1
|
Tenner AJ, Petrisko TJ. Knowing the enemy: strategic targeting of complement to treat Alzheimer disease. Nat Rev Neurol 2025; 21:250-264. [PMID: 40128350 DOI: 10.1038/s41582-025-01073-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 03/26/2025]
Abstract
The complement system protects against infection, positively responds to tissue damage, clears cell debris, directs and modulates the adaptive immune system, and functions in neuronal development, normal synapse elimination and intracellular metabolism. However, complement also has a role in aberrant synaptic pruning and neuroinflammation - processes that lead to a feedforward loop of inflammation, injury and neuronal death that can contribute to neurodegenerative and neurological disorders, including Alzheimer disease. This Review provides justification, largely from preclinical mouse models but also from correlates with human tissue and biomarkers, for targeting specific complement components for therapeutic intervention in Alzheimer disease. We discuss promising strategies to slow the progression of cognitive loss with minimal undesired effects. The diverse interactions and functions of complement system components can influence biological processes in the healthy and diseased brain; here, these functions are described as a prerequisite to selecting appropriate, safe and effective therapeutic targets for translation to the clinic.
Collapse
Affiliation(s)
- Andrea J Tenner
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA.
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA.
| | - Tiffany J Petrisko
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
2
|
Viiklepp K, Knuutila JS, Nissinen L, Siljamäki E, Rappu P, Suwal U, Pellinen T, Kallajoki M, Meri S, Heino J, Kähäri VM, Riihilä P. Expression of C1q by Macrophages and Fibroblasts in Tumor Microenvironment Is Associated with Progression and Metastasis of Cutaneous Squamous Cell Carcinoma. J Invest Dermatol 2025:S0022-202X(25)00446-4. [PMID: 40311866 DOI: 10.1016/j.jid.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 03/10/2025] [Accepted: 04/02/2025] [Indexed: 05/03/2025]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the most common metastatic skin cancer, with poor prognosis for metastatic cases. We demonstrated previously that cSCC cells in culture express C1r and C1s components of the complement C1qr2s2 complex but not C1q. In this study, significantly higher mRNA levels of C1QA, C1QB, and C1QC variants 1 and 2 were found in cSCC tumors than in normal skin. Analysis of single-cell RNA-sequencing data of cSCC revealed expression of mRNAs for C1QA, C1QB, and C1QC in macrophages and activated fibroblasts. C1q staining was detected on the surface of cSCC tumor cells, in peritumoral and intratumoral macrophages, and in peritumoral activated fibroblasts using immunohistochemistry and multiplexed immunofluorescence. Expression was higher in cSCCs than in normal skin, actinic keratoses, and cSCC in situ. C1q production was induced in 3-dimensional spheroid cocultures of cSCC cells, fibroblasts, and macrophages. C1q stimulated the growth of cSCC cells in culture. C1q expression was significantly more prevalent in metastatic primary cSCCs and in metastases than in non-metastatic cSCCs. High C1q expression in cSCC correlated with poor prognosis. These findings provide evidence for macrophage- and fibroblast-derived C1q in the progression of cSCC. They also suggest stromal C1q as a marker for cSCC metastasis and poor prognosis.
Collapse
Affiliation(s)
- Kristina Viiklepp
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Jaakko S Knuutila
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Liisa Nissinen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Elina Siljamäki
- MediCity Research Laboratory, University of Turku, Turku, Finland; Department of Life Technologies, University of Turku, Turku, Finland; InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Pekka Rappu
- MediCity Research Laboratory, University of Turku, Turku, Finland; Department of Life Technologies, University of Turku, Turku, Finland; InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Ujjwal Suwal
- MediCity Research Laboratory, University of Turku, Turku, Finland; Department of Life Technologies, University of Turku, Turku, Finland; InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Markku Kallajoki
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Seppo Meri
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Jyrki Heino
- MediCity Research Laboratory, University of Turku, Turku, Finland; Department of Life Technologies, University of Turku, Turku, Finland; InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Veli-Matti Kähäri
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland.
| | - Pilvi Riihilä
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland.
| |
Collapse
|
3
|
Wang C, Chen B, Yu X, Guan X. Macrophages Unmasked: Their Pivotal Role in Driving Atherosclerosis in Systemic Lupus Erythematosus. Clin Rev Allergy Immunol 2025; 68:10. [PMID: 39920534 DOI: 10.1007/s12016-025-09025-6] [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] [Accepted: 01/20/2025] [Indexed: 02/09/2025]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that significantly increases the risk of cardiovascular diseases, particularly atherosclerosis (AS). Understanding the shared pathogenic mechanisms underlying SLE and AS is crucial for developing effective therapeutic strategies. Macrophages, as pivotal immune cells, play a critical role in the initiation and progression of atherosclerotic plaques within the context of SLE. This review delves into the molecular and cellular mechanisms governing macrophage activation and differentiation in response to SLE-related inflammatory mediators, highlighting their roles in lipid metabolism, plaque stability, and immune regulation. Additionally, we discussed the current treatment modalities for SLE and their impact on macrophage functionality, exploring these effects for atherosclerotic progression. By elucidating the intricate relationship between macrophages, SLE pathophysiology, and AS progression, this review underscores the need for a multidisciplinary approach in managing SLE and its cardiovascular complications, aiming to improve patient survival and quality of life through tailored therapeutic interventions addressing both autoimmune and cardiovascular pathologies.
Collapse
Affiliation(s)
- Chao Wang
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, NanGang, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Bingxing Chen
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, NanGang, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Xiaochen Yu
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, NanGang, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Xiuru Guan
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, NanGang, Harbin, 150001, Heilongjiang, People's Republic of China.
| |
Collapse
|
4
|
Goldbach-Mansky R, Alehashemi S, de Jesus AA. Emerging concepts and treatments in autoinflammatory interferonopathies and monogenic systemic lupus erythematosus. Nat Rev Rheumatol 2025; 21:22-45. [PMID: 39623155 DOI: 10.1038/s41584-024-01184-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 12/22/2024]
Abstract
Over the past two decades, the number of genetically defined autoinflammatory interferonopathies has steadily increased. Aicardi-Goutières syndrome and proteasome-associated autoinflammatory syndromes (PRAAS, also known as CANDLE) are caused by genetic defects that impair homeostatic intracellular nucleic acid and protein processing respectively. Research into these genetic defects revealed intracellular sensors that activate type I interferon production. In SAVI and COPA syndrome, genetic defects that cause chronic activation of the dinucleotide sensor stimulator of interferon genes (STING) share features of lung inflammation and fibrosis; and selected mutations that amplify interferon-α/β receptor signalling cause central nervous system manifestations resembling Aicardi-Goutières syndrome. Research into the monogenic causes of childhood-onset systemic lupus erythematosus (SLE) demonstrates the pathogenic role of autoantibodies to particle-bound extracellular nucleic acids that distinguishes monogenic SLE from the autoinflammatory interferonopathies. This Review introduces a classification for autoinflammatory interferonopathies and discusses the divergent and shared pathomechanisms of interferon production and signalling in these diseases. Early success with drugs that block type I interferon signalling, new insights into the roles of cytoplasmic DNA or RNA sensors, pathways in type I interferon production and organ-specific pathology of the autoinflammatory interferonopathies and monogenic SLE, reveal novel drug targets that could personalize treatment approaches.
Collapse
Affiliation(s)
- Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Sara Alehashemi
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Adriana A de Jesus
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Zhao YF, Zuo ZA, Li ZY, Yuan Y, Hong SC, Fu WG, Zhou B, Wang LX. Integrated multi-omics profiling reveals neutrophil extracellular traps potentiate Aortic dissection progression. Nat Commun 2024; 15:10736. [PMID: 39737994 PMCID: PMC11686284 DOI: 10.1038/s41467-024-55038-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Adverse aortic remodeling increases the risk of aorta-related adverse events (AAEs) after thoracic endovascular aortic repair (TEVAR) and affects the overall prognosis of aortic dissection (AD). It is imperative to delve into the exploration of prognostic indicators to streamline the identification of individuals at elevated risk for postoperative AAEs, and therapeutic targets to optimize the efficacy of TEVAR for patients with AD. Here, we perform proteomic and single-cell transcriptomic analyses of peripheral blood and aortic lesions, respectively, from patients with AD and healthy subjects. The integrated multi-omics profiling identifies that highly phenotype-associated macrophages orchestrate neutrophil extracellular traps (NETs) through CXCL3/CXCR2 axis, thereby promoting the development of AD. Increased NETs formation is a defining feature of systemic immunity and aortic microenvironment of AD. Inhibiting NETs formation through the blockade of citrullinated histone H3 or CXCL3/CXCR2 axis ameliorates the progression and rupture of aortic dissection in male mice. The plasma level of citrullinated histone H3 predicts AAEs following endovascular therapy, facilitating the risk stratification and prognostic evaluation for patients with AD.
Collapse
Affiliation(s)
- Yu-Fei Zhao
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
- Vascular Surgery Institute of Fudan University, Fudan University, 200032, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 200032, Shanghai, China
| | - Zi-Ang Zuo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
- Vascular Surgery Institute of Fudan University, Fudan University, 200032, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Zhe-Yun Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
- Vascular Surgery Institute of Fudan University, Fudan University, 200032, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Ye Yuan
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
- Vascular Surgery Institute of Fudan University, Fudan University, 200032, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Shi-Chai Hong
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, 361015, Xiamen, China
- Xiamen Municipal Vascular Disease Precise Diagnose & Treatment Lab, Xiamen, China
| | - Wei-Guo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
- Vascular Surgery Institute of Fudan University, Fudan University, 200032, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, 361015, Xiamen, China
- Xiamen Municipal Vascular Disease Precise Diagnose & Treatment Lab, Xiamen, China
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Li-Xin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
- Vascular Surgery Institute of Fudan University, Fudan University, 200032, Shanghai, China.
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, 361015, Xiamen, China.
- Xiamen Municipal Vascular Disease Precise Diagnose & Treatment Lab, Xiamen, China.
| |
Collapse
|
6
|
Watanabe H, Rana M, Son M, Chiu PY, Fei-Bloom Y, Choi K, Diamond B, Sherry B. Single cell RNA-seq reveals cellular and transcriptional heterogeneity in the splenic CD11b +Ly6C high monocyte population expanded in sepsis-surviving mice. Mol Med 2024; 30:202. [PMID: 39506629 PMCID: PMC11539566 DOI: 10.1186/s10020-024-00970-0] [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: 05/01/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Sepsis survivors exhibit immune dysregulation that contributes to poor long-term outcomes. Phenotypic and functional alterations within the myeloid compartment are believed to be a contributing factor. Here we dissect the cellular and transcriptional heterogeneity of splenic CD11b+Ly6Chigh myeloid cells that are expanded in mice that survive the cecal ligation and puncture (CLP) murine model of polymicrobial sepsis to better understand the basis of immune dysregulation in sepsis survivors. METHODS Sham or CLP surgeries were performed on C57BL/6J and BALB/c mice. Four weeks later splenic CD11b+Ly6Chigh cells from both groups were isolated for phenotypic (flow cytometry) and functional (phagocytosis and glycolysis) characterization and RNA was obtained for single-cell RNA-seq (scRNA-seq) and subsequent analysis. RESULTS CD11b+Ly6Chigh cells from sham and CLP surviving mice exhibit phenotypic and functional differences that relate to immune function, some of which are observed in both C57BL/6J and BALB/c strains and others that are not. To dissect disease-specific and strain-specific distinctions within the myeloid compartment, scRNA-seq analysis was performed on CD11b+Ly6Chigh cells from C57BL/6J and BALB/c sham and CLP mice. Uniform Manifold Approximation and Projection from both strains identified 13 distinct clusters of sorted CD11b+Ly6Chigh cells demonstrating significant transcriptional heterogeneity and expressing gene signatures corresponding to classical-monocytes, non-classical monocytes, M1- or M2-like macrophages, dendritic-like cells, monocyte-derived dendritic-like cells, and proliferating monocytic myeloid-derived suppressor cells (M-MDSCs). Frequency plots showed that the percentages of proliferating M-MDSCs (clusters 8, 11 and 12) were increased in CLP mice compared to sham mice in both strains. Pathway and UCell score analysis in CLP mice revealed that cell cycle and glycolytic pathways were upregulated in proliferating M-MDSCs in both strains. Notably, granule protease genes were upregulated in M-MDSCs from CLP mice. ScRNA-seq analyses also showed that phagocytic pathways were upregulated in multiple clusters including the classical monocyte cluster, confirming the increased phagocytic capacity in CD11b+Ly6Chigh cells from CLP mice observed in ex vivo functional assays in C57BL/6J mice. CONCLUSION The splenic CD11b+Ly6Chigh myeloid populations expanded in survivors of CLP sepsis correspond to proliferating cells that have an increased metabolic demand and gene signatures consistent with M-MDSCs, a population known to have immunosuppressive capacity.
Collapse
Affiliation(s)
- Haruki Watanabe
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Minakshi Rana
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery at Weill Cornell Medicine, New York, New York, 10021, USA
| | - Myoungsun Son
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Pui Yan Chiu
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Yurong Fei-Bloom
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Betty Diamond
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA.
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA.
| | - Barbara Sherry
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA.
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA.
| |
Collapse
|
7
|
Pisu D, Johnston L, Mattila JT, Russell DG. The frequency of CD38 + alveolar macrophages correlates with early control of M. tuberculosis in the murine lung. Nat Commun 2024; 15:8522. [PMID: 39358361 PMCID: PMC11447019 DOI: 10.1038/s41467-024-52846-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024] Open
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis, remains an enduring global health challenge due to the limited efficacy of existing treatments. Although much research has focused on immune failure, the role of host macrophage biology in controlling the disease remains underappreciated. Here we show, through multi-modal single-cell RNA sequencing in a murine model, that different alveolar macrophage subsets play distinct roles in either advancing or controlling the disease. Initially, alveolar macrophages that are negative for the CD38 marker are the main infected population. As the infection progresses, CD38+ monocyte-derived and tissue-resident alveolar macrophages emerge as significant controllers of bacterial growth. These macrophages display a unique chromatin organization pre-infection, indicative of epigenetic priming for pro-inflammatory responses. Moreover, intranasal BCG immunization increases the numbers of CD38+ macrophages, enhancing their capability to restrict Mycobacterium tuberculosis growth. Our findings highlight the dynamic roles of alveolar macrophages in tuberculosis and open pathways for improved vaccines and therapies.
