1
|
Möller-Ramon Z, Aslani M, Sobczak N, Hristov M, Weber C, Rot A, Duchêne J. The 129 strain-derived passenger mutations in ACKR1-deficient mice alter the expression of PYHIN and Fc-gamma receptor genes. J Leukoc Biol 2025; 117:qiae208. [PMID: 39319406 DOI: 10.1093/jleuko/qiae208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/24/2024] [Indexed: 09/26/2024] Open
Abstract
Most genetically modified mice have been produced using 129 strain-derived embryonic stem cells. Despite ample backcrosses with other strains, these may retain characteristics for 129 passenger mutations, leading to confounding phenotypes unrelated to targeted genes. Here we show that widely used Ackr1-/-129ES mice have approximately 6 Mb of the 129-derived genome retained adjacently to the Ackr1 locus on chromosome 1, including several characteristic polymorphisms. These most notably affect the expression of PYHIN and Fc-gamma receptor genes in myeloid cells, resulting in the overproduction of IL-1β by activated macrophages and the loss of Fc-gamma receptors on myeloid progenitor cells. Therefore, caution is warranted when interpreting Ackr1-/-129ES mouse phenotypes as being solely due to the ACKR1 deficiency. Our findings call for a careful reevaluation of data from previous studies using Ackr1-/-129ES mice and underscore the limitations and pitfalls inherent to mouse models produced using traditional genetic engineering techniques involving 129 embryonic stem cells.
Collapse
Affiliation(s)
- Zoe Möller-Ramon
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University Munich, Pettenkoferstraße 9, 80336 Munich, Germany
| | - Maria Aslani
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University Munich, Pettenkoferstraße 9, 80336 Munich, Germany
| | - Nikola Sobczak
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University Munich, Pettenkoferstraße 9, 80336 Munich, Germany
| | - Michael Hristov
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University Munich, Pettenkoferstraße 9, 80336 Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University Munich, Pettenkoferstraße 9, 80336 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Pettenkoferstraße 8a, 80336 Munich, Germany
- Cardiovascular Research Institute Maastricht, University of Maastricht, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Antal Rot
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University Munich, Pettenkoferstraße 9, 80336 Munich, Germany
- Centre for Microvascular Research, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, EC1M 6BQ London, United Kingdom
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, Charterhouse Square, EC1M 6BQ London, United Kingdom
| | - Johan Duchêne
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University Munich, Pettenkoferstraße 9, 80336 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Pettenkoferstraße 8a, 80336 Munich, Germany
| |
Collapse
|
2
|
Cheng P, Quertermous T. Spatial transcriptomic mapping of coronary atherosclerosis in the luminal plaque and beyond. NATURE CARDIOVASCULAR RESEARCH 2025; 4:8-10. [PMID: 39747456 DOI: 10.1038/s44161-024-00568-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Affiliation(s)
- Paul Cheng
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
3
|
Song J, Xie D, Wei X, Liu B, Yao F, Ye W. A cuproptosis-related lncRNAs signature predicts prognosis and reveals pivotal interactions between immune cells in colon cancer. Heliyon 2024; 10:e34586. [PMID: 39114018 PMCID: PMC11305305 DOI: 10.1016/j.heliyon.2024.e34586] [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: 09/17/2023] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Copper-mediated cell death presents distinct pathways from established apoptosis processes, suggesting alternative therapeutic approaches for colon cancer. Our research aims to develop a predictive framework utilizing long-noncoding RNAs (lncRNAs) related to cuproptosis to predict colon cancer outcomes while examining immune interactions and intercellular signaling. We obtained colon cancer-related human mRNA expression profiles and clinical information from the Cancer Genome Atlas repository. To isolate lncRNAs involved in cuproptosis, we applied Cox proportional hazards modeling alongside the least absolute shrinkage and selection operator technique. We elucidated the underlying mechanisms by examining the tumor mutational burden, the extent of immune cell penetration, and intercellular communication dynamics. Based on the model, drugs were predicted and validated with cytological experiments. A 13 lncRNA-cuproptosis-associated risk model was constructed. Two colon cancer cell lines were used to validate the predicted representative mRNAs with high correlation coefficients with copper-induced cell death. Survival enhancement in the low-risk cohort was evidenced by the trends in Kaplan-Meier survival estimates. Analysis of immune cell infiltration suggested that survival was induced by the increased infiltration of naïve CD4+ T cells and a reduction of M2 macrophages within the low-risk faction. Decreased infiltration of naïve B cells, resting NK cells, and M0 macrophages was significantly associated with better overall survival. Combined single-cell analysis suggested that CCL5-ACKR1, CCL2-ACKR1, and CCL5-CCR1 pathways play key roles in mediating intercellular dialogues among immune constituents within the neoplastic microhabitat. We identified three drugs with a high sensitivity in the high-risk group. In summary, this discovery establishes the possibility of using 13 cuproptosis-associated lncRNAs as a risk model to assess the prognosis, unravel the immune mechanisms and cell communication, and improve treatment options, which may provide a new idea for treating colon cancer.
Collapse
Affiliation(s)
- Jingru Song
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| | - Dong Xie
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xia Wei
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| | - Binbin Liu
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| | - Fang Yao
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| | - Wei Ye
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| |
Collapse
|
4
|
Yang R, Lin F, Wang W, Dai G, Ke X, Wu G. Investigating the therapeutic effects and mechanisms of Carthamus tinctorius L.-derived nanovesicles in atherosclerosis treatment. Cell Commun Signal 2024; 22:178. [PMID: 38475787 PMCID: PMC10936069 DOI: 10.1186/s12964-024-01561-6] [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] [Received: 07/07/2023] [Accepted: 03/05/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Carthamus tinctorius L., a traditional herbal medicine used for atherosclerosis (AS), lacks a clear understanding of its therapeutic mechanisms. This study aimed to investigate the therapeutic effects and mechanisms of Carthamus tinctorius L.-derived nanovesicles (CDNVs) in AS treatment. METHODS CDNVs were isolated and characterized using improved isolation methods. Transmission electron microscopy, nanoparticle tracking analysis, and protein analysis confirmed their morphology, size, and protein composition. Small RNA sequencing was performed to identify the miRNA profile of CDNVs, and bioinformatics analysis was used to determine their potential biological roles. In vivo biodistribution and toxicity studies were conducted in mice to assess the stability and safety of orally administered CDNVs. The anti-atherosclerotic effects of CDNVs were evaluated in ApoE-/- mice through plaque burden analysis. The protective effects of CDNVs on ox-LDL-treated endothelial cells were assessed through proliferation, apoptosis, reactive oxygen species activation, and monocyte adhesion assays. miRNA and mRNA sequencing of CDNV-treated endothelial cells were performed to explore their regulatory effects and potential target genes. RESULTS CDNVs were successfully isolated and purified from Carthamus tinctorius L. tissue lysates. They exhibited a saucer-shaped or cup-shaped morphology, with an average particle size of 142.6 ± 0.7 nm, and expressed EV markers CD63 and TSG101. CDNVs contained proteins, small RNAs, and metabolites, including the therapeutic compound HSYA. Small RNA sequencing identified 95 miRNAs, with 10 common miRNAs accounting for 72.63% of the total miRNAs. These miRNAs targeted genes involved in cell adhesion, apoptosis, and cell proliferation, suggesting their relevance in cardiovascular disease. Orally administered CDNVs were stable in the gastrointestinal tract, absorbed into the bloodstream, and accumulated in the liver, lungs, heart, and aorta. They significantly reduced the burden of atherosclerotic plaques in ApoE-/- mice and exhibited superior effects compared to HSYA. In vitro studies demonstrated that CDNVs were taken up by HUVECs, promoted proliferation, attenuated ox-LDL-induced apoptosis and ROS activation, and reduced monocyte adhesion. CDNV treatment resulted in significant changes in miRNA and mRNA expression profiles of HUVECs, with enrichment in inflammation-related genes. CXCL12 was identified as a potential direct target of miR166a-3p. CONCLUSION CDNVs isolated from Carthamus tinctorius L. tissue lysates represent a promising oral therapeutic option for cardiovascular diseases. The delivery of miRNAs by CDNVs regulates inflammation-related genes, including CXCL12, in HUVECs, suggesting their potential role in modulating endothelial inflammation. These findings provide valuable insights into the therapeutic potential of CDNVs and their miRNAs in cardiovascular disease.
