1
|
Wang Y, Xu F, Li G, Cheng C, Yu B, Zhang Z, Kong D, Chen F, Liu Y, Fang Z, Cao L, Yu Y, Gu Y, He Y. Structure of scavenger receptor SCARF1 and its interaction with lipoproteins. eLife 2024; 13:RP93428. [PMID: 39541158 PMCID: PMC11563577 DOI: 10.7554/elife.93428] [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] [Indexed: 11/16/2024] Open
Abstract
SCARF1 (scavenger receptor class F member 1, SREC-1 or SR-F1) is a type I transmembrane protein that recognizes multiple endogenous and exogenous ligands such as modified low-density lipoproteins (LDLs) and is important for maintaining homeostasis and immunity. But the structural information and the mechanisms of ligand recognition of SCARF1 are largely unavailable. Here, we solve the crystal structures of the N-terminal fragments of human SCARF1, which show that SCARF1 forms homodimers and its epidermal growth factor (EGF)-like domains adopt a long-curved conformation. Then, we examine the interactions of SCARF1 with lipoproteins and are able to identify a region on SCARF1 for recognizing modified LDLs. The mutagenesis data show that the positively charged residues in the region are crucial for the interaction of SCARF1 with modified LDLs, which is confirmed by making chimeric molecules of SCARF1 and SCARF2. In addition, teichoic acids, a cell wall polymer expressed on the surface of gram-positive bacteria, are able to inhibit the interactions of modified LDLs with SCARF1, suggesting the ligand binding sites of SCARF1 might be shared for some of its scavenging targets. Overall, these results provide mechanistic insights into SCARF1 and its interactions with the ligands, which are important for understanding its physiological roles in homeostasis and the related diseases.
Collapse
Affiliation(s)
- Yuanyuan Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Fan Xu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Guangyi Li
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of SciencesShanghaiChina
| | - Chen Cheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bowen Yu
- Department of Immunology, School of Basic Medical Sciences, Weifang Medical UniversityWeifangChina
| | - Ze Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Dandan Kong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fabao Chen
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yali Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhen Fang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Longxing Cao
- School of Life Science, Westlake UniversityHangzhouChina
| | - Yang Yu
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of SciencesShanghaiChina
| | - Yijun Gu
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of SciencesShanghaiChina
| | - Yongning He
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical TranslationShanghaiChina
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
2
|
Ravi S, Martin LC, Krishnan M, Kumaresan M, Manikandan B, Ramar M. Interactions between macrophage membrane and lipid mediators during cardiovascular diseases with the implications of scavenger receptors. Chem Phys Lipids 2024; 258:105362. [PMID: 38006924 DOI: 10.1016/j.chemphyslip.2023.105362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
The onset and progression of cardiovascular diseases with the major underlying cause being atherosclerosis, occur during chronic inflammatory persistence in the vascular system, especially within the arterial wall. Such prolonged maladaptive inflammation is driven by macrophages and their key mediators are generally attributed to a disparity in lipid metabolism. Macrophages are the primary cells of innate immunity, endowed with expansive membrane domains involved in immune responses with their signalling systems. During atherosclerosis, the membrane domains and receptors control various active organisations of macrophages. Their scavenger/endocytic receptors regulate the trafficking of intracellular and extracellular cargo. Corresponding influence on lipid metabolism is mediated by their dynamic interaction with scavenger membrane receptors and their integrated mechanisms such as pinocytosis, phagocytosis, cholesterol export/import, etc. This interaction not only results in the functional differentiation of macrophages but also modifies their structural configurations. Here, we reviewed the association of macrophage membrane biomechanics and their scavenger receptor families with lipid metabolites during the event of atherogenesis. In addition, the membrane structure of macrophages and the signalling pathways involved in endocytosis integrated with lipid metabolism are detailed. This article establishes future insights into the scavenger receptors as potential targets for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Manikandan Kumaresan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni's College for Women, Chennai 600 015, India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India.
| |
Collapse
|
3
|
Qiao X, Lin H, Zhang Y, Lu D. A novel scavenger receptor (EcSRECII) as a lipopolysaccharide recognition molecule involved in regulating NF-κB activation through extracellular EGF-like cysteine-rich repeat domains with lysosomes in Epinephelus coioides. Int J Biol Macromol 2023:125111. [PMID: 37257531 DOI: 10.1016/j.ijbiomac.2023.125111] [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: 04/12/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
Scavenger receptors (SRs), as multifunctional pattern recognition receptors, play an important role in innate immunity in mammals, however, their function in fish is limited. Herein, scavenger receptor F2 in Epinephelus coioides (EcSRECII) induced an innate immune response to LPS in GS cells. EcSRECII markedly enhanced LPS-induced NF-κB and IFN-β signaling pathways, whereas knockdown of EcSRECII significantly inhibited LPS-induced NF-κB and IFN-β promoter activation. Interestingly, only retain of epidermal growth factor (EGF)/EGF-like domain in EcSRECII resulted in a punctate cytoplasmic distribution, while the C-terminal domain exhibited a distinct cytoskeletal cytoplasmic distribution. Moreover, this EGF/EGF-like domain fragment more sharply impaired its ability to activate EcSRECII-induced NF-κB activation than the C-terminal domain region, but both domains significantly induced IFN-β promoter activation. Full-length EcSRECII and the delete mutant of C-terminal domain could partly colocalize with lysosomes by LPS derived from V. parahaemolyticus (V.p. LPS) in GS cells, but there was no similar distribution in the delete mutant of EGF/EGF-like domain. This finding firstly suggested that the N-terminal EGF/EGF-like domain was necessary for the NF-κB signaling pathway to trigger resistance to vibrio infection and its functional exertion may be associated with lysosomes, thus providing insights into the regulation of vibrio infection resistance in teleosts.
Collapse
Affiliation(s)
- Xifeng Qiao
- State Key Laboratory of Biocontrol and School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, Sun Yat-Sen University, Guangzhou, PR China; Guangzhou Laboratory, Guangzhou 510005, PR China
| | - Haoran Lin
- State Key Laboratory of Biocontrol and School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, Sun Yat-Sen University, Guangzhou, PR China; College of Ocean, Hainan University, Haikou 570228, PR China
| | - Yong Zhang
- State Key Laboratory of Biocontrol and School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, Sun Yat-Sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (ZhanJiang), Fisheries College, Guangdong Ocean University, Zhanjiang 524088, PR China
| | - Danqi Lu
- State Key Laboratory of Biocontrol and School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, Sun Yat-Sen University, Guangzhou, PR China.
| |
Collapse
|
4
|
Proteomic profiling of concurrently isolated primary microvascular endothelial cells, pericytes, and vascular smooth muscle cells from adult mouse heart. Sci Rep 2022; 12:8835. [PMID: 35614104 PMCID: PMC9132906 DOI: 10.1038/s41598-022-12749-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
The microcirculation serves crucial functions in adult heart, distinct from those carried out by epicardial vessels. Microvessels are governed by unique regulatory mechanisms, impairment of which leads to microvessel-specific pathology. There are few treatment options for patients with microvascular heart disease, primarily due to limited understanding of underlying pathology. High throughput mRNA sequencing and protein expression profiling in specific cells can improve our understanding of microvessel biology and disease at the molecular level. Understanding responses of individual microvascular cells to the same physiological or pathophysiological stimuli requires the ability to isolate the specific cell types that comprise the functional units of the microcirculation in the heart, preferably from the same heart, to ensure that different cells have been exposed to the same in-vivo conditions. We developed an integrated process for simultaneous isolation and culture of the main cell types comprising the microcirculation in adult mouse heart: endothelial cells, pericytes, and vascular smooth muscle cells. These cell types were characterized with isobaric labeling quantitative proteomics and mRNA sequencing. We defined microvascular cell proteomes, identified novel protein markers, and confirmed established cell-specific markers. Our results allow identification of unique markers and regulatory proteins that govern microvascular physiology and pathology.
Collapse
|
5
|
Cheng C, Zheng E, Yu B, Zhang Z, Wang Y, Liu Y, He Y. Recognition of lipoproteins by scavenger receptor class A members. J Biol Chem 2021; 297:100948. [PMID: 34252459 PMCID: PMC8353498 DOI: 10.1016/j.jbc.2021.100948] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/22/2021] [Accepted: 07/08/2021] [Indexed: 12/15/2022] Open
Abstract
Scavenger receptor class A (SR-A) proteins are type II transmembrane glycoproteins that form homotrimers on the cell surface. This family has five known members (SCARA1 to 5, or SR-A1 to A5) that recognize a variety of ligands and are involved in multiple biological pathways. Previous reports have shown that some SR-A family members can bind modified low-density lipoproteins (LDLs); however, the mechanisms of the interactions between the SR-A members and these lipoproteins are not fully understood. Here, we systematically characterize the recognition of SR-A receptors with lipoproteins and report that SCARA1 (SR-A1, CD204), MARCO (SCARA2), and SCARA5 recognize acetylated or oxidized LDL and very-low-density lipoprotein in a Ca2+-dependent manner through their C-terminal scavenger receptor cysteine-rich (SRCR) domains. These interactions occur specifically between the SRCR domains and the modified apolipoprotein B component of the lipoproteins, suggesting that they might share a similar mechanism for lipoprotein recognition. Meanwhile, SCARA4, a SR-A member with a carbohydrate recognition domain instead of the SRCR domain at the C terminus, shows low affinity for modified LDL and very-low-density lipoprotein but binds in a Ca2+-independent manner. SCARA3, which does not have a globular domain at the C terminus, was found to have no detectable binding with these lipoproteins. Taken together, these results provide mechanistic insights into the interactions between SR-A family members and lipoproteins that may help us understand the roles of SR-A receptors in lipid transport and related diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Chen Cheng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Enlin Zheng
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Bowen Yu
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ze Zhang
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yuanyuan Wang
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yingbin Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Biliary Tract Disease, Shanghai, China
| | - Yongning He
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China; Shanghai Key Laboratory of Biliary Tract Disease, Shanghai, China.
| |
Collapse
|
6
|
Wicker-Planquart C, Tacnet-Delorme P, Preisser L, Dufour S, Delneste Y, Housset D, Frachet P, Thielens NM. Insights into the ligand binding specificity of SREC-II (scavenger receptor expressed by endothelial cells). FEBS Open Bio 2021; 11:2693-2704. [PMID: 34328698 PMCID: PMC8487046 DOI: 10.1002/2211-5463.13260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/24/2021] [Accepted: 07/29/2021] [Indexed: 11/12/2022] Open
Abstract
SREC-II (scavenger receptor expressed by endothelial cells-II) is a membrane protein encoded by the SCARF2 gene, with high homology to class F scavenger receptor SR-F1, but no known scavenging function. We produced the extracellular domain of SREC-II in a recombinant form and investigated its capacity to interact with common scavenger receptor ligands, including acetylated low density lipoprotein (AcLDL) and maleylated or acetylated BSA (MalBSA or AcBSA). Whereas no binding was observed for AcLDL, SREC-II ectodomain interacted strongly with MalBSA and bound with high affinity to AcBSA, a property shared with the SR-F1 ectodomain. SREC-II ectodomain also interacted with two SR-F1 specific ligands, complement C1q and calreticulin, with affinities in the 100 nM range. We proceeded to generate a stable CHO cell line overexpressing full-length SREC-II; binding of MalBSA to these cells was significantly increased compared to non-transfected CHO cells. In contrast, no increase in binding could be detected for C1q and calreticulin. We show for the first time that SREC-II has the capacity to interact with the common scavenger receptor ligand MalBSA. In addition, our data highlight similarities and differences in the ligand binding properties of SREC-II in soluble form and at the cell surface, and show that endogenous protein ligands of the ectodomain of SREC-II, such as C1q and calreticulin, are shared with the corresponding domain of SR-F1.