Collapse
Affiliation(s)
- Davide Pisu
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Luana Johnston
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
8
|
Lee S, Yoo I, Cheon Y, Ka H. Complement system molecules: Expression and regulation at the maternal-conceptus interface during pregnancy in pigs. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 159:105229. [PMID: 39004297 DOI: 10.1016/j.dci.2024.105229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
The complement system, composed of complement components and complement control proteins, plays an essential role in innate immunity. Complement system molecules are expressed at the maternal-conceptus interface, and inappropriate activation of the complement system is associated with various adverse pregnancy outcomes in humans and rodents. However, the expression, regulation, and function of the complement system at the maternal-conceptus interface in pigs have not been studied. In this study, we investigated the expression, localization, and regulation of complement system molecules at the maternal-conceptus interface in pigs. Complement components and complement control proteins were expressed in the endometrium, early-stage conceptus, and chorioallantoic tissues during pregnancy. The expression of complement components acting on the early stage of complement activation increased in the endometrium on Day 15 of pregnancy, with greater levels on that day compared with the estrous cycle. Localization of several complement components and complement control proteins was cell-type specific in the endometrium. The expression of C1QC, C2, C3, C4A, CFI, ITGB2, MASP1, and SERPING1 was increased by IFNG in endometrial explant tissues. Furthermore, cleaved C3 fragments were detected in endometrial tissues and uterine flushings on Day 15 of the estrous cycle and Day 15 of pregnancy, with greater levels on Day 15 of pregnancy. These results suggest that complement system molecules in pigs expressed at the maternal-conceptus interface play important roles in the establishment and maintenance of pregnancy by regulating innate immunity and modulating the maternal immune environment during pregnancy.
Collapse
Affiliation(s)
- Soohyung Lee
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea.
| | - Inkyu Yoo
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea
| | - Yugyeong Cheon
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea
| | - Hakhyun Ka
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea.
| |
Collapse
|
9
|
Lim J, La J, Kim HC, Kang I, Kang BH, Ku KB, Kim Y, Kwon MS, Lee HK. Type I interferon signaling regulates myeloid and T cell crosstalk in the glioblastoma tumor microenvironment. iScience 2024; 27:110810. [PMID: 39286510 PMCID: PMC11404196 DOI: 10.1016/j.isci.2024.110810] [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: 04/08/2024] [Revised: 08/02/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Downstream interferon signaling through the type I interferon (IFN) receptor, IFNAR, is crucial for the proper production of type I IFNs in mounting anti-tumor immune responses. Our study investigates the role of type I IFN signaling in the glioblastoma (GBM) tumor microenvironment by leveraging single-cell RNA sequencing to analyze tumor-infiltrating lymphocytes. We investigate how type I IFN signaling within the myeloid compartment contributes to the crosstalk with T cells in the tumor microenvironment. Through the use of the Gl261 murine GBM model, we find that the lack of proper type I IFN response results in enhanced PD-L1 interactions among myeloid cells, thereby affecting T cell functionality. Additionally, we also characterize how anti-PD1 treatment induces transcriptional changes in tumor-associated monocytes and macrophages by analyzing intercellular communication networks and propose how immune checkpoint blockade therapy could possibly relieve some of the immunosuppression derived from the lack of proper type I IFN production.
Collapse
Affiliation(s)
- Juhee Lim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jeongwoo La
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Hyeon Cheol Kim
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
- Life Science Institute, KAIST, Daejeon 34141, Republic of Korea
| | - In Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Byeong Hoon Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Keun Bon Ku
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
- Center for Infectious Disease Vaccine and Diagnosis Innovation, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Yumin Kim
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Myoung Seung Kwon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Heung Kyu Lee
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
- KAIST Institute of Health Science and Technology, KAIST, Daejeon 34141, Republic of Korea
| |
Collapse
|
10
|
Russell D, Pisu D, Mattila J, Johnston L. CD38+ Alveolar macrophages mediate early control of M. tuberculosis proliferation in the lung. RESEARCH SQUARE 2024:rs.3.rs-3934768. [PMID: 39070650 PMCID: PMC11275981 DOI: 10.21203/rs.3.rs-3934768/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Tuberculosis, caused by M.tuberculosis (Mtb), remains an enduring global health challenge, especially given the limited efficacy of current therapeutic interventions. Much of existing research has focused on immune failure as a driver of tuberculosis. However, the crucial role of host macrophage biology in controlling the disease remains underappreciated. While we have gained deeper insights into how alveolar macrophages (AMs) interact with Mtb, the precise AM subsets that mediate protection and potentially prevent tuberculosis progression have yet to be identified. In this study, we employed multi-modal scRNA-seq analyses to evaluate the functional roles of diverse macrophage subpopulations across different infection timepoints, allowing us to delineate the dynamic landscape of controller and permissive AM populations during the course of infection. Our analyses at specific time-intervals post-Mtb challenge revealed macrophage populations transitioning between distinct anti- and pro-inflammatory states. Notably, early in Mtb infection, CD38- AMs showed a muted response. As infection progressed, we observed a phenotypic shift in AMs, with CD38+ monocyte-derived AMs (moAMs) and a subset of tissue-resident AMs (TR-AMs) emerging as significant controllers of bacterial growth. Furthermore, scATAC-seq analysis of naïve lungs demonstrated that CD38+ TR-AMs possessed a distinct chromatin signature prior to infection, indicative of epigenetic priming and predisposition to a pro-inflammatory response. BCG intranasal immunization increased the numbers of CD38+ macrophages, substantially enhancing their capability to restrict Mtb growth. Collectively, our findings emphasize the pivotal, dynamic roles of different macrophage subsets in TB infection and reveal rational pathways for the development of improved vaccines and immunotherapeutic strategies.
Collapse
|
11
|
Kajimura Y, Taguchi A, Nagao Y, Yamamoto K, Masuda K, Shibata K, Asaoka Y, Furutani-Seiki M, Tanizawa Y, Ohta Y. E4BP4 in macrophages induces an anti-inflammatory phenotype that ameliorates the severity of colitis. Commun Biol 2024; 7:527. [PMID: 38714733 PMCID: PMC11076557 DOI: 10.1038/s42003-024-06099-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 03/22/2024] [Indexed: 05/10/2024] Open
Abstract
Macrophages are versatile cells of the innate immune system that work by altering their pro- or anti-inflammatory features. Their dysregulation leads to inflammatory disorders such as inflammatory bowel disease. We show that macrophage-specific upregulation of the clock output gene and transcription factor E4BP4 reduces the severity of colitis in mice. RNA-sequencing and single-cell analyses of macrophages revealed that increased expression of E4BP4 leads to an overall increase in expression of anti-inflammatory genes including Il4ra with a concomitant reduction in pro-inflammatory gene expression. In contrast, knockout of E4BP4 in macrophages leads to increased proinflammatory gene expression and decreased expression of anti-inflammatory genes. ChIP-seq and ATAC-seq analyses further identified Il4ra as a target of E4BP4, which drives anti-inflammatory polarization in macrophages. Together, these results reveal a critical role for E4BP4 in regulating macrophage inflammatory phenotypes and resolving inflammatory bowel diseases.
Collapse
Affiliation(s)
- Yasuko Kajimura
- Division of Endocrinology, Metabolism, Hematological Science and Therapeutics, Department of Bio-Signal Analysis, Yamaguchi University, Graduate School of Medicine, 1-1-1, Minami Kogushi, Ube, 755-8505, Japan
| | - Akihiko Taguchi
- Division of Endocrinology, Metabolism, Hematological Science and Therapeutics, Department of Bio-Signal Analysis, Yamaguchi University, Graduate School of Medicine, 1-1-1, Minami Kogushi, Ube, 755-8505, Japan.
| | - Yuko Nagao
- Division of Endocrinology, Metabolism, Hematological Science and Therapeutics, Department of Bio-Signal Analysis, Yamaguchi University, Graduate School of Medicine, 1-1-1, Minami Kogushi, Ube, 755-8505, Japan
| | - Kaoru Yamamoto
- Division of Endocrinology, Metabolism, Hematological Science and Therapeutics, Department of Bio-Signal Analysis, Yamaguchi University, Graduate School of Medicine, 1-1-1, Minami Kogushi, Ube, 755-8505, Japan
| | - Konosuke Masuda
- Division of Endocrinology, Metabolism, Hematological Science and Therapeutics, Department of Bio-Signal Analysis, Yamaguchi University, Graduate School of Medicine, 1-1-1, Minami Kogushi, Ube, 755-8505, Japan
| | - Kensuke Shibata
- Department of Microbiology and Immunology, Yamaguchi University, School of Medicine, 1-1-1, Minami Kogushi, Ube, 755-8505, Japan
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871, Japan
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yoichi Asaoka
- Department of Systems Biochemistry in Pathology and Regeneration, Yamaguchi University, School of Medicine, 1-1-1, Minami Kogushi, Ube, 755-8505, Japan
| | - Makoto Furutani-Seiki
- Department of Systems Biochemistry in Pathology and Regeneration, Yamaguchi University, School of Medicine, 1-1-1, Minami Kogushi, Ube, 755-8505, Japan
| | - Yukio Tanizawa
- Yamaguchi University, 1677-1, Yoshida, Yamaguchi, 753-8511, Japan
| | - Yasuharu Ohta
- Division of Endocrinology, Metabolism, Hematological Science and Therapeutics, Department of Bio-Signal Analysis, Yamaguchi University, Graduate School of Medicine, 1-1-1, Minami Kogushi, Ube, 755-8505, Japan
| |
Collapse
|
12
|
Wang Y, Zheng J, Li Q, Ma Y, Liu C, Deng J, Gao D. The relationship between complement C1q and coronary plaque vulnerability based on optical coherence tomography analysis. Sci Rep 2024; 14:9477. [PMID: 38658599 PMCID: PMC11043360 DOI: 10.1038/s41598-024-60128-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
To determine the association between complement C1q and vulnerable plaque morphology among coronary artery disease (CAD) patients. We conducted a retrospective observational study of 221 CAD patients admitted to The Second Affiliated Hospital of Xi'an Jiaotong University. Intravascular optical coherence tomography was utilized to describe the culprit plaques' morphology. Using logistic regression analysis to explore the correlation between C1q and vulnerable plaques, and receiver operator characteristic (ROC) analysis assess the predictive accuracy. As reported, the complement C1q level was lower in ACS patients than CCS patients (18.25 ± 3.88 vs. 19.18 ± 4.25, P = 0.045). The low complement-C1q-level group was more prone to develop vulnerable plaques. In lipid-rich plaques, the complement C1q level was positively correlated with the thickness of fibrous cap (r = 0.480, P = 0.041). Univariate and multivariate logistic regression analyses suggested that complement C1q could be an independent contributor to plaques' vulnerability. For plaque rupture, erosion, thrombus, and cholesterol crystals, the areas under the ROC curve of complement C1q level were 0.873, 0.816, 0.785, and 0.837, respectively (P < 0.05 for all). In CAD patients, the complement C1q could be a valuable indicator of plaque vulnerability.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an, 710000, Shaanxi, People's Republic of China
| | - Jiawei Zheng
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an, 710000, Shaanxi, People's Republic of China
| | - Qing Li
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an, 710000, Shaanxi, People's Republic of China
| | - Yao Ma
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an, 710000, Shaanxi, People's Republic of China
| | - Chang Liu
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an, 710000, Shaanxi, People's Republic of China
| | - Jie Deng
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an, 710000, Shaanxi, People's Republic of China
| | - Dengfeng Gao
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an, 710000, Shaanxi, People's Republic of China.
| |
Collapse
|
13
|
Ghosh M, Lee J, Burke AN, Strong TA, Sagen J, Pearse DD. Sex Dependent Disparities in the Central Innate Immune Response after Moderate Spinal Cord Contusion in Rat. Cells 2024; 13:645. [PMID: 38607084 PMCID: PMC11011714 DOI: 10.3390/cells13070645] [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: 01/31/2024] [Revised: 03/12/2024] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
Subacute spinal cord injury (SCI) displays a complex pathophysiology associated with pro-inflammation and ensuing tissue damage. Microglia, the resident innate immune cells of the CNS, in concert with infiltrating macrophages, are the primary contributors to SCI-induced inflammation. However, subpopulations of activated microglia can also possess immunomodulatory activities that are essential for tissue remodeling and repair, including the production of anti-inflammatory cytokines and growth factors that are vital for SCI recovery. Recently, reports have provided convincing evidence that sex-dependent differences exist in how microglia function during CNS pathologies and the extent to which these cells contribute to neurorepair and endogenous recovery. Herein we employed flow cytometry and immunohistochemical methods to characterize the phenotype and population dynamics of activated innate immune cells within the injured spinal cord of age-matched male and female rats within the first week (7 days) following thoracic SCI contusion. This assessment included the analysis of pro- and anti-inflammatory markers, as well as the expression of critical immunomodulatory kinases, including P38 MAPK, and transcription factors, such as NFκB, which play pivotal roles in injury-induced inflammation. We demonstrate that activated microglia from the injured spinal cord of female rats exhibited a significantly diminutive pro-inflammatory response, but enhanced anti-inflammatory activity compared to males. These changes included lower levels of iNOS and TLR4 expression but increased levels of ARG-1 and CD68 in females after SCI. The altered expression of these markers is indicative of a disparate secretome between the microglia of males and females after SCI and that the female microglia possesses higher phagocytic capabilities (increased CD68). The examination of immunoregulatory kinases and transcription factors revealed that female microglia had higher levels of phosphorylated P38Thr180/Tyr182 MAPK and nuclear NFκB pp50Ser337 but lower amounts of nuclear NFκB pp65Ser536, suggestive of an attenuated pro-inflammatory phenotype in females compared to males after SCI. Collectively, this work provides novel insight into some of the sex disparities that exist in the innate immune response after SCI and indicates that sex is an important variable when designing and testing new therapeutic interventions or interpretating positive or negative responses to an intervention.