Collapse
Affiliation(s)
- Rongfeng Yang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Shenzhen, China
| | - Fengxia Lin
- Department of Cardiology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Wenlin Wang
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences (Shenzhen Sun Yat-sen Cardiovascular Hospital), Shenzhen, China
- Department of Clinical Medicine, University of South China, Hengyang, China
| | - Gang Dai
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Shenzhen, China
| | - Xiao Ke
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences (Shenzhen Sun Yat-sen Cardiovascular Hospital), Shenzhen, China.
| | - Guifu Wu
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
- Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Shenzhen, China.
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Shenzhen, China.
| |
Collapse
|
5
|
Xiao YY, Zhang Q, Huang F, Rao L, Yao TX, Yang SY, Xie L, Zou XX, Cai LP, Yang JW, Yang B, Huang LS. Single-cell profiling of the pig cecum at various developmental stages. Zool Res 2024; 45:55-68. [PMID: 38114433 PMCID: PMC10839672 DOI: 10.24272/j.issn.2095-8137.2023.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 09/12/2023] [Indexed: 12/21/2023] Open
Abstract
The gastrointestinal tract is essential for food digestion, nutrient absorption, waste elimination, and microbial defense. Single-cell transcriptome profiling of the intestinal tract has greatly enriched our understanding of cellular diversity, functional heterogeneity, and their importance in intestinal tract development and disease. Although such profiling has been extensively conducted in humans and mice, the single-cell gene expression landscape of the pig cecum remains unexplored. Here, single-cell RNA sequencing was performed on 45 572 cells obtained from seven cecal samples in pigs at four different developmental stages (days (D) 30, 42, 150, and 730). Analysis revealed 12 major cell types and 38 subtypes, as well as their distinctive genes, transcription factors, and regulons, many of which were conserved in humans. An increase in the relative proportions of CD8 + T and Granzyme A (low expression) natural killer T cells (GZMA low NKT) cells and a decrease in the relative proportions of epithelial stem cells, Tregs, RHEX + T cells, and plasmacytoid dendritic cells (pDCs) were noted across the developmental stages. Moreover, the post-weaning period exhibited an up-regulation in mitochondrial genes, COX2 and ND2, as well as genes involved in immune activation in multiple cell types. Cell-cell crosstalk analysis indicated that IBP6 + fibroblasts were the main signal senders at D30, whereas IBP6 - fibroblasts assumed this role at the other stages. NKT cells established interactions with epithelial cells and IBP6 + fibroblasts in the D730 cecum through mediation of GZMA-F2RL1/F2RL2 pairs. This study provides valuable insights into cellular heterogeneity and function in the pig cecum at different development stages.
Collapse
Affiliation(s)
- Yan-Yuan Xiao
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Qing Zhang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Fei Huang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Lin Rao
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Tian-Xiong Yao
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Si-Yu Yang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Lei Xie
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Xiao-Xiao Zou
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Li-Ping Cai
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Jia-Wen Yang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Bin Yang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China. E-mail:
| | - Lu-Sheng Huang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China. E-mail:
| |
Collapse
|
6
|
Lindsay HG, Hendrix CJ, Gonzalez Murcia JD, Haynie C, Weber KS. The Role of Atypical Chemokine Receptors in Neuroinflammation and Neurodegenerative Disorders. Int J Mol Sci 2023; 24:16493. [PMID: 38003682 PMCID: PMC10671188 DOI: 10.3390/ijms242216493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Neuroinflammation is associated with several neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Neuroinflammation provides protection in acute situations but results in significant damage to the nervous system if chronic. Overexpression of chemokines within the brain results in the recruitment and activation of glial and peripheral immune cells which can propagate a cascading inflammatory response, resulting in neurodegeneration and the onset of neurodegenerative disorders. Recent work has identified the role of atypical chemokine receptors (ACKRs) in neurodegenerative conditions. ACKRs are seven-transmembrane domain receptors that do not follow canonical G protein signaling, but regulate inflammatory responses by modulating chemokine abundance, location, and availability. This review summarizes what is known about the four ACKRs and three putative ACKRs within the brain, highlighting their known expression and discussing the current understanding of each ACKR in the context of neurodegeneration. The ability of ACKRs to alter levels of chemokines makes them an appealing therapeutic target for neurodegenerative conditions. However, further work is necessary to understand the expression of several ACKRs within the neuroimmune system and the effectiveness of targeted drug therapies in the prevention and treatment of neurodegenerative conditions.
Collapse
Affiliation(s)
- Hunter G. Lindsay
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| | - Colby J. Hendrix
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| | | | - Christopher Haynie
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| | - K. Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| |
Collapse
|
7
|
Yang L, Zhang S, Pu P. Comprehensive analysis of ACKR family members in breast cancer using prognostic values. Oncol Lett 2023; 26:425. [PMID: 37664667 PMCID: PMC10472033 DOI: 10.3892/ol.2023.14011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/25/2023] [Indexed: 09/05/2023] Open
Abstract
Breast cancer (BC) is by far the most prevalent malignancy found in the female population. Atypical chemokine receptors (ACKRs) are a subclass of G-protein-coupled receptors, which are characterized by disrupted ligand binding and a breakdown of signaling following ligand binding. The evolution and function of multiple ACKRs in BC have yet to be fully elucidated, although certain findings on this family have been reported in several studies in Homo sapiens and other species. The present study identified that the expression level of ACKRs was significantly lower in breast carcinoma (BRCA) tissues compared with normal breast tissues through searches of the Tumor Immune Estimation Resource, UALCAN and Gene Expression Profiling Interactive Analysis databases. Additionally, when comparing BRCA tissues with normal breast tissues, it was found that there was obvious hypomethylation in the promoters of ACKR1, ACKR3 and ACKR5, as well as a marked hypermethylation in the promoters of ACKR2 and ACKR6. In determining the prognosis of patients with BRCA, the expression levels of ACKR1, ACKR2, ACKR3, ACKR4 and ACKR6 were all found to be important factors. The values for distant metastasis-free survival (DMFS), overall survival (OS) and recurrence-free survival (RFS) were all found to be lower in patients with BRCA who had a low expression level of ACKR1. In addition, the RFS rates for patients with BRCA were lower when the expression of ACKR2 was low, and worse values for DMFS, OS and RFS were found to be highly correlated with higher expression levels of ACKR3. Moreover, the DMFS, OS, RFS and predictive power score values were worse in those patients with low ACKR4 expression, and the RFS values for patients with BRCA were also found to be lower when the expression level of ACKR6 was low. Additionally, dendritic cells, macrophages, neutrophils, T cells with CD4+ status, T cells with CD8+ status and B cells were all substantially linked with ACKR expression, as well as immune cell infiltration. Taken together, the findings of the present study may offer a theoretical foundation for the creation of novel targets and prognostic indicators for BRCA therapy.
