1
|
Gallerand A, Dolfi B, Stunault MI, Caillot Z, Castiglione A, Strazzulla A, Chen C, Heo GS, Luehmann H, Batoul F, Vaillant N, Dumont A, Pilot T, Merlin J, Zair FN, Gilleron J, Bertola A, Carmeliet P, Williams JW, Arguello RJ, Masson D, Dombrowicz D, Yvan-Charvet L, Doyen D, Haschemi A, Liu Y, Guinamard RR, Ivanov S. Glucose metabolism controls monocyte homeostasis and migration but has no impact on atherosclerosis development in mice. Nat Commun 2024; 15:9027. [PMID: 39424804 PMCID: PMC11489573 DOI: 10.1038/s41467-024-53267-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
Monocytes directly contribute to atherosclerosis development by their recruitment to plaques in which they differentiate into macrophages. In the present study, we ask how modulating monocyte glucose metabolism could affect their homeostasis and their impact on atherosclerosis. Here we investigate how circulating metabolites control monocyte behavior in blood, bone marrow and peripheral tissues of mice. We find that serum glucose concentrations correlate with monocyte numbers. In diet-restricted mice, monocytes fail to metabolically reprogram from glycolysis to fatty acid oxidation, leading to reduced monocyte numbers in the blood. Mechanistically, Glut1-dependent glucose metabolism helps maintain CD115 membrane expression on monocytes and their progenitors, and regulates monocyte migratory capacity by modulating CCR2 expression. Results from genetic models and pharmacological inhibitors further depict the relative contribution of different metabolic pathways to the regulation of CD115 and CCR2 expression. Meanwhile, Glut1 inhibition does not impact atherosclerotic plaque development in mouse models despite dramatically reducing blood monocyte numbers, potentially due to the remaining monocytes having increased migratory capacity. Together, these data emphasize the role of glucose uptake and intracellular glucose metabolism in controlling monocyte homeostasis and functions.
Collapse
Affiliation(s)
- Alexandre Gallerand
- Université Côte d'Azur, CNRS, LP2M, Nice, France.
- Université Côte d'Azur, INSERM, C3M, Nice, France.
| | - Bastien Dolfi
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | | | - Alexia Castiglione
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | - Chuqiao Chen
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Gyu Seong Heo
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Flora Batoul
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | | | - Thomas Pilot
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France
| | | | | | | | | | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, 3000, Belgium
| | - Jesse W Williams
- Center for Immunology, Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Rafael J Arguello
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - David Masson
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France
| | - David Dombrowicz
- Univ.Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | | | - Denis Doyen
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Médecine Intensive Réanimation, Hôpital Pasteur, CHU de Nice, Nice, France
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rodolphe R Guinamard
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | - Stoyan Ivanov
- Université Côte d'Azur, CNRS, LP2M, Nice, France.
- Université Côte d'Azur, INSERM, C3M, Nice, France.
| |
Collapse
|
2
|
Swirski FK, Binder CJ. Lower your cholesterol early, and stick with it! Nat Rev Cardiol 2024:10.1038/s41569-024-01095-x. [PMID: 39424909 DOI: 10.1038/s41569-024-01095-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Affiliation(s)
- Filip K Swirski
- Cardiovascular Research Institute, Friedman Brain Institute, Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Costa ADSD, Vadym K, Park K. Engineered endothelium model enables recapitulation of vascular function and early atherosclerosis development. Biomaterials 2024; 314:122889. [PMID: 39423515 DOI: 10.1016/j.biomaterials.2024.122889] [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: 04/30/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Human health relies heavily on the vascular endothelium. Here, we propose a novel engineered endothelium model (EEM), which recapitulated both normal vascular function and pathology. An artificial basement membrane (aBM), where porous polyvinyl alcohol hydrogel was securely integrated with human fibroblast-derived, decellularized extracellular matrix on both sides was fabricated first and followed by endothelial cells (ECs) and pericytes (PCs) adhesion, respectively. Our EEM formed robust adherens junction (VE-cad) and built an impermeable barrier with time, along with the nitric oxide (NO) secretion. In our EEM, ECs and PCs interacted each other via aBM and led to hemoglobin alpha 1 (Hb-α1) development, which was involved in NO control and was strongly interconnected with VE-cad as well. A resilient property of EEM under inflammatory milieu was also confirmed by VE-cad and barrier recovery with time. In particular interest, foam cells formation, a hallmark of atherosclerotic initiation was successfully recapitulated in our EEM, where a series of sequential events were confirmed: human monocytes adhesion, transendothelial migration, and oxidized low-density lipoprotein uptake by macrophages. Collectively, our EEM is excellent in recapitulating not only normal endothelium but early pathologic one, thereby enabling EEM to be a physiologically relevant model for vascular study and disease modeling.
Collapse
Affiliation(s)
| | - Kopych Vadym
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
4
|
Döring Y, Bender A, Soehnlein O. Lack of Formyl-peptide Receptor 1 Mitigates Atherosclerosis in Hyperlipidemic Mice. Thromb Haemost 2024; 124:986-989. [PMID: 38782024 DOI: 10.1055/s-0044-1787264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Affiliation(s)
- Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), Bern University Hospital, University of Bern, Bern, Switzerland
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Munich Heart Alliance Partner Site, Munich, Germany
| | - Alexander Bender
- Institute of Experimental Pathology (ExPat), Center of Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| | - Oliver Soehnlein
- Institute of Experimental Pathology (ExPat), Center of Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| |
Collapse
|
5
|
Smith TKT, Ghorbani P, LeBlond ND, Nunes JRC, O'Dwyer C, Ambursley N, Fong-McMaster C, Minarrieta L, Burkovsky LA, El-Hakim R, Trzaskalski NA, Locatelli CAA, Stotts C, Pember C, Rayner KJ, Kemp BE, Loh K, Harper ME, Mulvihill EE, St-Pierre J, Fullerton MD. AMPK-mediated regulation of endogenous cholesterol synthesis does not affect atherosclerosis in a murine Pcsk9-AAV model. Atherosclerosis 2024; 397:117608. [PMID: 38880706 DOI: 10.1016/j.atherosclerosis.2024.117608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 05/09/2024] [Accepted: 05/30/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND AND AIMS Dysregulated cholesterol metabolism is a hallmark of atherosclerotic cardiovascular diseases, yet our understanding of how endogenous cholesterol synthesis affects atherosclerosis is not clear. The energy sensor AMP-activated protein kinase (AMPK) phosphorylates and inhibits the rate-limiting enzyme in the mevalonate pathway HMG-CoA reductase (HMGCR). Recent work demonstrated that when AMPK-HMGCR signaling was compromised in an Apoe-/- model of hypercholesterolemia, atherosclerosis was exacerbated due to elevated hematopoietic stem and progenitor cell mobilization and myelopoiesis. We sought to validate the significance of the AMPK-HMGCR signaling axis in atherosclerosis using a non-germline hypercholesterolemia model with functional ApoE. METHODS Male and female HMGCR S871A knock-in (KI) mice and wild-type (WT) littermate controls were made atherosclerotic by intravenous injection of a gain-of-function Pcsk9D374Y-adeno-associated virus followed by high-fat and high-cholesterol atherogenic western diet feeding for 16 weeks. RESULTS AMPK activation suppressed endogenous cholesterol synthesis in primary bone marrow-derived macrophages from WT but not HMGCR KI mice, without changing other parameters of cholesterol regulation. Atherosclerotic plaque area was unchanged between WT and HMGCR KI mice, independent of sex. Correspondingly, there were no phenotypic differences observed in hematopoietic progenitors or differentiated immune cells in the bone marrow, blood, or spleen, and no significant changes in systemic markers of inflammation. When lethally irradiated female mice were transplanted with KI bone marrow, there was similar plaque content relative to WT. CONCLUSIONS Given previous work, our study demonstrates the importance of preclinical atherosclerosis model comparison and brings into question the importance of AMPK-mediated control of cholesterol synthesis in atherosclerosis.
Collapse
Affiliation(s)
- Tyler K T Smith
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Peyman Ghorbani
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Nicholas D LeBlond
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Julia R C Nunes
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Conor O'Dwyer
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Nia Ambursley
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Claire Fong-McMaster
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Lucía Minarrieta
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Leah A Burkovsky
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Rama El-Hakim
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Natasha A Trzaskalski
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Cassandra A A Locatelli
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Cameron Stotts
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Ciara Pember
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Katey J Rayner
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Bruce E Kemp
- Protein Chemistry and Metabolism, St. Vincent's Institute of Medical Research, Fitzroy, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia; Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Kim Loh
- Diabetes and Metabolic Disease, St. Vincent's Institute of Medical Research, Fitzroy, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia; Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Erin E Mulvihill
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Julie St-Pierre
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Morgan D Fullerton
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Centre for Catalysis Research and Innovation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada.
| |
Collapse
|
6
|
Pi H, Wang G, Wang Y, Zhang M, He Q, Zheng X, Yin K, Zhao G, Jiang T. Immunological perspectives on atherosclerotic plaque formation and progression. Front Immunol 2024; 15:1437821. [PMID: 39399488 PMCID: PMC11466832 DOI: 10.3389/fimmu.2024.1437821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Atherosclerosis serves as the primary catalyst for numerous cardiovascular diseases. Growing evidence suggests that the immune response is involved in every stage of atherosclerotic plaque evolution. Rapid, but not specific, innate immune arms, including neutrophils, monocytes/macrophages, dendritic cells (DCs) and other innate immune cells, as well as pattern-recognition receptors and various inflammatory mediators, contribute to atherogenesis. The specific adaptive immune response, governed by T cells and B cells, antibodies, and immunomodulatory cytokines potently regulates disease activity and progression. In the inflammatory microenvironment, the heterogeneity of leukocyte subpopulations plays a very important regulatory role in plaque evolution. With advances in experimental techniques, the fine mechanisms of immune system involvement in atherosclerotic plaque evolution are becoming known. In this review, we examine the critical immune responses involved in atherosclerotic plaque evolution, in particular, looking at atherosclerosis from the perspective of evolutionary immunobiology. A comprehensive understanding of the interplay between plaque evolution and plaque immunity provides clues for strategically combating atherosclerosis.
Collapse
Affiliation(s)
- Hui Pi
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Guangliang Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Yu Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ming Zhang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Qin He
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Xilong Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ting Jiang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| |
Collapse
|
7
|
Tsuji S, Kuramoto Y, Rajbhandari S, Takeda Y, Yamahara K, Yoshimura S. Intravenous administration of human amnion-derived mesenchymal stem cells improves gait and sensory function in mouse models of spinal cord injury. Front Cell Dev Biol 2024; 12:1464727. [PMID: 39324071 PMCID: PMC11422150 DOI: 10.3389/fcell.2024.1464727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 08/21/2024] [Indexed: 09/27/2024] Open
Abstract
Introduction Spinal cord injury (SCI) leads to severe disabilities and remains a significant social and economic challenge. Despite advances in medical research, there are still no effective treatments for SCI. Human amnion-derived mesenchymal stem cells (hAMSCs) have shown potential due to their anti-inflammatory and neuroprotective effects. This study evaluates the therapeutic potential of intravenously administered hAMSCs in SCI models. Methods Three days after induction of SCI with forceps calibrated with a 0.2 mm gap, hAMSCs or vehicle were administered intravenously. Up to 4 weeks of SCI induction, motor function was assessed by scores on the Basso Mouse Locomotor Scale (BMS) and the Basso-Beattie-Bresnahan Scale (BBB), and sensory function by hindlimb withdrawal reflex using von Frey filaments. Six weeks after SCI induction, gait function was assessed using three-dimensional motion analysis. Immunohistochemistry, polymerase chain reaction (PCR), flow cytometry, and ELISA assay were performed to clarify the mechanisms of functional improvement. Results The hAMSC treatment significantly improved sensory response and gait function. In the SCI site, immunohistochemistry showed a reduction in Iba1-positive cells and PCR revealed decreased TNFα and increased BDNF levels in the hAMSC-treated group. In assessing the systemic inflammatory response, hAMSC treatment reduced monocytic bone marrow-derived suppressor cells (M-MDSCs) and Ly6C-positive inflammatory macrophages in the bone marrow by flow cytometry and serum NO levels by ELISA assay. Discussion This study demonstrates the therapeutic potential of the hAMSC in SCI, with improvements in gait and sensory functions and reduced inflammation both locally and systemically. The findings support further investigation of the hAMSC as a potential treatment for SCI, focusing on their ability to modulate inflammation and promote neuroprotection.
