1
|
Pandit R, Yurdagul A. The Atherosclerotic Plaque Microenvironment as a Therapeutic Target. Curr Atheroscler Rep 2025; 27:47. [PMID: 40172727 PMCID: PMC11965263 DOI: 10.1007/s11883-025-01294-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2025] [Indexed: 04/04/2025]
Abstract
PURPOSE OF REVIEW Atherosclerosis is traditionally viewed as a disease triggered by lipid accumulation, but growing evidence underscores the crucial role of the plaque microenvironment in disease progression. This review explores recent advances in understanding how cellular and extracellular components of the plaque milieu drive atherosclerosis, with a focus on leveraging these microenvironmental factors for therapeutic intervention. This review highlights recent advances in cell-cell crosstalk and matrix remodeling, offering insights into innovative therapeutic strategies for atherosclerotic cardiovascular disease. RECENT FINDINGS While atherosclerosis begins with the subendothelial retention of apolipoprotein B (ApoB)-containing lipoproteins, its progression is increasingly recognized as a consequence of complex cellular and extracellular dynamics within the plaque microenvironment. Soluble factors and extracellular matrix proteins shape mechanical properties and the biochemical landscape, directly influencing cell behavior and inflammatory signaling. For instance, the deposition of transitional matrix proteins, such as fibronectin, in regions of disturbed flow primes endothelial cells for inflammation. Likewise, impaired clearance of dead cells and chronic extracellular matrix remodeling contribute to lesion expansion and instability, further exacerbating disease severity. Targeting the plaque microenvironment presents a promising avenue for stabilizing atherosclerotic lesions. Approaches that enhance beneficial cellular interactions, such as boosting macrophage efferocytosis to resolve inflammation while mitigating proatherogenic signals like integrin-mediated endothelial activation, may promote fibrous cap formation and reduce plaque vulnerability. Harnessing these mechanisms may lead to novel therapeutic approaches aimed at modifying the plaque microenvironment to combat atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Rajan Pandit
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, Shreveport, LA, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, Shreveport, LA, USA.
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, Shreveport, LA, USA.
| |
Collapse
|
2
|
Shin SJ, Bayarkhangai B, Tsogtbaatar K, Yuxuan M, Kim S, Kim Y, Taghizadeh A, Kim D, Kim D, Lee J, Hyun J, Kim H. Matrix-Rigidity Cooperates With Biochemical Cues in M2 Macrophage Activation Through Increased Nuclear Deformation and Chromatin Accessibility. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2403409. [PMID: 39828979 PMCID: PMC11848612 DOI: 10.1002/advs.202403409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/19/2024] [Indexed: 01/22/2025]
Abstract
Macrophages encounter a myriad of biochemical and mechanical stimuli across various tissues and pathological contexts. Notably, matrix rigidity has emerged as a pivotal regulator of macrophage activation through mechanotransduction. However, the precise mechanisms underlying the interplay between mechanical and biochemical cues within the nuclear milieu remain elusive. Here We elucidate how the increased matrix rigidity drives macrophages to amplify alternatively-activated (M2 phenotype) signaling cooperatively with biochemical cues (e.g., IL4/13) through altered nuclear mechanics. We demonstrate that reconstructed podosome-like F-actins and contractility induce nucleus deformation, opening nuclear pores, which facilitates nuclear translocation of the key transcription factor STAT6. Furthermore, the altered nuclear mechanics increases chromatin accessibility induced by H3K9 methylation, particularly of M2-associated gene promoters. These cooperative events of the mechano-chemical signaling at the cytoskeletal-to-nuclear domains facilitate M2 transcriptional activation and cellular functions. We further evidence the rigidity-primed M2 macrophages are immunosuppressive and accumulated within stiffened tumors in patients. This study proposes a mechanism by which matrix mechanics crosstalks with biochemical signals to potentiate macrophage activation through nuclear mechanosensing and chromatin modifications, offering insights into macrophage mechanobiology and its therapeutic modulations.
Collapse
Affiliation(s)
- Seung Jae Shin
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Mechanobiology Dental Medicine Research CenterDankook UniversityCheonan31116Republic of Korea
| | - Buuvee Bayarkhangai
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Khaliunsarnai Tsogtbaatar
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Meng Yuxuan
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Sang‐Hyun Kim
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Department of ChemistryCollege of Science & TechnologyDankook UniversityCheonan31116Republic of Korea
| | - Yong‐Jae Kim
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Ali Taghizadeh
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Daesan Kim
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Dong‐Hwee Kim
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
- Department of Integrative Energy EngineeringCollege of EngineeringKorea UniversitySeoul02841Republic of Korea
- Biomedical Research CenterKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Jung‐Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Mechanobiology Dental Medicine Research CenterDankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
- UCL Eastman‐Korea Dental Medicine Innovation CentreDankook UniversityCheonan31116Republic of Korea
- Department of Biomaterials ScienceSchool of DentistryDankook UniversityCheonan31116Republic of Korea
- Cell & Matter InstituteDankook UniversityCheonan31116Republic of Korea
| | - Jeongeun Hyun
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Mechanobiology Dental Medicine Research CenterDankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
- UCL Eastman‐Korea Dental Medicine Innovation CentreDankook UniversityCheonan31116Republic of Korea
- Department of Regenerative Dental MedicineSchool of DentistryDankook UniversityCheonan31116Republic of Korea
| | - Hae‐Won Kim
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Mechanobiology Dental Medicine Research CenterDankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
- UCL Eastman‐Korea Dental Medicine Innovation CentreDankook UniversityCheonan31116Republic of Korea
- Cell & Matter InstituteDankook UniversityCheonan31116Republic of Korea
- Department of Regenerative Dental MedicineSchool of DentistryDankook UniversityCheonan31116Republic of Korea
| |
Collapse
|
3
|
Kim MJ, Chang JH. Structure simulation-based comparison of active site variations in fungal ornithine decarboxylases. Commun Integr Biol 2025; 18:2458872. [PMID: 39906711 PMCID: PMC11792860 DOI: 10.1080/19420889.2025.2458872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
Polyamines play crucial roles in various biological processes, including cell proliferation and differentiation, immune response modulation, and signal transduction. Ornithine decarboxylase (ODC) initiates polyamine biosynthesis by catalyzing the conversion of ornithine to putrescine in a pyridoxal phosphate (PLP)-dependent manner. While the structures of mammalian and protozoan ODCs have been elucidated, fungal ODCs remain uncharacterized. In this study, AlphaFold2 was employed to simulate the structures of ODCs from four fungi: Kluyveromyces lactis, Candida albicans, Debaryomyces hansenii, and Schizosaccharomyces pombe. The results indicated that, although these ODCs share α/β-barrel and β-sheet domains, their active site conformations exhibit subtle differences. Additionally, substrate selectivity among ODCs and related decarboxylases varied depending on the distance between the Cα of aspartate or glutamate residues within the specificity helix and the C4α of PLP. Notably, the bacterial Campylobacter jejuni decarboxylase (CjCANSDC), which binds the largest substrate, exhibits the longest distance, whereas fungal ODC, which binds the smallest substrate, displays the shortest distance. Furthermore, significant differences in the composition of amino acid residues within the active sites were also observed. This study provides insights into the structural diversity and catalytic activity of ODCs across a broad range of organisms, advancing the understanding of structure-dependent evolutionary processes.
Collapse
Affiliation(s)
- Min Jeong Kim
- Department of Biology Education, Kyungpook National University, Daegu, South Korea
| | - Jeong Ho Chang
- Department of Biology Education, Kyungpook National University, Daegu, South Korea
- Department of Biomedical Convergence Science and Technology, Kyungpook National University, Daegu, South Korea
- Science Education Research Institute, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
4
|
Bamgbose TT, Schilke RM, Igiehon OO, Nkadi EH, Binwal M, Custis D, Bharrhan S, Schwarz B, Bohrnsen E, Bosio CM, Scott RS, Yurdagul Jr. A, Finck BN, Woolard MD. Lipin-1 restrains macrophage lipid synthesis to promote inflammation resolution. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:85-103. [PMID: 40073265 PMCID: PMC11844145 DOI: 10.1093/jimmun/vkae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 10/24/2024] [Indexed: 03/14/2025]
Abstract
Macrophages are critical to maintaining and restoring tissue homeostasis during inflammation. The lipid metabolic state of macrophages influences their function and polarization, which is crucial to the resolution of inflammation. The contribution of lipid synthesis to proinflammatory macrophage responses is well understood. However, how lipid synthesis regulates proresolving macrophage responses needs to be better understood. Lipin-1 is a phosphatidic acid phosphatase with a transcriptional coregulatory activity that regulates lipid metabolism. We previously demonstrated that lipin-1 supports proresolving macrophage responses, and here, myeloid-associated lipin-1 is required for inflammation resolution, yet how lipin-1-regulated cellular mechanisms promote macrophage proresolution responses is unknown. We demonstrated that the loss of lipin-1 in macrophages led to increased free fatty acid, neutral lipid, and ceramide content and increased phosphorylation of acetyl-CoA carboxylase. The inhibition of the first step of lipid synthesis, the transport of citrate from the mitochondria, reduced lipid content and restored efferocytosis and inflammation resolution in lipin-1mKO mice and macrophages. Our findings suggest macrophage-associated lipin-1 restrains lipid synthesis, promoting proresolving macrophage function in response to proresolving stimuli.