Collapse
Affiliation(s)
| | | | - Laurence Preisser
- Univ Angers, Université de Nantes, CHU Angers, Inserm, CRCINA, SFR ICAT, F-49000, Angers, France
| | - Samy Dufour
- Univ. Grenoble Alpes, CNRS, CEA, IBS, F-38000, Grenoble, France
| | - Yves Delneste
- Univ Angers, Université de Nantes, CHU Angers, Inserm, CRCINA, SFR ICAT, F-49000, Angers, France
| | | | | | | |
Collapse
|
7
|
Wall Teichoic Acid in Staphylococcus aureus Host Interaction. Trends Microbiol 2020; 28:985-998. [DOI: 10.1016/j.tim.2020.05.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
|
8
|
Patten DA, Wilkinson AL, O'Rourke JM, Shetty S. Prognostic Value and Potential Immunoregulatory Role of SCARF1 in Hepatocellular Carcinoma. Front Oncol 2020; 10:565950. [PMID: 34354939 PMCID: PMC8336907 DOI: 10.3389/fonc.2020.565950] [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: 05/26/2020] [Accepted: 08/26/2020] [Indexed: 12/19/2022] Open
Abstract
Scavenger receptor class F member 1 (SCARF1) is thought to play an important role in the selective recruitment of CD4+ T cells to liver sinusoidal endothelial cells during chronic liver disease. However, the contribution of SCARF1 to hepatocellular carcinoma (HCC) is currently unknown. We utilized publically-available RNA-sequencing data from The Cancer Genome Atlas (TGCA) to explore SCARF1 expression in HCC and correlated it with a number of clinicopathological features. Flow adhesion assays were used to determine the role of SCARF1 in CD4+ T cell subset recruitment. SCARF1 expression was downregulated in HCC tumor tissues, compared to non-tumoral tissues, and loss of SCARF1 expression was associated with poorly differentiated/aggressive tumors. Additionally, higher SCARF1 expression in HCC tumor tissues was highly prognostic of better overall, disease-free and progression-free survival. SCARF1 within HCC was largely associated with tumor endothelial cells and adhesion studies suggested that it played a role in the specific recruitment of proinflammatory CD4+ T cells (CD4+CD25−) to HCC tumor tissues. Endothelial SCARF1 expression in tumor biopsies may provide critical prognostic information. Additionally, SCARF1 may also be a novel endothelial target that could help re-programme the microenvironment of HCC by promoting effector T cell tumor infiltration.
Collapse
Affiliation(s)
- Daniel A Patten
- National Institute for Health Research Birmingham Liver Biomedical Research Unit and Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Alex L Wilkinson
- National Institute for Health Research Birmingham Liver Biomedical Research Unit and Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Joanne M O'Rourke
- National Institute for Health Research Birmingham Liver Biomedical Research Unit and Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Shishir Shetty
- National Institute for Health Research Birmingham Liver Biomedical Research Unit and Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
9
|
Dietary Apigenin Reduces Induction of LOX-1 and NLRP3 Expression, Leukocyte Adhesion, and Acetylated Low-Density Lipoprotein Uptake in Human Endothelial Cells Exposed to Trimethylamine-N-Oxide. J Cardiovasc Pharmacol 2020; 74:558-565. [PMID: 31815868 DOI: 10.1097/fjc.0000000000000747] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
By inducing vascular inflammation, trimethylamine-N-oxide (TMAO) is associated with endothelial dysfunction, atherosclerosis, and enhanced risk of cardiovascular diseases in humans. However, the underlying mechanisms are unknown. Expression of several genes related to arteriosclerosis, inflammasomes, and endothelial dysfunction was quantified by polymerase chain reaction after exposure to TMAO. LOX-1, ICAM-1, and NLRP3 were also quantified by Western blot, whereas leukocytic adhesion was examined using fluorescently labeled U937 cells. Scavenger receptors, adhesion molecules, and other genes associated with atherosclerosis were induced in endothelial cells exposed to TMAO. On the other hand, apigenin, a flavonoid that is abundant in parsley and celery, prevents initial arteriosclerosis events in endothelial cells. Apigenin reversed the effects of TMAO on mRNA expression of LOX-1, SREC, SR-PSOX, NLRP3, ASC, TXNIP, VCAM-1, ICAM-1, and MCP-1, as well as protein expression of LOX-1, the adhesion molecule ICAM-1, and the inflammasome protein NLRP3. Apigenin also suppressed leukocyte adhesion and uptake of acetylated low-density lipoprotein. The data indicate that expression of scavenger receptors and adhesion molecules in response to TMAO, along with formation of NLRP3 inflammasomes, may drive endothelial dysfunction through uptake of acetylated low-density lipoprotein and lymphocyte adhesion. Apigenin reverses these effects, implying that it may also prevent arteriosclerosis.
Collapse
|
10
|
Shi Q, Carman CV, Chen Y, Sage PT, Xue F, Liang XM, Gilbert GE. Unexpected enhancement of FVIII immunogenicity by endothelial expression in lentivirus-transduced and transgenic mice. Blood Adv 2020; 4:2272-2285. [PMID: 32453842 PMCID: PMC7252558 DOI: 10.1182/bloodadvances.2020001468] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/24/2020] [Indexed: 12/16/2022] Open
Abstract
Factor VIII (FVIII) replacement therapy for hemophilia A is complicated by development of inhibitory antibodies (inhibitors) in ∼30% of patients. Because endothelial cells (ECs) are the primary physiologic expression site, we probed the therapeutic potential of genetically restoring FVIII expression selectively in ECs in hemophilia A mice (FVIIInull). Expression of FVIII was driven by the Tie2 promoter in the context of lentivirus (LV)-mediated in situ transduction (T2F8LV) or embryonic stem cell-mediated transgenesis (T2F8Tg). Both endothelial expression approaches were associated with a strikingly robust immune response. Following in situ T2F8LV-mediated EC transduction, all FVIIInull mice developed inhibitors but had no detectable plasma FVIII. In the transgenic approach, the T2F8Tg mice had normalized plasma FVIII levels, but showed strong sensitivity to developing an FVIII immune response upon FVIII immunization. A single injection of FVIII with incomplete Freund adjuvant led to high titers of inhibitors and reduction of plasma FVIII to undetectable levels. Because ECs are putative major histocompatibility complex class II (MHCII)-expressing nonhematopoietic, "semiprofessional" antigen-presenting cells (APCs), we asked whether they might directly influence the FVIII immune responses. Imaging and flow cytometric studies confirmed that both murine and human ECs express MHCII and efficiently bind and take up FVIII protein in vitro. Moreover, microvascular ECs preconditioned ex vivo with inflammatory cytokines could functionally present exogenously taken-up FVIII to previously primed CD4+/CXCR5+ T follicular helper (Tfh) cells to drive FVIII-specific proliferation. Our results show an unanticipated immunogenicity of EC-expressed FVIII and suggest a context-dependent role for ECs in the regulation of inhibitors as auxiliary APCs for Tfh cells.
Collapse
Affiliation(s)
- Qizhen Shi
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Children's Research Institute, Children's Wisconsin, Milwaukee, WI
- Midwest Athletes Against Childhood Cancer Fund Research Center, Milwaukee, WI
| | - Christopher V Carman
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA
| | - Yingyu Chen
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
| | - Peter T Sage
- Renal Division, Transplant Research Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; and
| | - Feng Xue
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
| | - Xin M Liang
- Department of Medicine, Veterans Affairs Boston Healthcare System and Harvard Medical School, Boston, MA
| | - Gary E Gilbert
- Department of Medicine, Veterans Affairs Boston Healthcare System and Harvard Medical School, Boston, MA
| |
Collapse
|
11
|
Wicker-Planquart C, Dufour S, Tacnet-Delorme P, Bally I, Delneste Y, Frachet P, Housset D, Thielens NM. Molecular and Cellular Interactions of Scavenger Receptor SR-F1 With Complement C1q Provide Insights Into Its Role in the Clearance of Apoptotic Cells. Front Immunol 2020; 11:544. [PMID: 32296440 PMCID: PMC7137648 DOI: 10.3389/fimmu.2020.00544] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/10/2020] [Indexed: 01/05/2023] Open
Abstract
The scavenger receptor SR-F1 binds to and mediates the internalization of a wide range of ligands, and is involved in several immunological processes. We produced recombinant SR-F1 ectodomain and fragments deleted from the last 2 or 5 C-terminal epidermal growth factor-like modules and investigated their role in the binding of acetylated low density lipoprotein (AcLDL), complement C1q, and calreticulin (CRT). C1q measured affinity was in the 100 nM range and C1q interaction occurs via its collagen-like region. We identified two different binding regions on SR-F1: the N-terminal moiety interacts with C1q and CRT whereas the C-terminal moiety binds AcLDL. The role of SR-F1 N-linked glycans was also tested by mutating each of the three glycosylated asparagines. The three mutants retained binding activities for both AcLDL and C1q. A stable THP-1 cell line overexpressing SR-F1 was generated and C1q was shown to bind more strongly to the surface of SR-F1 overexpressing macrophages, with C1q/SR-F1 colocalization observed in some membrane areas. We also observed a higher level of CRT internalization for THP-1 SR-F1 cells. Increasing SR-F1 negatively modulated the uptake of apoptotic cells. Indeed, THP-1 cells overexpressing SR-F1 displayed a lower phagocytic capacity as compared with mock-transfected cells, which could be partially restored by addition of C1q in the extracellular milieu. Our data shed some light on the role of SR-F1 in efferocytosis, through its capacity to bind C1q and CRT, two proteins involved in this process.
Collapse
Affiliation(s)
| | - Samy Dufour
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | | | - Isabelle Bally
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Yves Delneste
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,CHU Angers, Département d'Immunologie Allergologie, Angers, France
| | | | | | | |
Collapse
|
12
|
Qiao X, Li P, He J, Yu Z, Chen J, He L, Yu X, Lin H, Lu D, Zhang Y. Type F scavenger receptor expressed by endothelial cells (SREC)-II from Epinephelus coioides is a potential pathogen recognition receptor in the immune response to Vibrio parahaemolyticus infection. FISH & SHELLFISH IMMUNOLOGY 2020; 98:262-270. [PMID: 31899357 DOI: 10.1016/j.fsi.2019.12.086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/24/2019] [Accepted: 12/28/2019] [Indexed: 06/10/2023]
Abstract
Scavenger receptors play a central role in defending against infectious diseases in mammals. However, the function of SRECII remains unknown in teleost fish. In this study, type F scavenger receptor expressed by endothelial cells-II (SRECII) cDNA sequence was first identified from Epinephelus coioides, named EcSRECII, which contained an N-terminal signal peptide, eight EGF/EGF-like cysteine-rich motifs and a C-terminal low-complexity region. The gene location maps revealed that EcSRECII has the conservation of synteny among selected species. Subcellular localization showed that EcSRECII was mainly located in the cytoplasm in HEK293T cells and GS cells. In healthy E. coioides, EcSRECII mRNA was highly expressed in spleen, skin, gill, thymus and head kidney. The relative EcSRECII mRNA expression after Vibrio parahaemolyticus infection was significantly up-regulated at 12 h in spleen, head kidney and thymus, but downregulated at 1 d in skin and reduced at 3 d and 1 w in spleen. Furthermore, overexpression of EcSRECII activated NF-κB and IFN-β signaling pathway in vitro. Taken together, these results indicated that EcSRECII could be as the potential pathogen recognition receptor for involving in bacterial infection by regulating innate immunity responses in E. coioides.