Collapse
Affiliation(s)
- Lixian Yang
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei 054000, P.R. China
| | - Shiyu Zhang
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei 054000, P.R. China
| | - Pengpeng Pu
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei 054000, P.R. China
| |
Collapse
|
8
|
Tang C, Chen G, Wu F, Cao Y, Yang F, You T, Liu C, Li M, Hu S, Ren L, Lu Q, Deng W, Xu Y, Wang G, Jo H, Zhang Y, Wu Y, Zabel BA, Zhu L. Endothelial CCRL2 induced by disturbed flow promotes atherosclerosis via chemerin-dependent β2 integrin activation in monocytes. Cardiovasc Res 2023; 119:1811-1824. [PMID: 37279540 PMCID: PMC10405567 DOI: 10.1093/cvr/cvad085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/01/2023] [Indexed: 06/08/2023] Open
Abstract
AIMS Chemoattractants and their cognate receptors are essential for leucocyte recruitment during atherogenesis, and atherosclerotic plaques preferentially occur at predilection sites of the arterial wall with disturbed flow (d-flow). In profiling the endothelial expression of atypical chemoattractant receptors (ACKRs), we found that Ackr5 (CCRL2) was up-regulated in an endothelial subpopulation by atherosclerotic stimulation. We therefore investigated the role of CCRL2 and its ligand chemerin in atherosclerosis and the underlying mechanism. METHODS AND RESULTS By analysing scRNA-seq data of the left carotid artery under d-flow and scRNA-seq datasets GSE131776 of ApoE-/- mice from the Gene Expression Omnibus database, we found that CCRL2 was up-regulated in one subpopulation of endothelial cells in response to d-flow stimulation and atherosclerosis. Using CCRL2-/-ApoE-/- mice, we showed that CCRL2 deficiency protected against plaque formation primarily in the d-flow areas of the aortic arch in ApoE-/- mice fed high-fat diet. Disturbed flow induced the expression of vascular endothelial CCRL2, recruiting chemerin, which caused leucocyte adhesion to the endothelium. Surprisingly, instead of binding to monocytic CMKLR1, chemerin was found to activate β2 integrin, enhancing ERK1/2 phosphorylation and monocyte adhesion. Moreover, chemerin was found to have protein disulfide isomerase-like enzymatic activity, which was responsible for the interaction of chemerin with β2 integrin, as identified by a Di-E-GSSG assay and a proximity ligation assay. For clinical relevance, relatively high serum levels of chemerin were found in patients with acute atherothrombotic stroke compared to healthy individuals. CONCLUSIONS Our findings indicate that d-flow-induced CCRL2 promotes atherosclerotic plaque formation via a novel CCRL2-chemerin-β2 integrin axis, providing potential targets for the prevention or therapeutic intervention of atherosclerosis.
Collapse
Affiliation(s)
- Chaojun Tang
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, China
- JinFeng Laboratory, Chongqing, China
| | - Guona Chen
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Fan Wu
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Cambridge-Suda Genomic Resource Center, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Yiren Cao
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Fei Yang
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Tao You
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chu Liu
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Menglu Li
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Shuhong Hu
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Lijie Ren
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Qiongyu Lu
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Wei Deng
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Ying Xu
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Cambridge-Suda Genomic Resource Center, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Guixue Wang
- JinFeng Laboratory, Chongqing, China
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Yonghong Zhang
- Department of Epidemiology School of Public Health, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Yi Wu
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Brian A Zabel
- Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Li Zhu
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- The Ninth Affiliated Hospital, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, China
- JinFeng Laboratory, Chongqing, China
| |
Collapse
|
9
|
Crawford KS, Volkman BF. Prospects for targeting ACKR1 in cancer and other diseases. Front Immunol 2023; 14:1111960. [PMID: 37006247 PMCID: PMC10050359 DOI: 10.3389/fimmu.2023.1111960] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
The chemokine network is comprised of a family of signal proteins that encode messages for cells displaying chemokine G-protein coupled receptors (GPCRs). The diversity of effects on cellular functions, particularly directed migration of different cell types to sites of inflammation, is enabled by different combinations of chemokines activating signal transduction cascades on cells displaying a combination of receptors. These signals can contribute to autoimmune disease or be hijacked in cancer to stimulate cancer progression and metastatic migration. Thus far, three chemokine receptor-targeting drugs have been approved for clinical use: Maraviroc for HIV, Plerixafor for hematopoietic stem cell mobilization, and Mogalizumab for cutaneous T-cell lymphoma. Numerous compounds have been developed to inhibit specific chemokine GPCRs, but the complexity of the chemokine network has precluded more widespread clinical implementation, particularly as anti-neoplastic and anti-metastatic agents. Drugs that block a single signaling axis may be rendered ineffective or cause adverse reactions because each chemokine and receptor often have multiple context-specific functions. The chemokine network is tightly regulated at multiple levels, including by atypical chemokine receptors (ACKRs) that control chemokine gradients independently of G-proteins. ACKRs have numerous functions linked to chemokine immobilization, movement through and within cells, and recruitment of alternate effectors like β-arrestins. Atypical chemokine receptor 1 (ACKR1), previously known as the Duffy antigen receptor for chemokines (DARC), is a key regulator that binds chemokines involved in inflammatory responses and cancer proliferation, angiogenesis, and metastasis. Understanding more about ACKR1 in different diseases and populations may contribute to the development of therapeutic strategies targeting the chemokine network.
Collapse
Affiliation(s)
- Kyler S. Crawford
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | | |
Collapse
|
10
|
Chi G, Pei J, Li X. The imbalance of liver resident macrophages polarization promotes chronic autoimmune hepatitis development in mice. PeerJ 2023; 11:e14871. [PMID: 36778150 PMCID: PMC9912947 DOI: 10.7717/peerj.14871] [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: 12/05/2022] [Accepted: 01/18/2023] [Indexed: 02/10/2023] Open
Abstract
Background Autoimmune hepatitis (AIH) is a chronic immune-mediated inflammatory liver disease. At present, it is largely unknown how the innate immune cells influence AIH development. Objective To inquiry about mechanism of liver resident macrophages in AIH development, thus offering a new direction for AIH targeted treatment. Methods The liver resident macrophages were eliminated by clodronate liposomes in AIH liver tissues, followed by HE and Picrosirius assay to detect liver fibrosis and lymphocyte infiltration. The liver resident macrophages polarization was detected by Immunohistochemistry and qPCR. The collagenase digestion was used to isolate Kupffer cells from AIH mice liver tissues and pro-/anti-inflammatory cytokines were determined by qPCR. Results M2 macrophages were the dominant phenotype at early immune response stage and hepatic inflammation was progressively aggravated after depletion of liver resident macrophages. M2 macrophages could effectively delay the development of AIH and could be polarized to M1 macrophages at the disease progresses. TLR2 ligands could promote M2 macrophages producing anti-inflammatory cytokines, whereas TLR4 ligands could promote M1 macrophages producing proinflammatory cytokines. The change of TLR2 and TLR4 ligands could lead to continuous high expression of TLR4 and decreased expression of TLR2 in macrophages to further affect liver resident macrophages polarization state. Conclusion TLR2 and TLR4 ligands mediated liver resident macrophages polarization to favor chronic autoimmune hepatitis development.
Collapse
|
11
|
Tian Z, Zhang P, Li X, Jiang D. Analysis of immunogenic cell death in ascending thoracic aortic aneurysms based on single-cell sequencing data. Front Immunol 2023; 14:1087978. [PMID: 37207221 PMCID: PMC10191229 DOI: 10.3389/fimmu.2023.1087978] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 04/14/2023] [Indexed: 05/21/2023] Open
Abstract
Background At present, research on immunogenic cell death (ICD) is mainly associated with cancer therapy. Little is known about the role of ICD in cardiovascular disease, especially in ascending thoracic aortic aneurysms (ATAA). Method ATAA single-cell RNA (scRNA) sequencing data were analyzed to identify the involved cell types and determine their transcriptomic characteristics. The chi-square test, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, Gene Set Enrichment Analysis (GSEA), and CellChat for cell-to-cell communication analysis from the Gene Expression Omnibus (GEO) database were used. Result A total of 10 cell types were identified, namely, monocytes, macrophages, CD4 T/NK (CD4+ T cells and natural killer T cells), mast cells, B/Plasma B cells, fibroblasts, endothelial cells, cytotoxic T cells (CD8+ T cells, CTLs), vascular smooth muscle cells (vSMCs), and mature dendritic cells (mDCs). A large number of inflammation-related pathways were present in the GSEA results. A large number of ICD-related pathways were found in the KEGG enrichment analysis of differentially expressed genes in endothelial cells. The number of mDCs and CTLs in the ATAA group was significantly different from that in the control group. A total of 44 pathway networks were obtained, of which 9 were associated with ICD in endothelial cells (CCL, CXCL, ANNEXIN, CD40, IL1, IL6, TNF, IFN-II, GALECTIN). The most important ligand-receptor pair by which endothelial cells act on CD4 T/NK cells, CTLs and mDCs is CXCL12-CXCR4. The most important ligand-receptor pair by which endothelial cells act on monocytes and macrophages is ANXA1-FPR1. The most important ligand-receptor pair by which CD4 T/NK cells and CTLs act on endothelial cells is CCL5-ACKR1. The most important ligand-receptor pair that myeloid cells (macrophages, monocytes and mDCs) act on endothelial cells is CXCL8-ACKR1. Moreover, vSMCs and fibroblasts mainly promote inflammatory responses through the MIF signaling pathway. Conclusion ICD is present in ATAA and plays an important role in the development of ATAA. The target cells of ICD may be mainly endothelial cells, in which the aortic endothelial cell ACKR1 receptor can not only promote T-cell infiltration through the CCL5 ligand but also promote myeloid cell infiltration through the CXCL8 ligand. ACKR1 and CXCL12 may become target genes for ATAA drug therapy in the future.