Collapse
Affiliation(s)
- Temitayo T Bamgbose
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Robert M Schilke
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Oluwakemi O Igiehon
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Ebubechukwu H Nkadi
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Monika Binwal
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - David Custis
- Research Core Facility, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Sushma Bharrhan
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Benjamin Schwarz
- Proteins and Chemistry Section, Research and Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Eric Bohrnsen
- Proteins and Chemistry Section, Research and Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Catharine M Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Rona S Scott
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Arif Yurdagul Jr.
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Brian N Finck
- Division of Nutritional Sciences and Obesity Medicine, Washington University School of Medicine in St. Louis, St Louis, MO, United States
| | - Matthew D Woolard
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| |
Collapse
|
5
|
Zhang B, Zou Y, Yuan Z, Jiang K, Zhang Z, Chen S, Zhou X, Wu Q, Zhang X. Efferocytosis: the resolution of inflammation in cardiovascular and cerebrovascular disease. Front Immunol 2024; 15:1485222. [PMID: 39660125 PMCID: PMC11628373 DOI: 10.3389/fimmu.2024.1485222] [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: 08/23/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Cardiovascular and cerebrovascular diseases have surpassed cancer as significant global health challenges, which mainly include atherosclerosis, myocardial infarction, hemorrhagic stroke and ischemia stroke. The inflammatory response immediately following these diseases profoundly impacts patient prognosis and recovery. Efficient resolution of inflammation is crucial not only for halting the inflammatory process but also for restoring tissue homeostasis. Efferocytosis, the phagocytic clearance of dying cells by phagocytes, especially microglia and macrophages, plays a critical role in this resolution process. Upon tissue injury, phagocytes are recruited to the site of damage where they engulf and clear dying cells through efferocytosis. Efferocytosis suppresses the secretion of pro-inflammatory cytokines, stimulates the production of anti-inflammatory cytokines, modulates the phenotype of microglia and macrophages, accelerates the resolution of inflammation, and promotes tissue repair. It involves three main stages: recognition, engulfment, and degradation of dying cells. Optimal removal of apoptotic cargo by phagocytes requires finely tuned machinery and associated modifications. Key molecules in efferocytosis, such as 'Find-me signals', 'Eat-me signals', and 'Don't eat-me signals', have been shown to enhance efferocytosis following cardiovascular and cerebrovascular diseases. Moreover, various additional molecules, pathways, and mitochondrial metabolic processes have been identified to enhance prognosis and outcomes via efferocytosis in diverse experimental models. Impaired efferocytosis can lead to inflammation-associated pathologies and prolonged recovery periods. Therefore, this review consolidates current understanding of efferocytosis mechanisms and its application in cardiovascular and cerebrovascular diseases, proposing future research directions.
Collapse
Affiliation(s)
- Bingtao Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yan Zou
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zixuan Yuan
- Department of Neurosurgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Kun Jiang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhaoxiang Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shujuan Chen
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaoming Zhou
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qi Wu
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xin Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
6
|
Abstract
Cardiovascular disease is the leading cause of death worldwide, and it commonly results from atherosclerotic plaque progression. One of the increasingly recognized drivers of atherosclerosis is dysfunctional efferocytosis, a homeostatic mechanism responsible for the clearance of dead cells and the resolution of inflammation. In atherosclerosis, the capacity of phagocytes to participate in efferocytosis is hampered, leading to the accumulation of apoptotic and necrotic tissue within the plaque, which results in enlargement of the necrotic core, increased luminal stenosis and plaque inflammation, and predisposition to plaque rupture or erosion. In this Review, we describe the different forms of programmed cell death that can occur in the atherosclerotic plaque and highlight the efferocytic machinery that is normally implicated in cardiovascular physiology. We then discuss the mechanisms by which efferocytosis fails in atherosclerosis and other cardiovascular and cardiometabolic diseases, including myocardial infarction and diabetes mellitus, and discuss therapeutic approaches that might reverse this pathological process.
Collapse
Affiliation(s)
- Shaunak S Adkar
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Nicholas J Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford, CA, USA.
| |
Collapse
|
7
|
Bobrovskikh AV, Zubairova US, Naumenko LG, Doroshkov AV. Catching the Big Fish in Big Data: A Meta-Analysis of Zebrafish Kidney scRNA-Seq Datasets Highlights Conserved Molecular Profiles of Macrophages and Neutrophils in Vertebrates. BIOLOGY 2024; 13:773. [PMID: 39452082 PMCID: PMC11505477 DOI: 10.3390/biology13100773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
The innate immune system (IIS) is an ancient and essential defense mechanism that protects animals against a wide range of pathogens and diseases. Although extensively studied in mammals, our understanding of the IIS in other taxa remains limited. The zebrafish (Danio rerio) serves as a promising model organism for investigating IIS-related processes, yet the immunogenetics of fish are not fully elucidated. To address this gap, we conducted a meta-analysis of single-cell RNA sequencing (scRNA-seq) datasets from zebrafish kidney marrow, encompassing approximately 250,000 immune cells. Our analysis confirms the presence of key genetic pathways in zebrafish innate immune cells that are similar to those identified in mammals. Zebrafish macrophages specifically express genes encoding cathepsins, major histocompatibility complex class II proteins, integral membrane proteins, and the V-ATPase complex and demonstrate the enrichment of oxidative phosphorylation ferroptosis processes. Neutrophils are characterized by the significant expression of genes encoding actins, cytoskeleton organizing proteins, the Arp2/3 complex, and glycolysis enzymes and have demonstrated their involvement in GnRH and CLR signaling pathways, adherents, and tight junctions. Both macrophages and neutrophils highly express genes of NOD-like receptors, phagosomes, and lysosome pathways and genes involved in apoptosis. Our findings reinforce the idea about the existence of a wide spectrum of immune cell phenotypes in fish since we found only a small number of cells with clear pro- or anti-inflammatory signatures.
Collapse
Affiliation(s)
- Aleksandr V. Bobrovskikh
- Department of Physics, Novosibirsk State University, 630090 Novosibirsk, Russia;
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
| | - Ulyana S. Zubairova
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
- Department of Information Technologies, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Ludmila G. Naumenko
- Department of Physics, Novosibirsk State University, 630090 Novosibirsk, Russia;
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
| | - Alexey V. Doroshkov
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
- Department of Genomics and Bioinformatics, Institute of Fundamental Biology and Biotechnology, Siberian Federal University, 660036 Krasnoyarsk, Russia
| |
Collapse
|
8
|
Sukka SR, Ampomah PB, Darville LNF, Ngai D, Wang X, Kuriakose G, Xiao Y, Shi J, Koomen JM, McCusker RH, Tabas I. Efferocytosis drives a tryptophan metabolism pathway in macrophages to promote tissue resolution. Nat Metab 2024; 6:1736-1755. [PMID: 39242914 PMCID: PMC11734744 DOI: 10.1038/s42255-024-01115-7] [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: 02/03/2024] [Accepted: 07/24/2024] [Indexed: 09/09/2024]
Abstract
Macrophage efferocytosis prevents apoptotic cell (AC) accumulation and triggers inflammation-resolution pathways. The mechanisms linking efferocytosis to resolution often involve changes in macrophage metabolism, but many gaps remain in our understanding of these processes. We now report that efferocytosis triggers an indoleamine 2,3-dioxygenase-1 (IDO1)-dependent tryptophan (Trp) metabolism pathway that promotes several key resolution processes, including the induction of pro-resolving proteins, such interleukin-10, and further enhancement of efferocytosis. The process begins with upregulation of Trp transport and metabolism, and it involves subsequent activation of the aryl hydrocarbon receptor (AhR) by the Trp metabolite kynurenine (Kyn). Through these mechanisms, macrophage IDO1 and AhR contribute to a proper resolution response in several different mouse models of efferocytosis-dependent tissue repair, notably during atherosclerosis regression induced by plasma low-density lipoprotein (LDL) lowering. These findings reveal an integrated metabolism programme in macrophages that links efferocytosis to resolution, with possible therapeutic implications for non-resolving chronic inflammatory diseases, notably atherosclerosis.
