1
|
Xie W, Kong Y, Ren C, Wen Y, Ying M, Xing H. Chemistries on the inner leaflet of the cell membrane. Chem Commun (Camb) 2025; 61:2387-2402. [PMID: 39810742 DOI: 10.1039/d4cc05186f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The cell membrane, characterized by its inherent asymmetry, functions as a dynamic barrier that regulates numerous cellular activities. This Highlight aims to provide the chemistry community with a comprehensive overview of the intriguing and underexplored inner leaflet, encompassing both fundamental biology and emerging synthetic modification strategies. We begin by describing the asymmetric nature of the plasma membrane, with a focus on the distinct roles of lipids, proteins, and glycan chains, highlighting the composition and biofunctions of the inner leaflet and the biological mechanisms that sustain membrane asymmetry. Next, we explore chemical biological strategies for engineering the inner leaflet, including genetic engineering, transmembrane peptides, and liposome fusion-based transport. In the perspective section, we discuss the challenges in developing chemistries for the inner leaflet of the cell membrane, aiming to inspire researchers and collaborators to explore this field and address its unanswered biological questions.
Collapse
Affiliation(s)
- Wenxue Xie
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | - Yuhan Kong
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | - Cong Ren
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | - Yujian Wen
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | | | - Hang Xing
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
- Research Institute of Hunan University in Chongqing, Chongqing, 401100, China
| |
Collapse
|
2
|
Hu L, Chen Z, Lu J, Jiang S, Lin H, Zhou J, Wang N, Ding C, Ni W, Peng H, Li Y, He X, Li J, Jing C, Cao Y, Zhou H, Yan F, Chen G. Extracellular Vesicles From Bone Marrow-Derived Macrophages Enriched in ARG1 Enhance Microglial Phagocytosis and Haematoma Clearance Following Intracerebral Haemorrhage. J Extracell Vesicles 2025; 14:e70041. [PMID: 39868438 PMCID: PMC11770371 DOI: 10.1002/jev2.70041] [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: 08/06/2024] [Revised: 11/21/2024] [Accepted: 12/24/2024] [Indexed: 01/28/2025] Open
Abstract
Microglial phagocytosis of haematomas is crucial for neural functional recovery following intracerebral haemorrhage (ICH), a process regulated by various factors from within and outside the central nervous system (CNS). Extracellular vesicles (EVs), significant mediators of intercellular communication, have been demonstrated to play a pivotal role in the pathogenesis and progression of CNS diseases. However, the regulatory role of endogenous EVs on the phagocytic capacity of microglia post-ICH remains elusive. Utilising multi-omics analysis of brain tissue-derived EVs proteomics and single-cell RNA sequencing, this study identified that bone marrow-derived macrophages (BMDMs) potentially enhance microglial phagocytosis via EVs following ICH. By blocking BMDMs and reducing ARG1 in BMDM-derived EVs, we demonstrated that BMDMs facilitate erythrophagocytosis by delivering ARG1 to microglia via EVs post-ICH. EVs-carried ARG1 was found to augment phagocytosis by promoting RAC1-dependent cytoskeletal remodelling in microglia. Collectively, this research uncovers an intercellular communication pathway from BMDMs to microglia mediated by EVs post-ICH. This provides a novel paradigm for EV-mediated intercellular communication mechanisms and suggests a promising therapeutic potential for BMDM-derived EVs in the treatment of ICH.