Collapse
Affiliation(s)
- Xifeng Qiao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
| | - Pingchao Li
- Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences, Guangzhou, 510530, PR China
| | - Jianan He
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
| | - Zeshu Yu
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
| | - Jiaxing Chen
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
| | - Liangge He
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
| | - Xue Yu
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, PR China; College of Ocean, Hainan University, Haikou, 570228, PR China
| | - Danqi Lu
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China.
| | - Yong Zhang
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, PR China; Southern Marine Science and Engineering Guangdong Laboratory (ZhanJiang), Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, PR China.
| |
Collapse
|
13
|
Ahmed M, Thirunavukkarasu S, Rosa BA, Thomas KA, Das S, Rangel-Moreno J, Lu L, Mehra S, Mbandi SK, Thackray LB, Diamond MS, Murphy KM, Means T, Martin J, Kaushal D, Scriba TJ, Mitreva M, Khader SA. Immune correlates of tuberculosis disease and risk translate across species. Sci Transl Med 2020; 12:eaay0233. [PMID: 31996462 PMCID: PMC7354419 DOI: 10.1126/scitranslmed.aay0233] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/29/2019] [Accepted: 01/09/2020] [Indexed: 12/12/2022]
Abstract
One quarter of the world's population is infected with Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). Although most infected individuals successfully control or clear the infection, some individuals will progress to TB disease. Immune correlates identified using animal models are not always effectively translated to human TB, thus resulting in a slow pace of translational discoveries from animal models to human TB for many platforms including vaccines, therapeutics, biomarkers, and diagnostic discovery. Therefore, it is critical to improve our poor understanding of immune correlates of disease and protection that are shared across animal TB models and human TB. In this study, we have provided an in-depth identification of the conserved and diversified gene/immune pathways in TB models of nonhuman primate and diversity outbred mouse and human TB. Our results show that prominent differentially expressed genes/pathways induced during TB disease progression are conserved in genetically diverse mice, macaques, and humans. In addition, using gene-deficient inbred mouse models, we have addressed the functional role of individual genes comprising the gene signature of disease progression seen in humans with Mtb infection. We show that genes representing specific immune pathways can be protective, detrimental, or redundant in controlling Mtb infection and translate into identifying immune pathways that mediate TB immunopathology in humans. Together, our cross-species findings provide insights into modeling TB disease and the immunological basis of TB disease progression.
Collapse
Affiliation(s)
- Mushtaq Ahmed
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | | | - Bruce A Rosa
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kimberly A Thomas
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Shibali Das
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Javier Rangel-Moreno
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY 14624, USA
| | - Lan Lu
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Smriti Mehra
- Department of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433, USA
| | - Stanley Kimbung Mbandi
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Larissa B Thackray
- Department of Medicine, Division of Infectious Diseases, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Medicine, Division of Infectious Diseases, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Terry Means
- Autoimmunity Cluster, Immunology & Inflammation Therapeutic Area, Sanofi, Cambridge, MA 02139, USA
| | - John Martin
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Makedonka Mitreva
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63110, USA.
- Department of Medicine, Division of Infectious Diseases, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
14
|
Tousignant KD, Rockstroh A, Taherian Fard A, Lehman ML, Wang C, McPherson SJ, Philp LK, Bartonicek N, Dinger ME, Nelson CC, Sadowski MC. Lipid Uptake Is an Androgen-Enhanced Lipid Supply Pathway Associated with Prostate Cancer Disease Progression and Bone Metastasis. Mol Cancer Res 2019; 17:1166-1179. [PMID: 30808729 DOI: 10.1158/1541-7786.mcr-18-1147] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/03/2019] [Accepted: 02/21/2019] [Indexed: 11/16/2022]
Abstract
De novo lipogenesis is a well-described androgen receptor (AR)-regulated metabolic pathway that supports prostate cancer tumor growth by providing fuel, membrane material, and steroid hormone precursor. In contrast, our current understanding of lipid supply from uptake of exogenous lipids and its regulation by AR is limited, and exogenous lipids may play a much more significant role in prostate cancer and disease progression than previously thought. By applying advanced automated quantitative fluorescence microscopy, we provide the most comprehensive functional analysis of lipid uptake in cancer cells to date and demonstrate that treatment of AR-positive prostate cancer cell lines with androgens results in significantly increased cellular uptake of fatty acids, cholesterol, and low-density lipoprotein particles. Consistent with a direct, regulatory role of AR in this process, androgen-enhanced lipid uptake can be blocked by the AR-antagonist enzalutamide, but is independent of proliferation and cell-cycle progression. This work for the first time comprehensively delineates the lipid transporter landscape in prostate cancer cell lines and patient samples by analysis of transcriptomics and proteomics data, including the plasma membrane proteome. We show that androgen exposure or deprivation regulates the expression of multiple lipid transporters in prostate cancer cell lines and tumor xenografts and that mRNA and protein expression of lipid transporters is enhanced in bone metastatic disease when compared with primary, localized prostate cancer. Our findings provide a strong rationale to investigate lipid uptake as a therapeutic cotarget in the fight against advanced prostate cancer in combination with inhibitors of lipogenesis to delay disease progression and metastasis. IMPLICATIONS: Prostate cancer exhibits metabolic plasticity in acquiring lipids from uptake and lipogenesis at different disease stages, indicating potential therapeutic benefit by cotargeting lipid supply.
Collapse
Affiliation(s)
- Kaylyn D Tousignant
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Anja Rockstroh
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Atefeh Taherian Fard
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Melanie L Lehman
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Chenwei Wang
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Stephen J McPherson
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Lisa K Philp
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Nenad Bartonicek
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, Australia
| | - Marcel E Dinger
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, Australia
| | - Colleen C Nelson
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Martin C Sadowski
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
15
|
Patten DA, Shetty S. More Than Just a Removal Service: Scavenger Receptors in Leukocyte Trafficking. Front Immunol 2018; 9:2904. [PMID: 30631321 PMCID: PMC6315190 DOI: 10.3389/fimmu.2018.02904] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/27/2018] [Indexed: 12/15/2022] Open
Abstract
Scavenger receptors are a highly diverse superfamily of proteins which are grouped by their inherent ability to bind and internalize a wide array of structurally diverse ligands which can be either endogenous or exogenous in nature. Consequently, scavenger receptors are known to play important roles in host homeostasis, with common endogenous ligands including apoptotic cells, and modified low density lipoproteins (LDLs); additionally, scavenger receptors are key regulators of inflammatory diseases, such as atherosclerosis. Also, as a consequence of their affinity for a wide range of microbial products, their role in innate immunity is also being increasingly studied. However, in this review, a secondary function of a number of endothelial-expressed scavenger receptors is discussed. There is increasing evidence that some endothelial-expressed scavenger receptors are able to directly bind leukocyte-expressed ligands and subsequently act as adhesion molecules in the trafficking of leukocytes in lymphatic and vascular tissues. Here, we cover the current literature on this alternative role for endothelial-expressed scavenger receptors and also speculate on their therapeutic potential.
Collapse
Affiliation(s)
- Daniel A Patten
- National Institute for Health Research Birmingham Liver Biomedical Research Unit and Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Shishir Shetty
- National Institute for Health Research Birmingham Liver Biomedical Research Unit and Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
16
|
SCARF1: a multifaceted, yet largely understudied, scavenger receptor. Inflamm Res 2018; 67:627-632. [PMID: 29725698 PMCID: PMC6028831 DOI: 10.1007/s00011-018-1154-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/24/2018] [Accepted: 04/26/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND As is a prerequisite of belonging to the scavenger receptor super family, SCARF1 (scavenger receptor class F, member 1) is known to play a key role in the binding and endocytosis of a wide range of endogenous and exogenous ligands. FINDINGS Unlike most scavenger receptors, SCARF1 is an essential protein, as SCARF1-deficient mice exhibit a severe resting phenotype in which they develop systemic lupus erythematosus (SLE)-like disease, thus highlighting the importance of SCARF1-mediated clearance of apoptotic host cells in homeostasis. In addition, a number of other roles in homeostasis and disease pathology have also been suggested, including roles in both innate and adaptive immunity; however, the majority of these studies have utilised transfected cell lines engineered to ectopically express SCARF1 and very few have utilised in vivo or ex vivo approaches. CONCLUSION This review summarises our current knowledge on SCARF1 biology and reflects on future directions for research on this multifaceted, yet largely understudied, scavenger receptor.
Collapse
|
17
|
Belt H, Koponen JK, Kekarainen T, Puttonen KA, Mäkinen PI, Niskanen H, Oja J, Wirth G, Koistinaho J, Kaikkonen MU, Ylä-Herttuala S. Temporal Dynamics of Gene Expression During Endothelial Cell Differentiation From Human iPS Cells: A Comparison Study of Signalling Factors and Small Molecules. Front Cardiovasc Med 2018; 5:16. [PMID: 29594149 PMCID: PMC5861200 DOI: 10.3389/fcvm.2018.00016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/16/2018] [Indexed: 01/22/2023] Open
Abstract
Endothelial cell (EC) therapy may promote vascular growth or reendothelization in a variety of disease conditions. However, the production of a cell therapy preparation containing differentiated, dividing cells presenting typical EC phenotype, functional properties and chemokine profile is challenging. We focused on comparative analysis of seven small molecule-mediated differentiation protocols of ECs from human induced pluripotent stem cells. Differentiated cells showed a typical surface antigen pattern of ECs as characterized with flow cytometry analysis, functional properties, such as tube formation and ability to uptake acetylated LDL. Gene expression analysis by RNA sequencing revealed an efficient silencing of pluripotency genes and upregulation of genes related to cellular adhesion during differentiation. In addition, distinct patterns of transcription factor expression were identified during cellular reprogramming providing targets for more effective differentiation protocols in the future. Altogether, our results suggest that the most optimal EC differentiation protocol includes early inhibition of Rho-associated coiled-coil kinase and activation of cyclic AMP signaling, and inhibition of transforming growth factor beta signaling after mesodermal stage. These findings provide the first systematic characterization of the most potent signalling factors and small molecules used to generate ECs from human induced pluripotent stem cells and, consequently, this work improves the existing EC differentiation protocols and opens up new avenues for controlling cell fate for regenerative EC therapy.