Collapse
Affiliation(s)
- Zemin Tian
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Peng Zhang
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xinyang Li
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- *Correspondence: Delong Jiang, ; Xinyang Li,
| | - Delong Jiang
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- *Correspondence: Delong Jiang, ; Xinyang Li,
| |
Collapse
|
12
|
Zhu XM, Tan Y, Shi YH, Li Q, Zhu J, Liu XD, Tong QZ. TMT-based quantitative proteomics analysis of the effects of Jiawei Danshen decoction myocardial ischemia-reperfusion injury. Proteome Sci 2022; 20:17. [PMID: 36517846 PMCID: PMC9749149 DOI: 10.1186/s12953-022-00200-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Every year, approximately 17 million people worldwide die due to coronary heart disease, with China ranking second in terms of the death toll. Myocardial ischemia-reperfusion injury (MIRI) significantly influences cardiac function and prognosis in cardiac surgery patients. Jiawei Danshen Decoction (JWDSD) is a traditional Chinese herbal prescription that has been used clinically for many years in China to treat MIRI. The underlying molecular mechanisms, however, remain unknown. To investigate the proteomic changes in myocardial tissue of rats given JWDSD for MIRI therapy-based proteomics. METHODS MIRI rat model was created by ligating/releasing the left anterior descending coronary artery. For seven days, the drugs were administered twice daily. The model was created following the last drug administration. JWDSD's efficacy in improving MIRI was evaluated using biochemical markers and cardiac histology. Tandem mass tag-based quantitative proteomics (TMT) technology was also used to detect proteins in the extracted heart tissue. To analyze differentially expressed proteins (DEPs), bioinformatics analysis, including gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathways, were employed. Furthermore, western blotting confirmed the potential targets regulated by JWDSD. RESULTS The histopathologic characteristics and biochemical data showed JWDSD's protective effects on MIRI rats. A total of 4549 proteins were identified with FDR (false discovery rate) ≤1%. Twenty overlapping were identified (162 DEPs and 45 DEPs in Model/Control or JWDSD/Model group, respectively). Of these DEPs, 16 were regulated by JWDSD. GO analysis provided a summary of the deregulated protein expression in the categories of biological process (BP), cell component (CC), and molecular function (MF). KEGG enrichment analysis revealed that the signaling pathways of neutrophil extracellular trap formation, RNA polymerase, serotonergic synapse, and linoleic acid metabolism are all closely related to JWDSD effects in MIRI rats. Furthermore, T-cell lymphoma invasion and metastasis 1 (TIAM1) was validated using western blotting, and the results were consistent with proteomics data. CONCLUSIONS Our study suggests that JWDSD may exert therapeutic effects through multi-pathways regulation in MIRI treatment. This work may provide proteomics clues for continuing research on JWDSD in treating MIRI.
Collapse
Affiliation(s)
- Xiang-Mei Zhu
- grid.488482.a0000 0004 1765 5169Pharmacy of College, Hunan University of Chinese Medicine, Xueshi Road, Number 300, Changsha, Hunan 410208, People’s Republic of China ,grid.67293.39The Second Hospital of Hunan University of Chinese Medicine, Caie North Road, Number 233, Changsha, Hunan 410005, People’s Republic of China
| | - Yang Tan
- grid.488482.a0000 0004 1765 5169Pharmacy of College, Hunan University of Chinese Medicine, Xueshi Road, Number 300, Changsha, Hunan 410208, People’s Republic of China
| | - Yu-He Shi
- grid.488482.a0000 0004 1765 5169Pharmacy of College, Hunan University of Chinese Medicine, Xueshi Road, Number 300, Changsha, Hunan 410208, People’s Republic of China
| | - Qing Li
- grid.488482.a0000 0004 1765 5169Pharmacy of College, Hunan University of Chinese Medicine, Xueshi Road, Number 300, Changsha, Hunan 410208, People’s Republic of China
| | - Jue Zhu
- grid.488482.a0000 0004 1765 5169Pharmacy of College, Hunan University of Chinese Medicine, Xueshi Road, Number 300, Changsha, Hunan 410208, People’s Republic of China
| | - Xiang-Dan Liu
- grid.488482.a0000 0004 1765 5169Pharmacy of College, Hunan University of Chinese Medicine, Xueshi Road, Number 300, Changsha, Hunan 410208, People’s Republic of China ,Key Laboratory of Germplasm Resources and Standardized Planting of Bulk Authentic Medicinal Materials from Hunan, Xueshi Road, Number 300, Changsha, Hunan 410208, People’s Republic of China
| | - Qiao-Zhen Tong
- grid.488482.a0000 0004 1765 5169Pharmacy of College, Hunan University of Chinese Medicine, Xueshi Road, Number 300, Changsha, Hunan 410208, People’s Republic of China ,Key Laboratory of Germplasm Resources and Standardized Planting of Bulk Authentic Medicinal Materials from Hunan, Xueshi Road, Number 300, Changsha, Hunan 410208, People’s Republic of China
| |
Collapse
|
13
|
Brandt KJ, Burger F, Baptista D, Roth A, Fernandes da Silva R, Montecucco F, Mach F, Miteva K. Single-Cell Analysis Uncovers Osteoblast Factor Growth Differentiation Factor 10 as Mediator of Vascular Smooth Muscle Cell Phenotypic Modulation Associated with Plaque Rupture in Human Carotid Artery Disease. Int J Mol Sci 2022; 23:1796. [PMID: 35163719 PMCID: PMC8836240 DOI: 10.3390/ijms23031796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 12/18/2022] Open
Abstract
(1) Background: Vascular smooth muscle cells (VSMCs) undergo a complex phenotypic switch in response to atherosclerosis environmental triggers, contributing to atherosclerosis disease progression. However, the complex heterogeneity of VSMCs and how VSMC dedifferentiation affects human carotid artery disease (CAD) risk has not been clearly established. (2) Method: A single-cell RNA sequencing analysis of CD45- cells derived from the atherosclerotic aorta of Apolipoprotein E-deficient (Apoe-/-) mice on a normal cholesterol diet (NCD) or a high cholesterol diet (HCD), respecting the site-specific predisposition to atherosclerosis was performed. Growth Differentiation Factor 10 (GDF10) role in VSMCs phenotypic switch was investigated via flow cytometry, immunofluorescence in human atherosclerotic plaques. (3) Results: scRNAseq analysis revealed the transcriptomic profile of seven clusters, five of which showed disease-relevant gene signature of VSMC macrophagic calcific phenotype, VSMC mesenchymal chondrogenic phenotype, VSMC inflammatory and fibro-phenotype and VSMC inflammatory phenotype. Osteoblast factor GDF10 involved in ossification and osteoblast differentiation emerged as a hallmark of VSMCs undergoing phenotypic switch. Under hypercholesteremia, GDF10 triggered VSMC osteogenic switch in vitro. The abundance of GDF10 expressing osteogenic-like VSMCs cells was linked to the occurrence of carotid artery disease (CAD) events. (4) Conclusions: Taken together, these results provide evidence about GDF10-mediated VSMC osteogenic switch, with a likely detrimental role in atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Karim J. Brandt
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Daniela Baptista
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Aline Roth
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Rafaela Fernandes da Silva
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 6627, Brazil
- Swiss Institute for Translational and Entrepreneurial Medicine, Freiburgstrasse 3, 3010 Bern, Switzerland
| | - Fabrizio Montecucco
- Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy;
- First Clinic of Internal Medicine, Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - Francois Mach
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Kapka Miteva
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| |
Collapse
|
14
|
Guha A, Wang X, Harris RA, Nelson AG, Stepp D, Klaassen Z, Raval P, Cortes J, Coughlin SS, Bogdanov VY, Moore JX, Desai N, Miller DD, Lu XY, Kim HW, Weintraub NL. Obesity and the Bidirectional Risk of Cancer and Cardiovascular Diseases in African Americans: Disparity vs. Ancestry. Front Cardiovasc Med 2021; 8:761488. [PMID: 34733899 PMCID: PMC8558482 DOI: 10.3389/fcvm.2021.761488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/21/2021] [Indexed: 12/28/2022] Open
Abstract