Collapse
Affiliation(s)
- Santosh R Sukka
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | - Patrick B Ampomah
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Lancia N F Darville
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - David Ngai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - George Kuriakose
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Yuling Xiao
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - John M Koomen
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Robert H McCusker
- Department of Animal Sciences, Integrative Immunology and Behavior Program and Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Departments of Physiology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
9
|
Wu X, Wang Z, Croce KR, Li F, Cui J, D’Agati VD, Soni RK, Khalid S, Saleheen D, Tabas I, Yamamoto A, Zhang H. Macrophage WDFY3, a protector against autoimmunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.17.608411. [PMID: 39229152 PMCID: PMC11370343 DOI: 10.1101/2024.08.17.608411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Efficient efferocytosis is essential for maintaining homeostasis. Excessive apoptotic cell (AC) death and impaired macrophage efferocytosis lead to autoantigen release and autoantibody production, immune activation, and organ damage. It remains unclear whether these immunogenic autoantigens are the sole cause of increased autoimmunity or if efferocytosis of ACs directly influences macrophage function, impacting their ability to activate T cells and potentially amplifying autoimmune responses. Additionally, it has not been established if enhancing macrophage efferocytosis or modulating macrophage responses to AC engulfment can be protective in autoimmune-like disorders. Our previous work showed WDFY3 is crucial for efficient macrophage efferocytosis. This study reveals that myeloid knockout of Wdfy3 exacerbates autoimmunity in young mice with increased AC burden by systemic injections of ACs and in middle-aged mice developing spontaneous autoimmunity, whereas ectopic overexpression of WDFY3 suppresses autoimmunity in these models. Macrophages, as efferocytes, can activate T cells and the inflammasome upon engulfing ACs, which are suppressed by overexpressing WDFY3. This work uncovered the role of WDFY3 as a protector against autoimmunity by promoting macrophage efferocytosis thus limiting autoantigen production, as well as mitigating T cell activation and inflammasome activation.
Collapse
Affiliation(s)
- Xun Wu
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ziyi Wang
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Fang Li
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Jian Cui
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Vivette D. D’Agati
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Renal Pathology Laboratory, Columbia University Irving Medical Center, New York, NY, USA
| | - Rajesh K. Soni
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Shareef Khalid
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Danish Saleheen
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ira Tabas
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Ai Yamamoto
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Hanrui Zhang
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
10
|
Shahror RA, Shosha E, Morris C, Wild M, Mu S, Csanyi G, Boerma M, Rusch NJ, Fouda AY. Deletion of myeloid HDAC3 promotes efferocytosis to ameliorate retinal ischemic injury. J Neuroinflammation 2024; 21:170. [PMID: 38997746 PMCID: PMC11241909 DOI: 10.1186/s12974-024-03159-8] [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: 03/19/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
Ischemia-induced retinopathy is a hallmark finding of common visual disorders including diabetic retinopathy (DR) and central retinal artery and vein occlusions. Treatments for ischemic retinopathies fail to improve clinical outcomes and the design of new therapies will depend on understanding the underlying disease mechanisms. Histone deacetylases (HDACs) are an enzyme class that removes acetyl groups from histone and non-histone proteins, thereby regulating gene expression and protein function. HDACs have been implicated in retinal neurovascular injury in preclinical studies in which nonspecific HDAC inhibitors mitigated retinal injury. Histone deacetylase 3 (HDAC3) is a class I histone deacetylase isoform that plays a central role in the macrophage inflammatory response. We recently reported that myeloid cells upregulate HDAC3 in a mouse model of retinal ischemia-reperfusion (IR) injury. However, whether this cellular event is an essential contributor to retinal IR injury is unknown. In this study, we explored the role of myeloid HDAC3 in ischemia-induced retinal neurovascular injury by subjecting myeloid-specific HDAC3 knockout (M-HDAC3 KO) and floxed control mice to retinal IR. The M-HDAC3 KO mice were protected from retinal IR injury as shown by the preservation of inner retinal neurons, vascular integrity, and retinal thickness. Electroretinography confirmed that this neurovascular protection translated to improved retinal function. The retinas of M-HDAC3 KO mice also showed less proliferation and infiltration of myeloid cells after injury. Interestingly, myeloid cells lacking HDAC3 more avidly engulfed apoptotic cells in vitro and after retinal IR injury in vivo compared to wild-type myeloid cells, suggesting that HDAC3 hinders the reparative phagocytosis of dead cells, a process known as efferocytosis. Further mechanistic studies indicated that although HDAC3 KO macrophages upregulate the reparative enzyme arginase 1 (A1) that enhances efferocytosis, the inhibitory effect of HDAC3 on efferocytosis is not solely dependent on A1. Finally, treatment of wild-type mice with the HDAC3 inhibitor RGFP966 ameliorated the retinal neurodegeneration and thinning caused by IR injury. Collectively, our data show that HDAC3 deletion enhances macrophage-mediated efferocytosis and protects against retinal IR injury, suggesting that inhibiting myeloid HDAC3 holds promise as a novel therapeutic strategy for preserving retinal integrity after ischemic insult.
Collapse
Affiliation(s)
- Rami A Shahror
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA
| | - Esraa Shosha
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA
- Clinical Pharmacy Department, Cairo University, Cairo, Egypt
| | - Carol Morris
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA
| | - Melissa Wild
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA
| | - Shengyu Mu
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA
| | - Gabor Csanyi
- Department of Pharmacology and Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Marjan Boerma
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Nancy J Rusch
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA
| | - Abdelrahman Y Fouda
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA.
- Clinical Pharmacy Department, Cairo University, Cairo, Egypt.
| |
Collapse
|
11
|
Shu LX, Cao LL, Guo X, Wang ZB, Wang SZ. Mechanism of efferocytosis in atherosclerosis. J Mol Med (Berl) 2024; 102:831-840. [PMID: 38727748 DOI: 10.1007/s00109-024-02439-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 06/29/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease that occurs in the intima of large and medium-sized arteries with the immune system's involvement. It is a common pathological basis for high morbidity and mortality of cardiovascular diseases. Abnormal proliferation of apoptotic cells and necrotic cells leads to AS plaque expansion, necrotic core formation, and rupture. In the early stage of AS, macrophages exert an efferocytosis effect to engulf and degrade apoptotic, dead, damaged, or senescent cells by efferocytosis, thus enabling the regulation of the organism. In the early stage of AS, macrophages rely on this effect to slow down the process of AS. However, in the advanced stage of AS, the efferocytosis of macrophages within the plaque is impaired, which leads to the inability of macrophages to promptly remove the apoptotic cells (ACs) from the organism promptly, causing exacerbation of AS. Moreover, upregulation of CD47 expression in AS plaques also protects ACs from phagocytosis by macrophages, resulting in a large amount of residual ACs in the plaque, further expanding the necrotic core. In this review, we discussed the molecular mechanisms involved in the process of efferocytosis and how efferocytosis is impaired and regulated during AS, hoping to provide new insights for treating AS.
Collapse
Affiliation(s)
- Li-Xia Shu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Liu-Li Cao
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Xin Guo
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Zong-Bao Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China.
| |
Collapse
|
12
|
Li Q, Liu H, Yin G, Xie Q. Efferocytosis: Current status and future prospects in the treatment of autoimmune diseases. Heliyon 2024; 10:e28399. [PMID: 38596091 PMCID: PMC11002059 DOI: 10.1016/j.heliyon.2024.e28399] [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: 01/08/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024] Open
Abstract
Billions of apoptotic cells are swiftly removed from the human body daily. This clearance process is regulated by efferocytosis, an active anti-inflammatory process during which phagocytes engulf and remove apoptotic cells. However, impaired clearance of apoptotic cells is associated with the development of various autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus, and inflammatory bowel disease. In this review, we conducted a comprehensive search of relevant studies published from January 1, 2000, to the present, focusing on efferocytosis, autoimmune disease pathogenesis, regulatory mechanisms governing efferocytosis, and potential treatments targeting this process. Our review highlights the key molecules involved in different stages of efferocytosis-namely, the "find me," "eat me," and "engulf and digest" phases-while elucidating their relevance to autoimmune disease pathology. Furthermore, we explore the therapeutic potential of modulating efferocytosis to restore immune homeostasis and mitigate autoimmune responses. By providing theoretical underpinnings for the targeting of efferocytosis in the treatment of autoimmune diseases, this review contributes to the advancement of therapeutic strategies in this field.