Collapse
Affiliation(s)
- Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Jinyoung Lee
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
| | - Ashley N. Burke
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
| | - Thomas A. Strong
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
| | - Jacqueline Sagen
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
14
|
Wang J, Zhu N, Su X, Gao Y, Yang R. Novel tumor-associated macrophage populations and subpopulations by single cell RNA sequencing. Front Immunol 2024; 14:1264774. [PMID: 38347955 PMCID: PMC10859433 DOI: 10.3389/fimmu.2023.1264774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/30/2023] [Indexed: 02/15/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are present in almost all solid tumor tissues. 16They play critical roles in immune regulation, tumor angiogenesis, tumor stem cell activation, tumor invasion and metastasis, and resistance to therapy. However, it is unclear how TAMs perform these functions. With the application of single-cell RNA sequencing (scRNA-seq), it has become possible to identify TAM subpopulations associated with distinct functions. In this review, we discuss four novel TAM subpopulations in distinct solid tumors based on core gene signatures by scRNA-seq, including FCN1 +, SPP1 +, C1Q + and CCL18 + TAMs. Functional enrichment and gene expression in scRNA-seq data from different solid tumor tissues found that FCN1 + TAMs may induce inflammation; SPP1 + TAMs are potentially involved in metastasis, angiogenesis, and cancer cell stem cell activation, whereas C1Q + TAMs participate in immune regulation and suppression; And CCL18 + cells are terminal immunosuppressive macrophages that not only have a stronger immunosuppressive function but also enhance tumor metastasis. SPP1 + and C1Q + TAM subpopulations can be further divided into distinct populations with different functions. Meanwhile, we will also present emerging evidence highlighting the separating macrophage subpopulations associated with distinct functions. However, there exist the potential disconnects between cell types and subpopulations identified by scRNA-seq and their actual function.
Collapse
Affiliation(s)
- Juanjuan Wang
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Ningning Zhu
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Xiaomin Su
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yunhuan Gao
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Rongcun Yang
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
15
|
Xu C, Sarver DC, Lei X, Sahagun A, Zhong J, Na CH, Rudich A, Wong GW. CTRP6 promotes the macrophage inflammatory response, and its deficiency attenuates LPS-induced inflammation. J Biol Chem 2024; 300:105566. [PMID: 38103643 PMCID: PMC10789631 DOI: 10.1016/j.jbc.2023.105566] [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: 05/20/2023] [Revised: 11/27/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023] Open
Abstract
Macrophages play critical roles in inflammation and tissue homeostasis, and their functions are regulated by various autocrine, paracrine, and endocrine factors. We have previously shown that CTRP6, a secreted protein of the C1q family, targets both adipocytes and macrophages to promote obesity-linked inflammation. However, the gene programs and signaling pathways directly regulated by CTRP6 in macrophages remain unknown. Here, we combine transcriptomic and phosphoproteomic analyses to show that CTRP6 activates inflammatory gene programs and signaling pathways in mouse bone marrow-derived macrophages (BMDMs). Treatment of BMDMs with CTRP6 upregulated proinflammatory, and suppressed the antiinflammatory, gene expression. We also showed that CTRP6 activates p44/42-MAPK, p38-MAPK, and NF-κB signaling pathways to promote inflammatory cytokine secretion from BMDMs, and that pharmacologic inhibition of these signaling pathways markedly attenuated the effects of CTRP6. Pretreatment of BMDMs with CTRP6 also sensitized and potentiated the BMDMs response to lipopolysaccharide (LPS)-induced inflammatory signaling and cytokine secretion. Consistent with the metabolic phenotype of proinflammatory macrophages, CTRP6 treatment induced a shift toward aerobic glycolysis and lactate production, reduced oxidative metabolism, and elevated mitochondrial reactive oxygen species production in BMDMs. Importantly, in accordance with our in vitro findings, BMDMs from CTRP6-deficient mice were less inflammatory at baseline and showed a marked suppression of LPS-induced inflammatory gene expression and cytokine secretion. Finally, loss of CTRP6 in mice also dampened LPS-induced inflammation and hypothermia. Collectively, our findings suggest that CTRP6 regulates and primes the macrophage response to inflammatory stimuli and thus may have a role in modulating tissue inflammatory tone in different physiological and disease contexts.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xia Lei
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Ageline Sahagun
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jun Zhong
- Delta Omics Inc, Baltimore, Maryland, USA
| | - Chan Hyun Na
- Department of Neurology, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Assaf Rudich
- Faculty of Health Sciences, Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
16
|
Zhu ZX, Liu Y, Wang J, Xie Y, Li RY, Ma Q, Tu Q, Melhem NA, Couldwell S, El-Araby RE, Tai A, Van Dyke TE, Karimbux N, Jeong YN, Chen JJ. A novel lncRNA-mediated epigenetic regulatory mechanism in periodontitis. Int J Biol Sci 2023; 19:5187-5203. [PMID: 37928259 PMCID: PMC10620817 DOI: 10.7150/ijbs.87977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/01/2023] [Indexed: 11/07/2023] Open
Abstract
Periodontitis is a highly prevalent chronic inflammatory disease with an exaggerated host immune response, resulting in periodontal tissue destruction and potential tooth loss. The long non-coding RNA, LncR-ANRIL, located on human chromosome 9p21, is recognized as a genetic risk factor for various conditions, including atherosclerosis, periodontitis, diabetes, and cancer. LncR-APDC is an ortholog of ANRIL located on mouse genome chr4. This study aims to comprehend the regulatory role of lncR-APDC in periodontitis progression. Our experimental findings, obtained from lncR-APDC gene knockout (KO) mice with induced experimental periodontitis (EP), revealed exacerbated bone loss and disrupted pro-inflammatory cytokine regulation. Downregulation of osteogenic differentiation occurred in bone marrow stem cells harvested from lncR-APDC-KO mice. Furthermore, single-cell RNA sequencing of periodontitis gingival tissue revealed alterations in the proportion and function of immune cells, including T and B cells, macrophages, and neutrophils, due to lncR-APDC silencing. Our findings also unveiled a previously unidentified epithelial cell subset that is distinctively presenting in the lncR-APDC-KO group. This epithelial subset, characterized by the positive expression of Krt8 and Krt18, engages in interactions with immune cells through a variety of ligand-receptor pairs. The expression of Tff2, now recognized for its role in chronic inflammatory conditions, exhibited a notable increase across various tissue and cell types in lncR-APDC deficient mice. Additionally, our investigation revealed the potential for a direct binding interaction between lncR-APDC and Tff2. Intra-gingival administration of AAV9-lncR-APDC was shown to have therapeutic effects in the EP model. In conclusion, our results suggest that lncR-APDC plays a critical role in the progression of periodontal disease and holds therapeutic potential for periodontitis. Furthermore, the presence of the distinctive epithelial subpopulation and significantly elevated Tff2 levels in the lncR-APDC-silenced EP model offer new perspectives on the epigenetic regulation of periodontitis pathogenesis.
Collapse
Affiliation(s)
- Zoe Xiaofang Zhu
- Division of Oral Biology, Tufts University School of Dental Medicine, 136 Harrison Ave, M&V Building Room 830, Boston, MA 02111, United States
- Department of Periodontology, Tufts University School of Dental Medicine, Boston, MA, 02211, United States
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jinghao Wang
- Division of Oral Biology, Tufts University School of Dental Medicine, 136 Harrison Ave, M&V Building Room 830, Boston, MA 02111, United States
| | - Ying Xie
- Division of Oral Biology, Tufts University School of Dental Medicine, 136 Harrison Ave, M&V Building Room 830, Boston, MA 02111, United States
| | - Rachel Yuantong Li
- Division of Oral Biology, Tufts University School of Dental Medicine, 136 Harrison Ave, M&V Building Room 830, Boston, MA 02111, United States
| | - Qian Ma
- Division of Oral Biology, Tufts University School of Dental Medicine, 136 Harrison Ave, M&V Building Room 830, Boston, MA 02111, United States
| | - Qisheng Tu
- Division of Oral Biology, Tufts University School of Dental Medicine, 136 Harrison Ave, M&V Building Room 830, Boston, MA 02111, United States
- Department of Periodontology, Tufts University School of Dental Medicine, Boston, MA, 02211, United States
| | - Neiman A Melhem
- Division of Oral Biology, Tufts University School of Dental Medicine, 136 Harrison Ave, M&V Building Room 830, Boston, MA 02111, United States
| | - Sandrine Couldwell
- Division of Oral Biology, Tufts University School of Dental Medicine, 136 Harrison Ave, M&V Building Room 830, Boston, MA 02111, United States
- Department of Periodontology, Tufts University School of Dental Medicine, Boston, MA, 02211, United States
| | - Rady E. El-Araby
- Division of Oral Biology, Tufts University School of Dental Medicine, 136 Harrison Ave, M&V Building Room 830, Boston, MA 02111, United States
| | - Albert Tai
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
- Data Intensive Studies Center, Tufts University, Medford, MA, United States
| | - Thomas E. Van Dyke
- Clinical and Translational Research, Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Forsyth Institute, Boston, MA, United States
| | - Nadeem Karimbux
- Department of Periodontology, Tufts University School of Dental Medicine, Boston, MA, 02211, United States
| | - Y. Natalie Jeong
- Department of Periodontology, Tufts University School of Dental Medicine, Boston, MA, 02211, United States
| | - Jake Jinkun Chen
- Division of Oral Biology, Tufts University School of Dental Medicine, 136 Harrison Ave, M&V Building Room 830, Boston, MA 02111, United States
- Department of Periodontology, Tufts University School of Dental Medicine, Boston, MA, 02211, United States
- Department of Genetics, Molecular and Cell Biology, Tufts University School of Medicine, Tufts School of Graduate Biomedical Sciences, 136 Harrison Ave, M&V Room 811, Boston, MA 02111, United States
| |
Collapse
|
17
|
Ye J, Wen Z, Wu T, Chen L, Sheng L, Wang C, Teng C, Wu B, Xu J, Wei W. Single-Cell Sequencing Reveals the Optimal Time Window for Anti-Inflammatory Treatment in Spinal Cord Injury. Adv Biol (Weinh) 2023; 7:e2300098. [PMID: 37085744 DOI: 10.1002/adbi.202300098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/02/2023] [Indexed: 04/23/2023]
Abstract
Though the occurrence of neuroinflammation after spinal cord injury (SCI) is essential for antigen clearance and tissue repair, excessive inflammation results in cell death and axon dieback. The effect of anti-inflammatory medicine used in clinical treatment remains debatable owing to the inappropriate therapeutic schedule that does not align with the biological process of immune reaction. A better understanding of the immunity process is critical to promote effective anti-inflammatory therapeutics. However, cellular heterogeneity, which results in complex cellular functions, is a major challenge. This study performs single-cell RNA sequencing by profiling the tissue proximity to the injury site at different time points after SCI. Depending on the analysis of single-cell data and histochemistry observation, an appropriate time window for anti-inflammatory medicine treatment is proposed. This work also verifies the mechanism of typical anti-inflammatory medicine methylprednisolone sodium succinate (MPSS), which is found attributable to the activation inhibition of cells with pro-inflammatory phenotype through the downregulation of pathways such as TNF, IL2, and MIF. These pathways can also be provided as targets for anti-inflammatory treatment. Collectively, this work provides a therapeutic schedule of 1-3 dpi (days post injury) to argue against classical early pulse therapy and provides some pathways for target therapy in the future.
Collapse
Affiliation(s)
- Jingjia Ye
- International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Zhengfa Wen
- International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Tianxin Wu
- International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Liangliang Chen
- College of Computer Science and Technology, Zhejiang University, Hangzhou, 310000, China
| | - Lingchao Sheng
- International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Chenhuan Wang
- International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Chong Teng
- International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Bingbing Wu
- International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Jian Xu
- International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Wei Wei
- International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| |
Collapse
|
18
|
Zhang Y, Xiang G, Jiang AY, Lynch A, Zeng Z, Wang C, Zhang W, Fan J, Kang J, Gu SS, Wan C, Zhang B, Liu XS, Brown M, Meyer CA. MetaTiME integrates single-cell gene expression to characterize the meta-components of the tumor immune microenvironment. Nat Commun 2023; 14:2634. [PMID: 37149682 PMCID: PMC10164163 DOI: 10.1038/s41467-023-38333-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 04/26/2023] [Indexed: 05/08/2023] Open
Abstract
Recent advances in single-cell RNA sequencing have shown heterogeneous cell types and gene expression states in the non-cancerous cells in tumors. The integration of multiple scRNA-seq datasets across tumors can indicate common cell types and states in the tumor microenvironment (TME). We develop a data driven framework, MetaTiME, to overcome the limitations in resolution and consistency that result from manual labelling using known gene markers. Using millions of TME single cells, MetaTiME learns meta-components that encode independent components of gene expression observed across cancer types. The meta-components are biologically interpretable as cell types, cell states, and signaling activities. By projecting onto the MetaTiME space, we provide a tool to annotate cell states and signature continuums for TME scRNA-seq data. Leveraging epigenetics data, MetaTiME reveals critical transcriptional regulators for the cell states. Overall, MetaTiME learns data-driven meta-components that depict cellular states and gene regulators for tumor immunity and cancer immunotherapy.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Guanjue Xiang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Alva Yijia Jiang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Allen Lynch
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Zexian Zeng
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Chenfei Wang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Wubing Zhang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Jingyu Fan
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Jiajinlong Kang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Shengqing Stan Gu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Changxin Wan
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Boning Zhang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - X Shirley Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Clifford A Meyer
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
19
|
Association of serum complement C1q with cardiovascular outcomes among patients with acute coronary syndrome undergoing percutaneous coronary intervention. J Geriatr Cardiol 2022; 19:949-959. [PMID: 36632200 PMCID: PMC9807401 DOI: 10.11909/j.issn.1671-5411.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE To determine the association of serum complement C1q levels with cardiovascular outcomes among patients with acute coronary syndrome (ACS) undergoing percutaneous coronary intervention (PCI), and evaluate the value of C1q modified by high-sensitivity C-reactive protein (hs-CRP) levels as an independent predictor. METHODS As a single-center prospective observational study, we analyzed 1701 patients who had received primary or elective PCI for ACS at Beijing Anzhen Hospital, Capital Medical University, Beijing, China between June 1, 2016 and November 30, 2017. The associations of C1q modified by hs-CRP with major adverse cardiovascular events (MACE) were determined in survival analysis. RESULTS Patients with the lowest C1q tertile had the highest cumulative risk of MACE (log-rank P = 0.007). In fully adjusted Cox regression models, stratifying the total population according to hs-CRP dichotomy, C1q was significantly associated with MACE in patients with hs-CRP levels less than 2 mg/L but not in those with 2 mg/L or more (P interaction = 0.02). In patients with hs-CRP levels less than 2 mg/L, with the lowest C1q tertile as reference, the risk of MACE was reduced by 40.0% in the middle C1q tertile [hazard ratio (HR) = 0.600, 95% CI: 0.423-0.852, P = 0.004] and by 43.9% in the highest C1q tertile (HR = 0.561, 95% CI: 0.375-0.840, P = 0.005). CONCLUSIONS Serum complement C1q is significantly associated with cardiovascular outcomes in patients with ACS undergoing PCI, only when hs-CRP levels are less than 2 mg/L. This finding implicates the usefulness of C1q for the risk stratification in ACS patients with reduced systemic inflammation.
