food and Agricultural Research, University of Florida, Gainesville, FL 32611, USA; (T.L.); (L.B.-N.)
| | - Leandro Balzano-Nogueira
- Microbiology and Cell Science, Institute for Food and Agricultural Research, University of Florida, Gainesville, FL 32611, USA; (T.L.); (L.B.-N.)
| | - Ana Lleo
- Internal Medicine and Hepatology, Humanitas Clinical and Research Center-IRCCS, Department of Biomedical Sciences, Humanitas University, MI 20089 Rozzano, Italy;
| | - Ana Conesa
- Microbiology and Cell Science, Institute for Food and Agricultural Research, University of Florida, Gainesville, FL 32611, USA; (T.L.); (L.B.-N.)
- Genetics Institute, University of Florida, Gainesville, FL 32608, USA
- Correspondence: ; Tel.: +1-352-273-8127
| |
Collapse
|
15
|
Trypanosoma cruzi Induces the PARP1/AP-1 Pathway for Upregulation of Metalloproteinases and Transforming Growth Factor β in Macrophages: Role in Cardiac Fibroblast Differentiation and Fibrosis in Chagas Disease. mBio 2020; 11:mBio.01853-20. [PMID: 33172999 PMCID: PMC7667027 DOI: 10.1128/mbio.01853-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiomyopathy is the most important clinical manifestation of T. cruzi-driven CD. Recent studies have suggested the detrimental role of the matrix metalloproteinases MMP2 and MMP9 in extracellular matrix (ECM) degradation during cardiac remodeling in T. cruzi infection. Peripheral TGF-β levels are increased in clinically symptomatic CD patients over those in clinically asymptomatic seropositive individuals. We provide the first evidence that during T. cruzi infection, Mϕ release of MMP2 and MMP9 plays an active role in activation of TGF-β signaling of ECM remodeling and cardiac fibroblast-to-myofibroblast differentiation. We also determined that PARP1 signals c-Fos- and JunB-mediated AP-1 transcriptional activation of profibrotic gene expression and demonstrated the significance of PARP1 inhibition in controlling chronic fibrosis in Chagas disease. Our study provides a promising therapeutic approach for controlling T. cruzi-driven fibroblast differentiation in CD by PARP1 inhibitors through modulation of the Mϕ signaling of the AP-1–MMP9–TGF-β pathway. Chagas disease (CD), caused by Trypanosoma cruzi, is a degenerative heart condition. In the present study, we investigated the role of poly [ADP-ribose] polymerase 1/activator protein 1 (PARP1/AP-1) in upregulation of profibrotic macrophages (Mϕ) and subsequent development of cardiac fibrosis in CD. We used in vitro and in vivo models of T. cruzi infection and chemical and genetic inhibition of Parp1 to examine the molecular mechanisms by which Mϕ might augment profibrotic events in CD. Cultured (RAW 264.7 and THP-1) Mϕ infected with T. cruzi and primary cardiac and splenic Mϕ of chronically infected mice exhibited a significant increase in the expression, activity, and release of metalloproteinases (MMP2, MMP9, and MMP12) and the cytokine transforming growth factor β (TGF-β). Mϕ release of MMPs and TGF-β signaled the cardiac fibroblast to myofibroblast differentiation, as evidenced by a shift from S100A4 to alpha smooth muscle actin (α-SMA) expression. Incubation of infected Mϕ with MMP2 and MMP9 inhibitors resulted in 60 to 74% decline in TGF-β release, and MMP9 and PARP1 inhibitors resulted in 57 to 70% decline in Mϕ TGF-β-driven cardiac fibroblast differentiation. Likewise, histological studies showed a 12- to 16-fold increase in myocardial expression of CD68 (Mϕ marker) and its colocalization with MMP9/TGF-β, galectin-3, and vimentin in wild-type mice with CD. In comparison, chronically infected Parp1−/− mice exhibited a >50% decline in myocardial levels of Mϕ and associated fibrosis markers. Further study showed that PARP1 synergized with c-Fos and JunB AP-1 family members for transcriptional activation of profibrotic response after T. cruzi infection. We conclude that PARP1 inhibition offers a potential therapy for controlling the T. cruzi-driven fibroblast differentiation in CD through modulation of the Mϕ signaling of the AP-1–MMP9–TGF-β pathway.