Collapse
Affiliation(s)
- Shoichiro Tsuji
- Department of Neurosurgery, Hyogo Medical University, Hyogo, Japan
| | - Yoji Kuramoto
- Department of Neurosurgery, Hyogo Medical University, Hyogo, Japan
| | | | - Yuki Takeda
- Department of Neurosurgery, Hyogo Medical University, Hyogo, Japan
| | - Kenichi Yamahara
- Laboratory of Molecular and Cellular Therapy, Institute for Advanced Medical Sciences, Hyogo Medical University, Hyogo, Japan
| | | |
Collapse
|
8
|
Dutka M, Zimmer K, Ćwiertnia M, Ilczak T, Bobiński R. The role of PCSK9 in heart failure and other cardiovascular diseases-mechanisms of action beyond its effect on LDL cholesterol. Heart Fail Rev 2024; 29:917-937. [PMID: 38886277 PMCID: PMC11306431 DOI: 10.1007/s10741-024-10409-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
Proprotein convertase subtilisin/kexin type-9 (PCSK9) is a protein that regulates low-density lipoprotein (LDL) cholesterol metabolism by binding to the hepatic LDL receptor (LDLR), ultimately leading to its lysosomal degradation and an increase in LDL cholesterol (LDLc) levels. Treatment strategies have been developed based on blocking PCSK9 with specific antibodies (alirocumab, evolocumab) and on blocking its production with small regulatory RNA (siRNA) (inclisiran). Clinical trials evaluating these drugs have confirmed their high efficacy in reducing serum LDLc levels and improving the prognosis in patients with atherosclerotic cardiovascular diseases. Most studies have focused on the action of PCSK9 on LDLRs and the subsequent increase in LDLc concentrations. Increasing evidence suggests that the adverse cardiovascular effects of PCSK9, particularly its atherosclerotic effects on the vascular wall, may also result from mechanisms independent of its effects on lipid metabolism. PCSK9 induces the expression of pro-inflammatory cytokines contributing to inflammation within the vascular wall and promotes apoptosis, pyroptosis, and ferroptosis of cardiomyocytes and is thus involved in the development and progression of heart failure. The elimination of PCSK9 may, therefore, not only be a treatment for hypercholesterolaemia but also for atherosclerosis and other cardiovascular diseases. The mechanisms of action of PCSK9 in the cardiovascular system are not yet fully understood. This article reviews the current understanding of the mechanisms of PCSK9 action in the cardiovascular system and its contribution to cardiovascular diseases. Knowledge of these mechanisms may contribute to the wider use of PCSK9 inhibitors in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland.
| | - Karolina Zimmer
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Michał Ćwiertnia
- Department of Emergency Medicine, Faculty of Health Sciences, University of Bielsko-Biala, 43-309, Bielsko-Biała, Poland
| | - Tomasz Ilczak
- Department of Emergency Medicine, Faculty of Health Sciences, University of Bielsko-Biala, 43-309, Bielsko-Biała, Poland
| | - Rafał Bobiński
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| |
Collapse
|
9
|
Bashore AC, Xue C, Kim E, Yan H, Zhu LY, Pan H, Kissner M, Ross LS, Zhang H, Li M, Reilly MP. Monocyte Single-Cell Multimodal Profiling in Cardiovascular Disease Risk States. Circ Res 2024; 135:685-700. [PMID: 39105287 PMCID: PMC11430373 DOI: 10.1161/circresaha.124.324457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 07/11/2024] [Accepted: 07/28/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Monocytes are a critical innate immune system cell type that serves homeostatic and immunoregulatory functions. They have been identified historically by the cell surface expression of CD14 and CD16. However, recent single-cell studies have revealed that they are much more heterogeneous than previously realized. METHODS We utilized cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) and single-cell RNA sequencing to describe the comprehensive transcriptional and phenotypic landscape of 437 126 monocytes. RESULTS This high-dimensional multimodal approach identified vast phenotypic diversity and functionally distinct subsets, including IFN-responsive, MHCIIhi (major histocompatibility complex class II), monocyte-platelet aggregates, as well as nonclassical, and several subpopulations of classical monocytes. Using flow cytometry, we validated the existence of MHCII+CD275+ MHCIIhi, CD42b+ monocyte-platelet aggregates, CD16+CD99- nonclassical monocytes, and CD99+ classical monocytes. Each subpopulation exhibited unique characteristics, developmental trajectories, transcriptional regulation, and tissue distribution. In addition, alterations associated with cardiovascular disease risk factors, including race, smoking, and hyperlipidemia were identified. Moreover, the effect of hyperlipidemia was recapitulated in mouse models of elevated cholesterol. CONCLUSIONS This integrative and cross-species comparative analysis provides a new perspective on the comparison of alterations in monocytes in pathological conditions and offers insights into monocyte-driven mechanisms in cardiovascular disease and the potential for monocyte subpopulation targeted therapies.
Collapse
Affiliation(s)
- Alexander C Bashore
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Chenyi Xue
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Eunyoung Kim
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Hanying Yan
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia (H.Y., M.L.)
| | - Lucie Y Zhu
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Huize Pan
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (H.P.)
| | - Michael Kissner
- Columbia Stem Cell Initiative, Department of Genetics and Development (M.K.), Columbia University Irving Medical Center, New York
| | - Leila S Ross
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Hanrui Zhang
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia (H.Y., M.L.)
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York (M.P.R.)
| |
Collapse
|
10
|
Hu Q, Luo Y, He H, Chen H, Liao D. Comprehensive analysis of shared risk genes and immunity-metabolisms between non-alcoholic fatty liver disease and atherosclerosis via bulk and single-cell transcriptome analyses. Heliyon 2024; 10:e35453. [PMID: 39165965 PMCID: PMC11334902 DOI: 10.1016/j.heliyon.2024.e35453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/22/2024] Open
Abstract
Objective and design: Considering the clinical link between non-alcoholic fatty liver disease (NAFLD) and atherosclerosis (AS), we performed bioinformatics analysis to uncover their pathogenic interrelationship. Methods and results Data from the U.S. National Health and Nutritional Examination Survey (NHANES) 1999-2018 were included. Among 4851 participants in NHANES, NAFLD was significantly associated with atherosclerotic cardiovascular disease risk (ASCVD risk) (OR = 2.32, 95%CI: 2.04-2.65, P < 0.0001). We conducted WGCNA analysis for NAFLD (GSE130970) and AS (GSE28829) and identified three modules positively related to NAFLD severity and two modules accelerating atherosclerosis plaque progression. 198 key-modules genes were obtained via overlapping these modules. Next, we mined the disease-controlled differentially expressed genes (DEGs) from NAFLD (GSE89632) and AS (GSE100927), respectively. The final common risk genes (ACP5, TP53I3, RPS6KA1, TYMS, TREM2, CA12, and IFI27) were defined by intersecting the upregulated DEGs with 198 genes and validated in new datasets (GSE48452 and GSE43292). Importantly, they showed good diagnostic ability for NAFLD and AS. Immune infiltration analysis showed both illnesses have dysregulated immunity. Analysis of single-cell sequencing datasets NAFLD (GSE179886) and AS (GSE159677) uncovered different abnormal expressions of seven common genes in different immune cells while highlighting metabolic disturbances including upregulation of fatty acid biosynthesis, downregulation of fatty acid degradation and elongation. Conclusion We found 7 shared hub genes with good diagnostic ability and depicted the landscapes of immune and metabolism involved in NAFLD and AS. Our results provided a comprehensive association between them and may contribute to developing potential intervention strategies for targeting both disorders based on these risk factors.
Collapse
Affiliation(s)
- Qian Hu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Key Laboratory of Medical Genetics of Hunan Province, Central South University, Changsha, Hunan, China
| | - Yunfang Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
| | - Hao He
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hua Chen
- Department of Neurosurgery, the First people's Hospital of Changde City, Changde, Hunan, China
| | - Di Liao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
| |
Collapse
|
11
|
Annink ME, Kraaijenhof JM, Stroes ESG, Kroon J. Moving from lipids to leukocytes: inflammation and immune cells in atherosclerosis. Front Cell Dev Biol 2024; 12:1446758. [PMID: 39161593 PMCID: PMC11330886 DOI: 10.3389/fcell.2024.1446758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the most important cause of morbidity and mortality worldwide. While it is traditionally attributed to lipid accumulation in the vascular endothelium, recent research has shown that plaque inflammation is an important additional driver of atherogenesis. Though clinical outcome trials utilizing anti-inflammatory agents have proven promising in terms of reducing ASCVD risk, it is imperative to identify novel actionable targets that are more specific to atherosclerosis to mitigate adverse effects associated with systemic immune suppression. To that end, this review explores the contributions of various immune cells from the innate and adaptive immune system in promoting and mitigating atherosclerosis by integrating findings from experimental studies, high-throughput multi-omics technologies, and epidemiological research.
Collapse
Affiliation(s)
- Maxim E. Annink
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jordan M. Kraaijenhof
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Erik S. G. Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jeffrey Kroon
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, Netherlands
| |
Collapse
|
12
|
Gerhardt T, Huynh P, McAlpine CS. Neuroimmune circuits in the plaque and bone marrow regulate atherosclerosis. Cardiovasc Res 2024:cvae167. [PMID: 39086175 DOI: 10.1093/cvr/cvae167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/02/2024] [Accepted: 06/01/2024] [Indexed: 08/02/2024] Open
Abstract
Atherosclerosis remains the leading cause of death globally. Although its focal pathology is atheroma that develops in arterial walls, atherosclerosis is a systemic disease involving contributions by many organs and tissues. It is now established that the immune system causally contributes to all phases of atherosclerosis. Recent and emerging evidence positions the nervous system as a key modulator of inflammatory processes that underly atherosclerosis. This neuro-immune crosstalk, we are learning, is bidirectional, and immune regulated afferent signaling is becoming increasingly recognized in atherosclerosis. Here, we summarize data and concepts that link the immune and nervous systems in atherosclerosis by focusing on two important sites, the arterial vessel and the bone marrow.
Collapse
Affiliation(s)
- Teresa Gerhardt
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friede Springer Center for Cardiovascular Prevention at Charité, Berlin, Germany
| | - Pacific Huynh
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
13
|
Schäfer S, Gogiraju R, Rösch M, Kerstan Y, Beck L, Garbisch J, Saliba AE, Gisterå A, Hermanns HM, Boon L, Kastenmüller W, Schäfer K, Cochain C, Zernecke A. CD8 + T Cells Drive Plaque Smooth Muscle Cell Dedifferentiation in Experimental Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:1852-1872. [PMID: 38868941 DOI: 10.1161/atvbaha.123.320084] [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: 09/01/2023] [Accepted: 05/15/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Atherosclerosis is driven by the infiltration of the arterial intima by diverse immune cells and smooth muscle cells (SMCs). CD8+ T cells promote lesion growth during atherosclerotic lesion development, but their role in advanced atherosclerosis is less clear. Here, we studied the role of CD8+ T cells and their effects on SMCs in established atherosclerosis. METHODS CD8+ T cells were depleted in (SMC reporter) low-density lipoprotein receptor-deficient (Ldlr-/-) mice with established atherosclerotic lesions. Atherosclerotic lesion formation was examined, and single-cell RNA sequencing of aortic SMCs and their progeny was performed. Additionally, coculture experiments with primary aortic SMCs and CD8+ T cells were conducted. RESULTS Although we could not detect differences in atherosclerotic lesion size, an increased plaque SMC content was noted in mice after CD8+ T-cell depletion. Single-cell RNA sequencing of aortic lineage-traced SMCs revealed contractile SMCs and a modulated SMC cluster, expressing macrophage- and osteoblast-related genes. CD8+ T-cell depletion was associated with an increased contractile but decreased macrophage and osteoblast-like gene signature in this modulated aortic SMC cluster. Conversely, exposure of isolated aortic SMCs to activated CD8+ T cells decreased the expression of genes indicative of a contractile SMC phenotype and induced a macrophage and osteoblast-like cell state. Notably, CD8+ T cells triggered calcium deposits in SMCs under osteogenic conditions. Mechanistically, we identified transcription factors highly expressed in modulated SMCs, including Runx1, to be induced by CD8+ T cells in cultured SMCs in an IFNγ (interferon-γ)-dependent manner. CONCLUSIONS We here uncovered CD8+ T cells to control the SMC phenotype in atherosclerosis. CD8+ T cells promote SMC dedifferentiation and drive SMCs to adopt features of macrophage-like and osteoblast-like, procalcifying cell phenotypes. Given the critical role of SMCs in atherosclerotic plaque stability, CD8+ T cells could thus be explored as therapeutic target cells during lesion progression.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/immunology
- Cell Dedifferentiation
- Plaque, Atherosclerotic
- Mice
- Disease Models, Animal
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/immunology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/immunology
- Mice, Inbred C57BL
- Mice, Knockout
- Cells, Cultured
- Male
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Phenotype
- Core Binding Factor Alpha 2 Subunit/genetics
- Core Binding Factor Alpha 2 Subunit/metabolism
- Aorta/pathology
- Aorta/immunology
- Aorta/metabolism
- Coculture Techniques
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/immunology
- Aortic Diseases/metabolism
Collapse
Affiliation(s)
- Sarah Schäfer
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| | - Rajinikanth Gogiraju
- Department of Cardiology, Cardiology I, University Medicine Mainz, Germany (R.G., K.S.)
| | - Melanie Rösch
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| | - Yvonne Kerstan
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| | - Lina Beck
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| | - Janine Garbisch
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Institute of Molecular Infection Biology Faculty of Medicine, University of Würzburg, Germany (A.-E.S.)
| | - Anton Gisterå
- Center for Molecular Medicine, Department of Medicine, Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (A.G.)
| | - Heike M Hermanns
- Medical Clinic II, Division of Hepatology (H.M.H.), University Hospital of Würzburg, Germany
| | | | | | - Katrin Schäfer
- Department of Cardiology, Cardiology I, University Medicine Mainz, Germany (R.G., K.S.)
| | - Clément Cochain
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| |
Collapse
|
14
|
Shang R, Rodrigues B. Lipoprotein lipase as a target for obesity/diabetes related cardiovascular disease. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13199. [PMID: 39081272 PMCID: PMC11286490 DOI: 10.3389/jpps.2024.13199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024]
Abstract
Worldwide, the prevalence of obesity and diabetes have increased, with heart disease being their leading cause of death. Traditionally, the management of obesity and diabetes has focused mainly on weight reduction and controlling high blood glucose. Unfortunately, despite these efforts, poor medication management predisposes these patients to heart failure. One instigator for the development of heart failure is how cardiac tissue utilizes different sources of fuel for energy. In this regard, the heart switches from using various substrates, to predominantly using fatty acids (FA). This transformation to using FA as an exclusive source of energy is helpful in the initial stages of the disease. However, over the progression of diabetes this has grave end results. This is because toxic by-products are produced by overuse of FA, which weaken heart function (heart disease). Lipoprotein lipase (LPL) is responsible for regulating FA delivery to the heart, and its function during diabetes has not been completely revealed. In this review, the mechanisms by which LPL regulates fuel utilization by the heart in control conditions and following diabetes will be discussed in an attempt to identify new targets for therapeutic intervention. Currently, as treatment options to directly target diabetic heart disease are scarce, research on LPL may assist in drug development that exclusively targets fuel utilization by the heart and lipid accumulation in macrophages to help delay, prevent, or treat cardiac failure, and provide long-term management of this condition during diabetes.