Cardiovascular disease (CVD) and cancer often occur in the same individuals, in part due to the shared risk factors such as obesity. Obesity promotes adipose inflammation, which is pathogenically linked to both cardiovascular disease and cancer. Compared with Caucasians, the prevalence of obesity is significantly higher in African Americans (AA), who exhibit more pronounced inflammation and, in turn, suffer from a higher burden of CVD and cancer-related mortality. The mechanisms that underlie this association among obesity, inflammation, and the bidirectional risk of CVD and cancer, particularly in AA, remain to be determined. Socio-economic disparities such as lack of access to healthy and affordable food may promote obesity and exacerbate hypertension and other CVD risk factors in AA. In turn, the resulting pro-inflammatory milieu contributes to the higher burden of CVD and cancer in AA. Additionally, biological factors that regulate systemic inflammation may be contributory. Mutations in atypical chemokine receptor 1 (ACKR1), otherwise known as the Duffy antigen receptor for chemokines (DARC), confer protection against malaria. Many AAs carry a mutation in the gene encoding this receptor, resulting in loss of its expression. ACKR1 functions as a decoy chemokine receptor, thus dampening chemokine receptor activation and inflammation. Published and preliminary data in humans and mice genetically deficient in ACKR1 suggest that this common gene mutation may contribute to ethnic susceptibility to obesity-related disease, CVD, and cancer. In this narrative review, we present the evidence regarding obesity-related disparities in the bidirectional risk of CVD and cancer and also discuss the potential association of gene polymorphisms in AAs with emphasis on ACKR1.
Collapse
Affiliation(s)
- Avirup Guha
- Harrington Heart and Vascular Institute, Case Western Reserve University, Cleveland, OH, United States
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Xiaoling Wang
- Georgia Prevention Institute, Augusta University, Augusta, GA, United States
| | - Ryan A. Harris
- Georgia Prevention Institute, Augusta University, Augusta, GA, United States
| | - Anna-Gay Nelson
- Department of Chemistry, Paine College, Augusta, GA, United States
| | - David Stepp
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Zachary Klaassen
- Section of Urology, Department of Surgery, Medical College of Georgia at Augusta University, Georgia Cancer Center, Augusta, GA, United States
| | - Priyanka Raval
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
| | - Jorge Cortes
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
| | - Steven S. Coughlin
- Department of Population Health Sciences, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | | | - Justin X. Moore
- Cancer Prevention, Control, and Population Health Program, Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Nihar Desai
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Center for Outcomes Research and Evaluation, New Haven, CT, United States
| | - D. Douglas Miller
- Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Xin-Yun Lu
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Ha Won Kim
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Neal L. Weintraub
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
15
|
Nucera F, Lo Bello F, Shen SS, Ruggeri P, Coppolino I, Di Stefano A, Stellato C, Casolaro V, Hansbro PM, Adcock IM, Caramori G. Role of Atypical Chemokines and Chemokine Receptors Pathways in the Pathogenesis of COPD. Curr Med Chem 2021; 28:2577-2653. [PMID: 32819230 DOI: 10.2174/0929867327999200819145327] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 11/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) represents a heightened inflammatory response in the lung generally resulting from tobacco smoking-induced recruitment and activation of inflammatory cells and/or activation of lower airway structural cells. Several mediators can modulate activation and recruitment of these cells, particularly those belonging to the chemokines (conventional and atypical) family. There is emerging evidence for complex roles of atypical chemokines and their receptors (such as high mobility group box 1 (HMGB1), antimicrobial peptides, receptor for advanced glycosylation end products (RAGE) or toll-like receptors (TLRs)) in the pathogenesis of COPD, both in the stable disease and during exacerbations. Modulators of these pathways represent potential novel therapies for COPD and many are now in preclinical development. Inhibition of only a single atypical chemokine or receptor may not block inflammatory processes because there is redundancy in this network. However, there are many animal studies that encourage studies for modulating the atypical chemokine network in COPD. Thus, few pharmaceutical companies maintain a significant interest in developing agents that target these molecules as potential antiinflammatory drugs. Antibody-based (biological) and small molecule drug (SMD)-based therapies targeting atypical chemokines and/or their receptors are mostly at the preclinical stage and their progression to clinical trials is eagerly awaited. These agents will most likely enhance our knowledge about the role of atypical chemokines in COPD pathophysiology and thereby improve COPD management.
Collapse
Affiliation(s)
- Francesco Nucera
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Federica Lo Bello
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Sj S Shen
- Faculty of Science, Centre for Inflammation, Centenary Institute, University of Technology, Ultimo, Sydney, Australia
| | - Paolo Ruggeri
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Irene Coppolino
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Antonino Di Stefano
- Division of Pneumology, Cyto- Immunopathology Laboratory of the Cardio-Respiratory System, Clinical Scientific Institutes Maugeri IRCCS, Veruno, Italy
| | - Cristiana Stellato
- Department of Medicine, Surgery and Dentistry, Salerno Medical School, University of Salerno, Salerno, Italy
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry, Salerno Medical School, University of Salerno, Salerno, Italy
| | - Phil M Hansbro
- Faculty of Science, Centre for Inflammation, Centenary Institute, University of Technology, Ultimo, Sydney, Australia
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Gaetano Caramori
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| |
Collapse
|
16
|
Inhibition of fibroblast IL-6 production by ACKR4 deletion alleviates cardiac remodeling after myocardial infarction. Biochem Biophys Res Commun 2021; 547:139-147. [PMID: 33610913 DOI: 10.1016/j.bbrc.2021.02.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
Fibrotic scarring is tightly linked to the development of heart failure in patients with post-myocardial infarction (MI). Atypical chemokine receptor 4 (ACKR4) can eliminate chemokines, such as C-C chemokine ligand 21 (CCL21), which is independently associated with heart failure mortality. However, the role of ACKR4 in the heart during MI is unrevealed. This study aimed to determine whether ACKR4 modulates cardiac remodeling following MI and to illuminate the potential molecular mechanisms. The expression of ACKR4 was upregulated in the border/infarct area, and ACKR4 was predominantly expressed in cardiac fibroblasts (CFs). Knockout of ACKR4 protected against adverse ventricular remodeling in mice post-MI. These protective effects of ACKR4 deficiency were independent of dendritic cell immune response but could be attributed to downregulated CF-derived IL-6, affecting CF proliferation and endothelial cell (EC) functions, which consequently inhibited cardiac fibrosis. ACKR4 promoted IL-6 generation and proliferation of CFs. Besides, ACKR4 induced endothelial-to-mesenchymal transition (EndMT) in ECs through IL-6 paracrine effect. The p38 MAPK/NF-κB signaling pathway was involved in ACKR4 facilitated IL-6 generation. Moreover, ACKR4 overexpression in vivo via AAV9 carrying a periostin promoter aggravated heart functional impairment post-MI, which was abolished by IL-6 neutralizing antibody. Therefore, our study established a novel link between ACKR4 and IL-6 post-MI, indicating that ACKR4 may be a novel therapeutic target to ameliorate cardiac remodeling.