Collapse
|
20
|
Alic L, Binder CJ, Papac-Milicevic N. The OSE complotype and its clinical potential. Front Immunol 2022; 13:1010893. [PMID: 36248824 PMCID: PMC9561429 DOI: 10.3389/fimmu.2022.1010893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Cellular death, aging, and tissue damage trigger inflammation that leads to enzymatic and non-enzymatic lipid peroxidation of polyunsaturated fatty acids present on cellular membranes and lipoproteins. This results in the generation of highly reactive degradation products, such as malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE), that covalently modify free amino groups of proteins and lipids in their vicinity. These newly generated neoepitopes represent a unique set of damage-associated molecular patterns (DAMPs) associated with oxidative stress termed oxidation-specific epitopes (OSEs). OSEs are enriched on oxidized lipoproteins, microvesicles, and dying cells, and can trigger sterile inflammation. Therefore, prompt recognition and removal of OSEs is required to maintain the homeostatic balance. This is partially achieved by various humoral components of the innate immune system, such as natural IgM antibodies, pentraxins and complement components that not only bind OSEs but in some cases modulate their pro-inflammatory potential. Natural IgM antibodies are potent complement activators, and 30% of them recognize OSEs such as oxidized phosphocholine (OxPC-), 4-HNE-, and MDA-epitopes. Furthermore, OxPC-epitopes can bind the complement-activating pentraxin C-reactive protein, while MDA-epitopes are bound by C1q, C3a, complement factor H (CFH), and complement factor H-related proteins 1, 3, 5 (FHR-1, FHR-3, FHR-5). In addition, CFH and FHR-3 are recruited to 2-(ω-carboxyethyl)pyrrole (CEP), and full-length CFH also possesses the ability to attenuate 4-HNE-induced oxidative stress. Consequently, alterations in the innate humoral defense against OSEs predispose to the development of diseases associated with oxidative stress, as shown for the prototypical OSE, MDA-epitopes. In this mini-review, we focus on the mechanisms of the accumulation of OSEs, the pathophysiological consequences, and the interactions between different OSEs and complement components. Additionally, we will discuss the clinical potential of genetic variants in OSE-recognizing complement proteins – the OSE complotype - in the risk estimation of diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Lejla Alic
- Department of Medical Biochemistry, Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Nikolina Papac-Milicevic
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- *Correspondence: Nikolina Papac-Milicevic,
| |
Collapse
|
21
|
Nederlof R, Reidel S, Spychala A, Gödecke S, Heinen A, Lautwein T, Petzsch P, Köhrer K, Gödecke A. Insulin-Like Growth Factor 1 Attenuates the Pro-Inflammatory Phenotype of Neutrophils in Myocardial Infarction. Front Immunol 2022; 13:908023. [PMID: 35911749 PMCID: PMC9334797 DOI: 10.3389/fimmu.2022.908023] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Acute myocardial infarction (MI) induces an extensive sterile inflammation, which is dominated in the early phase by invading neutrophils and monocytes/macrophages. The inflammatory response after MI critically affects infarct healing and cardiac remodeling. Therefore, modulation of cardiac inflammation may improve outcome post MI. Insulin-like growth factor 1 (IGF1) treatment reduces infarct size and improves cardiac function after MI via IGF1 receptor mediated signaling in myeloid cells. Our study aimed to investigate the effect of IGF1 on neutrophil phenotype both in vitro and in vivo after MI. We show that IGF1 induces an anti-inflammatory phenotype in bone marrow derived neutrophils. On the molecular and functional level IGF1 treated neutrophils were indistinguishable from those induced by IL4. Surprisingly, insulin, even though it is highly similar to IGF1 did not create anti-inflammatory neutrophils. Notably, the IGF1 effect was independent of the canonical Ras/Raf/ERK or PI3K/AKT pathway, but depended on activation of the JAK2/STAT6 pathway, which was not activated by insulin treatment. Single cell sequencing analysis 3 days after MI also showed that 3 day IGF1 treatment caused a downregulation of pro-inflammatory genes and upstream regulators in most neutrophil and many macrophage cell clusters whereas anti-inflammatory genes and upstream regulators were upregulated. Thus, IGF1 acts like an anti-inflammatory cytokine on myeloid cells in vitro and attenuates the pro-inflammatory phenotype of neutrophils and macrophages in vivo after MI. IGF1 treatment might therefore represent an effective immune modulatory therapy to improve the outcome after MI.
Collapse
Affiliation(s)
- Rianne Nederlof
- Institut für Herz- und Kreislaufphysiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Sophia Reidel
- Institut für Herz- und Kreislaufphysiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - André Spychala
- Institut für Herz- und Kreislaufphysiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Stefanie Gödecke
- Institut für Herz- und Kreislaufphysiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - André Heinen
- Institut für Herz- und Kreislaufphysiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Tobias Lautwein
- Biologisch-Medizinisches Forschungszentrum (BMFZ), Genomics and Transcriptomics Labor, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Patrick Petzsch
- Biologisch-Medizinisches Forschungszentrum (BMFZ), Genomics and Transcriptomics Labor, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Karl Köhrer
- Biologisch-Medizinisches Forschungszentrum (BMFZ), Genomics and Transcriptomics Labor, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Axel Gödecke
- Institut für Herz- und Kreislaufphysiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
- *Correspondence: Axel Gödecke,
| |
Collapse
|
22
|
Son M. Understanding the contextual functions of C1q and LAIR-1 and their applications. Exp Mol Med 2022; 54:567-572. [PMID: 35562585 PMCID: PMC9098383 DOI: 10.1038/s12276-022-00774-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/03/2022] [Indexed: 11/09/2022] Open
Abstract
The importance of the complement component C1q has been highlighted by its involvement in autoimmunity, infection, inflammatory diseases, and tumors. The unique tulip-like structure of C1q has both a collagen-like stalk (C1q tail) and heterotrimeric globular head (gC1q), each with different binding specificities, and the binding of these components to their respective receptors leads to functional complexities in the body and bridges innate and adaptive immunity. This review describes the fundamental roles of C1q in various microenvironments and focuses on the importance of the interactions of C1q and its receptors with the inhibitory receptor LAIR-1 in maintaining homeostasis. Current therapeutic opportunities modulating LAIR-1 are also discussed. Research into the activities of the protein C1q, involved in a cascade of molecular interactions of the immune response called complement activation, is revealing new details of the protein’s role and opening up possible new therapeutic opportunities. Myoungsun Son at Feinstein Institutes for Medical Research in Manhasset, USA, reviews the involvement of C1q in infection, autoimmunity, inflammatory diseases and tumors. The interaction of C1q with a receptor protein called LAIR-1 seems to be particularly significant. LAIR-1 is present in the membrane of most blood-forming cells and is involved in maintaining the healthy balance of cellular activities referred to as homeostasis. Emerging research suggests that targeting the interactions between C1q and LAIR-1 could enable the development of new treatments for many diseases, including inflammatory diseases, the autoimmune condition lupus, a variety of cancers, and possibly Covid-19.
Collapse
Affiliation(s)
- Myoungsun Son
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, New York, USA. .,Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA.
| |
Collapse
|
23
|
Ye X, Wang L, Nie M, Wang Y, Dong S, Ren W, Li G, Li ZM, Wu K, Pan-Hammarström Q. A single-cell atlas of diffuse large B cell lymphoma. Cell Rep 2022; 39:110713. [PMID: 35443163 DOI: 10.1016/j.celrep.2022.110713] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/11/2022] [Accepted: 03/29/2022] [Indexed: 02/08/2023] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is one of the most common yet aggressive types of B cell lymphoma and remains incurable in 40% of patients. Herein, we profile the transcriptomes of 94,324 cells from 17 DLBCLs and 3 control samples using single-cell RNA sequencing. Altogether, 73 gene expression programs are identified in malignant cells, demonstrating high intra- and intertumor heterogeneity. Furthermore, 2,754 pairs of suggestive cell-cell interactions are predicted, indicating a complex and highly dynamic tumor microenvironment. Especially for B cell lymphomas, a strong costimulatory CD70-CD27 interaction is predicted between malignant and T cells. Furthermore, coinhibitory signals mediated by TIM3 or TIGIT seem to be the main driving force for T cell exhaustion. Finally, we find that chronic hepatitis B virus infection may have a significant impact on tumor cell survival and immune evasion in DLBCL. Our results provide insights into B cell lymphomagenesis and may facilitate the design of targeted immunotherapy strategies.
Collapse
Affiliation(s)
- Xiaofei Ye
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Stockholm, Sweden
| | - Lei Wang
- BGI-Shenzhen, Shenzhen 518000, China; Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI-Shenzhen, Shenzhen 518000, China
| | - Man Nie
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | | | | | - Weicheng Ren
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Stockholm, Sweden
| | - Guibo Li
- BGI-Shenzhen, Shenzhen 518000, China; Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518000, China
| | - Zhi-Ming Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Kui Wu
- BGI-Shenzhen, Shenzhen 518000, China; Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI-Shenzhen, Shenzhen 518000, China.
| | - Qiang Pan-Hammarström
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Stockholm, Sweden.
| |
Collapse
|
24
|
Revel M, Sautès-Fridman C, Fridman WH, Roumenina LT. C1q+ macrophages: passengers or drivers of cancer progression. Trends Cancer 2022; 8:517-526. [DOI: 10.1016/j.trecan.2022.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/24/2022]
|
25
|
Theocharidis G, Thomas BE, Sarkar D, Mumme HL, Pilcher WJR, Dwivedi B, Sandoval-Schaefer T, Sîrbulescu RF, Kafanas A, Mezghani I, Wang P, Lobao A, Vlachos IS, Dash B, Hsia HC, Horsley V, Bhasin SS, Veves A, Bhasin M. Single cell transcriptomic landscape of diabetic foot ulcers. Nat Commun 2022; 13:181. [PMID: 35013299 PMCID: PMC8748704 DOI: 10.1038/s41467-021-27801-8] [Citation(s) in RCA: 197] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetic foot ulceration (DFU) is a devastating complication of diabetes whose pathogenesis remains incompletely understood. Here, we profile 174,962 single cells from the foot, forearm, and peripheral blood mononuclear cells using single-cell RNA sequencing. Our analysis shows enrichment of a unique population of fibroblasts overexpressing MMP1, MMP3, MMP11, HIF1A, CHI3L1, and TNFAIP6 and increased M1 macrophage polarization in the DFU patients with healing wounds. Further, analysis of spatially separated samples from the same patient and spatial transcriptomics reveal preferential localization of these healing associated fibroblasts toward the wound bed as compared to the wound edge or unwounded skin. Spatial transcriptomics also validates our findings of higher abundance of M1 macrophages in healers and M2 macrophages in non-healers. Our analysis provides deep insights into the wound healing microenvironment, identifying cell types that could be critical in promoting DFU healing, and may inform novel therapeutic approaches for DFU treatment.
Collapse
Affiliation(s)
- Georgios Theocharidis
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics and Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Beena E Thomas
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Department of Pediatrics and Biomedical Informatics, Emory University, Atlanta, GA, USA
| | - Debasree Sarkar
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Department of Pediatrics and Biomedical Informatics, Emory University, Atlanta, GA, USA
| | - Hope L Mumme
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Department of Pediatrics and Biomedical Informatics, Emory University, Atlanta, GA, USA
| | - William J R Pilcher
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Department of Pediatrics and Biomedical Informatics, Emory University, Atlanta, GA, USA
| | - Bhakti Dwivedi
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | | | - Ruxandra F Sîrbulescu
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Antonios Kafanas
- Lincoln County Hospital, Northern Lincolnshire and Goole NHS Foundation Trust, Scunthorpe, UK
| | - Ikram Mezghani
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics and Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Peng Wang
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics and Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Antonio Lobao
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics and Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Ioannis S Vlachos
- Department of Pathology, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, MA, USA
| | - Biraja Dash
- Yale Plastic and Reconstructive Surgery-Wound Center, Yale School of Medicine, New Haven, CT, USA
| | - Henry C Hsia
- Yale Plastic and Reconstructive Surgery-Wound Center, Yale School of Medicine, New Haven, CT, USA
| | - Valerie Horsley
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Swati S Bhasin
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Department of Pediatrics and Biomedical Informatics, Emory University, Atlanta, GA, USA
| | - Aristidis Veves
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics and Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Manoj Bhasin
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Department of Pediatrics and Biomedical Informatics, Emory University, Atlanta, GA, USA.
| |
Collapse
|
26
|
Ramanjaneya M, Diboun I, Rizwana N, Dajani Y, Ahmed L, Butler AE, Almarzooqi TA, Shahata M, Al Bader MK, Elgassim E, Burjaq H, Atkin SL, Abou-Samra AB, Elrayess MA. Elevated Adipsin and Reduced C5a Levels in the Maternal Serum and Follicular Fluid During Implantation Are Associated With Successful Pregnancy in Obese Women. Front Endocrinol (Lausanne) 2022; 13:918320. [PMID: 35909516 PMCID: PMC9326155 DOI: 10.3389/fendo.2022.918320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Complement factors mediate the recruitment and activation of immune cells and are associated with metabolic changes during pregnancy. The aim of this study was to determine whether complement factors in the maternal serum and follicular fluid (FF) are associated with in vitro fertilization (IVF) outcomes in overweight/obese women. METHODS Forty overweight/obese (BMI = 30.8 ± 5.2 kg/m2) female patients, 33.6 ± 6.3 years old, undergoing IVF treatment for unexplained infertility were recruited. Baseline demographic information, including biochemical hormonal, metabolic, and inflammatory markers, and pregnancy outcome, was collected. Levels of 14 complement markers (C2, C4b, C5, C5a, C9, adipsin, mannose-binding lectin, C1q, C3, C3b/iC3b, C4, factor B, factor H, and properdin) were assessed in the serum and FF and compared to IVF outcome, inflammatory, and metabolic markers using multivariate and univariate models. RESULTS Out of 40 IVF cycles, 14 (35%) resulted in pregnancy. Compared to women with failed pregnancies, women with successful pregnancies had higher levels of adipsin in the serum and FF (p = 0.01) but lower C5a levels (p = 0.05). Serum adipsin levels were positively correlated with circulating levels of vitamin D (R = 0.5, p = 0.02), glucagon (R = 0.4, p = 0.03), leptin (R = 0.4, p = 0.01), resistin (R = 0.4, p = 0.02), and visfatin (R = 0.4, p = 0.02), but negatively correlated with total protein (R = -0.5, p = 0.03). Higher numbers of top-quality embryos were associated with increased levels of C3, properdin, C1q, factors H and B, C4, and adipsin, but with reduced C2 and C5a levels (p ≤ 0.01). CONCLUSIONS Higher adipsin and lower C5a levels in the maternal serum during implantation are potential markers of successful outcome in obese women undergoing IVF-assisted pregnancies.