Collapse
Affiliation(s)
- Rui Shang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Brian Rodrigues
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
15
|
Zhang J, Chang J, Chen V, Beg MA, Huang W, Vick L, Wang Y, Zhang H, Yttre E, Gupta A, Castleberry M, Zhang Z, Dai W, Song S, Zhu J, Yang M, Brown AK, Xu Z, Ma YQ, Smith BC, Zielonka J, Traylor JG, Dhaou CB, Orr AW, Cui W, Zheng Z, Chen Y. Oxidized LDL regulates efferocytosis through the CD36-PKM2-mtROS pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.07.556574. [PMID: 39071358 PMCID: PMC11275753 DOI: 10.1101/2023.09.07.556574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Macrophage efferocytosis, the process by which phagocytes engulf and remove apoptotic cells (ACs), plays a critical role in maintaining tissue homeostasis. Efficient efferocytosis prevents secondary necrosis, mitigates chronic inflammation, and impedes atherosclerosis progression. However, the regulatory mechanisms of efferocytosis under atherogenic conditions remain poorly understood. We previously demonstrated that oxidized LDL (oxLDL), an atherogenic lipoprotein, induces mitochondrial reactive oxygen species (mtROS) in macrophages via CD36. In this study, we demonstrate that macrophage mtROS facilitate continual efferocytosis through a positive feedback mechanism. However, oxLDL disrupts continual efferocytosis by dysregulating the internalization of ACs. This disruption is mediated by an overproduction of mtROS. Mechanistically, oxLDL/CD36 signaling promotes the translocation of cytosolic PKM2 to mitochondria, facilitated by the chaperone GRP75. Mitochondrial PKM2 then binds to Complex III of the electron transport chain, inducing mtROS production. This study elucidates a novel regulatory mechanism of efferocytosis in atherosclerosis, providing potential therapeutic targets for intervention. SUMMARY Macrophages clear apoptotic cells through a process called efferocytosis, which involves mitochondrial ROS. However, the atherogenic oxidized LDL overstimulates mitochondrial ROS via the CD36-PKM2 pathway, disrupting continual efferocytosis. This finding elucidates a novel molecular mechanism that explains defects in efferocytosis, driving atherosclerosis progression.
Collapse
|
16
|
Chen Q, Zhang JL, Yang JS, Jin Q, Yang J, Xue Q, Guang XF. Novel Diagnostic Biomarkers Related to Necroptosis and Immune Infiltration in Coronary Heart Disease. J Inflamm Res 2024; 17:4525-4548. [PMID: 39006493 PMCID: PMC11246668 DOI: 10.2147/jir.s457469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/11/2024] [Indexed: 07/16/2024] Open
Abstract
Purpose Necroptosis, a monitored form of inflammatory cell death, contributes to coronary heart disease (CHD) progression. This study examined the potential of using necroptosis genes as diagnostic markers for CHD and sought to elucidate the underlying roles. Methods Through bioinformatic analysis of GSE20680 and GSE20681, we first identified the differentially expressed genes (DEGs) related to necroptosis in CHD. Hub genes were identified using least absolute shrinkage and selection operator (LASSO) regression and random forest analysis after studying immune infiltration and transcription factor-miRNA interaction networks according to the DEGs. Quantitative polymerase chain reaction and immunohistochemistry were used to further investigate hub gene expression in vivo, for which a diagnostic model was constructed and the predictive efficacy was validated. Finally, the CHD group was categorized into high- and low-score groups in accordance with the single-sample gene set enrichment analysis (ssGSEA) score of the necroptosis genes. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, GSEA, and further immune infiltration analyses were performed on the two groups to explore the possible roles of hub genes. Results Based on the results of the LASSO regression and random forest analyses, four genes were used to construct a diagnostic model to establish a nomogram. Additionally, an extensive analysis of all seventeen necroptosis genes revealed notable distinctions in expression between high-risk and low-risk groups. Evaluation of immune infiltration revealed that neutrophils, monocytes, B cells, and activated dendritic cells were highly distributed in the peripheral blood of patients with CHD. Specifically, the high CHD score group exhibited greater neutrophil and monocyte infiltration. Conversely, the high-score group showed lower infiltration of M0 and M2 macrophages, CD8+ T, plasma, and resting mast cells. Conclusion TLR3, MLKL, HMGB1, and NDRG2 may be prospective biomarkers for CHD diagnosis. These findings offer plausible explanations for the role of necroptosis in CHD progression through immune infiltration and inflammatory response.
Collapse
Affiliation(s)
- Qiu Chen
- Department of Cardiology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| | - Ji-Lei Zhang
- Department of Cardiology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| | - Jie-Shun Yang
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming, People's Republic of China
| | - Qing Jin
- Department of Cardiology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| | - Jun Yang
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| | - Qiang Xue
- Department of Cardiology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| | - Xue-Feng Guang
- Department of Cardiology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| |
Collapse
|
17
|
Jiao J, Hu B, Mou T, Li Q, Tian Y, Zhang N, Zhang Y, Yun M, Nan N, Tian J, Yu W, Mi H, Dong W, Song X. Translocator Protein 18 kDa Tracer 18F-FDPA PET/CTA Imaging for the Evaluation of Inflammation in Vulnerable Plaques. Mol Pharm 2024; 21:3623-3633. [PMID: 38819959 DOI: 10.1021/acs.molpharmaceut.4c00344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Inflammation induced by activated macrophages within vulnerable atherosclerotic plaques (VAPs) constitutes a significant risk factor for plaque rupture. Translocator protein (TSPO) is highly expressed in activated macrophages. This study investigated the effectiveness of TSPO radiotracers, 18F-FDPA, in detecting VAPs and quantifying plaque inflammation in rabbits. 18 New Zealand rabbits were divided into 3 groups: sham group A, VAP model group B, and evolocumab treatment group C. 18F-FDPA PET/CTA imaging was performed at 12, 16, and 24 weeks in all groups. Optical coherence tomography (OCT) was performed on the abdominal aorta at 24 weeks. The VAP was defined through OCT images, and ex vivo aorta PET imaging was also performed at 24 weeks. The SUVmax and SUVmean of 18F-FDPA were measured on the target organ, and the target-to-background ratio (TBRmax) was calculated as SUVmax/SUVblood pool. The arterial sections of the isolated abdominal aorta were analyzed by HE staining, CD68 and TSPO immunofluorescence staining, and TSPO Western blot. The results showed that at 24 weeks, the plaque TBRmax of 18F-FDPA in group B was significantly higher than in groups A and C. Immunofluorescence staining of CD68 and TSPO, as well as Western blot, confirmed the increased expression of macrophages and TSPO in the corresponding regions of group B. HE staining revealed an increased presence of the lipid core, multiple foam cells, and inflammatory cell infiltration in the area with high 18F-FDPA uptake. This indicates a correlation between 18F-FDPA uptake, inflammation severity, and VAPs. The TSPO-targeted tracer 18F-FDPA shows specific uptake in macrophage-rich regions of atherosclerotic plaques, making it a valuable tool for assessing inflammation in VAPs.
Collapse
Affiliation(s)
- Jian Jiao
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Biao Hu
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Tiantian Mou
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Quan Li
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yi Tian
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Nan Zhang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ying Zhang
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mingkai Yun
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Nan Nan
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Jing Tian
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Wei Yu
- Department of Pathology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Hongzhi Mi
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Wei Dong
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
18
|
Li CX, Yue L. The Multifaceted Nature of Macrophages in Cardiovascular Disease. Biomedicines 2024; 12:1317. [PMID: 38927523 PMCID: PMC11201197 DOI: 10.3390/biomedicines12061317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/01/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
As the leading cause of mortality worldwide, cardiovascular disease (CVD) represents a variety of heart diseases and vascular disorders, including atherosclerosis, aneurysm, ischemic injury in the heart and brain, arrythmias, and heart failure. Macrophages, a diverse population of immune cells that can promote or suppress inflammation, have been increasingly recognized as a key regulator in various processes in both healthy and disease states. In healthy conditions, these cells promote the proper clearance of cellular debris, dead and dying cells, and provide a strong innate immune barrier to foreign pathogens. However, macrophages can play a detrimental role in the progression of disease as well, particularly those inflammatory in nature. This review will focus on the current knowledge regarding the role of macrophages in cardiovascular diseases.
Collapse
Affiliation(s)
- Cindy X. Li
- Department of Cell Biology, Pat and Jim Calhoun Cardiovascular Center, University of Connecticut Health Center, Farmington, CT 06030, USA;
- Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Lixia Yue
- Department of Cell Biology, Pat and Jim Calhoun Cardiovascular Center, University of Connecticut Health Center, Farmington, CT 06030, USA;
- Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
19
|
Getz GS, Reardon CA. Insights from Murine Studies on the Site Specificity of Atherosclerosis. Int J Mol Sci 2024; 25:6375. [PMID: 38928086 PMCID: PMC11204064 DOI: 10.3390/ijms25126375] [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: 05/06/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Atherosclerosis is an inflammatory reaction that develops at specific regions within the artery wall and at specific sites of the arterial tree over a varying time frame in response to a variety of risk factors. The mechanisms that account for the interaction of systemic factors and atherosclerosis-susceptible regions of the arterial tree to mediate this site-specific development of atherosclerosis are not clear. The dynamics of blood flow has a major influence on where in the arterial tree atherosclerosis develops, priming the site for interactions with atherosclerotic risk factors and inducing cellular and molecular participants in atherogenesis. But how this accounts for lesion development at various locations along the vascular tree across differing time frames still requires additional study. Currently, murine models are favored for the experimental study of atherogenesis and provide the most insight into the mechanisms that may contribute to the development of atherosclerosis. Based largely on these studies, in this review, we discuss the role of hemodynamic shear stress, SR-B1, and other factors that may contribute to the site-specific development of atherosclerosis.
Collapse
Affiliation(s)
- Godfrey S. Getz
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA;
| | | |
Collapse
|
20
|
Zhen J, Li X, Yu H, Du B. High-density lipoprotein mimetic nano-therapeutics targeting monocytes and macrophages for improved cardiovascular care: a comprehensive review. J Nanobiotechnology 2024; 22:263. [PMID: 38760755 PMCID: PMC11100215 DOI: 10.1186/s12951-024-02529-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/03/2024] [Indexed: 05/19/2024] Open
Abstract
The prevalence of cardiovascular diseases continues to be a challenge for global health, necessitating innovative solutions. The potential of high-density lipoprotein (HDL) mimetic nanotherapeutics in the context of cardiovascular disease and the intricate mechanisms underlying the interactions between monocyte-derived cells and HDL mimetic showing their impact on inflammation, cellular lipid metabolism, and the progression of atherosclerotic plaque. Preclinical studies have demonstrated that HDL mimetic nanotherapeutics can regulate monocyte recruitment and macrophage polarization towards an anti-inflammatory phenotype, suggesting their potential to impede the progression of atherosclerosis. The challenges and opportunities associated with the clinical application of HDL mimetic nanotherapeutics, emphasize the need for additional research to gain a better understanding of the precise molecular pathways and long-term effects of these nanotherapeutics on monocytes and macrophages to maximize their therapeutic efficacy. Furthermore, the use of nanotechnology in the treatment of cardiovascular diseases highlights the potential of nanoparticles for targeted treatments. Moreover, the concept of theranostics combines therapy and diagnosis to create a selective platform for the conversion of traditional therapeutic medications into specialized and customized treatments. The multifaceted contributions of HDL to cardiovascular and metabolic health via highlight its potential to improve plaque stability and avert atherosclerosis-related problems. There is a need for further research to maximize the therapeutic efficacy of HDL mimetic nanotherapeutics and to develop targeted treatment approaches to prevent atherosclerosis. This review provides a comprehensive overview of the potential of nanotherapeutics in the treatment of cardiovascular diseases, emphasizing the need for innovative solutions to address the challenges posed by cardiovascular diseases.