Collapse
Affiliation(s)
- Heini Belt
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jonna K Koponen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | - Katja A Puttonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Kuopio Center for Gene and Cell Therapy, Kuopio, Finland
| | - Petri I Mäkinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Henri Niskanen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Joni Oja
- FinVector Vision Therapies Oy, Kuopio, Finland
| | - Galina Wirth
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jari Koistinaho
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Minna U Kaikkonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
18
|
Zhang B, Nguyen LXT, Li L, Zhao D, Kumar B, Wu H, Lin A, Pellicano F, Hopcroft L, Su YL, Copland M, Holyoake TL, Kuo CJ, Bhatia R, Snyder DS, Ali H, Stein AS, Brewer C, Wang H, McDonald T, Swiderski P, Troadec E, Chen CC, Dorrance A, Pullarkat V, Yuan YC, Perrotti D, Carlesso N, Forman SJ, Kortylewski M, Kuo YH, Marcucci G. Bone marrow niche trafficking of miR-126 controls the self-renewal of leukemia stem cells in chronic myelogenous leukemia. Nat Med 2018; 24:450-462. [PMID: 29505034 PMCID: PMC5965294 DOI: 10.1038/nm.4499] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/22/2018] [Indexed: 12/16/2022]
Abstract
Leukemia stem cells (LSCs) in individuals with chronic myelogenous leukemia (CML) (hereafter referred to as CML LSCs) are responsible for initiating and maintaining clonal hematopoiesis. These cells persist in the bone marrow (BM) despite effective inhibition of BCR-ABL kinase activity by tyrosine kinase inhibitors (TKIs). Here we show that although the microRNA (miRNA) miR-126 supported the quiescence, self-renewal and engraftment capacity of CML LSCs, miR-126 levels were lower in CML LSCs than in long-term hematopoietic stem cells (LT-HSCs) from healthy individuals. Downregulation of miR-126 levels in CML LSCs was due to phosphorylation of Sprouty-related EVH1-domain-containing 1 (SPRED1) by BCR-ABL, which led to inhibition of the RAN-exportin-5-RCC1 complex that mediates miRNA maturation. Endothelial cells (ECs) in the BM supply miR-126 to CML LSCs to support quiescence and leukemia growth, as shown using mouse models of CML in which Mir126a (encoding miR-126) was conditionally knocked out in ECs and/or LSCs. Inhibition of BCR-ABL by TKI treatment caused an undesired increase in endogenous miR-126 levels, which enhanced LSC quiescence and persistence. Mir126a knockout in LSCs and/or ECs, or treatment with a miR-126 inhibitor that targets miR-126 expression in both LSCs and ECs, enhanced the in vivo anti-leukemic effects of TKI treatment and strongly diminished LSC leukemia-initiating capacity, providing a new strategy for the elimination of LSCs in individuals with CML.
Collapse
Affiliation(s)
- Bin Zhang
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Le Xuan Truong Nguyen
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA.,Department of Medical Biotechnology, Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Ling Li
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Dandan Zhao
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Bijender Kumar
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Herman Wu
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Allen Lin
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Francesca Pellicano
- Paul O' Gorman Leukemia Research Centre, College of Medical, Veterinary and Life Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Lisa Hopcroft
- Paul O' Gorman Leukemia Research Centre, College of Medical, Veterinary and Life Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Yu-Lin Su
- Department of Immuno-oncology, City of Hope Medical Center, Duarte, California, USA
| | - Mhairi Copland
- Paul O' Gorman Leukemia Research Centre, College of Medical, Veterinary and Life Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Tessa L Holyoake
- Paul O' Gorman Leukemia Research Centre, College of Medical, Veterinary and Life Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Calvin J Kuo
- Stanford University School of Medicine, Stanford, California, USA
| | - Ravi Bhatia
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David S Snyder
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Haris Ali
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Anthony S Stein
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Casey Brewer
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Huafeng Wang
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA.,Department of Hematology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tinisha McDonald
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Piotr Swiderski
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Estelle Troadec
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Ching-Cheng Chen
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Adrienne Dorrance
- Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio, USA
| | - Vinod Pullarkat
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Yate-Ching Yuan
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Danilo Perrotti
- Department of Medicine, Biochemistry and Molecular Biology and the Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine Baltimore, Baltimore, Maryland, USA.,Deparment of Hematology, Hammersmith Hospital, Imperial College London, London, UK
| | - Nadia Carlesso
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Stephen J Forman
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Marcin Kortylewski
- Department of Immuno-oncology, City of Hope Medical Center, Duarte, California, USA
| | - Ya-Huei Kuo
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Guido Marcucci
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| |
Collapse
|
19
|
PRR Function of Innate Immune Receptors in Recognition of Bacteria or Bacterial Ligands. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1112:255-280. [DOI: 10.1007/978-981-13-3065-0_18] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
20
|
SCARF-1 promotes adhesion of CD4 + T cells to human hepatic sinusoidal endothelium under conditions of shear stress. Sci Rep 2017; 7:17600. [PMID: 29242513 PMCID: PMC5730566 DOI: 10.1038/s41598-017-17928-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 11/30/2017] [Indexed: 02/07/2023] Open
Abstract
Liver-resident cells are constantly exposed to gut-derived antigens via portal blood and, as a consequence, they express a unique repertoire of scavenger receptors. Whilst there is increasing evidence that the gut contributes to chronic inflammatory liver disease, the role of scavenger receptors in regulating liver inflammation remains limited. Here, we describe for the first time the expression of scavenger receptor class F, member 1 (SCARF-1) on hepatic sinusoidal endothelial cells (HSEC). We report that SCARF-1 shows a highly localised expression pattern and co-localised with endothelial markers on sinusoidal endothelium. Analysis of chronically inflamed liver tissue demonstrated accumulation of SCARF-1 at sites of CD4+ T cell aggregation. We then studied the regulation and functional role of SCARF-1 in HSEC and showed that SCARF-1 expression by HSEC is regulated by proinflammatory cytokines and bacterial lipopolysaccharide (LPS). Furthermore, SCARF-1 expression by HSEC, induced by proinflammatory and gut-derived factors acts as a novel adhesion molecule, present in adhesive cup structures, that specifically supports CD4+ T cells under conditions of physiological shear stress. In conclusion, we show that SCARF-1 contributes to lymphocyte subset adhesion to primary human HSEC and could play an important role in regulating the inflammatory response during chronic liver disease.
Collapse
|
21
|
Penberthy KK, Ravichandran KS. Apoptotic cell recognition receptors and scavenger receptors. Immunol Rev 2016; 269:44-59. [PMID: 26683144 DOI: 10.1111/imr.12376] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Phosphatidylserine recognition receptors are a highly diverse set of receptors grouped by their ability to recognize the 'eat-me' signal phosphatidylserine on apoptotic cells. Most of the phosphatidylserine recognition receptors dampen inflammation by inducing the production of anti-inflammatory mediators during the phagocytosis of apoptotic corpses. However, many phosphatidylserine receptors are also capable of recognizing other ligands, with some receptors being categorized as scavenger receptors. It is now appreciated that these receptors can elicit different downstream events for particular ligands. Therefore, how phosphatidylserine recognition receptors mediate specific signals during recognition of apoptotic cells versus other ligands, and how this might help regulate the inflammatory state of a tissue is an important question that is not fully understood. Here, we revisit the work on signaling downstream of the phosphatidylserine recognition receptor BAI1, and evaluate how these and other signaling modules mediate signaling downstream from other receptors, including Stabilin-2, MerTK, and αvβ5. We also propose the concept that phosphatidylserine recognition receptors could be viewed as a subset of scavenger receptors that are capable of eliciting anti-inflammatory responses to apoptotic cells.
Collapse
Affiliation(s)
- Kristen K Penberthy
- Department of Microbiology, Immunology, and Cancer Biology, Center for Cell Clearance, University of Virginia, Charlottesville, VA, USA
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer Biology, Center for Cell Clearance, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
22
|
He J, Liu W, Wang S, Liu W, Liu H. The SREC-I and SREC-II associated with epidermal growth factor in scavenger receptor family are the potential regulative transmembrane receptors in Larimichthys crocea. FISH & SHELLFISH IMMUNOLOGY 2015; 47:182-195. [PMID: 26343178 DOI: 10.1016/j.fsi.2015.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 07/03/2015] [Accepted: 08/14/2015] [Indexed: 06/05/2023]
Abstract
In innate immunity, the regulation of the immunologic gene expression plays a vital role in defense against pathogenic threat. The class F scavenger receptors (SCARFs), a kind of crucial immunologic type I transmembrane receptors, mainly involve in the signal transmission and eliminating pathogens in host immune system. In this study, the SREC-I and SREC-II of SCARFs in Larimichthys crocea (designated as LycSREC1 and LycSREC2 respectively) were first identified, the potential genetic locus relationships with other species were depicted and the features of gene expression after Vibrio alginolyticus stimulation were tested. The results demonstrated that the complete ORF sequences of two candidates were 3024 bp and 2832 bp (KM884873 and KM884874) respectively including some important domains and motifs, such as EGF/EGF-like domains, TRAF2-binding consensus motif, generic motif and atipical motif. The gene location maps and genetic locus interpreted that the DNA sequences of LycSREC1 and LycSREC2 were 7603 bp and 4883 bp, and some locus had changed compared with human being, but three more crucial genetic locus were conservative among ten species. Furthermore, quantitative real-time PCR (qRT-PCR) analysis indicated that the highest mRNA expression of LycSREC1 and LycSREC2 were both in liver among eight detected tissues, and their expression were up-regulated by V. alginolyticus stimulation. All these findings would contribute to better understanding the biologic function of SCARFs in defending against pathogenic bacteria challenge and further exploring the innate immune of sciaenidae fish.
Collapse
Affiliation(s)
- Jianyu He
- National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan 316022, PR China
| | - Wei Liu
- National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan 316022, PR China
| | - Shaoping Wang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Wan Liu
- National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan 316022, PR China
| | - Huihui Liu
- National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan 316022, PR China.
| |
Collapse
|
23
|
Jung JY, Suh CH. Incomplete clearance of apoptotic cells in systemic lupus erythematosus: pathogenic role and potential biomarker. Int J Rheum Dis 2015; 18:294-303. [PMID: 25884792 DOI: 10.1111/1756-185x.12568] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with repeated inflammation against multiple organs. Although its pathophysiology is not yet unveiled, uncleared apoptotic cells and their accumulation in tissue contribute to the autoimmune disturbance in SLE. Apoptosis is a programmed cell death process, which maintains tissue homeostasis and inhibits the development of any further immune response against apoptotic remnants. Earlier studies revealed that various 'eat-me' signals on apoptotic cells, bridging molecules and their receptors on phagocytes play a role in such a complicated process. Tyro3-Axl-Mer receptors, their bridging molecules, milk fat globulin epidermal growth factor-8, T-cell immunoglobulin mucin domain protein family, scavenger receptors, C1q, and pentraxins were found to be abnormal in SLE. In this review, apoptosis and clearance of its remnants are summarized, and the molecules involved in the incomplete clearance of apoptotic cells in SLE are discussed.
Collapse
Affiliation(s)
- Ju-Yang Jung
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Korea
| | | |
Collapse
|
24
|
Biedroń R, Konopiński MK, Marcinkiewicz J, Józefowski S. Oxidation by neutrophils-derived HOCl increases immunogenicity of proteins by converting them into ligands of several endocytic receptors involved in antigen uptake by dendritic cells and macrophages. PLoS One 2015; 10:e0123293. [PMID: 25849867 PMCID: PMC4388828 DOI: 10.1371/journal.pone.0123293] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/26/2015] [Indexed: 11/21/2022] Open
Abstract
The initiation of adaptive immune responses to protein antigens has to be preceded by their uptake by antigen presenting cells and intracellular proteolytic processing. Paradoxically, endocytic receptors involved in antigen uptake do not bind the majority of proteins, which may be the main reason why purified proteins stimulate at most weak immune responses. A shared feature of different types of adjuvants, capable of boosting immunogenicity of protein vaccines, is their ability to induce acute inflammation, characterized by early influx of activated neutrophils. Neutrophils are also rapidly recruited to sites of tissue injury or infection. These cells are the source of potent oxidants, including hypochlorous acid (HOCl), causing oxidation of proteins present in inflammatory foci. We demonstrate that oxidation of proteins by endogenous, neutrophils-derived HOCl increases their immunogenicity. Upon oxidation, different, randomly chosen simple proteins (yeast alcohol dehydrogenase, human and bovine serum albumin) and glycoproteins (human apo-transferrin, ovalbumin) gain the ability to bind with high affinity to several endocytic receptors on antigen presenting cells, which seems to be the major mechanism of their increased immunogenicity. The mannose receptor (CD206), scavenger receptors A (CD204) and CD36 were responsible for the uptake and presentation of HOCl-modified proteins by murine dendritic cells and macrophages. Other scavenger receptors, SREC-I and LOX-1, as well as RAGE were also able to bind HOCl-modified proteins, but they did not contribute significantly to these ligands uptake by dendritic cells because they were either not expressed or exhibited preference for more heavily oxidised proteins. Our results indicate that oxidation by neutrophils-derived HOCl may be a physiological mechanism of conferring immunogenicity on proteins which in their native forms do not bind to endocytic receptors. This mechanism might enable the immune system to detect infections caused by pathogens not recognized by pattern recognition receptors.