Collapse
Affiliation(s)
- Libin Hu
- Department of Neurosurgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical DiseasesHangzhouChina
- State Key Laboratory of Transvascular Implantation DevicesHangzhouChina
| | - Zihang Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical DiseasesHangzhouChina
- State Key Laboratory of Transvascular Implantation DevicesHangzhouChina
| | - Jianglong Lu
- Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Shandong Jiang
- Department of Neurosurgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical DiseasesHangzhouChina
| | - Haopu Lin
- Department of Neurosurgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical DiseasesHangzhouChina
| | - Jiayin Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical DiseasesHangzhouChina
| | - Ning Wang
- Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Chao Ding
- Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Weifang Ni
- Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Haitao Peng
- Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Yin Li
- Department of Neurosurgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical DiseasesHangzhouChina
| | - Xuchao He
- Department of Neurosurgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical DiseasesHangzhouChina
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical DiseasesHangzhouChina
| | - Chaohui Jing
- Department of NeurosurgeryXinHua Hospital affiliated to Shanghai JiaoTong University School of MedicineShanghaiChina
| | - Yang Cao
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of MedicineWestlake UniversityHangzhouChina
| | - Hang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical DiseasesHangzhouChina
- State Key Laboratory of Transvascular Implantation DevicesHangzhouChina
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical DiseasesHangzhouChina
- State Key Laboratory of Transvascular Implantation DevicesHangzhouChina
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical DiseasesHangzhouChina
- State Key Laboratory of Transvascular Implantation DevicesHangzhouChina
| |
Collapse
|
3
|
Chavula T, To S, Smith J, Pedroza M, Nimri J, Devaraj S, Wenderfer S, Vogel TP, Agarwal SK. CADHERIN-11 regulation of myeloid phagocytes and autoimmune inflammation in murine lupus. J Autoimmun 2024; 145:103197. [PMID: 38447248 DOI: 10.1016/j.jaut.2024.103197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND AND OBJECTIVE Understanding the regulation of efferocytosis by myeloid phagocytes is important in identifying novel targets in systemic lupus erythematosus (SLE). Cadherin-11 (CDH11), a cell adhesion molecule, is implicated in inflammatory arthritis and fibrosis and recently been shown to regulate macrophage phagocytosis. The extent and mechanism of this regulation is unknown. Our objective was to examine the extent to which CDH11 regulates myeloid phagocytes and contributes to autoimmunity and tissue inflammation. METHODS We analyzed efferocytosis in macrophages and dendritic cells (DCs) from WT and Cdh11-/- mice and investigated the mechanisms in vitro. We investigated the role of CDH11 in disease development in vivo using the pristane induced lupus model. To translate the clinical relevance of CDH11 in human disease, we measured serum CDH11 levels in two independent pediatric SLE (pSLE) cohorts and healthy controls. RESULTS Using bone marrow derived macrophages (BMDMs) and DCs (BMDCs), we found impaired efferocytosis in phagocytes from Cdh11-/- mice, mediated by downregulated efferocytosis receptor expression and RhoGTPase activation. Specifically, loss of CDH11 downregulated Mertk expression and Rac1 activation in BMDMs, and integrin αVβ3 expression and Cdc42 activation in BMDCs, highlighting distinct pathways. In vivo, Cdh11-/- mice displayed defective efferocytosis and increased accumulation of apoptotic debris in pristane-induced lupus. Further, Cdh11-/- mice had enhanced systemic inflammation and autoimmune inflammation with increased anti-dsDNA autoantibodies, splenomegaly, type I interferons, and inflammatory cytokines. Paradoxically, at the tissue level, Cdh11-/- mice were protected against glomerulonephritis, indicating a dual role in murine lupus. Finally, SLE patients had increased serum CDH11 compared to controls. CONCLUSION This study highlights a novel role of CDH11 in regulating myeloid cells and efferocytosis and its potential as a contributor to development in autoimmunity murine lupus. Despite the increase in autoimmunity, Cdh11-/- mice developed decreased tissue inflammation and damage.