Collapse
Affiliation(s)
- Juan Zhen
- The First Hospital of Jilin University, Changchun, 130021, China
| | - Xiangjun Li
- School of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Haitao Yu
- The First Hospital of Jilin University, Changchun, 130021, China
| | - Bing Du
- The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
21
|
Mulholland M, Depuydt MAC, Jakobsson G, Ljungcrantz I, Grentzmann A, To F, Bengtsson E, Jaensson Gyllenbäck E, Grönberg C, Rattik S, Liberg D, Schiopu A, Björkbacka H, Kuiper J, Bot I, Slütter B, Engelbertsen D. Interleukin-1 receptor accessory protein blockade limits the development of atherosclerosis and reduces plaque inflammation. Cardiovasc Res 2024; 120:581-595. [PMID: 38563353 PMCID: PMC11074796 DOI: 10.1093/cvr/cvae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 04/04/2024] Open
Abstract
AIMS The interleukin-1 receptor accessory protein (IL1RAP) is a co-receptor required for signalling through the IL-1, IL-33, and IL-36 receptors. Using a novel anti-IL1RAP-blocking antibody, we investigated the role of IL1RAP in atherosclerosis. METHODS AND RESULTS Single-cell RNA sequencing data from human atherosclerotic plaques revealed the expression of IL1RAP and several IL1RAP-related cytokines and receptors, including IL1B and IL33. Histological analysis showed the presence of IL1RAP in both the plaque and adventitia, and flow cytometry of murine atherosclerotic aortas revealed IL1RAP expression on plaque leucocytes, including neutrophils and macrophages. High-cholesterol diet fed apolipoprotein E-deficient (Apoe-/-) mice were treated with a novel non-depleting IL1RAP-blocking antibody or isotype control for the last 6 weeks of diet. IL1RAP blockade in mice resulted in a 20% reduction in subvalvular plaque size and limited the accumulation of neutrophils and monocytes/macrophages in plaques and of T cells in adventitia, compared with control mice. Indicative of reduced plaque inflammation, the expression of several genes related to leucocyte recruitment, including Cxcl1 and Cxcl2, was reduced in brachiocephalic arteries of anti-IL1RAP-treated mice, and the expression of these chemokines in human plaques was mainly restricted to CD68+ myeloid cells. Furthermore, in vitro studies demonstrated that IL-1, IL-33, and IL-36 induced CXCL1 release from both macrophages and fibroblasts, which could be mitigated by IL1RAP blockade. CONCLUSION Limiting IL1RAP-dependent cytokine signalling pathways in atherosclerotic mice reduces plaque burden and plaque inflammation, potentially by limiting plaque chemokine production.
Collapse
Affiliation(s)
- Megan Mulholland
- Department of Clinical Sciences, Cardiovascular Research—Immune Regulation, Lund University, Malmö, Sweden
| | - Marie A C Depuydt
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, The Netherlands
| | - Gabriel Jakobsson
- Department of Translational Medicine, Cardiac Inflammation, Lund University, Malmö, Sweden
| | - Irena Ljungcrantz
- Department of Clinical Sciences, Cardiovascular Research—Immune Regulation, Lund University, Malmö, Sweden
| | - Andrietta Grentzmann
- Department of Clinical Sciences, Cardiovascular Research—Immune Regulation, Lund University, Malmö, Sweden
| | - Fong To
- Department of Clinical Sciences, Cardiovascular Research—Matrix and Inflammation in Atherosclerosis, Lund University, Malmö, Sweden
| | - Eva Bengtsson
- Department of Clinical Sciences, Cardiovascular Research—Matrix and Inflammation in Atherosclerosis, Lund University, Malmö, Sweden
- Department of Biomedical Science, Malmö University, Malmö, Sweden
- Biofilms—Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | | | | | - Sara Rattik
- Department of Clinical Sciences, Cardiovascular Research—Immune Regulation, Lund University, Malmö, Sweden
- Cantargia AB, Lund, Sweden
| | | | - Alexandru Schiopu
- Department of Translational Medicine, Cardiac Inflammation, Lund University, Malmö, Sweden
| | - Harry Björkbacka
- Department of Clinical Sciences, Cardiovascular Research—Cellular Metabolism and Inflammation, Lund University, Malmö, Sweden
| | - Johan Kuiper
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, The Netherlands
| | - Ilze Bot
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, The Netherlands
| | - Bram Slütter
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, The Netherlands
| | - Daniel Engelbertsen
- Department of Clinical Sciences, Cardiovascular Research—Immune Regulation, Lund University, Malmö, Sweden
| |
Collapse
|
22
|
Nguyen TK, Paone S, Baxter AA, Mayfosh AJ, Phan TK, Chan E, Peter K, Poon IKH, Thomas SR, Hulett MD. Heparanase promotes the onset and progression of atherosclerosis in apolipoprotein E gene knockout mice. Atherosclerosis 2024; 392:117519. [PMID: 38581737 DOI: 10.1016/j.atherosclerosis.2024.117519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/31/2024] [Accepted: 03/12/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND AND AIMS Atherosclerosis is the primary underlying cause of myocardial infarction and stroke, which are the major causes of death globally. Heparanase (Hpse) is a pro-inflammatory extracellular matrix degrading enzyme that has been implicated in atherogenesis. However, to date the precise roles of Hpse in atherosclerosis and its mechanisms of action are not well defined. This study aims to provide new insights into the contribution of Hpse in different stages of atherosclerosis in vivo. METHODS We generated Hpse gene-deficient mice on the atherosclerosis-prone apolipoprotein E gene knockout (ApoE-/-) background to investigate the impact of Hpse gene deficiency on the initiation and progression of atherosclerosis after 6 and 14 weeks high-fat diet feeding, respectively. Atherosclerotic lesion development, blood serum profiles, lesion composition and aortic immune cell populations were evaluated. RESULTS Hpse-deficient mice exhibited significantly reduced atherosclerotic lesion burden in the aortic sinus and aorta at both time-points, independent of changes in plasma cholesterol levels. A significant reduction in the necrotic core size and an increase in smooth muscle cell content were also observed in advanced atherosclerotic plaques of Hpse-deficient mice. Additionally, Hpse deficiency reduced circulating and aortic levels of VCAM-1 at the initiation and progression stages of disease and circulating MCP-1 levels in the initiation but not progression stage. Moreover, the aortic levels of total leukocytes and dendritic cells in Hpse-deficient ApoE-/- mice were significantly decreased compared to control ApoE-/-mice at both disease stages. CONCLUSIONS This study identifies Hpse as a key pro-inflammatory enzyme driving the initiation and progression of atherosclerosis and highlighting the potential of Hpse inhibitors as novel anti-inflammatory treatments for cardiovascular disease.
Collapse
Affiliation(s)
- Tien K Nguyen
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Stephanie Paone
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Amy A Baxter
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Alyce J Mayfosh
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Thanh Kha Phan
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Enoch Chan
- Department of Pathology, School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia
| | - Ivan K H Poon
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Shane R Thomas
- Department of Pathology, School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Mark D Hulett
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia.
| |
Collapse
|
23
|
Rasheed A, Robichaud S, Dennison T, Nguyen MA, Geoffrion M, Reed JN, Wyatt HJ, Marouf Y, Baxi A, Lee R, Kazan H, Civelek M, van Solingen C, Ouimet M, Rayner KJ. Hyperlipidemia-induced hematopoiesis is repressed by MLKL in endothelial cells of the splenic niche. NATURE CARDIOVASCULAR RESEARCH 2024; 3:594-611. [PMID: 39195940 DOI: 10.1038/s44161-024-00470-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/04/2024] [Indexed: 08/29/2024]
Abstract
Dysregulation of the hematopoietic niche during hyperlipidemia facilitates pathologic leukocyte production, driving atherogenesis. Although definitive hematopoiesis occurs primarily in the bone marrow, during atherosclerosis this also occurs in the spleen. Cells of the bone marrow niche, particularly endothelial cells, have been studied in atherosclerosis, although little is known about how splenic endothelial cells respond to the atherogenic environment. Here we show unique dysregulated pathways in splenic compared to bone marrow endothelial cells during atherosclerosis, including perturbations of lipid metabolism and endocytic trafficking pathways. As part of this response, we identify the mixed lineage kinase domain-like (MLKL) protein as a repressor of splenic, but not bone marrow, myelopoiesis. Silencing MLKL in splenic endothelial cells results in inefficient endosomal trafficking and lipid accumulation, ultimately promoting the production of myeloid cells that participate in plaque development. These studies identify endocytic trafficking by MLKL as a key mechanism of splenic endothelial cell maintenance, splenic hematopoiesis and, subsequently, atherosclerosis.
Collapse
Affiliation(s)
- Adil Rasheed
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada.
- Department of Physiology, Immunology Center of Georgia, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| | - Sabrina Robichaud
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Taylor Dennison
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - My-Anh Nguyen
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | | | - Jordan N Reed
- University of Virginia Center for Public Health Genomics, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Hailey J Wyatt
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Yacine Marouf
- Electrical and Computer Engineering Graduate Program, Antalya Bilim University, Antalya, Turkey
| | - Adir Baxi
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Richard Lee
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, CA, USA
| | - Hilal Kazan
- Department of Computer Engineering, Antalya Bilim University, Antalya, Turkey
| | - Mete Civelek
- University of Virginia Center for Public Health Genomics, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Coen van Solingen
- Department of Medicine, Leon H. Charney Division of Cardiology, NYU Cardiovascular Research Center, New York University Langone Health, New York, NY, USA
| | - Mireille Ouimet
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Katey J Rayner
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
24
|
Zhao D, Huang ZK, Liang Y, Li ZJ, Zhang XW, Li KH, Wu H, Zhang XD, Li CS, An D, Sun X, An MX, Shi JX, Bao YJ, Tian L, Wang DF, Wu AH, Chen YH, Zhao WD. Monocytes Release Pro-Cathepsin D to Drive Blood-to-Brain Transcytosis in Diabetes. Circ Res 2024; 134:e17-e33. [PMID: 38420756 DOI: 10.1161/circresaha.123.323622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Microvascular complications are the major outcome of type 2 diabetes progression, and the underlying mechanism remains to be determined. METHODS High-throughput RNA sequencing was performed using human monocyte samples from controls and diabetes. The transgenic mice expressing human CTSD (cathepsin D) in the monocytes was constructed using CD68 promoter. In vivo 2-photon imaging, behavioral tests, immunofluorescence, transmission electron microscopy, Western blot analysis, vascular leakage assay, and single-cell RNA sequencing were performed to clarify the phenotype and elucidate the molecular mechanism. RESULTS Monocytes expressed high-level CTSD in patients with type 2 diabetes. The transgenic mice expressing human CTSD in the monocytes showed increased brain microvascular permeability resembling the diabetic microvascular phenotype, accompanied by cognitive deficit. Mechanistically, the monocytes release nonenzymatic pro-CTSD to upregulate caveolin expression in brain endothelium triggering caveolae-mediated transcytosis, without affecting the paracellular route of brain microvasculature. The circulating pro-CTSD activated the caveolae-mediated transcytosis in brain endothelial cells via its binding with low-density LRP1 (lipoprotein receptor-related protein 1). Importantly, genetic ablation of CTSD in the monocytes exhibited a protective effect against the diabetes-enhanced brain microvascular transcytosis and the diabetes-induced cognitive impairment. CONCLUSIONS These findings uncover the novel role of circulatory pro-CTSD from monocytes in the pathogenesis of cerebral microvascular lesions in diabetes. The circulatory pro-CTSD is a potential target for the intervention of microvascular complications in diabetes.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Zeng-Kang Huang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Yu Liang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Zhi-Jun Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Xue-Wei Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Kun-Hang Li
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Hao Wu
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Xu-Dong Zhang
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Chen-Sheng Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Dong An
- School of Mechanical Engineering, Shenyang Jianzhu University, China (D.A.)
| | - Xue Sun
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Ming-Xin An
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Jun-Xiu Shi
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Yi-Jun Bao
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Li Tian
- Department of Gerontology (L.T., D.-F.W.), Shengjing Hospital of China Medical University, Shenyang, China
| | - Di-Fei Wang
- Department of Gerontology (L.T., D.-F.W.), Shengjing Hospital of China Medical University, Shenyang, China
| | - An-Hua Wu
- Department of Neurosurgery (A.-H.W.), Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Hua Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Wei-Dong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| |
Collapse
|
25
|
Pande S, Vary C, Yang X, Liaw L, Gower L, Friesel R, Prudovsky I, Ryzhov S. Endothelial IL17RD promotes Western diet-induced aortic myeloid cell infiltration. Biochem Biophys Res Commun 2024; 701:149552. [PMID: 38335918 PMCID: PMC10936543 DOI: 10.1016/j.bbrc.2024.149552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024]
Abstract
The Interleukin-17 (IL17) family is a group of cytokines implicated in the etiology of several inflammatory diseases. Interleukin-17 receptor D (IL17RD), also known as Sef (similar expression to fibroblast growth factor) belonging to the family of IL17 receptors, has been shown to modulate IL17A-associated inflammatory phenotypes. The objective of this study was to test the hypothesis that IL17RD promotes endothelial cell activation and consequent leukocyte adhesion. We utilized primary human aortic endothelial cells and demonstrated that RNAi targeting of IL17RD suppressed transcript levels by 83 % compared to non-targeted controls. Further, RNAi knockdown of IL17RD decreased the adhesion of THP-1 monocytic cells onto a monolayer of aortic endothelial cells in response to IL17A. Additionally, we determined that IL17A did not significantly enhance the activation of canonical MAPK and NFκB pathways in endothelial cells, and further did not significantly affect the expression of VCAM-1 and ICAM-1 in aortic endothelial cells, which is contrary to previous findings. We also determined the functional relevance of our findings in vivo by comparing the expression of endothelial VCAM-1 and ICAM-1 and leukocyte infiltration in the aorta in Western diet-fed Il17rd null versus wild-type mice. Our results showed that although Il17rd null mice do not have significant alteration in aortic expression of VCAM-1 and ICAM-1 in endothelial cells, they exhibit decreased accumulation of proinflammatory monocytes and neutrophils, suggesting that endothelial IL17RD induced in vivo myeloid cell accumulation is not dependent on upregulation of VCAM-1 and ICAM-1 expression. We further performed proteomics analysis to identify potential molecular mediators of the IL17A/IL17RD signaling axis. Collectively, our results underscore a critical role for Il17rd in the regulation of aortic myeloid cell infiltration in the context of Western diet feeding.