Collapse
Affiliation(s)
- Rafał Biedroń
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
| | | | - Janusz Marcinkiewicz
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
| | - Szczepan Józefowski
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
- * E-mail:
| |
Collapse
|
25
|
Murshid A, Gong J, Prince T, Borges TJ, Calderwood SK. Scavenger receptor SREC-I mediated entry of TLR4 into lipid microdomains and triggered inflammatory cytokine release in RAW 264.7 cells upon LPS activation. PLoS One 2015; 10:e0122529. [PMID: 25836976 PMCID: PMC4383338 DOI: 10.1371/journal.pone.0122529] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 02/20/2015] [Indexed: 12/11/2022] Open
Abstract
Scavenger receptor associated with endothelial cells I (SREC-I) was shown to be expressed in immune cells and to play a role in the endocytosis of peptides and antigen presentation. As our previous studies indicated that SREC-I required intact Toll-like receptor 4 (TLR4) expression for its functions in tumor immunity, we examined potential interactions between these two receptors. We have shown here that SREC-I became associated with TLR4 on binding bacterial lipopolysaccharides (LPS) in RAW 264.7 and HEK 293 cells overexpressing these two receptors. The receptors then became internalized together in intracellular endosomes. SREC-I promoted TLR4-induced signal transduction through the NF-kB and MAP kinase pathways, leading to enhanced inflammatory cytokine release. Activation of inflammatory signaling through SREC-I/TLR4 complexes appeared to involve recruitment of the receptors into detergent-insoluble, cholesterol-rich lipid microdomains that contained the small GTPase Cdc42 and the non-receptor tyrosine kinase c-src. Under conditions of SREC-I activation by LPS, TLR4 activity required Cdc42 as well as cholesterol and actin polymerization for signaling through NF-kB and MAP kinase pathways in RAW 264.7 cells. SREC-I appeared to respond differently to another ligand, the molecular chaperone Hsp90 that, while triggering SREC-I-TLR4 binding caused only faint activation of the NF-kB pathway. Our experiments therefore indicated that SREC-I could bind LPS and might be involved in innate inflammatory immune responses to extracellular danger signals in RAW 264.7 cells or bone marrow-derived macrophages.
Collapse
Affiliation(s)
- Ayesha Murshid
- Molecular and Cellular Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 3 Blackfan Circle, Boston, Massachusetts, United States of America
| | - Jianlin Gong
- Stress Response Center, Boston University Medical Center, Boston, Massachusetts, United States of America
| | - Thomas Prince
- Molecular and Cellular Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 3 Blackfan Circle, Boston, Massachusetts, United States of America
| | - Thiago J. Borges
- Molecular and Cellular Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 3 Blackfan Circle, Boston, Massachusetts, United States of America
- School of Biosciences and Biomedical Research Institute, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Stuart K. Calderwood
- Molecular and Cellular Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 3 Blackfan Circle, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
26
|
Murshid A, Borges TJ, Calderwood SK. Emerging roles for scavenger receptor SREC-I in immunity. Cytokine 2015; 75:256-60. [PMID: 25767073 DOI: 10.1016/j.cyto.2015.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 02/09/2015] [Accepted: 02/10/2015] [Indexed: 10/23/2022]
Abstract
SREC-I is a class F scavenger receptor with key role in the immune response, particularly in antigen presenting cell (APC) such as macrophages and dendritic cells (DC). This receptor is able to mediate engulfment of dead cells as well as endocytosis of heat shock protein (HSP)-antigen complexes. SREC-I could thus potentially mediate the tolerizing influence of apoptotic cells or the immunostimulatory effects of HSP-peptide complexes, depending on context. This receptor was able to mediate presentation of external antigens, bound to HSPs through both the class II pathway as well as cross presentation via MHC class I complexes. In addition to its recently established role in adaptive immunity, emerging studies are indicating a broad role in innate immunity and regulation of cell signaling through Toll Like Receptors (TLR). SREC-I may thus play a key role in APC function by coordinating immune responses to internal and external antigens in APC.
Collapse
Affiliation(s)
- Ayesha Murshid
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, United States
| | - Thiago J Borges
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, United States; School of Biosciences and Biomedical Research Institute, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Stuart K Calderwood
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
27
|
Murshid A, Gong J, Ahmad R, Borges TJ, Calderwood SK. Scavenger receptor SREC-I promotes double stranded RNA-mediated TLR3 activation in human monocytes. Immunobiology 2014; 220:823-32. [PMID: 25641411 DOI: 10.1016/j.imbio.2014.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 11/25/2014] [Accepted: 12/22/2014] [Indexed: 10/24/2022]
Abstract
Scavenger receptor associated with endothelial cells (SREC-I) was previously shown to be expressed by immune cells and to play a role in CD8(+)-mediated T cell immunity. SREC-I was also shown to modulate the function of Toll like receptors with essential roles in innate immunity. Here we have shown that SREC-I enhanced double stranded RNA (dsRNA)-mediated Toll like receptor-3 (TLR3) activation. Viral double stranded RNA (dsRNA) was demonstrated to be a pathogen associated molecular pattern (PAMP) signaling viral infection. We found that in human monocyte/macrophage THP1 cells as well as murine bone marrow derived macrophages SREC-I led to elevated responses to the dsRNA-like molecule polyinosine-polycytidylic acid (Poly I:C) and enhanced production of inflammatory cytokines. Our data also showed that intracellular/endocytic TLR3 could directly interact with SREC-I in the presence of Poly I:C. The internalized ligand, along with TLR3 and SREC-I localized in endosomes within macrophages and in HEK293 cells engineered to express TLR3 and SREC-I. SREC-I also stimulated dsRNA-mediated TLR3 activation of signaling through the NFκβ, MAP kinase and interferon regulatory factor 3 (IRF3) pathways leading to expression of cytokines, most notably interleukin-8 and interferon-β. We therefore hypothesized that SREC-I could be a receptor capable of internalizing Poly I:C, boosting TLR3 mediated inflammatory signaling and stimulating cytokine production in macrophages.
Collapse
Affiliation(s)
- Ayesha Murshid
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | | | - Ridwan Ahmad
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Thiago J Borges
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; School of Biosciences and Biomedical Research Institute, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Stuart K Calderwood
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States.
| |
Collapse
|
28
|
SR-A and SREC-I binding peptides increase HDAd-mediated liver transduction. Gene Ther 2014; 21:950-7. [PMID: 25119377 PMCID: PMC4224584 DOI: 10.1038/gt.2014.71] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 06/05/2014] [Accepted: 07/09/2014] [Indexed: 12/19/2022]
Abstract
Helper-dependent adenoviral (HDAd) vectors can mediate long-term, high-level transgene expression from transduced hepatocytes without inducing chronic toxicity. However, vector therapeutic index is narrow because of a toxic acute response with potentially lethal consequences elicited by high vector doses. Kupffer cells and liver sinusoidal endothelial cells (LSECs) are major barriers to efficient hepatocyte transduction. We investigated two small peptides (PP1 and PP2) developed by phage display to block scavenger receptor type A (SR-A) and scavenger receptor expressed on endothelial cells type I (SREC-I) respectively, for enhancement of HDAd-mediated hepatocyte transduction efficiency. Pre-incubation of J774A.1 macrophages with either PP1 or PP2 prior to HDAd infection significantly reduced viral vector uptake. In vivo, fluorochrome-conjugated PP1 and PP2 injected intravenously into mice co-localized with both CD68 and CD31 on Kupffer cells and LSECs, respectively. Compared to saline pre-treated animals, intravenous injections of both peptides prior to the injection of an HDAd resulted in up to 3.7- and 2.9-fold increase of hepatic transgene expression with PP1 and PP2, respectively. In addition to hepatocyte transduction, compared to control saline injected mice, pre-treatment with either peptide resulted in no increased levels of serum interleukin-6 (IL-6), the major marker of adenoviral vector acute toxicity. In summary, we developed small peptides that significantly increase hepatocyte transduction efficacy and improve HDAd therapeutic index with potential for clinical applications.
Collapse
|
29
|
A nasal epithelial receptor for Staphylococcus aureus WTA governs adhesion to epithelial cells and modulates nasal colonization. PLoS Pathog 2014; 10:e1004089. [PMID: 24788600 PMCID: PMC4006915 DOI: 10.1371/journal.ppat.1004089] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 03/10/2014] [Indexed: 02/02/2023] Open
Abstract
Nasal colonization is a major risk factor for S. aureus infections. The mechanisms responsible for colonization are still not well understood and involve several factors on the host and the bacterial side. One key factor is the cell wall teichoic acid (WTA) of S. aureus, which governs direct interactions with nasal epithelial surfaces. We report here the first receptor for the cell wall glycopolymer WTA on nasal epithelial cells. In several assay systems this type F-scavenger receptor, termed SREC-I, bound WTA in a charge dependent manner and mediated adhesion to nasal epithelial cells in vitro. The impact of WTA and SREC-I interaction on epithelial adhesion was especially pronounced under shear stress, which resembles the conditions found in the nasal cavity. Most importantly, we demonstrate here a key role of the WTA-receptor interaction in a cotton rat model of nasal colonization. When we inhibited WTA mediated adhesion with a SREC-I antibody, nasal colonization in the animal model was strongly reduced at the early onset of colonization. More importantly, colonization stayed low over an extended period of 6 days. Therefore we propose targeting of this glycopolymer-receptor interaction as a novel strategy to prevent or control S. aureus nasal colonization. About 20% of the human population is colonized by Staphylococcus aureus. The reservoir of S. aureus is mainly the human nose. Usually, colonization does not lead to infection and is therefore without symptoms. However, when hospitalized patients exhibit a suppressed immune system, they are at risk of getting infected by their own nasal S. aureus strain. Therefore, it is important to understand the events and mechanisms underlying colonization. Until now S. aureus nasal colonization is only partially understood. One bacterial key factor is a sugar polymer of S. aureus, termed cell wall teichoic acid (WTA), which is involved in S. aureus adhesion to cellular surfaces in the inner part of the nasal cavity. We show here that a receptor-protein, which is expressed on such cells, binds WTA and is thereby involved in adhesion of S. aureus to nasal cells. This mechanism has a strong impact on nasal colonization in an animal model that resembles the situation in the human nose. Most importantly, inhibition of WTA mediated adhesion strongly reduces nasal colonization in the animal model. Therefore we propose that targeting of this glycopolymer-receptor interaction could serve as a novel strategy to control S. aureus nasal colonization.