Collapse
|
17
|
Parreira JR, Hernández-Castellano LE, Argüello A, Capote J, Castro N, de Sousa Araújo S, de Almeida AM. Understanding seasonal weight loss tolerance in dairy goats: a transcriptomics approach. BMC Genomics 2020; 21:629. [PMID: 32928114 PMCID: PMC7489022 DOI: 10.1186/s12864-020-06968-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
Background Seasonal weight loss (SWL) is a very important limitation to the production of ruminants in the Mediterranean and Tropical regions. In these areas, long dry seasons lead to poor pastures with low nutritional value. During the dry season, ruminants, particularly those raised in extensive production systems, lose around 30% of their body weight. Seasonal weight loss has important consequences on animal productive performance and health. In this study, RNA sequencing was used to characterize feed restriction effects in dairy goat of 2 breeds with different SWL tolerance: Majorera (tolerant) and Palmera (susceptible). Nine Majorera and ten Palmera goats were randomly distributed in a control and a restricted group: Majorera Control (adequately fed; MC; n = 4), Palmera Control (adequately fed; PC; n = 6), Majorera Restricted (feed restricted; ME; n = 5) and Palmera Restricted (feed restricted; PE; n = 4). On day 22 of the trial, mammary gland biopsies were collected for transcriptomics analysis. Results From these samples, 24,260 unique transcripts were identified. From those, 82 transcripts were differentially expressed between MC and ME, 99 between PC and PE, twelve between both control groups and twenty-nine between both restricted groups. Conclusions Feed restriction affected several biochemical pathways in both breeds such as: carbohydrate and lipid transport; intracellular trafficking, RNA processing and signal transduction. This research also highlights the importance or involvement of the genes in tolerance (ENPP1, S-LZ, MT2A and GPNB) and susceptibility (GPD1, CTPS1, ELOVL6 and NR4A1) to SWL with respectively higher expression in the Majorera restriced group and the Palmera restricted group in comparison to the control groups. In addition, results from the study may be extrapolated to other dairy ruminant species.
Collapse
Affiliation(s)
- José Ricardo Parreira
- IBET - Instituto de Biologia Experimental e Tecnológica, Av. da República, 2780-157, Oeiras, Portugal.,ITQB NOVA - Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | | | - Anastasio Argüello
- Animal Production and Biotechnology group, Institute of Animal Health and Food Safety, Universidad de Las Palmas de Gran Canaria, 35413, Arucas, Spain
| | - Juan Capote
- Unit of Animal Production, Pasture, and Forage in Arid and Subtropical Areas, Canary Islands Institute for Agricultural Research, 38270, La Laguna, Spain
| | - Noemí Castro
- Animal Production and Biotechnology group, Institute of Animal Health and Food Safety, Universidad de Las Palmas de Gran Canaria, 35413, Arucas, Spain
| | - Susana de Sousa Araújo
- ITQB NOVA - Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - André Martinho de Almeida
- LEAF - Linking Landscape, Environment, Agriculture And Food, Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 13409-017, Lisbon, Portugal.
| |
Collapse
|
18
|
Liu J, Li S, Lin L, Jiang Y, Wan Y, Zhou S, Cheng W. Co-expression network analysis identified atypical chemokine receptor 1 (ACKR1) association with lymph node metastasis and prognosis in cervical cancer. Cancer Biomark 2020; 27:213-223. [PMID: 32083574 DOI: 10.3233/cbm-190533] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cervical cancer (CC) is one kind of female cancer. With the development of bioinformatics, targeted specific biomarkers therapy has become much more valuable. GSE26511 was obtained from gene expression omnibus (GEO). We utilized a package called "WGCNA" to build co-expression network and choose the hub module. Search Tool for the Retrieval of Interacting Genes Database (STRING) was used to analyze protein-protein interaction (PPI) information of those genes in the hub module. A Plug-in called MCODE was utilized to choose hub clusters of PPI network, which was visualized in Cytoscape. Clusterprofiler was used to do functional analysis. Univariate and multivariate cox proportional hazards regression analysis were both conducted to predict the risk score of CC patients. Kaplan-Meier curve analysis was done to show the overall survival. Receiver operating characteristic (ROC) curve analysis was utilized to evaluate the predictive value of the patient outcome. Validation of the hub gene in databases, Gene set enrichment analysis (GSEA) and GEPIA were completed. We built co-expression network based on GSE26511 and one CC-related module was identified. Functional analysis of this module showed that extracellular space and Signaling pathways regulating pluripotency of stem cells were most related pathways. PPI network screened GNG11 as the most valuable protein. Cox analysis showed that ACKR1 was negatively correlated with CC progression, which was validated in Gene Expression Profiling Interactive Analysis (GEPIA) and datasets. Survival analysis was performed and showed the consistent result. GSEA set enrichment analysis was also completed. This study showed hub functional terms and gene participated in CC and then speculated that ACKR1 might be tumor suppressor for CC.
Collapse
Affiliation(s)
- Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Siyue Li
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lijuan Lin
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Jiang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yicong Wan
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shulin Zhou
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenjun Cheng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
19
|
Candidate SNP Markers of Atherogenesis Significantly Shifting the Affinity of TATA-Binding Protein for Human Gene Promoters show stabilizing Natural Selection as a Sum of Neutral Drift Accelerating Atherogenesis and Directional Natural Selection Slowing It. Int J Mol Sci 2020; 21:ijms21031045. [PMID: 32033288 PMCID: PMC7037642 DOI: 10.3390/ijms21031045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/15/2022] Open
Abstract
(1) Background: The World Health Organization (WHO) regards atherosclerosis-related myocardial infarction and stroke as the main causes of death in humans. Susceptibility to atherogenesis-associated diseases is caused by single-nucleotide polymorphisms (SNPs). (2) Methods: Using our previously developed public web-service SNP_TATA_Comparator, we estimated statistical significance of the SNP-caused alterations in TATA-binding protein (TBP) binding affinity for 70 bp proximal promoter regions of the human genes clinically associated with diseases syntonic or dystonic with atherogenesis. Additionally, we did the same for several genes related to the maintenance of mitochondrial genome integrity, according to present-day active research aimed at retarding atherogenesis. (3) Results: In dbSNP, we found 1186 SNPs altering such affinity to the same extent as clinical SNP markers do (as estimated). Particularly, clinical SNP marker rs2276109 can prevent autoimmune diseases via reduced TBP affinity for the human MMP12 gene promoter and therefore macrophage elastase deficiency, which is a well-known physiological marker of accelerated atherogenesis that could be retarded nutritionally using dairy fermented by lactobacilli. (4) Conclusions: Our results uncovered SNPs near clinical SNP markers as the basis of neutral drift accelerating atherogenesis and SNPs of genes encoding proteins related to mitochondrial genome integrity and microRNA genes associated with instability of the atherosclerotic plaque as a basis of directional natural selection slowing atherogenesis. Their sum may be stabilizing the natural selection that sets the normal level of atherogenesis.