Collapse
Affiliation(s)
- Qianwei Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Huan Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Geng Yin
- Department of General Practice, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
13
|
Lee EJ, Kim DJ, Kang DW, Yang W, Jeong HY, Kim JM, Ko SB, Lee SH, Yoon BW, Cho JY, Jung KH. Targeted Metabolomic Biomarkers for Stroke Subtyping. Transl Stroke Res 2024; 15:422-432. [PMID: 36764997 DOI: 10.1007/s12975-023-01137-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/11/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023]
Abstract
BACKGROUND AND PURPOSE Ischemic stroke is a heterogeneous disease with various etiologies. The current subtyping process is complicated, time-consuming, and costly. Metabolite-based biomarkers have the potential to improve classification and deliver optimal treatments. We here aimed to identify novel, targeted metabolomics-based biomarkers to discriminate between large-artery atherosclerosis (LAA) and cardioembolic (CE) stroke. METHODS We acquired serum samples and clinical data from a hospital-based acute stroke registry (ischemic stroke within 3 days from symptom onset). We included 346 participants (169 LAA, 147 CE, and 30 healthy older adults) and divided them into training and test sets. Targeted metabolomic analysis was performed using quantitative and quality-controlled liquid chromatography with tandem mass spectrometry. A multivariate regression model using metabolomic signatures was created that could independently distinguish between LAA and CE strokes. RESULTS The training set (n = 193) identified metabolomic signatures that were different in patients with LAA and CE strokes. Six metabolomic biomarkers, i.e., lysine, serine, threonine, kynurenine, putrescine, and lysophosphatidylcholine acyl C16:0, could discriminate between LAA and CE stroke after adjusting for sex, age, body mass index, stroke severity, and comorbidities. The enhanced diagnostic power of key metabolite combinations for discriminating between LAA and CE stroke was validated using the test set (n = 123). CONCLUSIONS We observed significant differences in metabolite profiles in LAA and CE strokes. Targeted metabolomics may provide enhanced diagnostic yield for stroke subtypes. The pathophysiological pathways of the identified metabolites should be explored in future studies.
Collapse
Affiliation(s)
- Eung-Joon Lee
- Department of Neurology, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Da Jung Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
- Metabolomics Core Facility, Department of Transdisciplinary Research and Collaboration, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong-Wan Kang
- Department of Neurology, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Wookjin Yang
- Department of Neurology, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Han-Yeong Jeong
- Department of Neurology, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Jeong-Min Kim
- Department of Neurology, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Sang-Bae Ko
- Department of Neurology, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Seung-Hoon Lee
- Department of Neurology, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Byung-Woo Yoon
- Department of Neurology, Uijeongbu Eulji Medical Center, Uijeongbu-si, Republic of Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Keun-Hwa Jung
- Department of Neurology, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea.
- Program in Neuroscience, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Anand SK, Governale TA, Zhang X, Razani B, Yurdagul A, Pattillo CB, Rom O. Amino Acid Metabolism and Atherosclerotic Cardiovascular Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:510-524. [PMID: 38171450 PMCID: PMC10988767 DOI: 10.1016/j.ajpath.2023.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/09/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024]
Abstract
Despite significant advances in medical treatments and drug development, atherosclerotic cardiovascular disease (ASCVD) remains a leading cause of death worldwide. Dysregulated lipid metabolism is a well-established driver of ASCVD. Unfortunately, even with potent lipid-lowering therapies, ASCVD-related deaths have continued to increase over the past decade, highlighting an incomplete understanding of the underlying risk factors and mechanisms of ASCVD. Accumulating evidence over the past decades indicates a correlation between amino acids and disease state. This review explores the emerging role of amino acid metabolism in ASCVD, uncovering novel potential biomarkers, causative factors, and therapeutic targets. Specifically, the significance of arginine and its related metabolites, homoarginine and polyamines, branched-chain amino acids, glycine, and aromatic amino acids, in ASCVD are discussed. These amino acids and their metabolites have been implicated in various processes characteristic of ASCVD, including impaired lipid metabolism, endothelial dysfunction, increased inflammatory response, and necrotic core development. Understanding the complex interplay between dysregulated amino acid metabolism and ASCVD provides new insights that may lead to the development of novel diagnostic and therapeutic approaches. Although further research is needed to uncover the precise mechanisms involved, it is evident that amino acid metabolism plays a role in ASCVD.
Collapse
Affiliation(s)
- Sumit Kumar Anand
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Theresea-Anne Governale
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Xiangyu Zhang
- Division of Cardiology and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Babak Razani
- Division of Cardiology and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Arif Yurdagul
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Christopher B Pattillo
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana.
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana.
| |
Collapse
|
15
|
Stroope C, Nettersheim FS, Coon B, Finney AC, Schwartz MA, Ley K, Rom O, Yurdagul A. Dysregulated cellular metabolism in atherosclerosis: mediators and therapeutic opportunities. Nat Metab 2024; 6:617-638. [PMID: 38532071 PMCID: PMC11055680 DOI: 10.1038/s42255-024-01015-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] [Received: 03/05/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
Accumulating evidence over the past decades has revealed an intricate relationship between dysregulation of cellular metabolism and the progression of atherosclerotic cardiovascular disease. However, an integrated understanding of dysregulated cellular metabolism in atherosclerotic cardiovascular disease and its potential value as a therapeutic target is missing. In this Review, we (1) summarize recent advances concerning the role of metabolic dysregulation during atherosclerosis progression in lesional cells, including endothelial cells, vascular smooth muscle cells, macrophages and T cells; (2) explore the complexity of metabolic cross-talk between these lesional cells; (3) highlight emerging technologies that promise to illuminate unknown aspects of metabolism in atherosclerosis; and (4) suggest strategies for targeting these underexplored metabolic alterations to mitigate atherosclerosis progression and stabilize rupture-prone atheromas with a potential new generation of cardiovascular therapeutics.
Collapse
Affiliation(s)
- Chad Stroope
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Felix Sebastian Nettersheim
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Brian Coon
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Cardiovascular Biology Research Program, OMRF, Oklahoma City, OK, USA
- Department of Cell Biology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
- Immunology Center of Georgia (IMMCG), Augusta University Immunology Center of Georgia, Augusta, GA, USA
| | - Oren Rom
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
16
|
Raju S, Botts SR, Blaser MC, Abdul-Samad M, Prajapati K, Khosraviani N, Ho TWW, Breda LC, Ching C, Galant NJ, Fiddes L, Wu R, Clift CL, Pham T, Lee WL, Singh SA, Aikawa E, Fish JE, Howe KL. Directional Endothelial Communication by Polarized Extracellular Vesicle Release. Circ Res 2024; 134:269-289. [PMID: 38174557 PMCID: PMC10826926 DOI: 10.1161/circresaha.123.322993] [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: 04/28/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Extracellular vesicles (EVs) contain bioactive cargo including miRNAs and proteins that are released by cells during cell-cell communication. Endothelial cells (ECs) form the innermost lining of all blood vessels, interfacing with cells in the circulation and vascular wall. It is unknown whether ECs release EVs capable of governing recipient cells within these 2 separate compartments. Given their boundary location, we propose ECs use bidirectional release of distinct EV cargo in quiescent (healthy) and activated (atheroprone) states to communicate with cells within the circulation and blood vessel wall. METHODS EVs were isolated from primary human aortic ECs (plate and transwell grown; ±IL [interleukin]-1β activation), quantified, visualized, and analyzed by miRNA transcriptomics and proteomics. Apical and basolateral EC-EV release was determined by miRNA transfer, total internal reflection fluorescence and electron microscopy. Vascular reprogramming (RNA sequencing) and functional assays were performed on primary human monocytes or smooth muscle cells±EC-EVs. RESULTS Activated ECs increased EV release, with miRNA and protein cargo related to atherosclerosis. EV-treated monocytes and smooth muscle cells revealed activated EC-EV altered pathways that were proinflammatory and atherogenic. ECs released more EVs apically, which increased with activation. Apical and basolateral EV cargo contained distinct transcriptomes and proteomes that were altered by EC activation. Notably, activated basolateral EC-EVs displayed greater changes in the EV secretome, with pathways specific to atherosclerosis. In silico analysis determined compartment-specific cargo released by the apical and basolateral surfaces of ECs can reprogram monocytes and smooth muscle cells, respectively, with functional assays and in vivo imaging supporting this concept. CONCLUSIONS Demonstrating that ECs are capable of polarized EV cargo loading and directional EV secretion reveals a novel paradigm for endothelial communication, which may ultimately enhance the design of endothelial-based therapeutics for cardiovascular diseases such as atherosclerosis where ECs are persistently activated.