Collapse
|
30
|
McNulty S, Colaco CA, Blandford LE, Bailey CR, Baschieri S, Todryk S. Heat-shock proteins as dendritic cell-targeting vaccines--getting warmer. Immunology 2013; 139:407-15. [PMID: 23551234 PMCID: PMC3719058 DOI: 10.1111/imm.12104] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 02/08/2013] [Accepted: 02/15/2013] [Indexed: 12/22/2022] Open
Abstract
Heat-shock proteins (hsp) provide a natural link between innate and adaptive immune responses by combining the ideal properties of antigen carriage (chaperoning), targeting and activation of antigen-presenting cells (APC), including dendritic cells (DC). Targeting is achieved through binding of hsp to distinct cell surface receptors and is followed by antigen internalization, processing and presentation. An improved understanding of the interaction of hsp with DC has driven the development of numerous hsp-containing vaccines, designed to deliver antigens directly to DC. Studies in mice have shown that for cancers, such vaccines generate impressive immune responses and protection from tumour challenge. However, translation to human use, as for many experimental immunotherapies, has been slow partly because of the need to perform trials in patients with advanced cancers, where demonstration of efficacy is challenging. Recently, the properties of hsp have been used for development of prophylactic vaccines against infectious diseases including tuberculosis and meningitis. These hsp-based vaccines, in the form of pathogen-derived hsp-antigen complexes, or recombinant hsp combined with selected antigens in vitro, offer an innovative approach against challenging diseases where broad antigen coverage is critical.
Collapse
Affiliation(s)
- Shaun McNulty
- ImmunoBiology Ltd., Babraham Research Campus, Babraham, Cambridge, UK.
| | | | | | | | | | | |
Collapse
|
31
|
The scavenger receptor SCARF1 mediates the clearance of apoptotic cells and prevents autoimmunity. Nat Immunol 2013; 14:917-26. [PMID: 23892722 PMCID: PMC3752698 DOI: 10.1038/ni.2670] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 06/21/2013] [Indexed: 12/14/2022]
Abstract
Clearance of apoptotic cells is critical for control of tissue homeostasis however the full range of receptor(s) on phagocytes responsible for recognition of apoptotic cells remains to be identified. Here we show that dendritic cells (DCs), macrophages and endothelial cells use scavenger receptor type F family member 1 (SCARF1) to recognize and engulf apoptotic cells via C1q. Loss of SCARF1 impairs uptake of apoptotic cells. Consequently, in SCARF1-deficient mice, dying cells accumulate in tissues leading to a lupus-like disease with the spontaneous generation of autoantibodies to DNA-containing antigens, immune cell activation, dermatitis and nephritis. The discovery of SCARF1 interactions with C1q and apoptotic cells provides insights into molecular mechanisms involved in maintenance of tolerance and prevention of autoimmune disease.
Collapse
|
32
|
Piccolo P, Vetrini F, Mithbaokar P, Grove NC, Bertin T, Palmer D, Ng P, Brunetti-Pierri N. SR-A and SREC-I are Kupffer and endothelial cell receptors for helper-dependent adenoviral vectors. Mol Ther 2013; 21:767-74. [PMID: 23358188 DOI: 10.1038/mt.2012.287] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Helper-dependent adenoviral (HDAd) vectors can mediate long-term, high-level transgene expression from transduced hepatocytes with no chronic toxicity. However, a toxic acute response with potentially lethal consequences has hindered their clinical applications. Liver sinusoidal endothelial cells (LSECs) and Kupffer cells are major barriers to efficient hepatocyte transduction. Understanding the mechanisms of adenoviral vector uptake by non-parenchymal cells may allow the development of strategies aimed at overcoming these important barriers and to achieve preferential hepatocyte gene transfer with reduced toxicity. Scavenger receptors on Kupffer cells bind adenoviral particles and remove them from the circulation, thus preventing hepatocyte transduction. In the present study, we show that HDAd particles interact in vitro and in vivo with scavenger receptor-A (SR-A) and with scavenger receptor expressed on endothelial cells-I (SREC-I) and we exploited this knowledge to increase the efficiency of hepatocyte transduction by HDAd vectors in vivo through blocking of SR-A and SREC-I with specific fragments antigen-binding (Fabs).
Collapse
|
33
|
Singh RP, Das M, Thakare V, Jain S. Functionalization density dependent toxicity of oxidized multiwalled carbon nanotubes in a murine macrophage cell line. Chem Res Toxicol 2012; 25:2127-37. [PMID: 22994501 DOI: 10.1021/tx300228d] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The present study investigates the effect of functionalization density on the toxicity and cellular uptake of oxidized multiwalled carbon nanotubes (f-MWCNTs) in vitro. The toxicity of f-MWCNTs at varying degrees of carboxylation was assessed in a murine macrophage RAW 264.7 cell line, a model for liver Kupffer cells. In vitro cytotoxicity of oxidized MWCNTs was directly proportional to their functionalization density. The increased cytotoxicity was associated with a concurrent increase in the number of apoptotic cells and production of reactive nitrogen species (RNS). In contrast, reactive oxygen species (ROS) generation was the highest in the case of pristine MWCNTs and decreased with increased functionalization density. Quantitative cellular uptake studies indicated that endogenous ROS production was independent of the concentration of CNTs internalized by a specific cell population and was directly proportional to their surface hydrophobicity. Mechanistic studies suggested that cellular uptake of CNTs was critically charge-dependent and mediated through scavenger receptors, albeit the involvement of nonscavenger receptor mechanisms at low CNT concentrations and their saturation at the experimental concentration cannot be ruled out. A mathematical model was established to correlate between the cellular uptake of CNTs with their length and zeta potential. In an attempt to correlate the results of in vitro toxicity experiments with those of the in vivo toxicity in the mouse model, we found that the toxicity trends in vitro and in vivo are rather opposing. The apparent anomaly was explained on the basis of different experimental conditions and doses associated with cells under in vivo and in vitro culture conditions.
Collapse
Affiliation(s)
- Raman Preet Singh
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-NIPER, SAS Nagar-Mohali Punjab, India 160062
| | | | | | | |
Collapse
|
34
|
Quarck R, Wynants M, Ronisz A, Sepulveda MR, Wuytack F, Van Raemdonck D, Meyns B, Delcroix M. Characterization of proximal pulmonary arterial cells from chronic thromboembolic pulmonary hypertension patients. Respir Res 2012; 13:27. [PMID: 22452949 PMCID: PMC3352254 DOI: 10.1186/1465-9921-13-27] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 03/27/2012] [Indexed: 12/02/2022] Open
Abstract
Background Chronic thromboembolic pulmonary hypertension (CTEPH) is associated with proximal pulmonary artery obstruction and vascular remodeling. We hypothesized that pulmonary arterial smooth muscle (PASMC) and endothelial cells (PAEC) may actively contribute to remodeling of the proximal pulmonary vascular wall in CTEPH. Our present objective was to characterize PASMC and PAEC from large arteries of CTEPH patients and investigate their potential involvement in vascular remodeling. Methods Primary cultures of proximal PAEC and PASMC from patients with CTEPH, with non-thromboembolic pulmonary hypertension (PH) and lung donors have been established. PAEC and PASMC have been characterized by immunofluorescence using specific markers. Expression of smooth muscle specific markers within the pulmonary vascular wall has been studied by immunofluorescence and Western blotting. Mitogenic activity and migratory capacity of PASMC and PAEC have been investigated in vitro. Results PAEC express CD31 on their surface, von Willebrand factor in Weibel-Palade bodies and take up acetylated LDL. PASMC express various differentiation markers including α-smooth muscle actin (α-SMA), desmin and smooth muscle myosin heavy chain (SMMHC). In vascular tissue from CTEPH and non-thromboembolic PH patients, expression of α-SMA and desmin is down-regulated compared to lung donors; desmin expression is also down-regulated in vascular tissue from CTEPH compared to non-thromboembolic PH patients. A low proportion of α-SMA positive cells express desmin and SMMHC in the neointima of proximal pulmonary arteries from CTEPH patients. Serum-induced mitogenic activity of PAEC and PASMC, as well as migratory capacity of PASMC, were increased in CTEPH only. Conclusions Modified proliferative and/or migratory responses of PASMC and PAEC in vitro, associated to a proliferative phenotype of PASMC suggest that PASMC and PAEC could contribute to proximal vascular remodeling in CTEPH.
Collapse
Affiliation(s)
- Rozenn Quarck
- Center for Pulmonary Vascular Diseases, Respiratory Disease Department, Katholieke Universiteit and Universitaire Ziekenhuizen Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Sano M, Korekane H, Ohtsubo K, Yamaguchi Y, Kato M, Shibukawa Y, Tajiri M, Adachi H, Wada Y, Asahi M, Taniguchi N. N-glycans of SREC-I (scavenger receptor expressed by endothelial cells): essential role for ligand binding, trafficking and stability. Glycobiology 2012; 22:714-24. [PMID: 22279061 DOI: 10.1093/glycob/cws010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Scavenger receptor expressed by endothelial cells (SREC-I) mediates the endocytosis of chemically modified lipoproteins such as acetylated low-density lipoprotein (Ac-LDL) and oxidized LDL and is implicated in atherogenesis. We produced recombinant SREC-I in Chinese hamster ovary-K1 cells and identified three potential glycosylation sites, Asn(289), Asn(382) and Asn(393), which were all glycosylated. To determine the function of N-glycans in SREC-I, we characterized SREC-I mutant proteins by intracellular distribution and the cellular incorporation rate of Ac-LDL. N382Q/N393Q and N289Q/N382Q/N393Q were sequestered in the endoplasmic reticulum, resulting in a severe reduction in the cellular incorporation of Ac-LDL. N382Q showed a normal cell surface residency and an enhanced affinity for Ac-LDL, resulting in an elevated Ac-LDL cellular incorporation. These results indicate that the N-glycan of Asn(393) regulates the intracellular sorting of SREC-I and that the N-glycan of Asn(382) controls ligand-binding affinity. Furthermore, we detected an enhanced trypsin sensitivity of the N289Q. Glycan structure analyses revealed that the core-fucosylated bi-antennary is the common major structure at all glycosylation sites. In addition, tri- and tetra-antennary were detected as minor constituents at Asn(289). A bisecting GlcNAc was also detected at Asn(382) and Asn(393). Structural analyses and homology modeling of SREC-I suggest that the N-glycan bearing a β1-6GlcNAc branch at Asn(289) protects from proteinase attack and thus confers a higher stability on SREC-I. These data indicate that Asn(289)-, Asn(382)- and Asn(393)-linked N-glycans of SREC-I have distinct functions in regulating proteolytic resistance, ligand-binding affinity and subcellular localization, all of which might be involved in the development of atherogenesis.