Collapse
Affiliation(s)
- Shivangi Pande
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04496, USA
| | - Calvin Vary
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04496, USA
| | - Xuehui Yang
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Lucy Liaw
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04496, USA
| | - Lindsey Gower
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Robert Friesel
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04496, USA.
| | - Igor Prudovsky
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04496, USA.
| | - Sergey Ryzhov
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04496, USA.
| |
Collapse
|
26
|
Volpatti LR, de Matos SN, Borjas G, Reda J, Watkins EA, Zhou Z, Nguyen M, Solanki A, Fang Y, Hubbell JA. LDL-Binding IL-10 Reduces Vascular Inflammation in Atherosclerotic Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.582839. [PMID: 38496521 PMCID: PMC10942346 DOI: 10.1101/2024.03.04.582839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease associated with the accumulation of low-density lipoprotein (LDL) in arterial walls. Higher levels of the anti-inflammatory cytokine IL-10 in serum are correlated with reduced plaque burden. However, cytokine therapies have not translated well to the clinic, partially due to their rapid clearance and pleiotropic nature. Here, we engineered IL-10 to overcome these challenges by hitchhiking on LDL to atherosclerotic plaques. Specifically, we constructed fusion proteins in which one domain is IL-10 and the other is an antibody fragment (Fab) that binds to protein epitopes of LDL. In murine models of atherosclerosis, we show that systemically administered Fab-IL-10 constructs bind circulating LDL and traffic to atherosclerotic plaques. One such construct, 2D03-IL-10, significantly reduces aortic immune cell infiltration to levels comparable to healthy mice, whereas non-targeted IL-10 has no therapeutic effect. Mechanistically, we demonstrate that 2D03-IL-10 preferentially associates with foamy macrophages and reduces pro-inflammatory activation markers. This platform technology can be applied to a variety of therapeutics and shows promise as a potential targeted anti-inflammatory therapy in atherosclerosis.
Collapse
Affiliation(s)
- Lisa R. Volpatti
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, United States
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, United States
| | - Salvador Norton de Matos
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
- Medical Scientist Training Program, Pritzker School of Medicine, University of Chicago, IL, 60637, United States
| | - Gustavo Borjas
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
| | - Joseph Reda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
| | - Elyse A. Watkins
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
| | - Zhengjie Zhou
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637, United States
| | - Mindy Nguyen
- Animal Resources Center, University of Chicago, Chicago, IL 60637, United States
| | - Ani Solanki
- Animal Resources Center, University of Chicago, Chicago, IL 60637, United States
| | - Yun Fang
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637, United States
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, USA
- Committee on Molecular Medicine, University of Chicago, Chicago, IL, 60637, United States
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
- Committee on Immunology, University of Chicago, Chicago, IL 60637, United States
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, United States
| |
Collapse
|
27
|
McQueen P, Molina D, Pinos I, Krug S, Taylor AJ, LaFrano MR, Kane MA, Amengual J. Finasteride delays atherosclerosis progression in mice and is associated with a reduction in plasma cholesterol in men. J Lipid Res 2024; 65:100507. [PMID: 38272355 PMCID: PMC10899056 DOI: 10.1016/j.jlr.2024.100507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/27/2024] Open
Abstract
Finasteride is commonly prescribed to treat benign prostate hyperplasia and male-pattern baldness in cis men and, more recently, trans individuals. However, the effect of finasteride on cardiovascular disease remains elusive. We evaluated the role of finasteride on atherosclerosis using low-density lipoprotein (LDL) receptor-deficient (Ldlr-/-) mice. Next, we examined the relevance to humans by analyzing the data deposited between 2009 and 2016 in the National Health and Nutrition Examination Survey. We show that finasteride reduces total plasma cholesterol and delays the development of atherosclerosis in Ldlr-/- mice. Finasteride reduced monocytosis, monocyte recruitment to the lesion, macrophage lesion content, and necrotic core area, the latter of which is an indicator of plaque vulnerability in humans. RNA sequencing analysis revealed a downregulation of inflammatory pathways and an upregulation of bile acid metabolism, oxidative phosphorylation, and cholesterol pathways in the liver of mice taking finasteride. Men reporting the use of finasteride showed lower plasma levels of cholesterol and LDL-cholesterol than those not taking the drug. Our data unveil finasteride as a potential treatment to delay cardiovascular disease in people by improving the plasma lipid profile.
Collapse
Affiliation(s)
- Patrick McQueen
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL, USA
| | - Donald Molina
- Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL, USA
| | - Ivan Pinos
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL, USA
| | - Samuel Krug
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Anna J Taylor
- Carver Metabolomics Core, Roy J. Carver Biotechnology Center, University of Illinois Urbana Champaign, Urbana, IL, USA
| | - Michael R LaFrano
- Carver Metabolomics Core, Roy J. Carver Biotechnology Center, University of Illinois Urbana Champaign, Urbana, IL, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Jaume Amengual
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL, USA; Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL, USA.
| |
Collapse
|
28
|
Janssen H, Koekkoek LL, Swirski FK. Effects of lifestyle factors on leukocytes in cardiovascular health and disease. Nat Rev Cardiol 2024; 21:157-169. [PMID: 37752350 DOI: 10.1038/s41569-023-00931-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/01/2023] [Indexed: 09/28/2023]
Abstract
Exercise, stress, sleep and diet are four distinct but intertwined lifestyle factors that influence the cardiovascular system. Abundant epidemiological, clinical and preclinical studies have underscored the importance of managing stress, having good sleep hygiene and responsible eating habits and exercising regularly. We are born with a genetic blueprint that can protect us against or predispose us to a particular disease. However, lifestyle factors build upon and profoundly influence those predispositions. Studies in the past 10 years have shown that the immune system in general and leukocytes in particular are particularly susceptible to environmental perturbations. Lifestyle factors such as stress, sleep, diet and exercise affect leukocyte behaviour and function and thus the immune system at large. In this Review, we explore the various mechanisms by which lifestyle factors modulate haematopoiesis and leukocyte migration and function in the context of cardiovascular health. We pay particular attention to the role of the nervous system as the key executor that connects environmental influences to leukocyte behaviour.
Collapse
Affiliation(s)
- Henrike Janssen
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura L Koekkoek
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Filip K Swirski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
29
|
Zhang T, Pang C, Xu M, Zhao Q, Hu Z, Jiang X, Guo M. The role of immune system in atherosclerosis: Molecular mechanisms, controversies, and future possibilities. Hum Immunol 2024; 85:110765. [PMID: 38369442 DOI: 10.1016/j.humimm.2024.110765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/20/2024]
Abstract
Numerous cardiovascular disorders have atherosclerosis as their pathological underpinning. Numerous studies have demonstrated that, with the aid of pattern recognition receptors, cytokines, and immunoglobulins, innate immunity, represented by monocytes/macrophages, and adaptive immunity, primarily T/B cells, play a critical role in controlling inflammation and abnormal lipid metabolism in atherosclerosis. Additionally, the finding of numerous complement components in atherosclerotic plaques suggests yet again how heavily the immune system controls atherosclerosis. Therefore, it is essential to have a thorough grasp of how the immune system contributes to atherosclerosis. The specific molecular mechanisms involved in the activation of immune cells and immune molecules in atherosclerosis, the controversy surrounding some immune cells in atherosclerosis, and the limitations of extrapolating from relevant animal models to humans were all carefully reviewed in this review from the three perspectives of innate immunity, adaptive immunity, and complement system. This could provide fresh possibilities for atherosclerosis research and treatment in the future.
Collapse
Affiliation(s)
- Tianle Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Chenxu Pang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Mengxin Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Qianqian Zhao
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhijie Hu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| |
Collapse
|
30
|
Robichaud S, Rochon V, Emerton C, Laval T, Ouimet M. Trehalose promotes atherosclerosis regression in female mice. Front Cardiovasc Med 2024; 11:1298014. [PMID: 38433753 PMCID: PMC10906268 DOI: 10.3389/fcvm.2024.1298014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/31/2024] [Indexed: 03/05/2024] Open
Abstract
Introduction Atherosclerosis is a chronic inflammatory disease caused by the deposition of lipids within the artery wall. During atherogenesis, efficient autophagy is needed to facilitate efferocytosis and cholesterol efflux, limit inflammation and lipid droplet buildup, and eliminate defective mitochondria and protein aggregates. Central to the regulation of autophagy is the transcription factor EB (TFEB), which coordinates the expression of lysosomal biogenesis and autophagy genes. In recent years, trehalose has been shown to promote TFEB activation and protect against atherogenesis. Here, we sought to investigate the role of autophagy activation during atherosclerosis regression. Methods and results Atherosclerosis was established in C57BL/6N mice by injecting AAV-PCSK9 and 16 weeks of Western diet feeding, followed by switching to a chow diet to induce atherosclerosis regression. During the regression period, mice were either injected with trehalose concomitant with trehalose supplementation in their drinking water or injected with saline for 6 weeks. Female mice receiving trehalose had reduced atherosclerosis burden, as evidenced by reduced plaque lipid content, macrophage numbers and IL-1β content in parallel with increased plaque collagen deposition, which was not observed in their male counterparts. In addition, trehalose-treated female mice had lower levels of circulating leukocytes, including inflammatory monocytes and CD4+ T cells. Lastly, we found that autophagy flux in male mice was basally higher than in female mice during atherosclerosis progression. Conclusions Our data demonstrate a sex-specific effect of trehalose in atherosclerosis regression, whereby trehalose reduced lipid content, inflammation, and increased collagen content in female mice but not in male mice. Furthermore, we discovered inherent differences in the autophagy flux capacities between the sexes: female mice exhibited lower plaque autophagy than males, which rendered the female mice more responsive to atherosclerosis regression. Our work highlights the importance of understanding sex differences in atherosclerosis to personalize the development of future therapies to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Sabrina Robichaud
- Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Cardiovascular Metabolism and Cell Biology Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Valérie Rochon
- Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Cardiovascular Metabolism and Cell Biology Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Christina Emerton
- Cardiovascular Metabolism and Cell Biology Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Thomas Laval
- Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Cardiovascular Metabolism and Cell Biology Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Mireille Ouimet
- Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Cardiovascular Metabolism and Cell Biology Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
| |
Collapse
|
31
|
Schelemei P, Wagner E, Picard FSR, Winkels H. Macrophage mediators and mechanisms in cardiovascular disease. FASEB J 2024; 38:e23424. [PMID: 38275140 DOI: 10.1096/fj.202302001r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/27/2024]
Abstract
Macrophages are major players in myocardial infarction (MI) and atherosclerosis, two major cardiovascular diseases (CVD). Atherosclerosis is caused by the buildup of cholesterol-rich lipoproteins in blood vessels, causing inflammation, vascular injury, and plaque formation. Plaque rupture or erosion can cause thrombus formation resulting in inadequate blood flow to the heart muscle and MI. Inflammation, particularly driven by macrophages, plays a central role in both atherosclerosis and MI. Recent integrative approaches of single-cell analysis-based classifications in both murine and human atherosclerosis as well as experimental MI showed overlap in origin, diversity, and function of macrophages in the aorta and the heart. We here discuss differences and communalities between macrophages in the heart and aorta at steady state and in atherosclerosis or upon MI. We focus on markers, mediators, and functional states of macrophage subpopulations. Recent trials testing anti-inflammatory agents show a major benefit in reducing the inflammatory burden of CVD patients, but highlight a necessity for a broader understanding of immune cell ontogeny and heterogeneity in CVD. The novel insights into macrophage biology in CVD represent exciting opportunities for the development of novel treatment strategies against CVD.
Collapse
Affiliation(s)
- Patrik Schelemei
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Elena Wagner
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Felix Simon Ruben Picard
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Holger Winkels
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
32
|
Song YJ, Ma Y, Meng T, Zhuang T, Ruan CC, Li Y, Zhang GN. The Characteristics of Macrophage Heterogeneity in Atherosclerotic Aortas. J Cardiovasc Transl Res 2024; 17:153-166. [PMID: 37713049 DOI: 10.1007/s12265-023-10434-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023]
Abstract
Macrophage is the main effector cell during atherosclerosis. We applied single-cell RNA sequencing (scRNA) data to investigate the role of macrophage subsets in atherosclerosis. Monocyte and macrophage clusters were divided into 6 subclusters. Each subcluster's markers were calculated and validated by immunofluorescence. Elevated macrophage subclusters in the WD group were subject to enrichment pathway analysis and exhibited different phenotypes. Pseudotime analysis shows the subclusters originate from monocytes. We cultured bone marrow-derived macrophages with CSF-1 and ox-LDL to simulate an atherosclerotic-like environment and detected the transformation of subclusters. Macrophage-Vegfa and Macrophage-C1qb increased in the WD group. Macrophage-Vegfa acquires the characteristics of phagocytosis and immune response, while Macrophage-C1qb is not involved in lipid metabolism. The two subclusters are both enriched in cell movement and migration pathways. Experimental verification proved Monocyte-Ly6C evolved into Macrophage-Vegfa and Macrophage-C1qb during atherosclerosis progression.