Collapse
Affiliation(s)
- Manjunath Ramanjaneya
- Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar
- Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | | | - Najeha Rizwana
- Biomedical Research Center (BRC), Qatar University, Doha, Qatar
| | | | | | | | - Thoraya Ali Almarzooqi
- Obstetrics and Gynecology Department, Women’s Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Mohammed Shahata
- Obstetrics and Gynecology Department, Women’s Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Moza Khalaf Al Bader
- Obstetrics and Gynecology Department, Women’s Hospital, Hamad Medical Corporation, Doha, Qatar
| | | | - Hasan Burjaq
- Obstetrics and Gynecology Department, Women’s Hospital, Hamad Medical Corporation, Doha, Qatar
| | | | | | - Mohamed A. Elrayess
- Biomedical Research Center (BRC), Qatar University, Doha, Qatar
- QU Health, Qatar University, Doha, Qatar
- *Correspondence: Mohamed A. Elrayess,
| |
Collapse
|
27
|
Jiang S, Zeng Q, Zhao K, Liu J, Sun Q, Huang K, He Y, Zhang X, Wang H, Shi X, Feng C, Deng X, Wei Y. Chirality Bias Tissue Homeostasis by Manipulating Immunological Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105136. [PMID: 34601779 DOI: 10.1002/adma.202105136] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/15/2021] [Indexed: 06/13/2023]
Abstract
The physiological chirality of extracellular environments is substantially affected by pathological diseases. However, how this stereochemical variation drives host immunity remains poorly understood. Here, it is reported that pathology-mimetic M-nanofibrils-but not physiology-mimetic P-nanofibrils-act as a defense mechanism that helps to restore tissue homeostasis by manipulating immunological response. Quantitative multi-omics in vivo and in vitro shows that M-nanofibrils significantly inhibit inflammation and promote tissue regeneration by upregulating M2 macrophage polarization and downstream immune signaling compared with P-nanofibrils. Molecular analysis and theoretical simulation demonstrate that M-chirality displays higher stereo-affinity to cellular binding, which induces higher cellular contractile stress and activates mechanosensitive ion channel PIEZOl to conduct Ca2+ influx. In turn, the nuclear transfer of STAT is biased by Ca2+ influx to promote M2 polarization. These findings underscore the structural mechanisms of disease, providing design basis for immunotherapy with bionic functional materials.
Collapse
Affiliation(s)
- Shengjie Jiang
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Qiang Zeng
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Kai Zhao
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Jinying Liu
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng, 475004, P. R. China
| | - Qiannan Sun
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Kang Huang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Ying He
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Xuehui Zhang
- Department of Dental Materials and Dental Medical Devices Testing Center, National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Hui Wang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Xinghua Shi
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Chuanliang Feng
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiaotong University, Shanghai, 200240, P. R. China
| | - Xuliang Deng
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Yan Wei
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| |
Collapse
|
28
|
Xiong Z, Wang Q, Li W, Huang L, Zhang J, Zhu J, Xie B, Wang S, Kuang H, Lin X, Lee C, Kumar A, Li X. Platelet-Derived Growth Factor-D Activates Complement System to Propagate Macrophage Polarization and Neovascularization. Front Cell Dev Biol 2021; 9:686886. [PMID: 34150781 PMCID: PMC8207142 DOI: 10.3389/fcell.2021.686886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/10/2021] [Indexed: 11/17/2022] Open
Abstract
Platelet-derived growth factor-D (PDGF-D) is highly expressed in immune cells. However, the potential role of PDGF-D in immune system remains thus far unclear. Here, we reveal a novel function of PDGF-D in activating both classical and alternative complement pathways that markedly increase chemokine and cytokine responses to promote macrophage polarization. Pharmacological targeting of the complement C3a receptor using SB290157 alleviated PDGF-D-induced neuroinflammation by blocking macrophage polarization and inhibited pathological choroidal neovascularization. Our study thus suggests that therapeutic strategies targeting both PDGF-D and the complement system may open up new possibilities for the treatment of neovascular diseases.
Collapse
Affiliation(s)
- Zhen Xiong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Qianqian Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wanhong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lijuan Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jianing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Juanhua Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Bingbing Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shasha Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Haiqing Kuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xianchai Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chunsik Lee
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Anil Kumar
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
29
|
Jia Y, Wen W, Yang Y, Huang M, Ning Y, Jiao X, Liu S, Qin Y, Zhang M. The clinical role of combined serum C1q and hsCRP in predicting coronary artery disease. Clin Biochem 2021; 93:50-58. [PMID: 33861985 DOI: 10.1016/j.clinbiochem.2021.04.004] [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: 01/26/2021] [Revised: 03/19/2021] [Accepted: 04/06/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE C1q has been shown to be associated with coronary heart disease (CAD) and can co-deposit with C-reactive protein (CRP) in atherosclerotic plaques. However, few studies have been conducted between C1q, CRP parameters and CAD. The aim of this study is to explore the relationship between C1q and CRP parameters and assess their clinical significance in CAD. METHODS 238 total patients who underwent coronary artery angiography were enrolled and divided into control group (n = 65), stable CAD group (n = 47) and unstable angina group (UA group, n = 126). Patients' data were collected from self-administered questionnaires and electrical medical records. The severity of coronary stenosis was presented by Gensini score. The relationship between C1q, CRP parameters and CAD were evaluated by multivariate regression analysis and their predicting performance were assessed by ROC analysis and odds ratio analysis. RESULTS Compared with control group, C1q was showed significantly lower in stable CAD (P = 0.004) and UA groups (P = 0.008), while hsCRP was higher in UA group (P = 0.024). Serum C1q was weakly positively associated with hsCRP (r = 0.24, P < 0.001) but not correlated with Gensini score. Logistic regression identified C1q (OR: 0.87 per 10 mg/L, 95% CI: 0.79-0.95, P = 0.001) and hsCRP (OR: 1.08 mg/L, 95% CI: 1.01-1.15, P = 0.032) as independent determinants of CAD. Furthermore, combined C1q and hsCRP level showed higher discriminatory accuracy in predicting CAD than C1q (AUC: 0.676 vs 0.585, P = 0.101; NRI: 10.4%, P = 0.049; IDI: 3.9%, P < 0.001) or hsCRP (AUC: 0.676 vs 0.585, P = 0.101; NRI: 16.7%, P = 0.006; IDI: 5.8%, P < 0.001). CONCLUSIONS Reduced serum C1q and increased hsCRP are independently associated with CAD and could be potential predictors for CAD diagnosis. Furthermore, combined C1q and hsCRP showed better performance in predicting CAD than using single one.
Collapse
Affiliation(s)
- Yifan Jia
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Wanwan Wen
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yunxiao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mengling Huang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yu Ning
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xiaolu Jiao
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Sheng Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yanwen Qin
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ming Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| |
Collapse
|
30
|
Miah M, Goh I, Haniffa M. Prenatal Development and Function of Human Mononuclear Phagocytes. Front Cell Dev Biol 2021; 9:649937. [PMID: 33898444 PMCID: PMC8060508 DOI: 10.3389/fcell.2021.649937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
The human mononuclear phagocyte (MP) system, which includes dendritic cells, monocytes, and macrophages, is a critical regulator of innate and adaptive immune responses. During embryonic development, MPs derive sequentially in yolk sac progenitors, fetal liver, and bone marrow haematopoietic stem cells. MPs maintain tissue homeostasis and confer protective immunity in post-natal life. Recent evidence - primarily in animal models - highlight their critical role in coordinating the remodeling, maturation, and repair of target organs during embryonic and fetal development. However, the molecular regulation governing chemotaxis, homeostasis, and functional diversification of resident MP cells in their respective organ systems during development remains elusive. In this review, we summarize the current understanding of the development and functional contribution of tissue MPs during human organ development and morphogenesis and its relevance to regenerative medicine. We outline how single-cell multi-omic approaches and next-generation ex-vivo organ-on-chip models provide new experimental platforms to study the role of human MPs during development and disease.
Collapse
Affiliation(s)
- Mohi Miah
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Issac Goh
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.,Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Wellcome Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
31
|
Agostinis C, Balduit A, Mangogna A, Zito G, Romano F, Ricci G, Kishore U, Bulla R. Immunological Basis of the Endometriosis: The Complement System as a Potential Therapeutic Target. Front Immunol 2021; 11:599117. [PMID: 33505394 PMCID: PMC7829336 DOI: 10.3389/fimmu.2020.599117] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Endometriosis (EM) is a chronic disease characterized by the presence and proliferation of functional endometrial glands and stroma outside the uterine cavity. Ovaries and pelvic peritoneum are the most common locations for endometrial ectopic tissue, followed by deep infiltrating EM sites. The cyclic and recurrent bleeding, the progressive fibrosis and the peritoneal adhesions of ectopic endometrial glands, may cause different symptoms depending on the origin involved. EM is a frequent clinical condition affecting around 10% of women of mainly reproductive age, as well as in post-menopausal women and adolescents, especially with uterine anomalies. The risk of developing EM depends on a complex interaction between genetic, immunological, hormonal, and environmental factors. It is largely considered to arise due to a dysfunction of immunological surveillance. In fact, women with EM exhibit altered functions of peritoneal macrophages, lymphocytes and natural killer cells, as well as levels of inflammatory mediators and growth factors in the peritoneal fluid. In EM patients, peritoneal macrophages are preponderant and highly active compared to healthy women. Peritoneal macrophages are able to regulate the events that determine the production of cytokines, prostaglandins, growth factors and complement components. Several studies have shown alteration in the regulation of the complement activation, leading to chronic inflammation characteristic of EM. Aberrant regulation/activation of the complement system has been observed in the peritoneal cavity of women affected by EM. Thus, complement inhibition may represent a new approach for the treatment of EM, given that a number of complement inhibitors are under pre-clinical and clinical development. Such an intervention may provide a broader therapeutic control of complement-mediated inflammatory damage in EM patients. This review will focus on our current understanding of the role of complement activation in EM and possible modalities available for complement-based therapy.
Collapse
Affiliation(s)
- Chiara Agostinis
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy
| | - Andrea Balduit
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy
| | - Gabriella Zito
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy
| | - Federico Romano
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy.,Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
32
|
Li H, Chen J, Hu Y, Cai X, Zhang P. Elevated Serum C1q Levels in Children With Sepsis. Front Pediatr 2021; 9:619899. [PMID: 33981650 PMCID: PMC8109246 DOI: 10.3389/fped.2021.619899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/24/2021] [Indexed: 12/29/2022] Open
Abstract
Objective: To analyze the serum complement C1q levels in children with sepsis, and explore the suggestive effect of serum C1q levels on the condition of children with sepsis. Methods: The clinical and laboratory data of children with sepsis (n = 95) and healthy children (n = 71) in Renmin Hospital of Wuhan University from January 2019 to October 2019 were collected, and each index of the two groups was compared. Then we divided children with sepsis into three subgroups based on the Pediatric Critical Illness Score (PCIS): non-critical group, critical group, and extremely critical group. The serum C1q and PCT levels of the three subgroups were analyzed, and the correlation analysis was carried out between the levels of serum C1q and PCT levels as well as PCIS among children with sepsis. Finally, we analyzed the serum C1q levels of septic children infected by different pathogens. Results: The serum C1q levels of children with sepsis were significantly higher than those of healthy children (median 198.4 vs. 186.2 mg/L, P < 0.001). In the analysis of subgroups, the serum C1q levels of non-critical group, critical group, and extremely critical group septic children were 182.80 (166.75, 195.85) mg/L, 219.90 (209.10, 246.40) mg/L and 249.95 (239.10, 272.25) mg/L, respectively, which were correlated with the severity of the disease. At the same time, we also found that serum C1q in children with sepsis was positively correlated with PCT levels (r = 0.5982, P < 0.001), and negatively correlated with PCIS score (r = -0.6607, P < 0.001). The serum C1q levels of septic children with bacterial infections, mycoplasma infections, viral infections, and co-infection were higher than those of the control group (P < 0.05). Conclusion: The serum levels of C1q in children with sepsis were increased and related to the severity of sepsis, suggesting that C1q may be involved in the occurrence and development of sepsis, which had reference value for the preliminary diagnosis and severity classification of sepsis.