Collapse
|
16
|
Steven S, Frenis K, Kalinovic S, Kvandova M, Oelze M, Helmstädter J, Hahad O, Filippou K, Kus K, Trevisan C, Schlüter KD, Boengler K, Chlopicki S, Frauenknecht K, Schulz R, Sorensen M, Daiber A, Kröller-Schön S, Münzel T. Exacerbation of adverse cardiovascular effects of aircraft noise in an animal model of arterial hypertension. Redox Biol 2020; 34:101515. [PMID: 32345536 PMCID: PMC7327989 DOI: 10.1016/j.redox.2020.101515] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023] Open
Abstract
Arterial hypertension is the most important risk factor for the development of cardiovascular disease. Recently, aircraft noise has been shown to be associated with elevated blood pressure, endothelial dysfunction, and oxidative stress. Here, we investigated the potential exacerbated cardiovascular effects of aircraft noise in combination with experimental arterial hypertension. C57BL/6J mice were infused with 0.5 mg/kg/d of angiotensin II for 7 days, exposed to aircraft noise for 7 days at a maximum sound pressure level of 85 dB(A) and a mean sound pressure level of 72 dB(A), or subjected to both stressors. Noise and angiotensin II increased blood pressure, endothelial dysfunction, oxidative stress and inflammation in aortic, cardiac and/or cerebral tissues in single exposure models. In mice subjected to both stressors, most of these risk factors showed potentiated adverse changes. We also found that mice exposed to both noise and ATII had increased phagocytic NADPH oxidase (NOX-2)-mediated superoxide formation, immune cell infiltration (monocytes, neutrophils and T cells) in the aortic wall, astrocyte activation in the brain, enhanced cytokine signaling, and subsequent vascular and cerebral oxidative stress. Exaggerated renal stress response was also observed. In summary, our results show an enhanced adverse cardiovascular effect between environmental noise exposure and arterial hypertension, which is mainly triggered by vascular inflammation and oxidative stress. Mechanistically, noise potentiates neuroinflammation and cerebral oxidative stress, which may be a potential link between both risk factors. The results indicate that a combination of classical (arterial hypertension) and novel (noise exposure) risk factors may be deleterious for cardiovascular health. Noise exposure causes non-auditory cardiovascular/cerebral adverse health effects by oxidative stress and inflammation. Aircraft noise causes exacerbated adverse effects on blood pressure and endothelial dysfunction in hypertensive mice. Aircraft noise and hypertension potentiate inflammation, ROS formation and oxidative damage in the brain, vessels and heart. Aircraft noise and hypertension seem to have enhanced adverse effects on stress responses in different organs.