Collapse
Affiliation(s)
- Yu-Jie Song
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Ma
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Meng
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Tao Zhuang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Cheng-Chao Ruan
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Yan Li
- Department of Cardiology, RuiJin Hospital/LuWan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Guan-Nan Zhang
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
33
|
Liu M, Chen X, Gu Z, He H, Chen M, Kuai L, Jia Z, Li Y, Chen Y, Hong M, Xiao F. Predictive Value of CFIm25 Expression in Peripheral Blood Monocytes for Coronary Atherosclerosis. Int J Med Sci 2024; 21:562-570. [PMID: 38322593 PMCID: PMC10845263 DOI: 10.7150/ijms.91148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/01/2024] [Indexed: 02/08/2024] Open
Abstract
Background: Cleavage factor Im25 (CFIm25) regulates cell function by affecting mRNA editing processes and plays diverse roles in various diseases. Studies have found that peripheral blood monocytes are valuable in diagnosing and prognosing coronary atherosclerosis. However, no studies have examined the predictive value of CFIm25 expression in peripheral blood monocytes for coronary atherosclerosis. Methods and Results: We collected the coronary angiography results of 267 patients and calculated the Gensini score to evaluate their degree of coronary atherosclerosis. We isolated peripheral blood monocytes and detected CFIm25 RNA expression. Based on their Gensini score, we divided the patients into negative (0, n = 46), mild lesion (≤ 8, n = 71), moderate lesion (8-23, n = 76), and severe lesion (≥ 23, n = 74) groups. Results showed that CFIm25 expression correlated negatively with the Gensini score and the number of involved coronary vessels. Univariate and multivariate binary logistic regression analyses showed that CFIm25 expression in peripheral blood monocytes was a protective factor for severe lesions, ≥ 50% stenosis, and three-vessel lesions. The areas under the receiver operating characteristic curve of CFIm25 expression for predicting lesions, severe lesions, ≥50% stenosis, and three-vessel lesions were 0.743, 0.735, 0.791, and 0.736, respectively. Conclusions: CFIm25 expression in peripheral blood monocytes correlates negatively with the degree of coronary atherosclerosis and helps predict the severity and number of coronary artery lesions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Mei Hong
- Department of Cardiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Fangping Xiao
- Department of Cardiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| |
Collapse
|
34
|
Govindarajah V, Sakabe M, Good S, Solomon M, Arasu A, Chen N, Zhang X, Grimes HL, Kendler A, Xin M, Reynaud D. Gestational diabetes in mice induces hematopoietic memory that affects the long-term health of the offspring. J Clin Invest 2024; 134:e169730. [PMID: 37988162 PMCID: PMC10786695 DOI: 10.1172/jci169730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023] Open
Abstract
Gestational diabetes is a common medical complication of pregnancy that is associated with adverse perinatal outcomes and an increased risk of metabolic diseases and atherosclerosis in adult offspring. The mechanisms responsible for this delayed pathological transmission remain unknown. In mouse models, we found that the development of atherosclerosis in adult offspring born to diabetic pregnancy can be in part linked to hematopoietic alterations. Although they do not show any gross metabolic disruptions, the adult offspring maintain hematopoietic features associated with diabetes, indicating the acquisition of a lasting diabetic hematopoietic memory. We show that the induction of this hematopoietic memory during gestation relies on the activity of the advanced glycation end product receptor (AGER) and the nucleotide binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, which lead to increased placental inflammation. In adult offspring, we find that this memory is associated with DNA methyltransferase 1 (DNMT1) upregulation and epigenetic changes in hematopoietic progenitors. Together, our results demonstrate that the hematopoietic system can acquire a lasting memory of gestational diabetes and that this memory constitutes a pathway connecting gestational health to adult pathologies.
Collapse
Affiliation(s)
| | | | - Samantha Good
- Division of Experimental Hematology and Cancer Biology and
| | | | - Ashok Arasu
- Division of Experimental Hematology and Cancer Biology and
| | - Nong Chen
- Division of Experimental Hematology and Cancer Biology and
| | - Xuan Zhang
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - H. Leighton Grimes
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
- Department of Pediatrics and
| | - Ady Kendler
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Mei Xin
- Division of Experimental Hematology and Cancer Biology and
- Department of Pediatrics and
| | - Damien Reynaud
- Division of Experimental Hematology and Cancer Biology and
- Department of Pediatrics and
| |
Collapse
|
35
|
Christ A, Maas SL, Jin H, Lu C, Legein B, Wijnands E, Temmerman L, Otten J, Isaacs A, Zenke M, Stoll M, Biessen EAL, van der Vorst EPC. In situ lipid-loading activates peripheral dendritic cell subsets characterized by cellular ROS accumulation but compromises their capacity to prime naïve T cells. Free Radic Biol Med 2024; 210:406-415. [PMID: 38061606 DOI: 10.1016/j.freeradbiomed.2023.11.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/22/2023]
Abstract
BACKGROUND AND AIMS Dendritic cells (DCs), professional antigen-presenting cells, play an important role in pathologies by controlling adaptive immune responses. However, their adaptation to and functionality in hypercholesterolemia, a driving factor in disease onset and progression of atherosclerosis remains to be established. METHODS In this study, we addressed the immediate impact of high fat diet-induced hypercholesterolemia in low-density lipoprotein receptor deficient (Ldlr-/-) mice on separate DC subsets, their compartmentalization and functionality. RESULTS While hypercholesterolemia induced a significant rise in bone marrow myeloid and dendritic cell progenitor (MDP) frequency and proliferation rate after high fat diet feeding, it did not affect DC subset numbers in lymphoid tissue. Hypercholesterolemia led to almost immediate and persistent augmentation in granularity of conventional DCs (cDCs), in particular cDC2, reflecting progressive lipid accumulation by these subsets. Plasmacytoid DCs were only marginally and transiently affected. Lipid loading increased co-stimulatory molecule expression and ROS accumulation by cDC2. Despite this hyperactivation, lipid-laden cDC2 displayed a profoundly reduced capacity to stimulate naïve CD4+ T cells. CONCLUSION Our data provide evidence that in hypercholesterolemic conditions, peripheral cDC2 subsets engulf lipids in situ, leading to a more activated status characterized by cellular ROS accumulation while, paradoxically, compromising their T cell priming ability. These findings will have repercussions not only for lipid driven cardiometabolic disorders like atherosclerosis, but also for adaptive immune responses to pathogens and/or endogenous (neo) antigens under conditions of hyperlipidemia.
Collapse
Affiliation(s)
- Anette Christ
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands; Health Office Frankfurt/Main, Frankfurt/Main, Germany.
| | - Sanne L Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany; Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
| | - Han Jin
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Chang Lu
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Bart Legein
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Erwin Wijnands
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Lieve Temmerman
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Jeroen Otten
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Aaron Isaacs
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Martin Zenke
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, Aachen, Germany; Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany; Department of Hematology, Oncology and Stem Cell Transplantation, RWTH Aachen University Medical School, 52074, Aachen, Germany
| | - Monika Stoll
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands; Genetic Epidemiology, Institute for Human Genetics, Westfälische Wilhelms-University, Münster, Germany
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands; Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| | - Emiel P C van der Vorst
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands; Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany; Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
36
|
Robinson KA, Akbar N, Baidžajevas K, Choudhury RP. Trained immunity in diabetes and hyperlipidemia: Emerging opportunities to target cardiovascular complications and design new therapies. FASEB J 2023; 37:e23231. [PMID: 37779347 PMCID: PMC10947360 DOI: 10.1096/fj.202301078r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023]
Abstract
Some metabolic diseases, such as diabetes and hyperlipidemia, are associated with a state of inflammation, which adversely affects cardiovascular health. Emerging evidence suggests that long-term hyperactivation of innate immune cells and their bone marrow progenitors, termed trained immunity, functions to accelerate atherosclerosis and its complications in cardiometabolic diseases. This review will focus on how trained immunity is established, particularly through metabolic and epigenetic reprogramming, to cause persistent and deleterious changes in immune cell function, even after the original stimulus has been corrected or removed. Understanding the mechanisms driving maladaptive trained immunity and its fundamental contribution to cardiovascular disease might enable the development of novel disease-modifying therapeutics for the reduction in cardiovascular risk in diabetes, hyperlipidemia, and related cardiometabolic states.
Collapse
Affiliation(s)
- Katherine A. Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Kajus Baidžajevas
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Robin P. Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
37
|
Hou P, Fang J, Liu Z, Shi Y, Agostini M, Bernassola F, Bove P, Candi E, Rovella V, Sica G, Sun Q, Wang Y, Scimeca M, Federici M, Mauriello A, Melino G. Macrophage polarization and metabolism in atherosclerosis. Cell Death Dis 2023; 14:691. [PMID: 37863894 PMCID: PMC10589261 DOI: 10.1038/s41419-023-06206-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/22/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of fatty deposits in the inner walls of vessels. These plaques restrict blood flow and lead to complications such as heart attack or stroke. The development of atherosclerosis is influenced by a variety of factors, including age, genetics, lifestyle, and underlying health conditions such as high blood pressure or diabetes. Atherosclerotic plaques in stable form are characterized by slow growth, which leads to luminal stenosis, with low embolic potential or in unstable form, which contributes to high risk for thrombotic and embolic complications with rapid clinical onset. In this complex scenario of atherosclerosis, macrophages participate in the whole process, including the initiation, growth and eventually rupture and wound healing stages of artery plaque formation. Macrophages in plaques exhibit high heterogeneity and plasticity, which affect the evolving plaque microenvironment, e.g., leading to excessive lipid accumulation, cytokine hyperactivation, hypoxia, apoptosis and necroptosis. The metabolic and functional transitions of plaque macrophages in response to plaque microenvironmental factors not only influence ongoing and imminent inflammatory responses within the lesions but also directly dictate atherosclerotic progression or regression. In this review, we discuss the origin of macrophages within plaques, their phenotypic diversity, metabolic shifts, and fate and the roles they play in the dynamic progression of atherosclerosis. It also describes how macrophages interact with other plaque cells, particularly T cells. Ultimately, targeting pathways involved in macrophage polarization may lead to innovative and promising approaches for precision medicine. Further insights into the landscape and biological features of macrophages within atherosclerotic plaques may offer valuable information for optimizing future clinical treatment for atherosclerosis by targeting macrophages.
Collapse
Affiliation(s)
- Pengbo Hou
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jiankai Fang
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Zhanhong Liu
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Massimiliano Agostini
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Pierluigi Bove
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Valentina Rovella
- Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Sica
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Qiang Sun
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Ying Wang
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Manuel Scimeca
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Massimo Federici
- Department of System Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy.
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
38
|
Li X, Li Z, Cai D, Li Y, Zhu Y, Jiao R, Lai C, Sun J, Bai W. Vitisin A, as a Type of Pyranoanthocyanin, Suppresses Inflammation by Restricting Hematopoietic Stem Cell Differentiation toward Monocytes in Bone Marrow. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15048-15063. [PMID: 37811833 DOI: 10.1021/acs.jafc.3c03119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) could be differentiated into mature myeloid and lymphoid cells, maintaining the requirements of immune cells. Atherosclerosis and ulcerative colitis (UC) drive HSPC homeostasis destruction, which triggers expansive HSPC proliferation and Ly6Chi monocyte production, contributing to aggravated inflammation. Vitisin A belongs to the anthocyanin derivatives with excellent stability and bioactivity in vitro. However, there is no report about the anti-inflammation of Vitisin A via reprogramming HSPC differentiation toward monocytes. In this study, we found that Vitisin A presents anti-inflammatory ability during the development of atherosclerosis and UC by depressing Ly6Chi monocyte production from bone marrow. This performance depended on restricted HSPC differentiation, which suggested that Vitisin A participated in monocyte generation and carried out the immunomodulation. Together, Vitisin A ameliorates inflammation during atherosclerosis and UC via the suppressed differentiation of HSPCs toward monocytes, which could be considered an ideal functional component with immunomodulatory effects.