Collapse
|
20
|
Chemokine (C-C motif) ligand 2 and coronary artery disease: Tissue expression of functional and atypical receptors. Cytokine 2019; 126:154923. [PMID: 31739217 DOI: 10.1016/j.cyto.2019.154923] [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: 04/24/2019] [Revised: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 12/12/2022]
Abstract
Chemokines, particularly chemokine (C-C- motif) ligand 2 (CCL2), control leukocyte migration into the wall of the artery and regulate the traffic of inflammatory cells. CCL2 is bound to functional receptors (CCR2), but also to atypical chemokine receptors (ACKRs), which do not induce cell migration but can modify chemokine gradients. Whether atherosclerosis alters CCL2 function by influencing the expression of these receptors remains unknown. In a necropsy study, we used immunohistochemistry to explore where and to what extent CCL2 and related receptors are present in diseased arteries that caused the death of men with coronary artery disease compared with unaffected arteries. CCL2 was marginally detected in normal arteries but was more frequently found in the intima. The expression of CCL2 and related receptors was significantly increased in diseased arteries with relative differences among the artery layers. The highest relative increases were those of CCL2 and ACKR1. CCL2 expression was associated with a significant predictive value of atherosclerosis. Findings suggest the need for further insight into receptor specificity or activity and the interplay among chemokines. CCL2-associated conventional and atypical receptors are overexpressed in atherosclerotic arteries, and these may suggest new potential therapeutic targets to locally modify the overall anti-inflammatory response.
Collapse
|
21
|
Ponomarenko MP, Rasskazov DA, Chadaeva IV, Sharypova EB, Drachkova IA, Ponomarenko PM, Oshchepkova EA, Savinkova LK, Kolchanov NA. Candidate SNP Markers of Atherosclerosis That May Significantly Change the Affinity of the TATA-Binding Protein for the Human Gene Promoters. RUSS J GENET+ 2019. [DOI: 10.1134/s1022795419090114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
22
|
van der Vorst EPC, Peters LJF, Müller M, Gencer S, Yan Y, Weber C, Döring Y. G-Protein Coupled Receptor Targeting on Myeloid Cells in Atherosclerosis. Front Pharmacol 2019; 10:531. [PMID: 31191301 PMCID: PMC6540917 DOI: 10.3389/fphar.2019.00531] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis, the underlying cause of the majority of cardiovascular diseases (CVDs), is a lipid-driven, inflammatory disease of the large arteries. Gold standard therapy with statins and the more recently developed proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors have improved health conditions among CVD patients by lowering low density lipoprotein (LDL) cholesterol. Nevertheless, a substantial part of these patients is still suffering and it seems that 'just' lipid lowering is insufficient. The results of the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) have now proven that inflammation is a key driver of atherosclerosis and that targeting inflammation improves CVD outcomes. Therefore, the identification of novel drug targets and development of novel therapeutics that block atherosclerosis-specific inflammatory pathways have to be promoted. The inflammatory processes in atherosclerosis are facilitated by a network of immune cells and their subsequent responses. Cell networking is orchestrated by various (inflammatory) mediators which interact, bind and induce signaling. Over the last years, G-protein coupled receptors (GPCRs) emerged as important players in recognizing these mediators, because of their diverse functions in steady state but also and specifically during chronic inflammatory processes - such as atherosclerosis. In this review, we will therefore highlight a selection of these receptors or receptor sub-families mainly expressed on myeloid cells and their role in atherosclerosis. More specifically, we will focus on chemokine receptors, both classical and atypical, formyl-peptide receptors, the chemerin receptor 23 and the calcium-sensing receptor. When information is available, we will also describe the consequences of their targeting which may hold promising options for future treatment of CVD.
Collapse
Affiliation(s)
- Emiel P. C. van der Vorst
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, Netherlands
- Institute for Molecular Cardiovascular Research/Interdisciplinary Center for Clinical Research, RWTH Aachen University, Aachen, Germany
- Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| | - Linsey J. F. Peters
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Madeleine Müller
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Selin Gencer
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Yi Yan
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
- Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Yvonne Döring
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
- Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| |
Collapse
|
23
|
Abstract
The neutrophil's journey through the vascular wall constitutes a critical step during inflammation. In this issue of Immunity, Girbl et al. (2018) demonstrate that neutrophil extravasation is mediated by sequential and compartmentalized chemokine action, endowing unexpected specificity to promiscuous chemokine receptors.
Collapse
|
24
|
Benson TW, Weintraub DS, Crowe M, Yiew NKH, Popoola O, Pillai A, Joseph J, Archer K, Greenway C, Chatterjee TK, Mintz J, Stepp DW, Stansfield BK, Chen W, Brittain J, Bogdanov VY, Gao Y, Wilson JG, Tang Y, Kim HW, Weintraub NL. Deletion of the Duffy antigen receptor for chemokines (DARC) promotes insulin resistance and adipose tissue inflammation during high fat feeding. Mol Cell Endocrinol 2018; 473:79-88. [PMID: 29341885 PMCID: PMC6045443 DOI: 10.1016/j.mce.2018.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Inflammation in adipose tissues in obesity promotes insulin resistance and metabolic disease. The Duffy antigen receptor for chemokines (DARC) is a promiscuous non-signaling receptor expressed on erythrocytes and other cell types that modulates tissue inflammation by binding chemokines such as monocyte chemoattractant protein-1 (MCP-1) and by acting as a chemokine reservoir. DARC allelic variants are common in humans, but the role of DARC in modulating obesity-related metabolic disease is unknown. METHODS We examined body weight gain, tissue adiposity, metabolic parameters and inflammatory marker expression in wild-type and DARC knockout mice fed a chow diet (CD) and high fat diet (HFD). RESULTS Compared to wild-type mice, HFD-fed DARC knockout mice developed glucose intolerance and insulin resistance independent of increases in body weight or adiposity. Interestingly, insulin sensitivity was also diminished in lean male DARC knockout mice fed a chow diet. Insulin production was not reduced by DARC gene deletion, and plasma leptin levels were similar in HFD fed wild-type and DARC knockout mice. MCP-1 levels in plasma rose significantly in the HFD fed wild-type mice, but not in the DARC knockout mice. Conversely, adipose tissue MCP-1 levels were higher, and more macrophage crown-like structures were detected, in the HFD fed DARC knockout mice as compared with the wild-type mice, consistent with augmented adipose tissue inflammation that is not accurately reflected by plasma levels of DARC-bound MCP-1 in these mice. CONCLUSIONS These findings suggest that DARC regulates metabolic function and adipose tissue inflammation, which may impact obesity-related disease in ethnic populations with high frequencies of DARC allelic variants.
Collapse
Affiliation(s)
- Tyler W Benson
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States
| | - Daniel S Weintraub
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States
| | - Matthew Crowe
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States
| | - Nicole K H Yiew
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States; Departments of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, United States
| | - Orishebawo Popoola
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States
| | - Ajay Pillai
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States
| | - Joel Joseph
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States
| | - Krystal Archer
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States
| | - Charlotte Greenway
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States
| | - Tapan K Chatterjee
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States
| | - James Mintz
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States
| | - David W Stepp
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States; Physiology, Medical College of Georgia at Augusta University, United States
| | - Brian K Stansfield
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States; Pediatrics, Medical College of Georgia at Augusta University, United States
| | - Weiqin Chen
- Physiology, Medical College of Georgia at Augusta University, United States
| | - Julia Brittain
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States; Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, United States
| | - Vladimir Y Bogdanov
- Department of Medicine, University of Cincinnati College of Medicine, United States
| | - Yan Gao
- Department of Physiology and Biophysics, University of Mississippi Medical Center, United States
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, United States
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States; Medicine, Medical College of Georgia at Augusta University, United States
| | - Ha Won Kim
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States; Medicine, Medical College of Georgia at Augusta University, United States.