Collapse
Affiliation(s)
- Sebastian Steven
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Katie Frenis
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Sanela Kalinovic
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Miroslava Kvandova
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Matthias Oelze
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Johanna Helmstädter
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Omar Hahad
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Konstantina Filippou
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Kamil Kus
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Chiara Trevisan
- Institute of Neuropathology, University Hospital, Zurich, Switzerland
| | | | - Kerstin Boengler
- Department of Physiology, Justus-Liebig University Gießen, Germany
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland; Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | | | - Rainer Schulz
- Department of Physiology, Justus-Liebig University Gießen, Germany
| | - Mette Sorensen
- Danish Cancer Society, Copenhagen, Denmark; Department of Natural Science and Environment, Roskilde University, Roskilde, Denmark
| | - Andreas Daiber
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| | - Swenja Kröller-Schön
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| |
Collapse
|
17
|
Koskimäki J, Zhang D, Li Y, Saadat L, Moore T, Lightle R, Polster SP, Carrión-Penagos J, Lyne SB, Zeineddine HA, Shi C, Shenkar R, Romanos S, Avner K, Srinath A, Shen L, Detter MR, Snellings D, Cao Y, Lopez-Ramirez MA, Fonseca G, Tang AT, Faber P, Andrade J, Ginsberg M, Kahn ML, Marchuk DA, Girard R, Awad IA. Transcriptome clarifies mechanisms of lesion genesis versus progression in models of Ccm3 cerebral cavernous malformations. Acta Neuropathol Commun 2019; 7:132. [PMID: 31426861 PMCID: PMC6699077 DOI: 10.1186/s40478-019-0789-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023] Open
Abstract
Cerebral cavernous malformations (CCMs) are dilated capillaries causing epilepsy and stroke. Inheritance of a heterozygous mutation in CCM3/PDCD10 is responsible for the most aggressive familial form of the disease. Here we studied the differences and commonalities between the transcriptomes of microdissected lesional neurovascular units (NVUs) from acute and chronic in vivo Ccm3/Pdcd10ECKO mice, and cultured brain microvascular endothelial cells (BMECs) Ccm3/Pdcd10ECKO.We identified 2409 differentially expressed genes (DEGs) in acute and 2962 in chronic in vivo NVUs compared to microdissected brain capillaries, as well as 121 in in vitro BMECs with and without Ccm3/Pdcd10 loss (fold change ≥ |2.0|; p < 0.05, false discovery rate corrected). A functional clustered dendrogram generated using the Euclidean distance showed that the DEGs identified only in acute in vivo NVUs were clustered in cellular proliferation gene ontology functions. The DEGs only identified in chronic in vivo NVUs were clustered in inflammation and immune response, permeability, and adhesion functions. In addition, 1225 DEGs were only identified in the in vivo NVUs but not in vitro BMECs, and these clustered within neuronal and glial functions. One miRNA mmu-miR-3472a was differentially expressed (FC = - 5.98; p = 0.07, FDR corrected) in the serum of Ccm3/Pdcd10+/- when compared to wild type mice, and this was functionally related as a putative target to Cand2 (cullin associated and neddylation dissociated 2), a DEG in acute and chronic lesional NVUs and in vitro BMECs. Our results suggest that the acute model is characterized by cell proliferation, while the chronic model showed inflammatory, adhesion and permeability processes. In addition, we highlight the importance of extra-endothelial structures in CCM disease, and potential role of circulating miRNAs as biomarkers of disease, interacting with DEGs. The extensive DEGs library of each model will serve as a validation tool for potential mechanistic, biomarker, and therapeutic targets.
Collapse
Affiliation(s)
- Janne Koskimäki
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Dongdong Zhang
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Yan Li
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Laleh Saadat
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Thomas Moore
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Sean P Polster
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Julián Carrión-Penagos
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Seán B Lyne
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Hussein A Zeineddine
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Changbin Shi
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Sharbel Romanos
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Kenneth Avner
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Abhinav Srinath
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Le Shen
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Matthew R Detter
- The Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC, USA
| | - Daniel Snellings
- The Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC, USA
| | - Ying Cao
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | | | - Gregory Fonseca
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Alan T Tang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Pieter Faber
- University of Chicago Genomics Facility, The University of Chicago, Chicago, IL, USA
| | - Jorge Andrade
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Mark Ginsberg
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas A Marchuk
- The Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC, USA
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA.
| |
Collapse
|
18
|
Guan C, Xiao Y, Li K, Wang T, Liang Y, Liao G. MMP-12 regulates proliferation of mouse macrophages via the ERK/P38 MAPK pathways during inflammation. Exp Cell Res 2019; 378:182-190. [DOI: 10.1016/j.yexcr.2019.03.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/28/2019] [Accepted: 03/10/2019] [Indexed: 12/25/2022]
|