Collapse
Affiliation(s)
- Huan Li
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Juanjuan Chen
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuanhui Hu
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Cai
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Pingan Zhang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
33
|
Sachs S, Niu L, Geyer P, Jall S, Kleinert M, Feuchtinger A, Stemmer K, Brielmeier M, Finan B, DiMarchi RD, Tschöp MH, Wewer Albrechtsen N, Mann M, Müller TD, Hofmann SM. Plasma proteome profiles treatment efficacy of incretin dual agonism in diet-induced obese female and male mice. Diabetes Obes Metab 2021; 23:195-207. [PMID: 33001570 DOI: 10.1111/dom.14215] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/21/2020] [Accepted: 09/26/2020] [Indexed: 02/06/2023]
Abstract
AIMS Unimolecular peptides targeting the receptors for glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) (GLP-1/GIP co-agonist) have been shown to outperform each single peptide in the treatment of obesity and cardiometabolic disease in preclinical and clinical trials. By combining physiological treatment endpoints with plasma proteomic profiling (PPP), we aimed to identify biomarkers to advance non-invasive metabolic monitoring of compound treatment success and exploration of ulterior treatment effects on an individual basis. MATERIALS AND METHODS We performed metabolic phenotyping along with PPP in body weight-matched male and female diet-induced obese (DIO) mice treated for 21 days with phosphate-buffered saline, single GIP and GLP-1 mono-agonists, or a GLP-1/GIP co-agonist. RESULTS GLP-1R/GIPR co-agonism improved obesity, glucose intolerance, non-alcoholic fatty liver disease (NAFLD) and dyslipidaemia with superior efficacy in both male and female mice compared with mono-agonist treatments. PPP revealed broader changes of plasma proteins after GLP-1/GIP co-agonist compared with mono-agonist treatments in both sexes, including established and potential novel biomarkers for systemic inflammation, NAFLD and atherosclerosis. Subtle sex-specific differences have been observed in metabolic phenotyping and PPP. CONCLUSIONS We herein show that a recently developed unimolecular GLP-1/GIP co-agonist is more efficient in improving metabolic disease than either mono-agonist in both sexes. PPP led to the identification of a sex-independent protein panel with the potential to monitor non-invasively the treatment efficacies on metabolic function of this clinically advancing GLP-1/GIP co-agonist.
Collapse
Affiliation(s)
- Stephan Sachs
- Institute for Diabetes and Regeneration, Helmholtz Diabetes Center at Helmholtz Centre Munich, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - Lili Niu
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Philipp Geyer
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Sigrid Jall
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - Maximilian Kleinert
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Neuherberg, Germany
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Center Munich, Neuherberg, Germany
| | - Kerstin Stemmer
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Markus Brielmeier
- Helmholtz Zentrum München-German Research Center for Environmental Health, Research Unit Comparative Medicine, Neuherberg, Germany
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, Indiana
| | - Richard D DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, Indiana
- Department of Chemistry, Indiana University, Bloomington, Indiana
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Nicolai Wewer Albrechtsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany
| | - Susanna M Hofmann
- Institute for Diabetes and Regeneration, Helmholtz Diabetes Center at Helmholtz Centre Munich, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV, Klinikum der LMU, Munich, Germany
| |
Collapse
|
34
|
Abundance of Non-Polarized Lung Macrophages with Poor Phagocytic Function in Chronic Obstructive Pulmonary Disease (COPD). Biomedicines 2020; 8:biomedicines8100398. [PMID: 33050042 PMCID: PMC7650830 DOI: 10.3390/biomedicines8100398] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 02/08/2023] Open
Abstract
Lung macrophages are the key immune effector cells in the pathogenesis of Chronic Obstructive Pulmonary Disease (COPD). Several studies have shown an increase in their numbers in bronchoalveolar lavage fluid (BAL) of subjects with COPD compared to controls, suggesting a pathogenic role in disease initiation and progression. Although reduced lung macrophage phagocytic ability has been previously shown in COPD, the relationship between lung macrophages' phenotypic characteristics and functional properties in COPD is still unclear. (1) Methods: Macrophages harvested from bronchoalveolar lavage (BAL) fluid of subjects with and without COPD (GOLD grades, I-III) were immuno-phenotyped, and their function and gene expression profiles were assessed using targeted assays. (2) Results: BAL macrophages from 18 COPD and 10 (non-COPD) control subjects were evaluated. The majority of macrophages from COPD subjects were non-polarized (negative for both M1 and M2 markers; 77.9%) in contrast to controls (23.9%; p < 0.001). The percentages of these non-polarized macrophages strongly correlated with the severity of COPD (p = 0.006) and current smoking status (p = 0.008). Non-polarized macrophages demonstrated poor phagocytic function in both the control (p = 0.02) and COPD (p < 0.001) subjects. Non-polarized macrophages demonstrated impaired ability to phagocytose Staphylococcus aureus (p < 0.001). They also demonstrated reduced gene expression for CD163, CD40, CCL13 and C1QA&B, which are involved in pathogen recognition and processing and showed an increased gene expression for CXCR4, RAF1, amphiregulin and MAP3K5, which are all involved in promoting the inflammatory response. (3) Conclusions: COPD is associated with an abundance of non-polarized airway macrophages that is related to the severity of COPD. These non-polarized macrophages are predominantly responsible for the poor phagocytic capacity of lung macrophages in COPD, having reduced capacity for pathogen recognition and processing. This could be a key risk factor for COPD exacerbation and could contribute to disease progression.
Collapse
|
35
|
Hosszu KK, Valentino A, Peerschke EI, Ghebrehiwet B. SLE: Novel Postulates for Therapeutic Options. Front Immunol 2020; 11:583853. [PMID: 33117397 PMCID: PMC7575694 DOI: 10.3389/fimmu.2020.583853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/10/2020] [Indexed: 12/19/2022] Open
Abstract
Genetic deficiency in C1q is a strong susceptibility factor for systemic lupus erythematosus (SLE). There are two major hypotheses that potentially explain the role of C1q in SLE. The first postulates that C1q deficiency abrogates apoptotic cell clearance, leading to persistently high loads of potentially immunogenic self-antigens that trigger autoimmune responses. While C1q undoubtedly plays an important role in apoptotic clearance, an essential biological process such as removal of self- waste is so critical for host survival that multiple ligand-receptor combinations do fortunately exist to ensure that proper disposal of apoptotic debris is accomplished even in the absence of C1q. The second hypothesis is based on the observation that locally synthesized C1q plays a critical role in regulating the earliest stages of monocyte to dendritic cell (DC) differentiation and function. Indeed, circulating C1q has been shown to keep monocytes in a pre-dendritic state by silencing key molecular players and ensuring that unwarranted DC-driven immune responses do not occur. Monocytes are also able to display macromolecular C1 on their surface, representing a novel mechanism for the recognition of circulating "danger." Translation of this danger signal in turn, provides the requisite "license" to trigger a differentiation pathway that leads to adaptive immune response. Based on this evidence, the second hypothesis proposes that deficiency in C1q dysregulates monocyte-to-DC differentiation and causes inefficient or defective maintenance of self-tolerance. The fact that C1q receptors (cC1qR and gC1qR) are also expressed on the surface of both monocytes and DCs, suggests that C1q/C1qR may regulate DC differentiation and function through specific cell-signaling pathways. While their primary ligand is C1q, C1qRs can also independently recognize a vast array of plasma proteins as well as pathogen-associated molecular ligands, indicating that these molecules may collaborate in antigen recognition and processing, and thus regulate DC-differentiation. This review will therefore focus on the role of C1q and C1qRs in SLE and explore the gC1qR/C1q axis as a potential target for therapy.
Collapse
Affiliation(s)
- Kinga K Hosszu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Alisa Valentino
- Department of Lab Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ellinor I Peerschke
- Department of Lab Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Berhane Ghebrehiwet
- The Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
36
|
Xu-Vanpala S, Deerhake ME, Wheaton JD, Parker ME, Juvvadi PR, MacIver N, Ciofani M, Shinohara ML. Functional heterogeneity of alveolar macrophage population based on expression of CXCL2. Sci Immunol 2020; 5:eaba7350. [PMID: 32769172 PMCID: PMC7717592 DOI: 10.1126/sciimmunol.aba7350] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 06/15/2020] [Indexed: 12/21/2022]
Abstract
Alveolar macrophages (AMs) are the major lung-resident macrophages and have contradictory functions. AMs maintain tolerance and tissue homeostasis, but they also initiate strong inflammatory responses. However, such opposing roles within the AM population were not known to be simultaneously generated and coexist. Here, we uncovered heterogeneous AM subpopulations generated in response to two distinct pulmonary fungal infections, Cryptococcus neoformans and Aspergillus fumigatus Some AMs are bona fide sentinel cells that produce chemoattractant CXCL2, which also serves as a marker for AM heterogeneity, in the context of pulmonary fungal infections. However, other AMs do not produce CXCL2 and other pro-inflammatory molecules. Instead, they highly produce anti-inflammatory molecules, including interleukin-10 (IL-10) and complement component 1q (C1q). These two AM subpopulations have distinct metabolic profiles and phagocytic capacities. We report that polarization of pro-inflammatory and anti-inflammatory AM subpopulations is regulated at both epigenetic and transcriptional levels and that these AM subpopulations are generally highly plastic. Our studies have uncovered the role of C1q expression in programming and sustaining anti-inflammatory AMs. Our finding of the AM heterogeneity upon fungal infections suggests a possible pharmacological intervention target to treat fungal infections by tipping the balance of AM subpopulations.
Collapse
Affiliation(s)
- Shengjie Xu-Vanpala
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - M Elizabeth Deerhake
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Joshua D Wheaton
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
- Amgen Research, Amgen Inc., South San Francisco, CA 94080, USA
| | - Morgan E Parker
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Praveen R Juvvadi
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Nancie MacIver
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Maria Ciofani
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
37
|
Ni XN, Yan SB, Zhang K, Sai WW, Zhang QY, Ti Y, Wang ZH, Zhang W, Zheng CY, Zhong M. Serum complement C1q level is associated with acute coronary syndrome. Mol Immunol 2020; 120:130-135. [PMID: 32120180 DOI: 10.1016/j.molimm.2020.02.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/24/2020] [Accepted: 02/18/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND OBJECTIVES The complement system plays an important role in the development of acute coronary syndrome (ACS). Complement C1q is an important initial component of the classical complement pathway and closely related to many chronic inflammatory diseases, including atherosclerosis (AS). We aimed to determine whether there was association between serum complement C1q and the severity of coronary stenosis. SUBJECTS AND METHODS 320 patients who underwent coronary arteriography (CAG) were stratified into non-ACS group (control group, n = 74), unstable angina group (UA group, n = 197) and acute myocardial infarction group (AMI group, n = 49) according to the severity of coronary stenosis and clinical manifestations. The severity of coronary stenosis was represented in Gensini score, and serum complement C1q level was compared using immunity transmission turbidity among three groups. RESULTS The level of complement C1q in AMI group was lower significantly than control group and UA group (P < 0.05), but there was no correlation between serum complement C1q and Gensini score (β=-0.086, P = 0.125). In nitrate-taking patients, serum complement C1q had a negative association with Gensini score (r=-0.275, P = 0.001), and in non-smokers, there was also a negative correlation (β=-0.159, P = 0.036). After calibrating smoking, drinking or statins, the serum complement C1q levels of control group, UA group and AMI group decreased in sequence (P < 0.05). Logistic regression analysis showed that the decreasing of serum complement C1q was an unfavorable factor for acute myocardial infarction (OR=0.984, 95 %CI=0.972∼0.997, P = 0.015) and for ACS (OR=0.984, 95 %CI=0.971∼0.984, P = 0.025) in drinking patients. Regrettably, ROC curve suggested that the accuracy in diagnosing coronary atherosclerotic heart disease by serum complement C1q was low (AUC=0.568, 95 %CI= 0.492-0.644, P = 0.076, sensitivity 73.6 %, specificity 58.1 %). CONCLUSION Serum complement C1q in ACS patients, in particular AMI patients, showed lower level. This finding suggests further decrease of complement C1q level in ACS patients may be a contributory factor to instability or rupture of atherosclerotic plaques. Combined with other clinical indicators, it can be helpful to predict the risk and severity of coronary stenosis.
Collapse
Affiliation(s)
- Xiao-Ning Ni
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Sen-Bo Yan
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ke Zhang
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, China
| | - Wen-Wen Sai
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qi-Yu Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yun Ti
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhi-Hao Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Department of Geriatric Medicine, Qilu Hospital of Shandong University, Key Laboratory of Cardiovascular Proteomics of Shandong Province, Jinan, China
| | - Wei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Chun-Yan Zheng
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, China.
| | - Ming Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
38
|
Espericueta V, Manughian-Peter AO, Bally I, Thielens NM, Fraser DA. Recombinant C1q variants modulate macrophage responses but do not activate the classical complement pathway. Mol Immunol 2019; 117:65-72. [PMID: 31739194 DOI: 10.1016/j.molimm.2019.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/03/2019] [Accepted: 10/08/2019] [Indexed: 11/25/2022]
Abstract
Complement protein C1q plays a dual role in a number of inflammatory diseases such as atherosclerosis. While in later stages classical complement pathway activation by C1q exacerbates disease progression, C1q also plays a beneficial role in early disease. Independent of its role in complement activation, we and others have identified a number of potentially beneficial interactions of C1q with phagocytes in vitro, including triggering phagocytosis of cellular and molecular debris and polarizing macrophages toward an anti-inflammatory phenotype. These interactions may also be important in preventing autoimmunity. Here, we characterize variants of recombinant human C1q (rC1q) which no longer initiate complement activation, through mutation of the C1r2C1s2 interaction site. For insight into the structural location of the site of C1q that is important for interaction with phagocytes, we investigated the effect of these mutations on phagocytosis and macrophage inflammatory polarization, as compared to wild-type C1q. Phagocytosis of antibody coated sheep erythrocytes and oxidized LDL was measured in human monocytes and monocyte-derived macrophages (HMDM) respectively that had interacted with rC1q wild-type or variants. Secreted levels of cytokines were also measured in C1q stimulated HMDM. All variants of C1q increased phagocytosis in HMDM compared to controls, similar to native or wild-type rC1q. In addition, levels of certain pro-inflammatory cytokines and chemokines secreted by HMDM were modulated in cells that interacted with C1q variants, similar to wild-type rC1q and native C1q. This includes downregulation of IL-1α, IL-1β, TNFα, MIP-1α, and IL-12p40 by native and rC1q in both resting and M1-polarized HMDM. This suggests that the site responsible for C1q interaction with phagocytes is independent of the C1r2C1s2 interaction site. Further studies with these classical pathway-null variants of C1q should provide greater understanding of the complement-independent role of C1q, and allow for potential therapeutic exploitation.