Collapse
Affiliation(s)
- Thandiwe Chavula
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sarah To
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer Smith
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mesias Pedroza
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jena Nimri
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sridevi Devaraj
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Pathology Department, Texas Children's Hospital, Houston, TX, USA
| | - Scott Wenderfer
- Department of Pediatric Nephrology, B.C. Children's Hospital, Vancouver, BC, Canada
| | - Tiphanie P Vogel
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics-Rheumatology, Baylor College of Medicine, Houston, TX, USA; Division of Rheumatology, Texas Children's Hospital, Houston, TX, USA
| | - Sandeep K Agarwal
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
4
|
Zhao Y, Li M, Mao J, Su Y, Huang X, Xia W, Leng X, Zan T. Immunomodulation of wound healing leading to efferocytosis. SMART MEDICINE 2024; 3:e20230036. [PMID: 39188510 PMCID: PMC11235971 DOI: 10.1002/smmd.20230036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/18/2023] [Indexed: 08/28/2024]
Abstract
Effectively eliminating apoptotic cells is precisely controlled by a variety of signaling molecules and a phagocytic effect known as efferocytosis. Abnormalities in efferocytosis may bring about the development of chronic conditions, including angiocardiopathy, chronic inflammatory diseases and autoimmune diseases. During wound healing, failure of efferocytosis leads to the collection of apoptosis, the release of necrotic material and chronic wounds that are difficult to heal. In addition to the traditional phagocytes-macrophages, other important cell species including dendritic cells, neutrophils, vascular endothelial cells, fibroblasts and keratinocytes contribute to wounding healing. This review summarizes how efferocytosis-mediated immunomodulation plays a repair-promoting role in wound healing, providing new insights for patients suffering from various cutaneous wounds.
Collapse
Affiliation(s)
- Yun Zhao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Cosmetic and Plastic SurgeryAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Minxiong Li
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jiayi Mao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yinghong Su
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xin Huang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenzheng Xia
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiangfeng Leng
- Department of Cosmetic and Plastic SurgeryAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Tao Zan
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
5
|
Socodato R, Almeida TO, Portugal CC, Santos ECS, Tedim-Moreira J, Galvão-Ferreira J, Canedo T, Baptista FI, Magalhães A, Ambrósio AF, Brakebusch C, Rubinstein B, Moreira IS, Summavielle T, Pinto IM, Relvas JB. Microglial Rac1 is essential for experience-dependent brain plasticity and cognitive performance. Cell Rep 2023; 42:113447. [PMID: 37980559 DOI: 10.1016/j.celrep.2023.113447] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/14/2023] [Accepted: 10/31/2023] [Indexed: 11/21/2023] Open
Abstract
Microglia, the largest population of brain immune cells, continuously interact with synapses to maintain brain homeostasis. In this study, we use conditional cell-specific gene targeting in mice with multi-omics approaches and demonstrate that the RhoGTPase Rac1 is an essential requirement for microglia to sense and interpret the brain microenvironment. This is crucial for microglia-synapse crosstalk that drives experience-dependent plasticity, a fundamental brain property impaired in several neuropsychiatric disorders. Phosphoproteomics profiling detects a large modulation of RhoGTPase signaling, predominantly of Rac1, in microglia of mice exposed to an environmental enrichment protocol known to induce experience-dependent brain plasticity and cognitive performance. Ablation of microglial Rac1 affects pathways involved in microglia-synapse communication, disrupts experience-dependent synaptic remodeling, and blocks the gains in learning, memory, and sociability induced by environmental enrichment. Our results reveal microglial Rac1 as a central regulator of pathways involved in the microglia-synapse crosstalk required for experience-dependent synaptic plasticity and cognitive performance.
Collapse
Affiliation(s)
- Renato Socodato
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal.
| | - Tiago O Almeida
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; ICBAS - School of Medicine and Biomedical Sciences, Porto, Portugal
| | - Camila C Portugal
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Evelyn C S Santos
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Joana Tedim-Moreira
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - João Galvão-Ferreira
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Teresa Canedo
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Filipa I Baptista
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra Institute for Clinical and Biomedical Research (iCBR), and Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
| | - Ana Magalhães
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - António F Ambrósio
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra Institute for Clinical and Biomedical Research (iCBR), and Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
| | - Cord Brakebusch
- Molecular Pathology Section, BRIC, Københavns Biocenter, Copenhagen, Denmark
| | | | - Irina S Moreira
- Department of Life Sciences, Center for Innovative Biomedicine and Biotechnology (CIBB) and CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Teresa Summavielle
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; ESS.PP, Escola Superior de Saúde do Politécnico do Porto, Porto, Portugal
| | - Inês Mendes Pinto
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - João B Relvas
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal.