Collapse
Affiliation(s)
- Masahiro Sano
- Department of Disease Glycomics, Institute of Scientific and Industrial Research, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka 567-0047, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Chaemchuen S, Rungpragayphan S, Poovorawan Y, Patarakul K. Identification of candidate host proteins that interact with LipL32, the major outer membrane protein of pathogenic Leptospira, by random phage display peptide library. Vet Microbiol 2011; 153:178-185. [PMID: 21592685 DOI: 10.1016/j.vetmic.2011.04.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 04/05/2011] [Accepted: 04/18/2011] [Indexed: 11/23/2022]
Abstract
Leptospirosis is a worldwide zoonotic disease caused by pathogenic Leptospira spp. Rodent species are the major reservoir hosts that can excrete leptospires in their urine leading to environmental contamination. After gaining entry into the host via skin breaks and mucosa, leptospires disseminate through the bloodstream to target organs causing a wide range of disease manifestations in susceptible mammalian hosts. The crucial step of infection requires host-pathogen interactions. LipL32, the major outer membrane protein (OMP) of pathogenic Leptospira, is conserved among pathogenic leptospires, immunogenic, and expressed in target organs during acute infection in animal models. Therefore, it may play a key role in host-microbe interactions. To identify host proteins that interact with LipL32, phage display technology was employed in our study. Recombinant LipL32 was used as a target molecule for biopanning with a random heptapeptide phage library to enrich for phages expressing peptides with high affinity to LipL32. After three rounds of panning, 44 plaques of eluted phages were subjected to pyrosequencing. Six different peptide sequences were identified and used to search for matching proteins in the database. Putative proteins with potential binding to LipL32 are proteins known to be expressed on the surface of target cells of pathogenic Leptospira such as chloride channel accessory 2, glycoprotein VI, scavenger receptor expressed by endothelial cell isoform I (SREC-I), coronin 2A, laminin alpha 5, collagen XX, and prostaglandin receptor EP1. However, interactions of LipL32 with these host proteins and their role in the pathogenesis of leptospirosis requires experimental confirmation.
Collapse
Affiliation(s)
- Suwittra Chaemchuen
- Medical Microbiology, Interdisciplinary Program, Graduate School, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | | | | | | |
Collapse
|
37
|
Means TK. Fungal pathogen recognition by scavenger receptors in nematodes and mammals. Virulence 2011; 1:37-41. [PMID: 21178411 DOI: 10.4161/viru.1.1.10228] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Macrophages are important cells in the host resistance to fungal infections, and fungal recognition by macrophages triggers phagocytosis, intracellular killing, induction of inflammatory cytokines and chemokines, and initiation of the adaptive immune response. All of the receptors that mediate binding and engulfment of fungal pathogens and the signaling pathways triggered by fungal pathogens that regulate anti-fungal immunity are not fully understood. Using an RNAi screen we recently demonstrated that the C. elegans receptors CED-1 and C03F11.3, and their mammalian orthologues, the scavenger receptors SCARF1 and CD36 mediate host defense against the fungal pathogen, Cryptococcus neoformans. Finally, SCARF1 and CD36 function as co-receptors by binding and engulfing fungal pathogens to facilitate Toll-like receptor 2 signaling. Here we will summarize and expand upon our previous findings.
Collapse
Affiliation(s)
- Terry K Means
- Center for Immunology and Inflammatory Diseases and Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
38
|
The zymogen granule protein 2 (GP2) binds to scavenger receptor expressed on endothelial cells I (SREC-I). Cell Immunol 2010; 267:88-93. [PMID: 21190681 PMCID: PMC3040788 DOI: 10.1016/j.cellimm.2010.12.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 11/29/2010] [Accepted: 12/02/2010] [Indexed: 12/11/2022]
Abstract
The pancreatic zymogen granule membrane protein (GP2) is expressed by pancreatic acinar cells and M cells of the ileum. GP2 is the closest related homologue of the urine resident Tamm-Horsfall protein (THP). Recently, it was shown that THP is a ligand of various scavenger receptors (SRs). Therefore, we were interested, if GP2 has similar properties. cDNA of different SRs was stably transfected into a murine thymoma cell line. GP2 was recombinantly expressed, purified and biotinylated. Binding or uptake of GP2 by transfected cells or monocyte-derived dendritic cells (moDCs) was analyzed by flow-cytometry. GP2 is a binding partner of the scavenger receptor expressed on endothelial cells I (SREC-I) but not of SR-AI and SR-BI. The dissociation constant (K(d)) of GP2 binding to SREC-I is 41.3nM. SREC transfected cells are able to internalize GP2. moDCs express SREC-I and also bind and internalize GP2. Inhibition of SREC-I on moDCs with anti-SREC-I antibodies does not result in a decreased GP2 binding. Interaction of GP2 with SREC-I and uptake might have profound effects in antigen clearance and mediation of the immune response. In addition to SREC-I other presently unknown receptors for GP2 on DCs might be involved in this process.
Collapse
|
39
|
Stephen SL, Freestone K, Dunn S, Twigg MW, Homer-Vanniasinkam S, Walker JH, Wheatcroft SB, Ponnambalam S. Scavenger receptors and their potential as therapeutic targets in the treatment of cardiovascular disease. Int J Hypertens 2010; 2010:646929. [PMID: 20981357 PMCID: PMC2958427 DOI: 10.4061/2010/646929] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Accepted: 07/07/2010] [Indexed: 12/12/2022] Open
Abstract
Scavenger receptors act as membrane-bound and soluble proteins that bind to macromolecular complexes and pathogens. This diverse supergroup of proteins mediates binding to modified lipoprotein particles which regulate the initiation and progression of atherosclerotic plaques. In vascular tissues, scavenger receptors are implicated in regulating intracellular signaling, lipid accumulation, foam cell development, and cellular apoptosis or necrosis linked to the pathophysiology of atherosclerosis. One approach is using gene therapy to modulate scavenger receptor function in atherosclerosis. Ectopic expression of membrane-bound scavenger receptors using viral vectors can modify lipid profiles and reduce the incidence of atherosclerosis. Alternatively, expression of soluble scavenger receptors can also block plaque initiation and progression. Inhibition of scavenger receptor expression using a combined gene therapy and RNA interference strategy also holds promise for long-term therapy. Here we review our current understanding of the gene delivery by viral vectors to cells and tissues in gene therapy strategies and its application to the modulation of scavenger receptor function in atherosclerosis.
Collapse
Affiliation(s)
- Sam L Stephen
- Endothelial Cell Biology Unit, Institute of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Cheng J, Wu Y, Mohler JL, Ip C. The transcriptomics of de novo androgen biosynthesis in prostate cancer cells following androgen reduction. Cancer Biol Ther 2010; 9:1033-42. [PMID: 20404538 DOI: 10.4161/cbt.9.12.11876] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The progression of prostate cancer to the castration-recurrent phenotype remains a major problem medically. The present study examined the transcriptomics of de novo androgen synthesis as a potential mechanism to escape from dependence on circulating androgen. VCaP, LNCaP and LAPC4 cells were acclimated to 1 nM testosterone for five generations before subjecting them to a reduced level of 0.03 nM testosterone. Changes in gene expression were quantified using qRT-PCR. Analyses of the cholesterol biosynthesis pathway and the Δ4, Δ5 and backdoor steroidogenic pathways were carried out. VCaP cells showed no change in the transcriptome of cholesterol biosynthesis. However, several receptors for cholesterol transport were upregulated. The Δ4 and Δ5 steroidogenic pathways, but not the backdoor pathway, were stimulated. Additionally, androgen receptor (AR) expression was increased. Taken together, the above changes might allow recovery of AR activity to a near normal level. In contrast, LNCaP cells showed only minimal adjustment in the transcriptome of steroidogenesis. LAPC4 cells were equally unresponsive to boosting the machinery of androgen production. In brief, our results suggest that the VCaP model is an appropriate model for further investigation of targeting the androgen-AR axis to block the emergence of castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Jinrong Cheng
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | | | | |
Collapse
|
41
|
Abstract
BACKGROUND Cells from the bone marrow contribute to ischemic neovascularization, but the identity of these cells remains unclear. The authors identify mesenchymal stem cells as a bone marrow-derived progenitor population that is able to engraft into peripheral tissue in response to ischemia. METHODS A murine model of skin ischemia was used. Bone marrow, blood, and skin were harvested at different time points and subjected to flow cytometric analysis for mesenchymal and hematopoietic markers (n = 3 to 7 per time point). Using a parabiotic model pairing donor green fluorescent protein (GFP)-positive with recipient wild-type mice, progenitor cell engraftment was examined in ischemic tissue by fluorescence microscopy, and engrafted cells were analyzed by flow cytometry for endothelial and mesenchymal markers. In vitro, the ability of both bone marrow- and adipose-derived mesenchymal stem cells to adopt endothelial characteristics was examined by analyzing (1) the ability of mesenchymal stem cells to take up DiI-acetylated low-density lipoprotein and Alexa Fluor lectin, and (2) phenotypic changes of mesenchymal stem cells co-cultured with GFP-labeled endothelial cells or under hypoxic/vascular endothelial growth factor stimulation. RESULTS In vivo, the bone marrow mesenchymal stem cell population decreased significantly immediately after surgery, with subsequent engraftment of these cells in ischemic tissue. Engrafted cells lacked the panhematopoietic antigen CD45, consistent with a mesenchymal origin. In vitro, bone marrow- and adipose-derived mesenchymal stem cells took up DiI-acetylated low-density lipoprotein and Alexa Fluor lectin, and expressed endothelial markers under hypoxic conditions. CONCLUSIONS The authors' data suggest that mesenchymal precursor cells can give rise to endothelial progenitors. Consequently, cell-based therapies augmenting the mesenchymal stem cell population could represent powerful alternatives to current therapies for ischemic vascular disease.
Collapse
|
42
|
Means TK, Mylonakis E, Tampakakis E, Colvin RA, Seung E, Puckett L, Tai MF, Stewart CR, Pukkila-Worley R, Hickman SE, Moore KJ, Calderwood SB, Hacohen N, Luster AD, El Khoury J. Evolutionarily conserved recognition and innate immunity to fungal pathogens by the scavenger receptors SCARF1 and CD36. ACTA ACUST UNITED AC 2009; 206:637-53. [PMID: 19237602 PMCID: PMC2699123 DOI: 10.1084/jem.20082109] [Citation(s) in RCA: 182] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Receptors involved in innate immunity to fungal pathogens have not been fully elucidated. We show that the Caenorhabditis elegans receptors CED-1 and C03F11.3, and their mammalian orthologues, the scavenger receptors SCARF1 and CD36, mediate host defense against two prototypic fungal pathogens, Cryptococcus neoformans and Candida albicans. CED-1 and C03F11.1 mediated antimicrobial peptide production and were necessary for nematode survival after C. neoformans infection. SCARF1 and CD36 mediated cytokine production and were required for macrophage binding to C. neoformans, and control of the infection in mice. Binding of these pathogens to SCARF1 and CD36 was β-glucan dependent. Thus, CED-1/SCARF1 and C03F11.3/CD36 are β-glucan binding receptors and define an evolutionarily conserved pathway for the innate sensing of fungal pathogens.
Collapse
Affiliation(s)
- Terry K Means
- Center for Immunology and Inflammatory Diseases and Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Shibata N, Glass CK. Regulation of macrophage function in inflammation and atherosclerosis. J Lipid Res 2008; 50 Suppl:S277-81. [PMID: 18987388 DOI: 10.1194/jlr.r800063-jlr200] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis can be considered as both a chronic inflammatory disease and a lipid metabolism disorder. Innate immunity pathways have long been suspected to contribute to the initiation and progression of atherosclerosis. This suggests that crosstalk between lipid metabolism and innate immunity pathways plays an important role for the development and/or the prevention of atherosclerosis. However, it is not fully defined how innate immunity affects lipid metabolism. Macrophages play a central role in atherogenesis through the accumulation of cholesterol and the production of inflammatory mediators and cytokines. Liver X receptors (LXRs) exert an important atheroprotective effect in the macrophage. In addition to regulating cholesterol metabolism, LXRs are also negative regulators of macrophage inflammatory gene responses. In this review, we will discuss the roles of LXRs in the macrophage as key factors that link innate immunity and lipid metabolism.