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia at Augusta University, United States; Medicine, Medical College of Georgia at Augusta University, United States.
| |
Collapse
|
25
|
Borroni EM, Savino B, Bonecchi R, Locati M. Chemokines sound the alarmin: The role of atypical chemokine in inflammation and cancer. Semin Immunol 2018; 38:63-71. [DOI: 10.1016/j.smim.2018.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/08/2018] [Indexed: 12/17/2022]
|
26
|
Chapman DG, Mougey EB, Van der Velden JL, Lahue KG, Aliyeva M, Daphtary N, George KL, Hoffman SM, Schneider RW, Tracy RP, Worthen GS, Poynter ME, Peters SP, Lima JJ, Janssen-Heininger YMW, Irvin CG. The Duffy antigen receptor for chemokines regulates asthma pathophysiology. Clin Exp Allergy 2017; 47:1214-1222. [PMID: 28471517 PMCID: PMC5578916 DOI: 10.1111/cea.12949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND The Duffy antigen receptor for chemokines (DARC) is an atypical receptor that regulates pro-inflammatory cytokines. However, the role of DARC in asthma pathophysiology is unknown. OBJECTIVE To determine the role of DARC in allergic airways disease in mice, and the association between DARC single nucleotide polymorphisms (SNPs) and clinical outcomes in patients with asthma. METHODS Mice with targeted disruption of the Darc gene (Darc∆E2 ) or WT mice were challenged over 3 weeks with house dust mite (HDM) antigen. Allergic airways disease was assessed 24 hours and 7 days following the final challenge. Additionally, associations between DARC SNPs and clinical outcomes were analysed in a cohort of poorly controlled asthmatics. RESULTS Total airway inflammation following HDM did not differ between Darc∆E2 and WT mice. At 24 hours, Darc∆E2 mice had increased airway hyperresponsiveness; however, at 7 days airway hyperresponsiveness had completely resolved in Darc∆E2 but persisted in WT mice. In poorly controlled asthmatics, DARC SNPs were associated with worse asthma control at randomization and subsequent increased risk of healthcare utilization (odds ratio 3.13(1.37-7.27), P=.0062). CONCLUSIONS AND CLINICAL RELEVANCE Our animal model and human patient data suggest a novel role for DARC in the temporal regulation in asthma pathophysiology and symptoms.
Collapse
Affiliation(s)
- D G Chapman
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
- Woolcock Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - E B Mougey
- Nemours Pharmacogenetics Center, Nemours Children's Clinic, Jacksonville, FL, USA
| | - J L Van der Velden
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA
| | - K G Lahue
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA
| | - M Aliyeva
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - N Daphtary
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - K L George
- Nemours Pharmacogenetics Center, Nemours Children's Clinic, Jacksonville, FL, USA
| | - S M Hoffman
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA
| | - R W Schneider
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA
| | - R P Tracy
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
- Department of Biochemistry, University of Vermont College of Medicine, Burlington, VT, USA
| | - G S Worthen
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - M E Poynter
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - S P Peters
- Section on Pulmonary, Critical Care, Allergy & Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - J J Lima
- Nemours Pharmacogenetics Center, Nemours Children's Clinic, Jacksonville, FL, USA
| | | | - C G Irvin
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| |
Collapse
|
27
|
Thiriot A, Perdomo C, Cheng G, Novitzky-Basso I, McArdle S, Kishimoto JK, Barreiro O, Mazo I, Triboulet R, Ley K, Rot A, von Andrian UH. Differential DARC/ACKR1 expression distinguishes venular from non-venular endothelial cells in murine tissues. BMC Biol 2017; 15:45. [PMID: 28526034 PMCID: PMC5438556 DOI: 10.1186/s12915-017-0381-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/26/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Intravascular leukocyte recruitment in most vertebrate tissues is restricted to postcapillary and collecting venules, whereas capillaries and arterioles usually support little or no leukocyte adhesion. This segmental restriction is thought to be mediated by endothelial, rather than hemodynamic, differences. The underlying mechanisms are largely unknown, in part because effective tools to distinguish, isolate, and analyze venular endothelial cells (V-ECs) and non-venular endothelial cells (NV-ECs) have been unavailable. We hypothesized that the atypical chemokine receptor DARC (Duffy Antigen Receptor for Chemokines, a.k.a. ACKR1 or CD234) may distinguish V-ECs versus NV-ECs in mice. METHODS We generated a rat-anti-mouse monoclonal antibody (MAb) that specifically recognizes the erythroid and endothelial forms of native, surface-expressed DARC. Using this reagent, we characterized DARC expression and distribution in the microvasculature of murine tissues. RESULTS DARC was exquisitely restricted to post-capillary and small collecting venules and completely absent from arteries, arterioles, capillaries, veins, and most lymphatics in every tissue analyzed. Accordingly, intravital microscopy showed that adhesive leukocyte-endothelial interactions were restricted to DARC+ venules. DARC was detectable over the entire circumference of V-ECs, but was more concentrated at cell-cell junctions. Analysis of single-cell suspensions suggested that the frequency of V-ECs among the total microvascular EC pool varies considerably between different tissues. CONCLUSIONS Immunostaining of endothelial DARC allows the identification and isolation of intact V-ECs from multiple murine tissues. This strategy may be useful to dissect the mechanisms underlying segmental microvascular specialization in healthy and diseased tissues and to characterize the role of EC subsets in tissue-homeostasis, immune surveillance, infection, inflammation, and malignancies.
Collapse
Affiliation(s)
- Aude Thiriot
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Carolina Perdomo
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Guiying Cheng
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Igor Novitzky-Basso
- Center for Immunology and Infection, Department of Biology, University of York, YO10 5DD, Heslington, York, UK
- Present address: Blood and Marrow Transplant Unit, Queen Elizabeth University Hospital, Glasgow, UK
| | - Sara McArdle
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Jamie K Kishimoto
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Olga Barreiro
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Irina Mazo
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | | | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Antal Rot
- Center for Immunology and Infection, Department of Biology, University of York, YO10 5DD, Heslington, York, UK
| | - Ulrich H von Andrian
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA.
| |
Collapse
|
28
|
Seilie ES, Bubeck Wardenburg J. Staphylococcus aureus pore-forming toxins: The interface of pathogen and host complexity. Semin Cell Dev Biol 2017; 72:101-116. [PMID: 28445785 DOI: 10.1016/j.semcdb.2017.04.003] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/22/2017] [Accepted: 04/18/2017] [Indexed: 12/20/2022]
Abstract
Staphylococcus aureus is a prominent human pathogen capable of infecting a variety of host species and tissue sites. This versatility stems from the pathogen's ability to secrete diverse host-damaging virulence factors. Among these factors, the S. aureus pore-forming toxins (PFTs) α-toxin and the bicomponent leukocidins, have garnered much attention for their ability to lyse cells at low concentrations and modulate disease severity. Although many of these toxins were discovered nearly a century ago, their host cell specificities have only been elucidated over the past five to six years, starting with the discovery of the eukaryotic receptor for α-toxin and rapidly followed by identification of the leukocidin receptors. The identification of these receptors has revealed the species- and cell type-specificity of toxin binding, and provided insight into non-lytic effects of PFT intoxication that contribute to disease pathogenesis.
Collapse
Affiliation(s)
- E Sachiko Seilie
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, United States; Department of Microbiology, The University of Chicago, Chicago, IL 60637, United States
| | | |
Collapse
|
29
|
Bonecchi R, Graham GJ. Atypical Chemokine Receptors and Their Roles in the Resolution of the Inflammatory Response. Front Immunol 2016; 7:224. [PMID: 27375622 PMCID: PMC4901034 DOI: 10.3389/fimmu.2016.00224] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 05/25/2016] [Indexed: 11/13/2022] Open
Abstract
Chemokines and their receptors are key mediators of the inflammatory process regulating leukocyte extravasation and directional migration into inflamed and infected tissues. The control of chemokine availability within inflamed tissues is necessary to attain a resolving environment and when this fails chronic inflammation ensues. Accordingly, vertebrates have adopted a number of mechanisms for removing chemokines from inflamed sites to help precipitate resolution. Over the past 15 years, it has become apparent that essential players in this process are the members of the atypical chemokine receptor (ACKR) family. Broadly speaking, this family is expressed on stromal cell types and scavenges chemokines to either limit their spatial availability or to remove them from in vivo sites. Here, we provide a brief review of these ACKRs and discuss their involvement in the resolution of inflammatory responses and the therapeutic implications of our current knowledge.
Collapse
Affiliation(s)
- Raffaella Bonecchi
- Humanitas Clinical and Research Center, Rozzano, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - Gerard J Graham
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow , Glasgow , UK
| |
Collapse
|