| |
Collapse
|
6
|
Wu M, Ge Y, Wang E, Liao Q, Ren Z, Yu Y, Zhu G, Liu C, Zhang M, Su H, Shen H, Chen Y, Wang L, Wang Y, Li M, Bian Z, Chai J, Ye RD, Lu J. Enhancement of efferocytosis through biased FPR2 signaling attenuates intestinal inflammation. EMBO Mol Med 2023; 15:e17815. [PMID: 37994307 PMCID: PMC10701612 DOI: 10.15252/emmm.202317815] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/24/2023] Open
Abstract
Efficient clearance of dying cells (efferocytosis) is an evolutionarily conserved process for tissue homeostasis. Genetic enhancement of efferocytosis exhibits therapeutic potential for inflammation resolution and tissue repair. However, pharmacological approaches to enhance efferocytosis remain sparse due to a lack of targets for modulation. Here, we report the identification of columbamine (COL) which enhances macrophage-mediated efferocytosis and attenuates intestinal inflammation in a murine colitis model. COL enhances efferocytosis by promoting LC3-associated phagocytosis (LAP), a non-canonical form of autophagy. Transcriptome analysis and pharmacological characterization revealed that COL is a biased agonist that occupies a part of the ligand binding pocket of formyl peptide receptor 2 (FPR2), a G-protein coupled receptor involved in inflammation regulation. Genetic ablation of the Fpr2 gene or treatment with an FPR2 antagonist abolishes COL-induced efferocytosis, anti-colitis activity and LAP. Taken together, our study identifies FPR2 as a potential target for modulating LC3-associated efferocytosis to alleviate intestinal inflammation and highlights the therapeutic value of COL, a natural and biased agonist of FPR2, in the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Ming‐Yue Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau SARChina
- Department of Gastroenterology, The First Affiliated Hospital (Southwest Hospital)Third Military Medical University (Army Medical University)ChongqingChina
| | - Yun‐Jun Ge
- Kobilka Institute of Innovative Drug Discovery, School of MedicineThe Chinese University of Hong KongShenzhenChina
- Department of Basic Medical Science, Wuxi School of MedicineJiangnan UniversityWuxiChina
| | - Er‐Jin Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau SARChina
| | - Qi‐Wen Liao
- Kobilka Institute of Innovative Drug Discovery, School of MedicineThe Chinese University of Hong KongShenzhenChina
| | - Zheng‐Yu Ren
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau SARChina
| | - Yang Yu
- Engineering Research Center of Cell and Therapeutic Antibody Medicine, Ministry of Education, School of PharmacyShanghai Jiao Tong UniversityShanghaiChina
| | - Guoyuan Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and HealthMacau University of Science and TechnologyMacau SARChina
| | - Chun‐Ping Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau SARChina
- Department of Cardiovascular MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchUniversity of MacauMacau SARChina
| | - Meng‐Ni Zhang
- Department of Gastroenterology, The First Affiliated Hospital (Southwest Hospital)Third Military Medical University (Army Medical University)ChongqingChina
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau SARChina
| | - Han‐Ming Shen
- Faculty of Health SciencesUniversity of MacauMacau SARChina
| | - Ye Chen
- Integrative Microecology Center, Department of Gastroenterology, Shenzhen HospitalSouthern Medical UniversityShenzhen, GuangzhouChina
| | - Lei Wang
- Department of Cardiovascular MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Yi‐Tao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau SARChina
| | - Min Li
- School of Chinese MedicineHong Kong Baptist UniversityHongkong SARChina
| | - Zhaoxiang Bian
- School of Chinese MedicineHong Kong Baptist UniversityHongkong SARChina
| | - Jin Chai
- Department of Gastroenterology, The First Affiliated Hospital (Southwest Hospital)Third Military Medical University (Army Medical University)ChongqingChina
| | - Richard D Ye
- Kobilka Institute of Innovative Drug Discovery, School of MedicineThe Chinese University of Hong KongShenzhenChina
- The Second Affiliated Hospital, School of MedicineThe Chinese University of Hong KongShenzhenChina
| | - Jia‐Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau SARChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchUniversity of MacauMacau SARChina
| |
Collapse
|
7
|
Schilperoort M, Ngai D, Sukka SR, Avrampou K, Shi H, Tabas I. The role of efferocytosis-fueled macrophage metabolism in the resolution of inflammation. Immunol Rev 2023; 319:65-80. [PMID: 37158427 PMCID: PMC10615666 DOI: 10.1111/imr.13214] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/20/2023] [Indexed: 05/10/2023]
Abstract