Collapse
Affiliation(s)
- Sneha Raju
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Institute of Medical Science (S.R., S.R.B., C.C., J.E.F., K.L.H.), University of Toronto, Toronto, ON, Canada
- Faculty of Medicine (S.R., S.R.B., L.F., K.L.H.), University of Toronto, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada (S.R., K.L.H.)
| | - Steven R. Botts
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Institute of Medical Science (S.R., S.R.B., C.C., J.E.F., K.L.H.), University of Toronto, Toronto, ON, Canada
- Faculty of Medicine (S.R., S.R.B., L.F., K.L.H.), University of Toronto, Toronto, ON, Canada
| | - Mark C. Blaser
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.C.B., C.L.C., T.P., S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Majed Abdul-Samad
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology (M.A.-S., N.K., R.W., J.E.F.), University of Toronto, Toronto, ON, Canada
| | - Kamalben Prajapati
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
| | - Negar Khosraviani
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology (M.A.-S., N.K., R.W., J.E.F.), University of Toronto, Toronto, ON, Canada
| | - Tse Wing Winnie Ho
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada (T.W.W.H., W.L.L.)
| | - Leandro C.D. Breda
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
| | - Crizza Ching
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Institute of Medical Science (S.R., S.R.B., C.C., J.E.F., K.L.H.), University of Toronto, Toronto, ON, Canada
| | | | - Lindsey Fiddes
- Faculty of Medicine (S.R., S.R.B., L.F., K.L.H.), University of Toronto, Toronto, ON, Canada
| | - Ruilin Wu
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology (M.A.-S., N.K., R.W., J.E.F.), University of Toronto, Toronto, ON, Canada
| | - Cassandra L. Clift
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.C.B., C.L.C., T.P., S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Tan Pham
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.C.B., C.L.C., T.P., S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Warren L. Lee
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada (T.W.W.H., W.L.L.)
| | - Sasha A. Singh
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.C.B., C.L.C., T.P., S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine (S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.C.B., C.L.C., T.P., S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine (S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Institute of Medical Science (S.R., S.R.B., C.C., J.E.F., K.L.H.), University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology (M.A.-S., N.K., R.W., J.E.F.), University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, ON, Canada (J.E.F., K.L.H.)
| | - Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Institute of Medical Science (S.R., S.R.B., C.C., J.E.F., K.L.H.), University of Toronto, Toronto, ON, Canada
- Faculty of Medicine (S.R., S.R.B., L.F., K.L.H.), University of Toronto, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada (S.R., K.L.H.)
- Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, ON, Canada (J.E.F., K.L.H.)
| |
Collapse
|
17
|
Cho S, Dadson K, Sung HK, Ayansola O, Mirzaesmaeili A, Noskovicova N, Zhao Y, Cheung K, Radisic M, Hinz B, Sater AAA, Hsu HH, Lopaschuk GD, Sweeney G. Cardioprotection by the adiponectin receptor agonist ALY688 in a preclinical mouse model of heart failure with reduced ejection fraction (HFrEF). Biomed Pharmacother 2024; 171:116119. [PMID: 38181714 DOI: 10.1016/j.biopha.2023.116119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
AIMS Adiponectin has been shown to mediate cardioprotective effects and levels are typically reduced in patients with cardiometabolic disease. Hence, there has been intense interest in developing adiponectin-based therapeutics. The aim of this translational research study was to examine the functional significance of targeting adiponectin signaling with the adiponectin receptor agonist ALY688 in a mouse model of heart failure with reduced ejection fraction (HFrEF), and the mechanisms of cardiac remodeling leading to cardioprotection. METHODS AND RESULTS Wild-type mice were subjected to transverse aortic constriction (TAC) to induce left ventricular pressure overload (PO), or sham surgery, with or without daily subcutaneous ALY688-SR administration. Temporal analysis of cardiac function was conducted via weekly echocardiography for 5 weeks and we observed that ALY688 attenuated the PO-induced dysfunction. ALY688 also reduced cardiac hypertrophic remodeling, assessed via LV mass, heart weight to body weight ratio, cardiomyocyte cross sectional area, ANP and BNP levels. ALY688 also attenuated PO-induced changes in myosin light and heavy chain expression. Collagen content and myofibroblast profile indicated that fibrosis was attenuated by ALY688 with TIMP1 and scleraxis/periostin identified as potential mechanistic contributors. ALY688 reduced PO-induced elevation in circulating cytokines including IL-5, IL-13 and IL-17, and the chemoattractants MCP-1, MIP-1β, MIP-1alpha and MIP-3α. Assessment of myocardial transcript levels indicated that ALY688 suppressed PO-induced elevations in IL-6, TLR-4 and IL-1β, collectively indicating anti-inflammatory effects. Targeted metabolomic profiling indicated that ALY688 increased fatty acid mobilization and oxidation, increased betaine and putrescine plus decreased sphingomyelin and lysophospholipids, a profile indicative of improved insulin sensitivity. CONCLUSION These results indicate that the adiponectin mimetic peptide ALY688 reduced PO-induced fibrosis, hypertrophy, inflammation and metabolic dysfunction and represents a promising therapeutic approach for treating HFrEF in a clinical setting.
Collapse
Affiliation(s)
- Sungji Cho
- Department of Biology, York University, Toronto, ON, Canada
| | - Keith Dadson
- Department of Biology, York University, Toronto, ON, Canada
| | | | | | - Ali Mirzaesmaeili
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Nina Noskovicova
- Faculty of Dentistry, University of Toronto, Toronto, ON M5S3E2, Canada
| | - Yimu Zhao
- Toronto General Hospital Research Institute, Toronto, ON M5G 2C4, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Krisco Cheung
- Department of Chemical Engineering and Applied Chemistry; University of Toronto, Toronto, ON M5S 3E5, Canada
| | - Milica Radisic
- Toronto General Hospital Research Institute, Toronto, ON M5G 2C4, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry; University of Toronto, Toronto, ON M5S 3E5, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5S3E2, Canada; Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON M5B 1T8, Canada
| | - Ali A Abdul Sater
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Henry H Hsu
- Allysta Pharmaceuticals Inc. Bellevue, WA, USA
| | - Gary D Lopaschuk
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada.
| |
Collapse
|
18
|
Bamgbose TT, Schilke RM, Igiehon OO, Nkadi EH, Custis D, Bharrhan S, Schwarz B, Bohrnsen E, Bosio CM, Scott RS, Yurdagul A, Finck BN, Woolard MD. Lipin-1 restrains macrophage lipid synthesis to promote inflammation resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563587. [PMID: 37961352 PMCID: PMC10634750 DOI: 10.1101/2023.10.23.563587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Macrophages are critical to maintaining and restoring tissue homeostasis during inflammation. The lipid metabolic state of macrophages influences their function, but a deeper understanding of how lipid metabolism is regulated in pro-resolving macrophage responses is needed. Lipin-1 is a phosphatidic acid phosphatase with a transcriptional coregulatory activity (TC) that regulates lipid metabolism. We previously demonstrated that lipin-1 supports pro-resolving macrophage responses, and here, myeloid-associated lipin-1 is required for inflammation resolution, yet how lipin-1-regulated cellular mechanisms promote macrophage pro-resolution responses is unknown. We demonstrated that the loss of lipin-1 in macrophages led to increased free fatty acid, neutral lipid, and ceramide content and increased phosphorylation of acetyl-CoA carboxylase. The inhibition of the first step of lipid synthesis and transport of citrate from the mitochondria in macrophages reduced lipid content and restored efferocytosis and inflammation resolution in lipin-1mKO macrophages and mice. Our findings suggest macrophage-associated lipin-1 restrains lipid synthesis, promoting pro-resolving macrophage function in response to pro-resolving stimuli.
Collapse
Affiliation(s)
- Temitayo T. Bamgbose
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Robert M. Schilke
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Oluwakemi O. Igiehon
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Ebubechukwu H. Nkadi
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - David Custis
- Research Core Facility, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Sushma Bharrhan
- Center for Applied Immunology and Pathological Processes (CAIPP), Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Benjamin Schwarz
- Proteins & Chemistry Section, Research and Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Disease, Hamilton, MT
| | - Eric Bohrnsen
- Proteins & Chemistry Section, Research and Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Disease, Hamilton, MT
| | - Catharine M. Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Disease, Hamilton, MT
| | - Rona S. Scott
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Center for Applied Immunology and Pathological Processes (CAIPP), Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Brian N. Finck
- Division of Nutritional Sciences and Obesity Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Matthew D. Woolard
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Center for Applied Immunology and Pathological Processes (CAIPP), Louisiana State University Health Sciences Center, Shreveport, LA, United States
| |
Collapse
|
19
|
Fredman G, Khan S. Specialized pro-resolving mediators enhance the clearance of dead cells. Immunol Rev 2023; 319:151-157. [PMID: 37787174 DOI: 10.1111/imr.13278] [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: 10/04/2023]
Abstract
The failure to resolve inflammation underpins to several prevalent diseases, like atherosclerosis, and so identifying ways to boost resolution is unmet clinical needs. The resolution of inflammation is governed by several factors such as specialized pro-resolving mediators (SPMs) that counter-regulate pro-inflammatory pathways and promote tissue repair without compromising host defense. A major function of nearly all SPMs is to enhance the clearance of dead cells or efferocytosis. As such, phagocytes, such as macrophages, are essential cellular players in the resolution of inflammation because of their ability to rapidly and efficiently clear dead cells. This review highlights the role of SPMs in the clearance of apoptotic and necroptotic cells and offers insights into how targeting efferocytosis may provide new treatments for non-resolving diseases, like atherosclerosis.