Collapse
Affiliation(s)
- Norihito Shibata
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0651, USA
| | | |
Collapse
|
44
|
Conjugated linoleic acid isomers reduce cholesterol accumulation in acetylated LDL-induced mouse RAW264.7 macrophage-derived foam cells. Lipids 2008; 43:913-23. [PMID: 18769950 DOI: 10.1007/s11745-008-3226-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Accepted: 08/13/2008] [Indexed: 10/21/2022]
Abstract
Synthetic activators of peroxisome proliferator-activated receptors (PPAR)-alpha and -gamma are capable of reducing macrophage foam cell cholesterol accumulation through the activation of genes involved in cholesterol homeostasis. Since conjugated linoleic acids (CLA) were also demonstrated to activate PPARalpha and PPARgamma in vivo and in vitro, we tested the hypothesis that CLA are also capable of reducing macrophage foam cell cholesterol accumulation. Thus, mouse RAW264.7 macrophage-derived foam cells were treated with CLA isomers, c9t11-CLA and t10c12-CLA, and linoleic acid (LA), as reference fatty acid, and analyzed for the concentrations of free and esterified cholesterol, cholesterol efflux and expression of genes involved in cholesterol homeostasis (CD36, ABCA1, LXRalpha, NPC-1, and NPC-2). Treatment with c9t11-CLA and t10c12-CLA, but not LA, lowered cholesterol accumulation, stimulated acceptor-dependent cholesterol efflux, and increased relative mRNA concentrations of CD36, ABCA1, LXRalpha, NPC-1, and NPC-2 (P < 0.05). In conclusion, the present study showed that CLA isomers reduce cholesterol accumulation in RAW264.7 macrophage-derived foam cells presumably by enhancing lipid acceptor-dependent cholesterol efflux.
Collapse
|
45
|
Macrophage scavenger receptors and host-derived ligands. Methods 2008; 43:207-17. [PMID: 17920517 DOI: 10.1016/j.ymeth.2007.06.004] [Citation(s) in RCA: 223] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Accepted: 06/25/2007] [Indexed: 02/07/2023] Open
Abstract
The scavenger receptors are a large family of molecules that are structurally diverse and have been implicated in a range of functions. They are expressed by myeloid cells, selected endothelial cells and some epithelial cells and recognise many different ligands, including microbial pathogens as well as endogenous and modified host-derived molecules. This review will focus on the eight classes of scavenger receptors (class A-H) in terms of their structure, expression and recognition of host-derived ligands. Scavenger receptors have been implicated in a range of physiological and pathological processes, such as atherosclerosis and Alzheimer's disease, and function in adhesion and tissue maintenance. More recently, some of the scavenger receptors have been shown to mediate binding and endocytosis of chaperone proteins, such as the heat shock proteins, thereby playing an important role in antigen cross-presentation.
Collapse
|
46
|
Yasuda T, Hirata KI, Ishida T, Kojima Y, Tanaka H, Okada T, Quertermous T, Yokoyama M. Endothelial lipase is increased by inflammation and promotes LDL uptake in macrophages. J Atheroscler Thromb 2007; 14:192-201. [PMID: 17726294 DOI: 10.5551/jat.e502] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Endothelial lipase (EL) is a member of the lipoprotein lipase family that regulates HDL metabolism. EL is known to act as a bridging molecule for monocytes or lipoproteins in vascular endothelial cells. We investigated the role and regulatory mechanisms of EL expression in macrophages. METHODS Macrophages originating from wild-type (EL+/+) and EL-deficient (EL-/-) mice were stimulated with lipopolysaccharide (LPS). The expression of EL mRNA was evaluated by northern blotting. DiI-LDL was used to measure the uptake of native low-density lipoprotein (nLDL). RESULTS LPS increased EL mRNA levels by increasing intracellular oxidative stress in the macrophages. LPS did not affect EL expression in macrophages derived from Toll-like receptor 4 (TLR4) gene mutant mice, C3H/HeJ. The uptake of nLDL after LPS-treatment was significantly lower in macrophages from EL-/- mice than those from EL+/+ mice. Simvastatin suppressed the LPS-induced upregulation of EL expression and uptake of nLDL. CONCLUSIONS EL expression is upregulated by LPS via TLR4 and promotes the uptake of nLDL by macrophages. Simvastatin inhibits the LPS-induced up-regulation and uptake in macrophages. Thus, our findings provide a novel role for EL in lipoprotein metabolism and would expand the range of anti-atherogenic effects of statins.
Collapse
Affiliation(s)
- Tomoyuki Yasuda
- Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Gottfried E, Kreutz M, Haffner S, Holler E, Iacobelli M, Andreesen R, Eissner G. Differentiation of human tumour-associated dendritic cells into endothelial-like cells: an alternative pathway of tumour angiogenesis. Scand J Immunol 2007; 65:329-35. [PMID: 17386023 DOI: 10.1111/j.1365-3083.2007.01903.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Until recently, the only accepted mechanism of tumour vascularization was the sprouting of endothelial cells (EC) from pre-existing vessels, while recent studies suggest the contribution of stem cell-derived endothelial progenitors as well as cells from the myeloid lineage. Here, we show a new way of endothelial differentiation that involves the specific modulation of monocytes by the tumour environment. The tumour milieu is characterized by the presence of cytokines and lactate which induce the differentiation of tumour-invading monocytes into tumour-associated dendritic cells (DC). Additional incubation of tumour-associated DC with pro-angiogenic factors, such as vascular endothelial growth factor and oncostatin M, led to transdifferentiation into endothelial-like cells. The cells showed strong expression of von Willebrand factor and VE-Cadherin, both classical EC markers, while leukocytic markers were reduced. In addition, they were able to form network-like structures on matrigel, which could be blocked by the DNA-based drug Defibrotide. This finding may be of great therapeutic relevance for tumour therapy.
Collapse
Affiliation(s)
- E Gottfried
- Department of Hematology and Oncology, Regensburg University Medical Center, Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
48
|
Liu X, Ukai T, Yumoto H, Davey M, Goswami S, Gibson FC, Genco CA. Toll-like receptor 2 plays a critical role in the progression of atherosclerosis that is independent of dietary lipids. Atherosclerosis 2007; 196:146-154. [PMID: 17466307 PMCID: PMC2243224 DOI: 10.1016/j.atherosclerosis.2007.03.025] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 03/05/2007] [Accepted: 03/13/2007] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Toll-like receptors (TLRs), a group of pathogen-associated microbial pattern recognition receptors, play an important role in innate immune signaling and are differentially regulated in chronic inflammatory diseases such as atherosclerosis. However, the involvement of TLRs in the progression of atherosclerosis is still unclear. METHODS AND RESULTS TLR2 and apolipoprotein E double knockout (Tlr2(-/-)Apoe(-/-)) mice were generated and the progressive formation of atherosclerotic plaque in the aortas was examined in mice fed a normal chow diet. We demonstrate that inactivation of TLR2 resulted in reduced progression of atherosclerosis in both male and female Apoe(-/-) mice. Likewise, TLR2 deficiency resulted in a reduction in lipid accumulation and decreased macrophage recruitment to the aortic sinus, as well as reduced monocyte chemoattractant protein-1 (MCP-1) levels. Furthermore, macrophages isolated from Tlr2(-/-)Apoe(-/-) mice demonstrated significantly reduced MCP-1 production upon stimulation with a TLR2 ligand. However, no differences in acetylated low-density lipoprotein uptake and foam cell formation were observed in macrophages isolated from Tlr2(-/-)Apoe(-/-) mice as compared to Apoe(-/-) mice. CONCLUSIONS TLR2 plays a critical role in the progression of atherosclerosis in Apoe(-/-) mice, which is independent of dietary lipids and macrophage lipid uptake.
Collapse
Affiliation(s)
- Xinyan Liu
- Department of Medicine, Section of Molecular Medicine, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, United States
| | - Takashi Ukai
- Department of Medicine, Section of Molecular Medicine, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, United States; Department of Periodontology, Unit of Translational Medicine, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Hiromichi Yumoto
- Department of Medicine, Section of Molecular Medicine, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, United States; Department of Conservative Dentistry, The Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8504, Japan
| | - Michael Davey
- Department of Periodontology and Oral Biology, Goldman School of Dental Medicine, Boston University, Boston, MA 02118, United States
| | - Sulip Goswami
- Department of Medicine, Section of Molecular Medicine, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, United States
| | - Frank C Gibson
- Department of Medicine, Section of Molecular Medicine, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, United States
| | - Caroline A Genco
- Department of Medicine, Section of Molecular Medicine, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, United States; Department of Periodontology and Oral Biology, Goldman School of Dental Medicine, Boston University, Boston, MA 02118, United States; Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, United States.
| |
Collapse
|
49
|
Stepanyan Z, Hughes B, Cliche DO, Camp D, Hekimi S. Genetic and molecular characterization of CLK-1/mCLK1, a conserved determinant of the rate of aging. Exp Gerontol 2006; 41:940-51. [PMID: 16889924 DOI: 10.1016/j.exger.2006.06.041] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2006] [Revised: 05/29/2006] [Accepted: 06/08/2006] [Indexed: 11/29/2022]
Abstract
The clk-1 gene of the nematode Caenorhabditis elegans encodes an evolutionarily conserved enzyme that is necessary for ubiquinone biosynthesis. Loss-of-function mutations in clk-1, as well as in its mouse orthologue mclk1, increase lifespan in both organisms. In nematodes, clk-1 extends lifespan by a mechanism that is distinct from the insulin signaling-like pathway but might have similarities to calorie restriction. The evolutionary conservation of the effect of clk-1/mclk1 on lifespan suggests that the gene affects a fundamental mechanism of aging. The clk-1/mclk1 system could allow for the understanding of this mechanism by combining genetic and molecular investigations in worms with studies in mice, where age-dependent disease processes relevant to human health can be modeled.
Collapse
Affiliation(s)
- Zaruhi Stepanyan
- Department of Biology, McGill University, Montréal, Que., Canada
| | | | | | | | | |
Collapse
|
50
|
Murphy JE, Tedbury PR, Homer-Vanniasinkam S, Walker JH, Ponnambalam S. Biochemistry and cell biology of mammalian scavenger receptors. Atherosclerosis 2006; 182:1-15. [PMID: 15904923 DOI: 10.1016/j.atherosclerosis.2005.03.036] [Citation(s) in RCA: 248] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2004] [Revised: 03/10/2005] [Accepted: 03/24/2005] [Indexed: 01/14/2023]
Abstract
Scavenger receptors are integral membrane proteins that bind a wide variety of ligands including modified or oxidised low-density lipoproteins, apoptotic cells and pathogens. Modified low-density lipoprotein accumulation is thought to be an early event in vascular disease and thus scavenger receptor function is critical in this context. The scavenger receptor family has at least eight different subclasses (A-H) which bear little sequence homology to each other but recognize common ligands. Here we review our current understanding of the scavenger receptor subclasses with emphasis on their genetics, protein structure, biochemical properties, membrane trafficking, intracellular signalling and links to disease states. We also highlight emerging areas where scavenger receptors play roles in cell and animal physiology.
Collapse
Affiliation(s)
- Jane E Murphy
- School of Biochemistry and Microbiology, University of Leeds, Leeds LS2 9JT, UK
| | | | | | | | | |
Collapse
|