The phagocytosis of dying cells by macrophages, termed efferocytosis, is a tightly regulated process that involves the sensing, binding, engulfment, and digestion of apoptotic cells. Efferocytosis not only prevents tissue necrosis and inflammation caused by secondary necrosis of dying cells, but it also promotes pro-resolving signaling in macrophages, which is essential for tissue resolution and repair following injury or inflammation. An important factor that contributes to this pro-resolving reprogramming is the cargo that is released from apoptotic cells after their engulfment and phagolysosomal digestion by macrophages. The apoptotic cell cargo contains amino acids, nucleotides, fatty acids, and cholesterol that function as metabolites and signaling molecules to bring about this re-programming. Here, we review efferocytosis-induced changes in macrophage metabolism that mediate the pro-resolving functions of macrophages. We also discuss various strategies, challenges, and future perspectives related to drugging efferocytosis-fueled macrophage metabolism as strategy to dampen inflammation and promote resolution in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Maaike Schilperoort
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - David Ngai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Santosh R Sukka
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kleopatra Avrampou
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hongxue Shi
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Physiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
8
|
Moon B, Yang S, Moon H, Lee J, Park D. After cell death: the molecular machinery of efferocytosis. Exp Mol Med 2023; 55:1644-1651. [PMID: 37612408 PMCID: PMC10474042 DOI: 10.1038/s12276-023-01070-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 08/25/2023] Open
Abstract
Cells constituting a multicellular organism die in a variety of ways throughout life, and most of them die via apoptosis under normal conditions. The occurrence of apoptosis is especially prevalent during development and in tissues with a high cellular turnover rate, such as the thymus and bone marrow. Interestingly, although the number of apoptotic cells produced daily is known to be innumerable in a healthy adult human body, apoptotic cells are rarely observed. This absence is due to the existence of a cellular process called efferocytosis that efficiently clears apoptotic cells. Studies over the past decades have focused on how phagocytes are able to remove apoptotic cells specifically, swiftly, and continuously, resulting in defined molecular and cellular events. In this review, we will discuss the current understanding of the clearance of apoptotic cells at the molecular level.
Collapse
Affiliation(s)
- Byeongjin Moon
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
- Cell Mechanobiology Laboratory, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Susumin Yang
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
- Cell Mechanobiology Laboratory, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Hyunji Moon
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
- Cell Mechanobiology Laboratory, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Juyeon Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
- Cell Mechanobiology Laboratory, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Daeho Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea.
- Cell Mechanobiology Laboratory, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea.
| |
Collapse
|
9
|
She Y, Xu X, Yu Q, Yang X, He J, Tang XX. Elevated expression of macrophage MERTK exhibits profibrotic effects and results in defective regulation of efferocytosis function in pulmonary fibrosis. Respir Res 2023; 24:118. [PMID: 37120511 PMCID: PMC10148433 DOI: 10.1186/s12931-023-02424-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 03/08/2023] [Indexed: 05/01/2023] Open
Abstract
Increased apoptosis of alveolar epithelial cells is a prominent feature of pulmonary fibrosis. Macrophage efferocytosis, phagocytosis of apoptotic cells by macrophages, is crucial for maintaining tissue homeostasis. Expression of Mer tyrosine kinase (MERTK, an important recognition receptor in efferocytosis) in macrophages is thought to be associated with fibrosis. However, how macrophage MERTK affects pulmonary fibrosis and whether it depends on efferocytosis are not yet clear. Here, we found elevated MERTK expression in lung macrophages from IPF patients and mice with bleomycin-induced pulmonary fibrosis. In vitro experiments showed that macrophages overexpressing MERTK exhibit profibrotic effects and that macrophage efferocytosis abrogates the profibrotic effect of MERTK by downregulating MERTK, forming a negative regulatory loop. In pulmonary fibrosis, this negative regulation is defective, and MERTK mainly exhibits profibrotic effects. Our study reveals a previously unsuspected profibrotic effect of elevated macrophage MERTK in pulmonary fibrosis and defective regulation of efferocytosis function as a result of that elevation, suggesting that targeting MERTK in macrophages may help to attenuate pulmonary fibrosis.