Collapse
Affiliation(s)
- Gabrielle Fredman
- The Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Sayeed Khan
- The Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| |
Collapse
|
20
|
Hu L, Lv Z, Gu Y, Zheng T, Kong Y, Mao W. A bibliometric analysis of efferocytosis in cardiovascular diseases from 2001 to 2022. Medicine (Baltimore) 2023; 102:e34366. [PMID: 37773819 PMCID: PMC10545234 DOI: 10.1097/md.0000000000034366] [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: 04/24/2023] [Accepted: 06/26/2023] [Indexed: 10/01/2023] Open
Abstract
INTRODUCTION In recent years, efferocytosis in cardiovascular diseases has become an intense area of research. However, only a few bibliometric analyses have been conducted in this area. In this review, we used CiteSpace 5.7. R2 and VOSviewer 1.6.17 software to perform text mining and knowledge map analysis. This study summarizes the latest progress, development paths, frontier research hotspots, and future research trends in this field. MATERIALS AND METHODS Studies on efferocytosis in cardiovascular diseases were downloaded from the Web of Science Core Collection. RESULTS In total, 327 studies published by 506 institutions across 42 countries and regions were identified. The number of studies on efferocytosis in cardiovascular diseases has increased over time. Arteriosclerosis Thrombosis and Vascular Biology published the highest number of articles and was the top co-cited journal. Tabas Ira. was the most prolific researcher and co-cited the most. The most productive countries were the United States and China. Columbia University, Harvard Medical School, and Brigham Women's Hospital were the 3 most productive institutions in the field of research. Keyword Co-occurrence, Clusters, and Burst analyses showed that inflammation, atherosclerosis, macrophages, and phagocytosis appeared with the highest frequency in these studies. CONCLUSION Multinational cooperation and multidisciplinary intersections are characteristic trends of development in the field, and the immune microenvironment, glycolysis, and lipid metabolism will be the focus of future research.
Collapse
Affiliation(s)
- Luoxia Hu
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, Hangzhou, China
| | - Zhengtian Lv
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, Hangzhou, China
| | - Yangyang Gu
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, Hangzhou, China
| | - Tiantian Zheng
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, Hangzhou, China
| | - Youjin Kong
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, Hangzhou, China
| | - Wei Mao
- Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, Hangzhou, China
- Department of Cardiology, Zhengjiang Hospital, Hangzhou, China
| |
Collapse
|
21
|
Seeley EH, Liu Z, Yuan S, Stroope C, Cockerham E, Rashdan NA, Delgadillo L, Finney AC, Kumar D, Das S, Razani B, Liu W, Traylor J, Orr AW, Rom O, Pattillo CB, Yurdagul A. Spatially Resolved Metabolites in Stable and Unstable Human Atherosclerotic Plaques Identified by Mass Spectrometry Imaging. Arterioscler Thromb Vasc Biol 2023; 43:1626-1635. [PMID: 37381983 PMCID: PMC10527524 DOI: 10.1161/atvbaha.122.318684] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/14/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Impairments in carbohydrate, lipid, and amino acid metabolism drive features of plaque instability. However, where these impairments occur within the atheroma remains largely unknown. Therefore, we sought to characterize the spatial distribution of metabolites within stable and unstable atherosclerosis in both the fibrous cap and necrotic core. METHODS Atherosclerotic tissue specimens from 9 unmatched individuals were scored based on the Stary classification scale and subdivided into stable and unstable atheromas. After performing mass spectrometry imaging on these samples, we identified over 850 metabolite-related peaks. Using MetaboScape, METASPACE, and Human Metabolome Database, we confidently annotated 170 of these metabolites and found over 60 of these were different between stable and unstable atheromas. We then integrated these results with an RNA-sequencing data set comparing stable and unstable human atherosclerosis. RESULTS Upon integrating our mass spectrometry imaging results with the RNA-sequencing data set, we discovered that pathways related to lipid metabolism and long-chain fatty acids were enriched in stable plaques, whereas reactive oxygen species, aromatic amino acid, and tryptophan metabolism were increased in unstable plaques. In addition, acylcarnitines and acylglycines were increased in stable plaques whereas tryptophan metabolites were enriched in unstable plaques. Evaluating spatial differences in stable plaques revealed lactic acid in the necrotic core, whereas pyruvic acid was elevated in the fibrous cap. In unstable plaques, 5-hydroxyindoleacetic acid was enriched in the fibrous cap. CONCLUSIONS Our work here represents the first step to defining an atlas of metabolic pathways involved in plaque destabilization in human atherosclerosis. We anticipate this will be a valuable resource and open new avenues of research in cardiovascular disease.
Collapse
Affiliation(s)
- Erin H. Seeley
- Department of Chemistry, University of Texas at Austin, TX, USA
| | - Zhipeng Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, IN, USA
| | - Shuai Yuan
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA, USA
| | - Chad Stroope
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Elizabeth Cockerham
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Nabil A Rashdan
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Luisa Delgadillo
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Dhananjay Kumar
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Sandeep Das
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Babak Razani
- Cardiovascular Division, Department of Medicine and Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
- John Cochran VA Medical Center, St. Louis, MO, USA
| | - Wanqing Liu
- Department of Pharmaceutical Sciences and Department of Pharmacology, Wayne State University, MI, USA
| | - James Traylor
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - A Wayne Orr
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Oren Rom
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Christopher B Pattillo
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| |
Collapse
|
22
|
Yang L, Zheng C, Xia YF, Dai Y, Wei ZF. 3, 3'-diindolylmethane enhances macrophage efferocytosis and subsequently relieves visceral pain via the AhR/Nrf2/Arg-1-mediated arginine metabolism pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154874. [PMID: 37216760 DOI: 10.1016/j.phymed.2023.154874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/28/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND 3, 3'-diindolylmethane (DIM), a classical aryl hydrocarbon receptor (AhR) agonist, has been shown to relieve neuropathic pain, but few studies have reported the efficacy of DIM in visceral pain under colitis condition. PURPOSE This study aimed to investigate the effect and mechanism of DIM on visceral pain under colitis condition. METHODS Cytotoxicity was performed using the MTT assay. RT-qPCR and ELISA assays were applied to determine the expression and release of algogenic substance P (SP), nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF). Flow cytometry was used to examine the apoptosis and efferocytosis. The expression of Arg-1-arginine metabolism-related enzymes was detected using western blotting assays. ChIP assays were used to examine the binding of Nrf2 to Arg-1. Mouse models of dextran sulfate sodium (DSS) were established to illustrate the effect of DIM and validate the mechanism in vivo. RESULTS DIM did not directly affect expressions and release of algogenic SP, NGF and BDNF in enteric glial cells (EGCs). However, when co-cultured with DIM-pre-treated RAW264.7 cells, the release of SP and NGF was decreased in lipopolysaccharides-stimulated EGCs. Furthermore, DIM increased the number of PKH67+ F4/80+ cells in the co-culture system of EGCs and RAW264.7 cells in vitro and alleviated visceral pain under colitis condition by regulating levels of SP and NGF as well as values of electromyogram (EMG), abdominal withdrawal reflex (AWR) and tail-flick latency (TFL) in vivo, which was significantly inhibited by efferocytosis inhibitor. Subsequently, DIM was found to down-regulate levels of intracellular arginine, up-regulate levels of ornithine, putrescine and Arg-1 but not extracellular arginine or other metabolic enzymes, and polyamine scavengers reversed the effect of DIM on efferocytosis and release of SP and NGF. Moving forward, Nrf2 transcription and the binding of Nrf2 to Arg-1-0.7 kb was enhanced by DIM, AhR antagonist CH223191 abolished the promotion of DIM on Arg-1 and efferocytosis. Finally, nor-NOHA validated the importance of Arg-1-dependent arginine metabolism in DIM-alleviated visceral pain. CONCLUSION DIM enhances macrophage efferocytosis in an arginine metabolism-dependent manner via "AhR-Nrf2/Arg-1" signals and inhibits the release of SP and NGF to relieve visceral pain under colitis condition. These findings provide a potential therapeutic strategy for the treatment of visceral pain in patients with colitis.