Collapse
Affiliation(s)
- Victoria Espericueta
- Department of Biological Sciences, California State University Long Beach, CA, USA
| | | | - Isabelle Bally
- Univ. Grenoble Alpes, CEA, CNRS, IBS, F-38000, Grenoble, France
| | | | - Deborah A Fraser
- Department of Biological Sciences, California State University Long Beach, CA, USA.
| |
Collapse
|
39
|
HMGB1-C1q complexes regulate macrophage function by switching between leukotriene and specialized proresolving mediator biosynthesis. Proc Natl Acad Sci U S A 2019; 116:23254-23263. [PMID: 31570601 DOI: 10.1073/pnas.1907490116] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Macrophage polarization is critical to inflammation and resolution of inflammation. We previously showed that high-mobility group box 1 (HMGB1) can engage receptor for advanced glycation end product (RAGE) to direct monocytes to a proinflammatory phenotype characterized by production of type 1 IFN and proinflammatory cytokines. In contrast, HMGB1 plus C1q form a tetramolecular complex cross-linking RAGE and LAIR-1 and directing monocytes to an antiinflammatory phenotype. Lipid mediators, as well as cytokines, help establish a milieu favoring either inflammation or resolution of inflammation. This study focuses on the induction of lipid mediators by HMGB1 and HMGB1 plus C1q and their regulation of IRF5, a transcription factor critical for the induction and maintenance of proinflammatory macrophages. Here, we show that HMGB1 induces leukotriene production through a RAGE-dependent pathway, while HMGB1 plus C1q induces specialized proresolving lipid mediators lipoxin A4, resolvin D1, and resolvin D2 through a RAGE- and LAIR-1-dependent pathway. Leukotriene exposure contributes to induction of IRF5 in a positive-feedback loop. In contrast, resolvins (at 20 nM) block IRF5 induction and prevent the differentiation of inflammatory macrophages. Finally, we have generated a molecular mimic of HMGB1 plus C1q, which cross-links RAGE and LAIR-1 and polarizes monocytes to an antiinflammatory phenotype. These findings may provide a mechanism to control nonresolving inflammation in many pathologic conditions.
Collapse
|
40
|
Gomez-Roca CA, Italiano A, Le Tourneau C, Cassier PA, Toulmonde M, D'Angelo SP, Campone M, Weber KL, Loirat D, Cannarile MA, Jegg AM, Ries C, Christen R, Meneses-Lorente G, Jacob W, Klaman I, Ooi CH, Watson C, Wonde K, Reis B, Michielin F, Rüttinger D, Delord JP, Blay JY. Phase I study of emactuzumab single agent or in combination with paclitaxel in patients with advanced/metastatic solid tumors reveals depletion of immunosuppressive M2-like macrophages. Ann Oncol 2019; 30:1381-1392. [PMID: 31114846 PMCID: PMC8887589 DOI: 10.1093/annonc/mdz163] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Emactuzumab is a monoclonal antibody against the colony-stimulating factor-1 receptor and targets tumor-associated macrophages (TAMs). This study assessed the safety, clinical activity, pharmacokinetics (PK) and pharmacodynamics (PD) of emactuzumab, as monotherapy and in combination with paclitaxel, in patients with advanced solid tumors. PATIENTS AND METHODS This open-label, phase Ia/b study comprised two parts (dose escalation and dose expansion), each containing two arms (emactuzumab, every 2 or 3 weeks, as monotherapy or in combination with paclitaxel 80 mg/m2 weekly). The dose-escalation part explored the maximum tolerated dose and optimal biological dose (OBD). The dose-expansion part extended the safety assessment and investigated the objective response rate. A PK/PD analysis of serial blood, skin and tumor biopsies was used to explore proof of mechanism and confirm the OBD. RESULTS No maximum tolerated dose was reached in either study arm, and the safety profile of emactuzumab alone and in combination does not appear to preclude its use. No patients receiving emactuzumab monotherapy showed an objective response; the objective response rate for emactuzumab in combination with paclitaxel was 7% across all doses. Skin macrophages rather than peripheral blood monocytes or circulating colony-stimulating factor-1 were identified as an optimal surrogate PD marker to select the OBD. Emactuzumab treatment alone and in combination with paclitaxel resulted in a plateau of immunosuppressive TAM reduction at the OBD of 1000 mg administered every 2 weeks. CONCLUSIONS Emactuzumab showed specific reduction of immunosuppressive TAMs at the OBD in both treatment arms but did not result in clinically relevant antitumor activity alone or in combination with paclitaxel. (ClinicalTrials.gov Identifier: NCT01494688).
Collapse
Affiliation(s)
- C A Gomez-Roca
- Department of Medicine & Clinical Research Unit, Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse (IUCT-Oncopole), Toulouse.
| | - A Italiano
- Department of Medical Oncology, Institut Bergonié, Bordeaux.
| | - C Le Tourneau
- Department of Drug Development and Innovation, Institut Curie, Paris & Saint-Cloud; INSERM U900 Research Unit, Saint-Cloud; Paris-Saclay University, Paris
| | - P A Cassier
- Department of Medicine, Centre Léon Bérard, Lyon, France
| | - M Toulmonde
- Department of Medical Oncology, Institut Bergonié, Bordeaux
| | - S P D'Angelo
- Memorial Sloan Kettering Cancer Center, New York; Weill Cornell Medical College, New York, USA
| | - M Campone
- ICO René Gauducheau, Saint-Herblain, France
| | - K L Weber
- Department of Orthopedic Oncology, Penn Medicine, Pennsylvania, USA
| | - D Loirat
- Department of Drug Development and Innovation, Institut Curie, Paris & Saint-Cloud
| | - M A Cannarile
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - A-M Jegg
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - C Ries
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - R Christen
- Licensing and Early Development (LEAD) Safety Science, Roche Innovation Center Basel, Basel, Switzerland
| | - G Meneses-Lorente
- Roche Innovation Center Welwyn, Roche Pharmaceutical Research and Early Development, Welwyn Garden City
| | - W Jacob
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - I Klaman
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - C-H Ooi
- Licensing and Early Development (LEAD) Safety Science, Roche Innovation Center Basel, Basel, Switzerland
| | - C Watson
- A4P Consulting Ltd, Sandwich, UK
| | - K Wonde
- Licensing and Early Development (LEAD) Safety Science, Roche Innovation Center Basel, Basel, Switzerland
| | - B Reis
- Licensing and Early Development (LEAD) Safety Science, Roche Innovation Center Basel, Basel, Switzerland
| | - F Michielin
- Licensing and Early Development (LEAD) Safety Science, Roche Innovation Center Basel, Basel, Switzerland
| | - D Rüttinger
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - J-P Delord
- Department of Drug Development and Innovation, Institut Curie, Paris & Saint-Cloud
| | - J-Y Blay
- Department of Medicine & Clinical Research Unit, Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse (IUCT-Oncopole), Toulouse
| |
Collapse
|
41
|
Casals C, García-Fojeda B, Minutti CM. Soluble defense collagens: Sweeping up immune threats. Mol Immunol 2019; 112:291-304. [DOI: 10.1016/j.molimm.2019.06.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/14/2022]
|
42
|
KONG LN, LIN X, HUANG C, MA TT, MENG XM, HU CJ, WANG QQ, LIU YH, SHI QP, LI J. Hesperetin derivative-12 (HDND-12) regulates macrophage polarization by modulating JAK2/STAT3 signaling pathway. Chin J Nat Med 2019; 17:122-130. [DOI: 10.1016/s1875-5364(19)30014-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Indexed: 12/17/2022]
|
43
|
Chen M, Ding M, Li Y, Zhong X, Liu S, Guo Z, Yin X, Fu S, Ye J. The complement component 1 q (C1q) in Nile tilapia (Oreochromis niloticus): Functional characterization in host defense against bacterial infection and effect on cytokine response in macrophages. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 87:98-108. [PMID: 29890197 DOI: 10.1016/j.dci.2018.05.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/31/2018] [Accepted: 05/31/2018] [Indexed: 06/08/2023]
Abstract
Complement component 1q (C1q) is the initial protein of the classical complement pathway and plays an important role in immune response against bacterial infection. In this study, the full-length of C1q subcomponent A, B and C chain genes (C1qA, C1qB and C1qC) were identified and characterized from Nile tilapia (Oreochromis niloticus). Molecular characterization of these three C1q subcomponents (OnC1qs) harbored conserved amino acids through analyses of multiple sequence alignment and phylogenetic tree, which were homologous to other teleost species. Expression analysis revealed that the OnC1qs were highly expressed in liver. After the in vivo challenges of Streptococcus agalactiae and LPS, the mRNA expressions of OnC1qs were significantly up-regulated in liver. Meanwhile, the concentration variation of OnC1qs at the protein level from tilapia serum after challenge with S. agalactiae were measured by a competitive-inhibition ELISA. In addition, the up-regulation expressions of OnC1qs were also demonstrated in head kidney adherent leukocytes and the cell culture medium in vitro stimulation with S. agalactiae, Aeromonas hydrophila and LPS, respectively. Moreover, the recombinant OnC1qs enhanced expression of cytokines IL-6, IL-8 and IL-10 in head kidney adherent leukocytes, and were able to bind both heat-aggregated mouse IgG and IgM. Taken together, the results of this study indicated that OnC1qs might be involved in host defense against bacterial infection in Nile tilapia.
Collapse
Affiliation(s)
- Meng Chen
- Guangdong Provincial Key Laboratory for Health and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Mingmei Ding
- Guangdong Provincial Key Laboratory for Health and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Yuan Li
- Guangdong Provincial Key Laboratory for Health and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Xiaofang Zhong
- Guangdong Provincial Key Laboratory for Health and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Shuo Liu
- Guangdong Provincial Key Laboratory for Health and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Zheng Guo
- Guangdong Provincial Key Laboratory for Health and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Xiaoxue Yin
- Guangdong Provincial Key Laboratory for Health and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Shengli Fu
- Guangdong Provincial Key Laboratory for Health and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Jianmin Ye
- Guangdong Provincial Key Laboratory for Health and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China.
| |
Collapse
|
44
|
Guo DB, Zhu XQ, Li QQ, Liu GMY, Ruan GP, Pang RQ, Chen YH, Wang Q, Wang JX, Liu JF, Chen Q, Pan XH. Efficacy and mechanisms underlying the effects of allogeneic umbilical cord mesenchymal stem cell transplantation on acute radiation injury in tree shrews. Cytotechnology 2018; 70:1447-1468. [PMID: 30066056 PMCID: PMC6214845 DOI: 10.1007/s10616-018-0239-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 07/21/2018] [Indexed: 12/30/2022] Open
Abstract
Umbilical cord mesenchymal stem cells (UC-MSCs) exert strong immunomodulatory effects and can repair organs. However, their roles in radiation injury remain unclear. We show that in tree shrews with acute radiation injury, injected UC-MSCs significantly improved survival rates, reduced lung inflammation and apoptosis, prevented pulmonary fibrotic processes, recovered hematopoiesis, and increased blood counts. A protein microarray analysis showed that serum levels of the anti-inflammatory cytokines IL-10 and IL-13 and the growth factors BMP-5, BMP-7, HGF, insulin, NT-4, VEGFR3, and SCF were significantly higher, while those of the inflammatory cytokines IL-2, TIMP-2, TNF-α, IFN-γ, IL-1ra, and IL-8 and the fibrosis-related factors PDGF-BB, PDGF-AA, TGF-β1, IGFBP-2, and IGFBP-4 were significantly lower in UC-MSC-injected animals. A transcriptome analysis of PBMCs showed that the mRNA expression of C1q was upregulated, while that of HLA-DP was downregulated after UC-MSC injection. These results confirm the immunohistochemistry results. eGFP-labeled UC-MSCs were traced in vivo and found in the heart, liver, spleen, lungs, kidneys, thymus, small intestine and bone marrow. Our findings suggest that UC-MSC transplantation may be a novel therapeutic approach for treating acute radiation injury.
Collapse
Affiliation(s)
- De-Bin Guo
- Department of Clinical Laboratory, Kunming General Hospital, Army Medical University, Kunming, 650032, Yunnan Province, China
| | - Xiang-Qing Zhu
- Department of Clinical Laboratory, Kunming General Hospital, Army Medical University, Kunming, 650032, Yunnan Province, China
- Cell Biological Therapy Center of Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Qing-Qing Li
- Kunming Medical University, Kunming, Yunnan Province, China
| | - Gao-Mi-Yang Liu
- Department of Clinical Laboratory, Kunming General Hospital, Army Medical University, Kunming, 650032, Yunnan Province, China
- Cell Biological Therapy Center of Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Guang-Ping Ruan
- Department of Clinical Laboratory, Kunming General Hospital, Army Medical University, Kunming, 650032, Yunnan Province, China
- Cell Biological Therapy Center of Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Rong-Qing Pang
- Department of Clinical Laboratory, Kunming General Hospital, Army Medical University, Kunming, 650032, Yunnan Province, China
- Cell Biological Therapy Center of Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Yu-Hao Chen
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Qiang Wang
- Department of Clinical Laboratory, Kunming General Hospital, Army Medical University, Kunming, 650032, Yunnan Province, China
- Cell Biological Therapy Center of Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Jin-Xiang Wang
- Department of Clinical Laboratory, Kunming General Hospital, Army Medical University, Kunming, 650032, Yunnan Province, China
- Cell Biological Therapy Center of Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Ju-Fen Liu
- Cell Biological Therapy Center of Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Qiang Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650093, Yunnan Province, China
| | - Xing-Hua Pan
- Department of Clinical Laboratory, Kunming General Hospital, Army Medical University, Kunming, 650032, Yunnan Province, China.