Collapse
Affiliation(s)
- Yixin She
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xin Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingyang Yu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiangsheng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianxing He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Xiao Xiao Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Laboratory, Bio-Island, Guangzhou, China.
| |
Collapse
|
10
|
Guo Y, Meng L, Wang M, Zhong Z, Li D, Zhang Y, Li H, Zhang H, Seim I, Li Y, Jiang A, Ji Q, Su X, Chen J, Fan G, Li C, Liu S. Hologenome analysis reveals independent evolution to chemosymbiosis by deep-sea bivalves. BMC Biol 2023; 21:51. [PMID: 36882766 PMCID: PMC9993606 DOI: 10.1186/s12915-023-01551-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/22/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND Bivalves have independently evolved a variety of symbiotic relationships with chemosynthetic bacteria. These relationships range from endo- to extracellular interactions, making them ideal for studies on symbiosis-related evolution. It is still unclear whether there are universal patterns to symbiosis across bivalves. Here, we investigate the hologenome of an extracellular symbiotic thyasirid clam that represents the early stages of symbiosis evolution. RESULTS We present a hologenome of Conchocele bisecta (Bivalvia: Thyasiridae) collected from deep-sea hydrothermal vents with extracellular symbionts, along with related ultrastructural evidence and expression data. Based on ultrastructural and sequencing evidence, only one dominant Thioglobaceae bacteria was densely aggregated in the large bacterial chambers of C. bisecta, and the bacterial genome shows nutritional complementarity and immune interactions with the host. Overall, gene family expansions may contribute to the symbiosis-related phenotypic variations in different bivalves. For instance, convergent expansions of gaseous substrate transport families in the endosymbiotic bivalves are absent in C. bisecta. Compared to endosymbiotic relatives, the thyasirid genome exhibits large-scale expansion in phagocytosis, which may facilitate symbiont digestion and account for extracellular symbiotic phenotypes. We also reveal that distinct immune system evolution, including expansion in lipopolysaccharide scavenging and contraction of IAP (inhibitor of apoptosis protein), may contribute to the different manners of bacterial virulence resistance in C. bisecta. CONCLUSIONS Thus, bivalves employ different pathways to adapt to the long-term co-existence with their bacterial symbionts, further highlighting the contribution of stochastic evolution to the independent gain of a symbiotic lifestyle in the lineage.
Collapse
Affiliation(s)
- Yang Guo
- Center of Deep-Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Lingfeng Meng
- BGI-Qingdao, BGI-Shenzhen, Qingdao, 266555, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Minxiao Wang
- Center of Deep-Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
- Pilot National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Zhaoshan Zhong
- Center of Deep-Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Denghui Li
- BGI-Qingdao, BGI-Shenzhen, Qingdao, 266555, China
| | - Yaolei Zhang
- BGI-Qingdao, BGI-Shenzhen, Qingdao, 266555, China
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Hanbo Li
- BGI-Qingdao, BGI-Shenzhen, Qingdao, 266555, China
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Huan Zhang
- Center of Deep-Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Inge Seim
- Integrative Biology Laboratory, College of Life Sciences, Nanjing Normal University, Nanjing, 210046, China
- School of Biology and Environmental Science, Queensland University of Technology, Brisbane, QLD, 4000, Australia
| | - Yuli Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Aijun Jiang
- BGI-Qingdao, BGI-Shenzhen, Qingdao, 266555, China
| | - Qianyue Ji
- BGI-Qingdao, BGI-Shenzhen, Qingdao, 266555, China
| | - Xiaoshan Su
- BGI-Qingdao, BGI-Shenzhen, Qingdao, 266555, China
| | - Jianwei Chen
- BGI-Qingdao, BGI-Shenzhen, Qingdao, 266555, China
| | - Guangyi Fan
- BGI-Qingdao, BGI-Shenzhen, Qingdao, 266555, China.