Collapse
Affiliation(s)
- Ling Yang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, China
| | - Chen Zheng
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, China
| | - Yu-Feng Xia
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, China
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, China.
| | - Zhi-Feng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, China.
| |
Collapse
|
23
|
Raju S, Botts SR, Blaser M, Prajapati K, Ho TWW, Ching C, Galant NJ, Fiddes L, Wu R, Clift CL, Pham T, Lee WL, Singh SA, Aikawa E, Fish JE, Howe KL. Endothelial cells secrete small extracellular vesicles bidirectionally containing distinct cargo to uniquely reprogram vascular cells in the circulation and vessel wall. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.28.538787. [PMID: 37162986 PMCID: PMC10168399 DOI: 10.1101/2023.04.28.538787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Rationale Extracellular vesicles (EVs) contain bioactive cargo including microRNAs (miRNAs) and proteins that are released by cells as a form of cell-cell communication. Endothelial cells (ECs) form the innermost lining of all blood vessels and thereby interface with cells in the circulation as well as cells residing in the vascular wall. It is unknown whether ECs have the capacity to release EVs capable of governing recipient cells within two separate compartments, and how this is affected by endothelial activation commonly seen in atheroprone regions. Objective Given their boundary location, we propose that ECs utilize bidirectional release of distinct EV cargo in quiescent and activated states to communicate with cells within the circulation and blood vessel wall. Methods and Results EVs were isolated from primary human aortic endothelial cells (ECs) (+/-IL-1β activation), quantified, and analysed by miRNA transcriptomics and proteomics. Compared to quiescent ECs, activated ECs increased EV release, with miRNA and protein cargo that were related to atherosclerosis. RNA sequencing of EV-treated monocytes and smooth muscle cells (SMCs) revealed that EVs from activated ECs altered pathways that were pro-inflammatory and atherogenic. Apical and basolateral EV release was assessed using ECs on transwells. ECs released more EVs apically, which increased with activation. Apical and basolateral EV cargo contained distinct transcriptomes and proteomes that were altered by EC activation. Notably, basolateral EC-EVs displayed greater changes in the EV secretome, with pathways specific to atherosclerosis. In silico analysis determined that compartment-specific cargo released by the apical and basolateral surfaces of ECs can reprogram monocytes and SMCs, respectively. Conclusions The demonstration that ECs are capable of polarized EV cargo loading and directional EV secretion reveals a novel paradigm for endothelial communication, which may ultimately enhance our ability to design endothelial-based therapeutics for cardiovascular diseases such as atherosclerosis where ECs are persistently activated.
Collapse
Affiliation(s)
- Sneha Raju
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Division of Vascular Surgery, Toronto General Hospital, Toronto, Canada
- Faculty of Medicine, University of Toronto, Toronto ON, Canada
| | - Steven R. Botts
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Faculty of Medicine, University of Toronto, Toronto ON, Canada
| | - Mark Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kamalben Prajapati
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Tse Wing Winnie Ho
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
| | - Crizza Ching
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | | | - Lindsey Fiddes
- Faculty of Medicine, University of Toronto, Toronto ON, Canada
| | - Ruilin Wu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Cassandra L. Clift
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Tan Pham
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Warren L Lee
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, Canada
| | - Kathryn L Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Division of Vascular Surgery, Toronto General Hospital, Toronto, Canada
- Faculty of Medicine, University of Toronto, Toronto ON, Canada
- Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, Canada
| |
Collapse
|
24
|
Kumar D, Pandit R, Yurdagul A. Mechanisms of continual efferocytosis by macrophages and its role in mitigating atherosclerosis. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00017. [PMID: 36710920 PMCID: PMC9869949 DOI: 10.1097/in9.0000000000000017] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/20/2022] [Indexed: 01/31/2023]
Abstract
Atherosclerotic cardiovascular disease is the leading cause of death worldwide. Rupture-prone atheromas that give rise to myocardial infarction and stroke are characterized by the presence of a necrotic core and a thin fibrous cap. During homeostasis, cellular debris and apoptotic cells are cleared quickly through a process termed "efferocytosis". However, clearance of apoptotic cells is significantly compromised in many chronic inflammatory diseases, including atherosclerosis. Emerging evidence suggests that impairments in efferocytosis drive necrotic core formation and contribute significantly to plaque vulnerability. Recently, it has been appreciated that successive rounds of efferocytosis, termed "continual efferocytosis", is mechanistically distinct from single efferocytosis and relies heavily on the metabolism and handling of apoptotic cell-derived cargo. In vivo, selective defects in continual efferocytosis drive secondary necrosis, impair inflammation resolution, and worsen atherosclerosis. This Mini Review focuses on our current understanding of the cellular and molecular mechanisms of continual efferocytosis and how dysregulations in this process mediate nonresolving inflammation. We will also discuss possible strategies to enhance efferocytosis when it fails.
Collapse
Affiliation(s)
- Dhananjay Kumar
- Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, USA
| | - Rajan Pandit
- Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, USA
| | - Arif Yurdagul
- Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, USA
| |
Collapse
|
25
|
Peng V, Cao S, Trsan T, Bando JK, Avila-Pacheco J, Cleveland JL, Clish C, Xavier RJ, Colonna M. Ornithine decarboxylase supports ILC3 responses in infectious and autoimmune colitis through positive regulation of IL-22 transcription. Proc Natl Acad Sci U S A 2022; 119:e2214900119. [PMID: 36279426 PMCID: PMC9659397 DOI: 10.1073/pnas.2214900119] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/27/2022] [Indexed: 01/14/2023] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) are RORγT+ lymphocytes that are predominately enriched in mucosal tissues and produce IL-22 and IL-17A. They are the innate counterparts of Th17 cells. While Th17 lymphocytes utilize unique metabolic pathways in their differentiation program, it is unknown whether ILC3s make similar metabolic adaptations. We employed single-cell RNA sequencing and metabolomic profiling of intestinal ILC subsets to identify an enrichment of polyamine biosynthesis in ILC3s, converging on the rate-limiting enzyme ornithine decarboxylase (ODC1). In vitro and in vivo studies demonstrated that exogenous supplementation with the polyamine putrescine or its biosynthetic substrate, ornithine, enhanced ILC3 production of IL-22. Conditional deletion of ODC1 in ILC3s impaired mouse antibacterial defense against Citrobacter rodentium infection, which was associated with a decrease in anti-microbial peptide production by the intestinal epithelium. Furthermore, in a model of anti-CD40 colitis, deficiency of ODC1 in ILC3s markedly reduced the production of IL-22 and severity of inflammatory colitis. We conclude that ILC3-intrinsic polyamine biosynthesis facilitates efficient defense against enteric pathogens as well as exacerbates autoimmune colitis, thus representing an attractive target to modulate ILC3 function in intestinal disease.
Collapse
Affiliation(s)
- Vincent Peng
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Siyan Cao
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO 63110
| | - Tihana Trsan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer K. Bando
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | | | - John L. Cleveland
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612
| | | | - Ramnik J. Xavier
- Broad Institute, Cambridge, MA 02412
- Center for Computational and Integrative Biology and Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Cambridge Street, Boston, MA 02114
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
26
|
Zhang J, Ding W, Zhao M, Liu J, Xu Y, Wan J, Wang M. Mechanisms of efferocytosis in determining inflammation resolution: Therapeutic potential and the association with cardiovascular disease. Br J Pharmacol 2022; 179:5151-5171. [PMID: 36028471 DOI: 10.1111/bph.15939] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/29/2022] Open
Abstract
Efferocytosis is defined as the clearance of apoptotic cells (ACs) in physiological and pathological states and is performed by efferocytes, such as macrophages. Efferocytosis can lead to the resolution of inflammation and restore tissue homoeostasis; however, the mechanisms of efferocytosis in determining inflammation resolution are still not completely understood, and the effects of efferocytosis on other proresolving properties need to be explored and explained. In this review, the process of efferocytosis will be summarized briefly, and then these mechanisms and effects will be thoroughly discussed. In addition, the association between the mechanisms of efferocytosis in determining inflammation resolution and cardiovascular diseases will also be reviewed, as an understanding of this association may provide information on novel treatment targets.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China.,department of radiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
27
|
Jarr KU, Kojima Y, Weissman IL, Leeper NJ. 2021 Jeffrey M. Hoeg Award Lecture: Defining the Role of Efferocytosis in Cardiovascular Disease: A Focus on the CD47 (Cluster of Differentiation 47) Axis. Arterioscler Thromb Vasc Biol 2022; 42:e145-e154. [PMID: 35387480 PMCID: PMC9183217 DOI: 10.1161/atvbaha.122.317049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/21/2022] [Indexed: 01/09/2023]
Abstract
A key feature of atherogenesis is the accumulation of diseased and dying cells within the lesional necrotic core. While the burden of intraplaque apoptotic cells may be driven in part by an increase in programmed cell death, mounting evidence suggests that their presence may primarily be dictated by a defect in programmed cell removal, or efferocytosis. In this brief review, we will summarize the evidence suggesting that inflammation-dependent changes within the plaque render target cells inedible and reduce the appetite of lesional phagocytes. We will present the genetic causation studies, which indicate these phenomena promote lesion expansion and plaque vulnerability, and the interventional data which suggest that these processes can be reversed. Particular emphasis is provided related to the antiphagocytic CD47 (cluster of differentiation 47) do not eat me axis, which has emerged as a novel antiatherosclerotic translational target that is predicted to provide benefit independent of traditional cardiovascular risk factors.