Collapse
Affiliation(s)
- Xusheng Li
- The Sixth Affiliated Hospital, Jinan University, Dongguan 523576, P. R. China
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, P. R. China
| | - Zhenhua Li
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, P. R. China
| | - Dongbao Cai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, P. R. China
| | - Yawen Li
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, P. R. China
| | - Yuanqin Zhu
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, P. R. China
| | - Rui Jiao
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, P. R. China
| | - Caiyong Lai
- The Sixth Affiliated Hospital, Jinan University, Dongguan 523576, P. R. China
- Department of Urology, Institute of Kidney Surgery, The First Affiliated Hospital, Jinan University, Guangzhou 510632, P. R. China
| | - Jianxia Sun
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, P. R. China
| | - Weibin Bai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, P. R. China
| |
Collapse
|
39
|
Prakhar P, Bhatt B, Lohia GK, Shah A, Mukherjee T, Kolthur-Seetharam U, Sundaresan NR, Rajmani RS, Balaji KN. G9a and Sirtuin6 epigenetically modulate host cholesterol accumulation to facilitate mycobacterial survival. PLoS Pathog 2023; 19:e1011731. [PMID: 37871034 PMCID: PMC10621959 DOI: 10.1371/journal.ppat.1011731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/02/2023] [Accepted: 10/02/2023] [Indexed: 10/25/2023] Open
Abstract
Cholesterol derived from the host milieu forms a critical factor for mycobacterial pathogenesis. However, the molecular circuitry co-opted by Mycobacterium tuberculosis (Mtb) to accumulate cholesterol in host cells remains obscure. Here, we report that the coordinated action of WNT-responsive histone modifiers G9a (H3K9 methyltransferase) and SIRT6 (H3K9 deacetylase) orchestrate cholesterol build-up in in vitro and in vivo mouse models of Mtb infection. Mechanistically, G9a, along with SREBP2, drives the expression of cholesterol biosynthesis and uptake genes; while SIRT6 along with G9a represses the genes involved in cholesterol efflux. The accumulated cholesterol in Mtb infected macrophages promotes the expression of antioxidant genes leading to reduced oxidative stress, thereby supporting Mtb survival. In corroboration, loss-of-function of G9a in vitro and pharmacological inhibition in vivo; or utilization of BMDMs derived from Sirt6-/- mice or in vivo infection in haplo-insufficient Sirt6-/+ mice; hampered host cholesterol accumulation and restricted Mtb burden. These findings shed light on the novel roles of G9a and SIRT6 during Mtb infection and highlight the previously unknown contribution of host cholesterol in potentiating anti-oxidative responses for aiding Mtb survival.
Collapse
Affiliation(s)
- Praveen Prakhar
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Bharat Bhatt
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Gaurav Kumar Lohia
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Awantika Shah
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Tanushree Mukherjee
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Ullas Kolthur-Seetharam
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra, India
| | - Nagalingam R. Sundaresan
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Raju S. Rajmani
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore–, Karnataka, India
| | | |
Collapse
|
40
|
Csoma B, Bikov A. The Role of the Circadian Rhythm in Dyslipidaemia and Vascular Inflammation Leading to Atherosclerosis. Int J Mol Sci 2023; 24:14145. [PMID: 37762448 PMCID: PMC10532147 DOI: 10.3390/ijms241814145] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiovascular diseases (CVD) are among the leading causes of death worldwide. Many lines of evidence suggest that the disturbances in circadian rhythm are responsible for the development of CVDs; however, circadian misalignment is not yet a treatable trait in clinical practice. The circadian rhythm is controlled by the central clock located in the suprachiasmatic nucleus and clock genes (molecular clock) located in all cells. Dyslipidaemia and vascular inflammation are two hallmarks of atherosclerosis and numerous experimental studies conclude that they are under direct influence by both central and molecular clocks. This review will summarise the results of experimental studies on lipid metabolism, vascular inflammation and circadian rhythm, and translate them into the pathophysiology of atherosclerosis and cardiovascular disease. We discuss the effect of time-respected administration of medications in cardiovascular medicine. We review the evidence on the effect of bright light and melatonin on cardiovascular health, lipid metabolism and vascular inflammation. Finally, we suggest an agenda for future research and recommend on clinical practice.
Collapse
Affiliation(s)
- Balazs Csoma
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester M23 9LT, UK;
- Department of Pulmonology, Semmelweis University, 1083 Budapest, Hungary
| | - Andras Bikov
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester M23 9LT, UK;
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
41
|
Rauch PJ, Gopakumar J, Silver AJ, Nachun D, Ahmad H, McConkey M, Nakao T, Bosse M, Rentz T, Vivanco Gonzalez N, Greenwald NF, McCaffrey EF, Khair Z, Gopakumar M, Rodrigues KB, Lin AE, Sinha E, Fefer M, Cohen DN, Vromman A, Shvartz E, Sukhova G, Bendall S, Angelo M, Libby P, Ebert BL, Jaiswal S. Loss-of-function mutations in Dnmt3a and Tet2 lead to accelerated atherosclerosis and concordant macrophage phenotypes. NATURE CARDIOVASCULAR RESEARCH 2023; 2:805-818. [PMID: 39196062 DOI: 10.1038/s44161-023-00326-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 07/27/2023] [Indexed: 08/29/2024]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is defined by the presence of a cancer-associated somatic mutation in white blood cells in the absence of overt hematological malignancy. It arises most commonly from loss-of-function mutations in the epigenetic regulators DNMT3A and TET2. CHIP predisposes to both hematological malignancies and atherosclerotic cardiovascular disease in humans. Here we demonstrate that loss of Dnmt3a in myeloid cells increased murine atherosclerosis to a similar degree as previously seen with loss of Tet2. Loss of Dnmt3a enhanced inflammation in macrophages in vitro and generated a distinct adventitial macrophage population in vivo which merges a resident macrophage profile with an inflammatory cytokine signature. These changes surprisingly phenocopy the effect of loss of Tet2. Our results identify a common pathway promoting heightened innate immune cell activation with loss of either gene, providing a biological basis for the excess atherosclerotic disease burden in carriers of these two most prevalent CHIP mutations.
Collapse
Affiliation(s)
- Philipp J Rauch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | | | - Alexander J Silver
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel Nachun
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Herra Ahmad
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Marie McConkey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Tetsushi Nakao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Marc Bosse
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thiago Rentz
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Noah F Greenwald
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Erin F McCaffrey
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Zumana Khair
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Manu Gopakumar
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Kameron B Rodrigues
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Amy E Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Eti Sinha
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Maia Fefer
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Drew N Cohen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amélie Vromman
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Eugenia Shvartz
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Galina Sukhova
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Sean Bendall
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Siddhartha Jaiswal
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
42
|
Choi SY, Lee EB, Kim JH, Lee JR. Over-Expression of p190RhoGEF Regulates the Formation of Atherosclerotic Plaques in the Aorta of ApoE -/- Mice via Macrophage Polarization. Int J Mol Sci 2023; 24:12785. [PMID: 37628966 PMCID: PMC10454661 DOI: 10.3390/ijms241612785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
The RhoA-specific guanine nucleotide exchange factor p190RhoGEF has been implicated in the control of cell morphology, focal adhesion formation, and cell motility. Previously, we reported that p190RhoGEF is also active in various immune cells. In this study, we examined whether over-expression of p190RhoGEF could affect atherosclerotic plaque formation in mouse aortae. For that purpose, transgenic (TG) mice over-expressing p190RhoGEF were cross-bred with atherosclerosis-prone apolipoprotein E (ApoE)-/- mice to obtain p190RhoGEF-TG mice with ApoE-/- backgrounds (TG/ApoE-/-). Aortic plaque formation was significantly increased in TG/ApoE mice-/- at 30 to 40 weeks of age compared to that in ApoE-/- mice. Serum concentrations of inflammatory cytokines (IL-6 and TNF-α) were greater in TG/ApoE-/- mice than in ApoE-/- mice at ~40 weeks of age. Furthermore, TG/ApoE-/- mice had a greater proportion of peritoneal macrophages within the M1 subset at 30 to 40 weeks of age, together with higher production of inflammatory cytokines and stronger responses to bacterial lipopolysaccharide than ApoE-/- mice. Collectively, these results highlight a crucial role of enhanced p190RhoGEF expression in atherosclerosis progression, including the activation of pro-inflammatory M1 macrophages.
Collapse
Affiliation(s)
- So-Yeon Choi
- Department of Bioinspired Science, The Graduate School, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Eun-Bi Lee
- Department of Bioinspired Science, The Graduate School, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jee-Hae Kim
- Department of Life Science, College of Natural Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jong Ran Lee
- Department of Life Science, College of Natural Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
43
|
Lee AG, Kang S, Yoon HJ, Im S, Oh SJ, Pak YK. Polystyrene Microplastics Exacerbate Systemic Inflammation in High-Fat Diet-Induced Obesity. Int J Mol Sci 2023; 24:12421. [PMID: 37569796 PMCID: PMC10419071 DOI: 10.3390/ijms241512421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Microplastics (MPs) are recognized as environmental pollutants with potential implications for human health. Considering the rapid increase in obesity rates despite stable caloric intake, there is a growing concern about the link between obesity and exposure to environmental pollutants, including MPs. In this study, we conducted a comprehensive investigation utilizing in silico, in vitro, and in vivo approaches to explore the brain distribution and physiological effects of MPs. Molecular docking simulations were performed to assess the binding affinity of three plastic polymers (ethylene, propylene, and styrene) to immune cells (macrophages, CD4+, and CD8+ lymphocytes). The results revealed that styrene exhibited the highest binding affinity for macrophages. Furthermore, in vitro experiments employing fluorescence-labeled PS-MPs (fPS-MPs) of 1 μm at various concentrations demonstrated a dose-dependent binding of fPS-MPs to BV2 murine microglial cells. Subsequent oral administration of fPS-MPs to high-fat diet-induced obese mice led to the co-existence of fPS-MPs with immune cells in the blood, exacerbating impaired glucose metabolism and insulin resistance and promoting systemic inflammation. Additionally, fPS-MPs were detected throughout the brain, with increased activation of microglia in the hypothalamus. These findings suggest that PS-MPs significantly contribute to the exacerbation of systemic inflammation in high-fat diet-induced obesity by activating peripheral and central inflammatory immune cells.
Collapse
Affiliation(s)
| | - Sora Kang
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
| | - Hye Ji Yoon
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
| | - Suyeol Im
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447,Republic of Korea; (S.I.); (S.J.O.)
| | - Seung Jun Oh
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447,Republic of Korea; (S.I.); (S.J.O.)
| | - Youngmi Kim Pak
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447,Republic of Korea; (S.I.); (S.J.O.)
- Department of Physiology, School of Medicine, Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
44
|
Dai D, Zhu Z, Han H, Xu T, Feng S, Zhang W, Ding F, Zhang R, Zhu J. Enhanced tyrosine sulfation is associated with chronic kidney disease-related atherosclerosis. BMC Biol 2023; 21:151. [PMID: 37424015 DOI: 10.1186/s12915-023-01641-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 06/02/2023] [Indexed: 07/11/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) accelerates atherosclerosis, but the mechanisms remain unclear. Tyrosine sulfation has been recognized as a key post-translational modification (PTM) in regulation of various cellular processes, and the sulfated adhesion molecules and chemokine receptors have been shown to participate in the pathogenesis of atherosclerosis via enhancement of monocyte/macrophage function. The levels of inorganic sulfate, the essential substrate for the sulfation reaction, are dramatically increased in patients with CKD, which indicates a change of sulfation status in CKD patients. Thus, in the present study, we detected the sulfation status in CKD patients and probed into the impact of sulfation on CKD-related atherosclerosis by targeting tyrosine sulfation function. RESULTS PBMCs from individuals with CKD showed higher amounts of total sulfotyrosine and tyrosylprotein sulfotransferase (TPST) type 1 and 2 protein levels. The plasma level of O-sulfotyrosine, the metabolic end product of tyrosine sulfation, increased significantly in CKD patients. Statistically, O-sulfotyrosine and the coronary atherosclerosis severity SYNTAX score positively correlated. Mechanically, more sulfate-positive nucleated cells in peripheral blood and more abundant infiltration of sulfated macrophages in deteriorated vascular plaques in CKD ApoE null mice were noted. Knockout of TPST1 and TPST2 decreased atherosclerosis and peritoneal macrophage adherence and migration in CKD condition. The sulfation of the chemokine receptors, CCR2 and CCR5, was increased in PBMCs from CKD patients. CONCLUSIONS CKD is associated with increased sulfation status. Increased sulfation contributes to monocyte/macrophage activation and might be involved in CKD-related atherosclerosis. Inhibition of sulfation may suppress CKD-related atherosclerosis and is worthy of further study.
Collapse
Affiliation(s)
- Daopeng Dai
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Shanghai, 200025, China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengbin Zhu
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Shanghai, 200025, China
| | - Hui Han
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Shanghai, 200025, China
| | - Tian Xu
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Feng
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenli Zhang
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Shanghai, 200025, China
| | - Fenghua Ding
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Shanghai, 200025, China
| | - Ruiyan Zhang
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Shanghai, 200025, China.
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jinzhou Zhu
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Shanghai, 200025, China.
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
45
|
DeBerge M, Chaudhary R, Schroth S, Thorp EB. Immunometabolism at the Heart of Cardiovascular Disease. JACC Basic Transl Sci 2023; 8:884-904. [PMID: 37547069 PMCID: PMC10401297 DOI: 10.1016/j.jacbts.2022.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 08/08/2023]
Abstract
Immune cell function among the myocardium, now more than ever, is appreciated to regulate cardiac function and pathophysiology. This is the case for both innate immunity, which includes neutrophils, monocytes, dendritic cells, and macrophages, as well as adaptive immunity, which includes T cells and B cells. This function is fueled by cell-intrinsic shifts in metabolism, such as glycolysis and oxidative phosphorylation, as well as metabolite availability, which originates from the surrounding extracellular milieu and varies during ischemia and metabolic syndrome. Immune cell crosstalk with cardiac parenchymal cells, such as cardiomyocytes and fibroblasts, is also regulated by complex cellular metabolic circuits. Although our understanding of immunometabolism has advanced rapidly over the past decade, in part through valuable insights made in cultured cells, there remains much to learn about contributions of in vivo immunometabolism and directly within the myocardium. Insight into such fundamental cell and molecular mechanisms holds potential to inform interventions that shift the balance of immunometabolism from maladaptive to cardioprotective and potentially even regenerative. Herein, we review our current working understanding of immunometabolism, specifically in the settings of sterile ischemic cardiac injury or cardiometabolic disease, both of which contribute to the onset of heart failure. We also discuss current gaps in knowledge in this context and therapeutic implications.