- Cell Biological Therapy Center of Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China.
| |
Collapse
|
45
|
Cavusoglu E, Kassotis JT, Anwar A, Marmur JD, Hussain SW, Yanamadala S, Hegde S, Parpas A, Eng C, Zhang M. Usefulness of Complement C1q to Predict 10-Year Mortality in Men With Diabetes Mellitus Referred for Coronary Angiography. Am J Cardiol 2018; 122:33-38. [PMID: 29703440 DOI: 10.1016/j.amjcard.2018.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 02/25/2018] [Accepted: 03/01/2018] [Indexed: 02/01/2023]
Abstract
The complement system consists of a family of proteins that play a critical role in the innate immune system. Complement activation has been implicated in many chronic inflammatory diseases, including atherosclerosis. However, a number of experimental studies have highlighted a beneficial role of component C1q in early atherosclerosis and in diabetes mellitus (DM). Despite these data, there have been no studies that have specifically examined the utility of plasma complement C1q as a clinical biomarker in patients with known or suspected coronary artery disease. In this study, baseline plasma complement C1q levels were measured in 159 men with DM who were referred for coronary angiography and who were followed up prospectively for the development of all-cause mortality for 10 years. After adjustment for baseline clinical, angiographic, and laboratory parameters, reduced plasma complement C1q levels were an independent predictor of all-cause mortality at 10 years (hazard ratio 0.66, 95% confidence interval 0.52 to 0.84, p = 0.0006). In additional multivariate models that adjusted for a variety of biomarkers with established prognostic efficacy, complement C1q remained an independent predictor of all-cause mortality at 10 years. In conclusion, reduced levels of complement C1q are associated with an increased risk of all-cause mortality at 10 years in patients with DM referred for coronary angiography. Furthermore, this association is independent of a variety of clinical, angiographic, laboratory variables, including biomarkers with established prognostic efficacy in the prediction of adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Erdal Cavusoglu
- Division of Cardiology, Department of Medicine, Bronx Veterans Affairs Medical Center, Bronx, New York; Division of Cardiology, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York.
| | - John T Kassotis
- Division of Cardiology, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Ayesha Anwar
- Division of Cardiology, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Jonathan D Marmur
- Division of Cardiology, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Syed Wasif Hussain
- Division of Cardiology, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Sunitha Yanamadala
- Division of Cardiology, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Sudhanva Hegde
- Division of Cardiology, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Alexander Parpas
- Division of Cardiology, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Calvin Eng
- Division of Cardiology, Department of Medicine, Bronx Veterans Affairs Medical Center, Bronx, New York
| | - Ming Zhang
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, New York; Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, New York
| |
Collapse
|
46
|
Kishimoto D, Kirino Y, Tamura M, Takeno M, Kunishita Y, Takase-Minegishi K, Nakano H, Kato I, Nagahama K, Yoshimi R, Igarashi K, Aoki I, Nakajima H. Dysregulated heme oxygenase-1 low M2-like macrophages augment lupus nephritis via Bach1 induced by type I interferons. Arthritis Res Ther 2018; 20:64. [PMID: 29636091 PMCID: PMC5894134 DOI: 10.1186/s13075-018-1568-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 03/15/2018] [Indexed: 12/17/2022] Open
Abstract
Background Innate immunity including macrophages (Mϕ) in lupus nephritis (LN) has been gaining attention, but roles of Mϕ in LN remain uncertain. Methods Immunohistochemical staining was performed to determine CD68, CD163, heme oxygenase (HO)-1 (a stress-inducible heme-degrading enzyme with anti-inflammatory property), pSTAT1, and CMAF-expressing Mϕ in the glomeruli of patients with LN. Effects of type I interferons on the expression levels of CD163, HO-1, BTB and CNC homology 1 (Bach1; a transcriptional HO-1 repressor), interleukin (IL)-6, and IL-10 by human M2-like Mϕ, which were differentiated in vitro from peripheral monocytes with macrophage colony-stimulating factor, were assessed by RT-PCR and immunocytostaining. Clinical manifestations, anti-double-stranded DNA (anti-dsDNA), and local HO-1 expression were compared in Bach1-deficient and wild-type MRL/lpr mice. Results The number of glomerular M2-like Mϕ correlated with the amounts of proteinuria in patients with LN. Unlike monocyte-derived M2-like Mϕ, HO-1 expression was defective in the majority of glomerular M2-like Mϕ of patients with LN. Stimulation of human M2-like Mϕ with type I interferons led to reduced HO-1 expression and increased Bach1 and IL-6 expression. Bach1-deficient MRL/lpr mice exhibited increased HO-1 expression in kidneys, prolonged survival, reduced urine proteins, and serum blood urea nitrogen levels, but serum anti-dsDNA antibody levels were comparable. Increased expression of CD163 and HO-1 was found in peritoneal Mϕ from Bach1-deficient MRL/lpr mice. Conclusions Our data suggest that dysregulated M2-like Mϕ play a proinflammatory role in LN. Bach1 is a potential therapeutic target that could restore the anti-inflammatory property of M2 Mϕ. Electronic supplementary material The online version of this article (10.1186/s13075-018-1568-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daiga Kishimoto
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Yohei Kirino
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan.
| | - Maasa Tamura
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Mitsuhiro Takeno
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Yosuke Kunishita
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Kaoru Takase-Minegishi
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Hiroto Nakano
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Ikuma Kato
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kiyotaka Nagahama
- Department of Pathology, Kyorin University School of Medicine, Tokyo, Japan
| | - Ryusuke Yoshimi
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University School of Medicine, Sendai, Japan.,Center for Regulatory Epigenome and Diseases, Tohoku University School of Medicine, Sendai, Japan
| | - Ichiro Aoki
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hideaki Nakajima
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| |
Collapse
|
47
|
Hertle E, Arts ICW, van der Kallen CJH, Feskens EJM, Schalkwijk CG, Stehouwer CDA, van Greevenbroek MMJ. Classical Pathway of Complement Activation: Longitudinal Associations of C1q and C1-INH With Cardiovascular Outcomes: The CODAM Study (Cohort on Diabetes and Atherosclerosis Maastricht)-Brief Report. Arterioscler Thromb Vasc Biol 2018; 38:1242-1244. [PMID: 29567681 DOI: 10.1161/atvbaha.118.310806] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 02/27/2018] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The classical complement pathway has been assigned both protective and pathological effects in cardiovascular disease (CVD), but human data are lacking. We determined the associations of the pattern recognition factor C1q and the regulator C1-INH (C1-inhibitor) with incident CVD, carotid intima-media thickness, endothelial dysfunction, and low-grade inflammation. APPROACH AND RESULTS Baseline concentrations of C1q and C1-INH were measured in the CODAM study (Cohort on Diabetes and Atherosclerosis Maastricht; n=574; 61% men; age, 60±7 years). The 7-year incidence of CVD in participants free of CVD at baseline was evaluated using logistic regression analyses (n=342; 73 cases). The lowest incidence of CVD was observed in the middle tertile of C1q (Tlow compared with Tmiddle: odds ratio, 2.38 [95% confidence interval, 1.14-4.95]; Thigh compared with Tmiddle: odds ratio, 1.96 [95% confidence interval, 0.94-4.07]). C1-INH was not associated with CVD. During the 7-year follow-up period, C1q and C1-INH were not, or inconsistently, associated with carotid intima-media thickness or with biomarker scores reflecting endothelial dysfunction and low-grade inflammation. CONCLUSIONS Our results suggest a nonlinear association between C1q and incident CVD. This supports the concept that early steps in classical pathway activation may have both protective and pathological effects on human CVD.
Collapse
Affiliation(s)
- Elisabeth Hertle
- From the CARIM School for Cardiovascular Diseases (E.H., I.C.W.A., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.).,Department of Internal Medicine (E.H., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.)
| | - Ilja C W Arts
- From the CARIM School for Cardiovascular Diseases (E.H., I.C.W.A., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.).,Department of Epidemiology (I.C.W.A.).,Maastricht Centre for Systems Biology (I.C.W.A.), Maastricht University, The Netherlands
| | - Carla J H van der Kallen
- From the CARIM School for Cardiovascular Diseases (E.H., I.C.W.A., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.).,Department of Internal Medicine (E.H., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.)
| | - Edith J M Feskens
- Division of Human Nutrition, Section of Nutrition and Epidemiology, Wageningen University, The Netherlands (E.J.M.F.)
| | - Casper G Schalkwijk
- From the CARIM School for Cardiovascular Diseases (E.H., I.C.W.A., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.).,Department of Internal Medicine (E.H., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.)
| | - Coen D A Stehouwer
- From the CARIM School for Cardiovascular Diseases (E.H., I.C.W.A., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.).,Department of Internal Medicine (E.H., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.)
| | - Marleen M J van Greevenbroek
- From the CARIM School for Cardiovascular Diseases (E.H., I.C.W.A., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.) .,Department of Internal Medicine (E.H., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.)
| |
Collapse
|
48
|
Complement C1q expression in Erythema nodosum leprosum. PLoS Negl Trop Dis 2018; 12:e0006321. [PMID: 29499046 PMCID: PMC5851649 DOI: 10.1371/journal.pntd.0006321] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/14/2018] [Accepted: 02/15/2018] [Indexed: 01/05/2023] Open
Abstract
Complement C1q is a soluble protein capable of initiating components of the classical pathway in host defence system. In earlier qualitative studies, C1q has been implicated in the pathogenesis of Erythema Nodosum Leprosum (ENL). However, little is known about the role of this complement in ENL reaction. In the present study we described the protein level of C1q production and its gene expression in the peripheral blood and skin biopsies in patients with ENL reaction and lepromatous leprosy (LL) patient controls before and after treatment. Thirty untreated patients with ENL reaction and 30 non-reactional LL patient controls were recruited at ALERT Hospital, Ethiopia. Peripheral blood and skin biopsies were obtained from each patient before and after treatment. The level of circulating C1q in the plasma was determined by enzyme-linked immunosorbent assay. The mRNA expression of the three C1q components, C1qA, C1qB, and C1qC in the peripheral blood and skin biopsies was determined by qPCR. Circulating C1q in the peripheral blood of untreated ENL patients was significantly decreased compared to LL patient controls. Untreated ENL patients had increased C1q gene expression in the peripheral blood compared to LL controls. Similarly, C1qA and C1qC gene expression were substantially increased in the skin biopsies of untreated ENL patients compared to LL controls. However, after treatment none of these genes show significant difference in both groups. In conclusion, while circulating C1q is inversely correlated with active ENL reactions, its gene expression is directly correlated with ENL. The decreased circulating C1q may suggest the utilization of C1q in immune-complex formation in these patients. Therefore, C1q could be a potential diagnostic marker for active ENL reactions as well as for monitoring ENL treatment.
Collapse
|
49
|
Ge X, Xu C, Liu Y, Zhu K, Zeng H, Su J, Huang J, Ji Y, Tan Y, Hou Y. Complement activation in the arteries of patients with severe atherosclerosis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:1-9. [PMID: 31938082 PMCID: PMC6957963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/03/2017] [Indexed: 06/10/2023]
Abstract
BACKGROUND Excessive complement activation plays an important role in the pathogenesis of atherosclerosis (AS). We therefore wanted to investigate whether complement is activated in areas of AS by detecting the deposition of C3b/iC3b and membrane attack complex (MAC). We also analyzed the relationships between C3b/iC3b and MAC levels and the clinicopathological features of patients with AS. METHODS The sample comprised 79 patients who had been diagnosed with AS. Their levels of C3b/iC3b and MAC deposition were evaluated by immunohistochemistry (IHC). The results were translated into scores, and the patients' clinical features were recorded. RESULTS Compared with normal arteries, significantly greater deposits of C3b/iC3b and MAC were found in AS arteries. In the group with more C3b/iC3b deposition, the ratio of patients with hypertension was higher. Moreover, in the group with more MAC deposition, the ratio of patients with hypertriglyceridemia was higher. CONCLUSIONS The finding of C3b/iC3b and MAC deposition in atherosclerotic arteries points to the activation of complement. Greater amounts of C3b/iC3b and MAC deposition imply excessive complement activation, which can lead to the development of AS. Hypertension and hypertriglyceridemia may, respectively, contribute to the activation of complement C3 or the formation of MAC.
Collapse
Affiliation(s)
- Xiaowen Ge
- Department of Pathology, Zhongshan Hospital, Fudan UniversityShanghai, P. R. China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan UniversityShanghai, P. R. China
| | - Yalan Liu
- Department of Pathology, Zhongshan Hospital, Fudan UniversityShanghai, P. R. China
| | - Kai Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan UniversityShanghai, P. R. China
| | - Haiying Zeng
- Department of Pathology, Zhongshan Hospital, Fudan UniversityShanghai, P. R. China
| | - Jieakesu Su
- Department of Pathology, Zhongshan Hospital, Fudan UniversityShanghai, P. R. China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan UniversityShanghai, P. R. China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan UniversityShanghai, P. R. China
| | - Yunshan Tan
- Department of Pathology, Zhongshan Hospital, Fudan UniversityShanghai, P. R. China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan UniversityShanghai, P. R. China
| |
Collapse
|
50
|
Pulanco MC, Cosman J, Ho MM, Huynh J, Fing K, Turcu J, Fraser DA. Complement Protein C1q Enhances Macrophage Foam Cell Survival and Efferocytosis. THE JOURNAL OF IMMUNOLOGY 2016; 198:472-480. [PMID: 27895181 DOI: 10.4049/jimmunol.1601445] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/01/2016] [Indexed: 12/12/2022]
Abstract
In the atherosclerotic lesion, macrophages ingest high levels of damaged modified low-density lipoproteins (LDLs), generating macrophage foam cells. Foam cells undergo apoptosis and, if not efficiently cleared by efferocytosis, can undergo secondary necrosis, leading to plaque instability and rupture. As a component of the innate immune complement cascade, C1q recognizes and opsonizes modified forms of LDL, such as oxidized or acetylated LDL, and promotes ingestion by macrophages in vitro. C1q was shown to be protective in an atherosclerosis model in vivo. Therefore, this study aimed to investigate whether ingestion of modified LDL in the presence of C1q alters macrophage foam cell survival or function. In an unbiased transcriptome analysis, C1q was shown to modulate expression of clusters of genes involved in cell death and apoptosis pathways in human monocyte-derived macrophages ingesting modified LDL; this was validated by quantitative PCR in human and murine macrophages. C1q downregulated levels and activity of active caspase-3 and PARP-1 in human and mouse macrophages during ingestion of modified LDL. This led to a measurable increase in survival and decrease in cell death, as measured by alamarBlue and propidium iodide assays, respectively. C1q opsonization also increased phagocytosis and efferocytosis in macrophage foam cells. These data suggest that C1q promotes macrophage survival during ingestion of excess cholesterol, as well as improves foam cell efferocytic function. This may be important in slowing disease progression and provides insight into the protective role of C1q in early atherosclerosis.
Collapse
Affiliation(s)
- Marc C Pulanco
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840
| | - Jason Cosman
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840
| | - Minh-Minh Ho
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840
| | - Jessica Huynh
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840
| | - Karina Fing
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840
| | - Jacqueline Turcu
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840
| | - Deborah A Fraser
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840
| |
Collapse
|