Collapse
Affiliation(s)
- Kai-Uwe Jarr
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yoko Kojima
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Irving L. Weissman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, United States of America
| | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, United States of America
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
28
|
Susser LI, Rayner KJ. Through the layers: how macrophages drive atherosclerosis across the vessel wall. J Clin Invest 2022; 132:157011. [PMID: 35499077 PMCID: PMC9057606 DOI: 10.1172/jci157011] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Leah I. Susser
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Katey J. Rayner
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
29
|
Kong P, Cui ZY, Huang XF, Zhang DD, Guo RJ, Han M. Inflammation and atherosclerosis: signaling pathways and therapeutic intervention. Signal Transduct Target Ther 2022; 7:131. [PMID: 35459215 PMCID: PMC9033871 DOI: 10.1038/s41392-022-00955-7] [Citation(s) in RCA: 449] [Impact Index Per Article: 149.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease driven by traditional and nontraditional risk factors. Genome-wide association combined with clonal lineage tracing and clinical trials have demonstrated that innate and adaptive immune responses can promote or quell atherosclerosis. Several signaling pathways, that are associated with the inflammatory response, have been implicated within atherosclerosis such as NLRP3 inflammasome, toll-like receptors, proprotein convertase subtilisin/kexin type 9, Notch and Wnt signaling pathways, which are of importance for atherosclerosis development and regression. Targeting inflammatory pathways, especially the NLRP3 inflammasome pathway and its regulated inflammatory cytokine interleukin-1β, could represent an attractive new route for the treatment of atherosclerotic diseases. Herein, we summarize the knowledge on cellular participants and key inflammatory signaling pathways in atherosclerosis, and discuss the preclinical studies targeting these key pathways for atherosclerosis, the clinical trials that are going to target some of these processes, and the effects of quelling inflammation and atherosclerosis in the clinic.
Collapse
Affiliation(s)
- Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Yang Cui
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Xiao-Fu Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Dan-Dan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Rui-Juan Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
30
|
Yurdagul A. Crosstalk Between Macrophages and Vascular Smooth Muscle Cells in Atherosclerotic Plaque Stability. Arterioscler Thromb Vasc Biol 2022; 42:372-380. [PMID: 35172605 PMCID: PMC8957544 DOI: 10.1161/atvbaha.121.316233] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Most acute cardiovascular events are due to plaque rupture, with atheromas containing large necrotic cores and thin fibrous caps being more susceptible to rupture and lesions with small necrotic cores and thick fibrous caps being more protected from rupture. Atherosclerotic plaques are comprised various extracellular matrix proteins, modified lipoprotein particles, and cells of different origins, that is, vascular cells and leukocytes. Although much has been revealed about the mechanisms that lead to plaque instability, several key areas remain incompletely understood. This In-Focus Review highlights processes related to cellular crosstalk and the role of the tissue microenvironment in determining cell function and plaque stability. Recent advances highlight critical underpinnings of atherosclerotic plaque vulnerability, particularly impairments in the ability of macrophages to clear dead cells and phenotypic switching of vascular smooth muscle cells. However, these processes do not occur in isolation, as crosstalk between macrophages and vascular smooth muscle cells and interactions with their surrounding microenvironment play a significant role in determining plaque stability. Understanding these aspects of cellular crosstalk within an atherosclerotic plaque may shed light on how to modify cell behavior and identify novel approaches to transform rupture-prone atheromas into stable lesions.
Collapse
Affiliation(s)
- Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences, Shreveport
| |
Collapse
|
31
|
Abstract
Resolution is an active and highly coordinated process that occurs in response to inflammation to limit tissue damage and promote repair. When the resolution program fails, inflammation persists. It is now understood that failed resolution is a major underlying cause of many chronic inflammatory diseases. Here, we will review the major failures of resolution in atherosclerosis, including the imbalance of proinflammatory to pro-resolving mediator production, impaired clearance of dead cells, and functional changes in immune cells that favor ongoing inflammation. In addition, we will briefly discuss new concepts that are emerging as possible regulators of resolution and highlight the translational significance for the field.
Collapse
Affiliation(s)
- Amanda C. Doran
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt Institute for Infection, Immunology, and Inflammation, Department of Molecular Physiology and Biophysics, Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
32
|
Decker C, Sadhu S, Fredman G. Pro-Resolving Ligands Orchestrate Phagocytosis. Front Immunol 2021; 12:660865. [PMID: 34177900 PMCID: PMC8222715 DOI: 10.3389/fimmu.2021.660865] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/06/2021] [Indexed: 12/18/2022] Open
Abstract
The resolution of inflammation is a tissue protective program that is governed by several factors including specialized pro-resolving mediators (SPMs), proteins, gasses and nucleotides. Pro-resolving mediators activate counterregulatory programs to quell inflammation and promote tissue repair in a manner that does not compromise host defense. Phagocytes like neutrophils and macrophages play key roles in the resolution of inflammation because of their ability to remove debris, microbes and dead cells through processes including phagocytosis and efferocytosis. Emerging evidence suggests that failed resolution of inflammation and defective phagocytosis or efferocytosis underpins several prevalent human diseases. Therefore, understanding factors and mechanisms associated with enhancing these processes is a critical need. SPMs enhance phagocytosis and efferocytosis and this review will highlight mechanisms associated with their actions.
Collapse
Affiliation(s)
- Christa Decker
- The Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Sudeshna Sadhu
- The Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Gabrielle Fredman
- The Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| |
Collapse
|
33
|
Dead cell and debris clearance in the atherosclerotic plaque: Mechanisms and therapeutic opportunities to promote inflammation resolution. Pharmacol Res 2021; 170:105699. [PMID: 34087352 DOI: 10.1016/j.phrs.2021.105699] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/05/2021] [Accepted: 05/28/2021] [Indexed: 01/08/2023]
Abstract
Phagocytic clearance of dead cells and debris is critical for inflammation resolution and maintenance of tissue homeostasis. Consequently, defective clearance of dead cells and debris is associated with initiation and exacerbation of several autoimmune disorders and chronic inflammatory diseases such as atherosclerosis. The progressive loss of dead cell clearance capacity within the atherosclerotic plaque leads to accumulation of necrotic cells, chronic non-resolving inflammation, and expansion of the necrotic core, which triggers atherosclerotic plaque rupture and clinical manifestation of acute thrombotic cardiovascular adverse events. In this review, we describe the fundamental molecular and cellular mechanisms of dead cell clearance and how it goes awry in atherosclerosis. Finally, we highlight novel therapeutic strategies that enhance dead cell and debris clearance within the atherosclerotic plaque to promote inflammation resolution and atherosclerotic plaque stabilization.
Collapse
|
34
|
Abstract
Billions of cells undergo apoptosis daily and are swiftly removed by macrophages through an evolutionarily conserved program termed "efferocytosis". Consequently, macromolecules within an apoptotic cell significantly burden a phagocyte with nutrients, such as lipids, oligonucleotides, and amino acids. In response to this nutrient overload, metabolic reprogramming must occur for the process of efferocytosis to remain non-phlogistic and to execute successive rounds of efferocytosis. The inability to undergo metabolic reprogramming after efferocytosis drives inflammation and impairs its resolution, often promoting many chronic inflammatory diseases. This is particularly evident for atherosclerosis, as metabolic reprogramming alters macrophage function in every stage of atherosclerosis, from the early formation of benign lesions to the progression of clinically relevant atheromas and during atherosclerosis regression upon aggressive lipid-lowering. This Review focuses on the metabolic pathways utilized upon apoptotic cell ingestion, the consequences of these metabolic pathways in macrophage function thereafter, and the role of metabolic reprogramming during atherosclerosis. Due to the growing interest in this new field, I introduce a new term, "efferotabolism", as a means to define the process by which macrophages break down, metabolize, and respond to AC-derived macromolecules. Understanding these aspects of efferotabolism will shed light on novel strategies to combat atherosclerosis and compromised inflammation resolution.
Collapse
|