Collapse
Affiliation(s)
| | | | - Samantha Schroth
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
46
|
Hsu CC, Fidler TP, Kanter JE, Kothari V, Kramer F, Tang J, Tall AR, Bornfeldt KE. Hematopoietic NLRP3 and AIM2 Inflammasomes Promote Diabetes-Accelerated Atherosclerosis, but Increased Necrosis Is Independent of Pyroptosis. Diabetes 2023; 72:999-1011. [PMID: 37083999 PMCID: PMC10281813 DOI: 10.2337/db22-0962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/15/2023] [Indexed: 04/22/2023]
Abstract
Serum apolipoprotein C3 (APOC3) predicts incident cardiovascular events in people with type 1 diabetes, and silencing of APOC3 prevents both lesion initiation and advanced lesion necrotic core expansion in a mouse model of type 1 diabetes. APOC3 acts by slowing the clearance of triglyceride-rich lipoproteins, but lipid-free APOC3 has recently been reported to activate an inflammasome pathway in monocytes. We therefore investigated the contribution of hematopoietic inflammasome pathways to atherosclerosis in mouse models of type 1 diabetes. LDL receptor-deficient diabetes mouse models were transplanted with bone marrow from donors deficient in NOD, LRR and pyrin domain-containing protein 3 (NLRP3), absent in melanoma 2 (AIM2) or gasdermin D (GSDMD), an inflammasome-induced executor of pyroptotic cell death. Mice with diabetes exhibited inflammasome activation and consistently, increased plasma interleukin-1β (IL-1β) and IL-18. Hematopoietic deletions of NLRP3, AIM2, or GSDMD caused smaller atherosclerotic lesions in diabetic mice. The increased lesion necrotic core size in diabetic mice was independent of macrophage pyroptosis because hematopoietic GSDMD deficiency failed to prevent necrotic core expansion in advanced lesions. Our findings demonstrate that AIM2 and NLRP3 inflammasomes contribute to atherogenesis in diabetes and suggest that necrotic core expansion is independent of macrophage pyroptosis. ARTICLE HIGHLIGHTS The contribution of hematopoietic cell inflammasome activation to atherosclerosis associated with type 1 diabetes is unknown. The goal of this study was to address whether hematopoietic NOD, LRR, and pyrin domain-containing protein 3 (NLRP3), absent in melanoma 2 (AIM2) inflammasomes, or the pyroptosis executioner gasdermin D (GSDMD) contributes to atherosclerosis in mouse models of type 1 diabetes. Diabetic mice exhibited increased inflammasome activation, with hematopoietic deletions of NLRP3, AIM2, or GSDMD causing smaller atherosclerotic lesions in diabetic mice, but the increased lesion necrotic core size in diabetic mice was independent of macrophage pyroptosis. Further studies on whether inflammasome activation contributes to cardiovascular complications in people with type 1 diabetes are warranted.
Collapse
Affiliation(s)
- Cheng-Chieh Hsu
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Trevor P. Fidler
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Jenny E. Kanter
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Vishal Kothari
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Farah Kramer
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Jingjing Tang
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Alan R. Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Karin E. Bornfeldt
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA
| |
Collapse
|
47
|
Abate E, Degef M, Melkie A, Gnanasekeran N, Mehdi M, Tolcha Y, Chala D. Haematological Parameters in People with Atherosclerotic Cardiovascular Disease versus Those Who are Only at Risk for Cardiovascular Disease: A Comparative Cross-Sectional Study. Diabetes Metab Syndr Obes 2023; 16:1869-1883. [PMID: 37384132 PMCID: PMC10296606 DOI: 10.2147/dmso.s407480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023] Open
Abstract
Introduction Cardiovascular disease (CVD) is the foremost killer disease worldwide. ASCVD is one of the most common types of CVD. It is mainly associated with a condition called atherosclerosis. Its occurrence is linked to several risk factors. Hypertension, diabetes, dyslipidemia, smoking, genetic factors, and so on are examples. The presence of ASCVD, as well as its risk factors, causes a variety of disruptions in the body's physiological and biological functions. The presence of abnormal physiological and biological functions, for example, tends to disrupt hematological parameters. Purpose The study's aim was to assess and compare the pattern of hematological parameters in people with established atherosclerotic cardiovascular disease (ASVD) versus people with ASCVD risks alone who attended TASH Addis Ababa, Ethiopia, as well as to correlate hematological parameters with the novel inflammatory marker hs-CRP. Methods A prospective cross-sectional comparative study with 100 study participants was conducted during where October 2019-March 2020 proposal development, sample collection, and lab analysis period, and from March 2020-June to 2021 data entry, analysis, and writing period. A serum sample was collected from each study participant for the lipid and hsCRP analyses and whole blood for hematological parameter determination. The socio-demographic characteristics of the study participants were obtained through a well-structured questionnaire. Results The ASCVD-risk group had significantly higher mean platelet volume (MPV), which was associated with the presence of the risk. Furthermore, hs-CRPs show a significant correlation with MPV in a correlation analysis of highly sensitive C-reactive protein (hs-CRP) with hematological parameters. Thus, using these affordable, routinely tested, and easily available tests may help to infer future ASCVD risk as well as the presence of ASCVD morbidity while hsCRP level in comparison group vs cases requires further study.
Collapse
Affiliation(s)
- Ebsitu Abate
- Department of Medical Biochemistry Addis Ababa University, Addis Ababa, Ethiopia
| | - Maria Degef
- Department of Medical Biochemistry Addis Ababa University, Addis Ababa, Ethiopia
| | - Addisu Melkie
- Department of Internal Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Natesan Gnanasekeran
- Department of Medical Biochemistry Addis Ababa University, Addis Ababa, Ethiopia
| | - Mohammed Mehdi
- Department of Medical Biochemistry Addis Ababa University, Addis Ababa, Ethiopia
| | - Yosef Tolcha
- Department of Diagnostic Laboratory, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Dawit Chala
- Department of Diagnostic Laboratory, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| |
Collapse
|
48
|
Abe JI, Imanishi M, Li S, Zhang A, Ae Ko K, Samanthapudi VSK, Lee LL, Bojorges AP, Gi YJ, Hobbs BP, Deswal A, Herrmann J, Lin SH, Chini EN, Shen YH, Schadler KL, Nguyen THM, Gupte AA, Reyes-Gibby C, Yeung SCJ, Abe RJ, Olmsted-Davis EA, Krishnan S, Dantzer R, Palaskas NL, Cooke JP, Pownall HJ, Yoshimoto M, Fujiwara K, Hamilton DJ, Burks JK, Wang G, Le NT, Kotla S. An ERK5-NRF2 Axis Mediates Senescence-Associated Stemness and Atherosclerosis. Circ Res 2023; 133:25-44. [PMID: 37264926 PMCID: PMC10357365 DOI: 10.1161/circresaha.122.322017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 05/17/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND ERK5 (extracellular signal-regulated kinase 5) is a dual kinase transcription factor containing an N-terminal kinase domain and a C-terminal transcriptional activation domain. Many ERK5 kinase inhibitors have been developed and tested to treat cancer and inflammatory diseases. However, recent data have raised questions about the role of the catalytic activity of ERK5 in proliferation and inflammation. We aimed to investigate how ERK5 reprograms myeloid cells to the proinflammatory senescent phenotype, subsequently leading to atherosclerosis. METHODS A ERK5 S496A (dephosphorylation mimic) knock in (KI) mouse model was generated using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9), and atherosclerosis was characterized by hypercholesterolemia induction. The plaque phenotyping in homozygous ERK5 S496A KI and wild type (WT) mice was studied using imaging mass cytometry. Bone marrow-derived macrophages were isolated from hypercholesterolemic mice and characterized using RNA sequencing and functional in vitro approaches, including senescence, mitochondria reactive oxygen species, and inflammation assays, as well as by metabolic extracellular flux analysis. RESULTS We show that atherosclerosis was inhibited in ERK5 S496A KI mice. Furthermore, ERK5 S496 phosphorylation mediates both senescence-associated secretory phenotype and senescence-associated stemness by upregulating AHR (aryl hydrocarbon receptor) in plaque and bone marrow-derived macrophages isolated from hypercholesterolemic mice. We also discovered that ERK5 S496 phosphorylation could induce NRF2 (NFE2-related factor 2) SUMOylation at a novel K518 site to inhibit NRF2 transcriptional activity without altering ERK5 catalytic activity and mediates oxidized LDL (low-density lipoprotein)-induced senescence-associated secretory phenotype. Specific ERK5 kinase inhibitors (AX15836 and XMD8-92) also inhibited ERK5 S496 phosphorylation, suggesting the involvement of ERK5 S496 phosphorylation in the anti-inflammatory effects of these ERK5 kinase inhibitors. CONCLUSIONS We discovered a novel mechanism by which the macrophage ERK5-NRF2 axis develops a unique senescence-associated secretory phenotype/stemness phenotype by upregulating AHR to engender atherogenesis. The finding of senescence-associated stemness phenotype provides a molecular explanation to resolve the paradox of senescence in proliferative plaque by permitting myeloid cells to escape the senescence-induced cell cycle arrest during atherosclerosis formation.
Collapse
Affiliation(s)
- Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- These authors contributed equally to this work and were designated as co-first authors
| | - Masaki Imanishi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- These authors contributed equally to this work and were designated as co-first authors
| | - Shengyu Li
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
- These authors contributed equally to this work and were designated as co-first authors
| | - Aijun Zhang
- Center for Bioenergetics, Houston Methodist Research Institute, Texas, and Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, Texas, USA
| | - Kyung Ae Ko
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Ling-Ling Lee
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Young Jin Gi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Brian P. Hobbs
- Department of Population Health, The University of Texas at Austin, Austin, Texas, USA
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Eduardo N. Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Ying H. Shen
- Division of Cardiothoracic Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Keri L. Schadler
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Thi-Hong-Minh Nguyen
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Anisha A. Gupte
- Center for Bioenergetics, Houston Methodist Research Institute, Texas, and Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, Texas, USA
| | - Cielito Reyes-Gibby
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sai-Ching J. Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rei J. Abe
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | | | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nicolas L. Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - John P. Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Henry J. Pownall
- Center for Bioenergetics, Houston Methodist Research Institute, Texas, and Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, Texas, USA
| | - Momoko Yoshimoto
- Center for Stem Cell & Regenerative Medicine, Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Keigi Fujiwara
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dale J. Hamilton
- Center for Bioenergetics, Houston Methodist Research Institute, Texas, and Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, Texas, USA
- These authors contributed equally to this work
| | - Jared K. Burks
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- These authors contributed equally to this work
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
- These authors were equivalent co-senior authors
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
- These authors were equivalent co-senior authors
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- These authors were equivalent co-senior authors
| |
Collapse
|
49
|
Cervantes J, Kanter JE. Monocyte and macrophage foam cells in diabetes-accelerated atherosclerosis. Front Cardiovasc Med 2023; 10:1213177. [PMID: 37378396 PMCID: PMC10291141 DOI: 10.3389/fcvm.2023.1213177] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Diabetes results in an increased risk of atherosclerotic cardiovascular disease. This minireview will discuss whether monocyte and macrophage lipid loading contribute to this increased risk, as monocytes and macrophages are critically involved in the progression of atherosclerosis. Both uptake and efflux pathways have been described as being altered by diabetes or conditions associated with diabetes, which may contribute to the increased accumulation of lipids seen in macrophages in diabetes. More recently, monocytes have also been described as lipid-laden in response to elevated lipids, including triglyceride-rich lipoproteins, the class of lipids often elevated in the setting of diabetes.
Collapse
Affiliation(s)
| | - Jenny E. Kanter
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA, United States
| |
Collapse
|
50
|
Wei YH, Bi RT, Qiu YM, Zhang CL, Li JZ, Li YN, Hu B. The gastrointestinal-brain-microbiota axis: a promising therapeutic target for ischemic stroke. Front Immunol 2023; 14:1141387. [PMID: 37342335 PMCID: PMC10277866 DOI: 10.3389/fimmu.2023.1141387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/22/2023] [Indexed: 06/22/2023] Open
Abstract
Ischemic stroke is a highly complex systemic disease characterized by intricate interactions between the brain and gastrointestinal tract. While our current understanding of these interactions primarily stems from experimental models, their relevance to human stroke outcomes is of considerable interest. After stroke, bidirectional communication between the brain and gastrointestinal tract initiates changes in the gastrointestinal microenvironment. These changes involve the activation of gastrointestinal immunity, disruption of the gastrointestinal barrier, and alterations in gastrointestinal microbiota. Importantly, experimental evidence suggests that these alterations facilitate the migration of gastrointestinal immune cells and cytokines across the damaged blood-brain barrier, ultimately infiltrating the ischemic brain. Although the characterization of these phenomena in humans is still limited, recognizing the significance of the brain-gastrointestinal crosstalk after stroke offers potential avenues for therapeutic intervention. By targeting the mutually reinforcing processes between the brain and gastrointestinal tract, it may be possible to improve the prognosis of ischemic stroke. Further investigation is warranted to elucidate the clinical relevance and translational potential of these findings.
Collapse
Affiliation(s)
| | | | | | | | | | - Ya-nan Li
- *Correspondence: Ya-nan Li, ; Bo Hu,
| | - Bo Hu
- *Correspondence: Ya-nan Li, ; Bo Hu,
| |
Collapse
|