- BGI-Shenzhen, Shenzhen, 518083, China.
| | - Chaolun Li
- Center of Deep-Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China.
- College of Marine Science, University of Chinese Academy of Sciences, Qingdao, 266400, China.
- South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, China.
| | - Shanshan Liu
- BGI-Qingdao, BGI-Shenzhen, Qingdao, 266555, China.
- Qingdao Key Laboratory of Marine Genomics, BGI-qingdao, Qingdao, China.
| |
Collapse
|
11
|
Chouinard FC, Davis L, Gilbert C, Bourgoin SG. Functional Role of AGAP2/PIKE-A in Fcγ Receptor-Mediated Phagocytosis. Cells 2022; 12:cells12010072. [PMID: 36611866 PMCID: PMC9818964 DOI: 10.3390/cells12010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
In phagocytes, cytoskeletal and membrane remodeling is finely regulated at the phagocytic cup. Various smaFll G proteins, including those of the Arf family, control these dynamic processes. Human neutrophils express AGAP2, an Arf GTPase activating protein (ArfGAP) that regulates endosomal trafficking and focal adhesion remodeling. We first examined the impact of AGAP2 on phagocytosis in CHO cells stably expressing the FcγRIIA receptor (CHO-IIA). In unstimulated CHO-IIA cells, AGAP2 only partially co-localized with cytoskeletal elements and intracellular compartments. In CHO-IIA cells, AGAP2 transiently accumulated at actin-rich phagocytic cups and increased Fcγ receptor-mediated phagocytosis. Enhanced phagocytosis was not dependent on the N-terminal GTP-binding protein-like (GLD) domain of AGAP2. AGAP2 deleted of its GTPase-activating protein (GAP) domain was not recruited to phagocytic cups and did not enhance the engulfment of IgG-opsonized beads. However, the GAP-deficient [R618K]AGAP2 transiently localized at the phagocytic cups and enhanced phagocytosis. In PLB-985 cells differentiated towards a neutrophil-like phenotype, silencing of AGAP2 reduced phagocytosis of opsonized zymosan. In human neutrophils, opsonized zymosan or monosodium urate crystals induced AGAP2 phosphorylation. The data indicate that particulate agonists induce AGAP2 phosphorylation in neutrophils. This study highlights the role of AGAP2 and its GAP domain but not GAP activity in FcγR-dependent uptake of opsonized particles.
Collapse
Affiliation(s)
- François C. Chouinard
- Centre de Recherche du CHU de Québec—Université Laval, Québec City, QC G1V 4G2, Canada
| | - Lynn Davis
- Centre de Recherche du CHU de Québec—Université Laval, Québec City, QC G1V 4G2, Canada
| | - Caroline Gilbert
- Centre de Recherche du CHU de Québec—Université Laval, Québec City, QC G1V 4G2, Canada
- Département de Microbiologie-Infectiologie et D’immunologie, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Sylvain G. Bourgoin
- Centre de Recherche du CHU de Québec—Université Laval, Québec City, QC G1V 4G2, Canada
- Département de Microbiologie-Infectiologie et D’immunologie, Université Laval, Québec City, QC G1V 0A6, Canada
- Centre ARThrite de L’université Laval, Québec City, QC G1V 4G2, Canada
- Correspondence:
| |
Collapse
|
12
|
Proteomic mapping and optogenetic manipulation of membrane contact sites. Biochem J 2022; 479:1857-1875. [PMID: 36111979 PMCID: PMC9555801 DOI: 10.1042/bcj20220382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022]
Abstract
Membrane contact sites (MCSs) mediate crucial physiological processes in eukaryotic cells, including ion signaling, lipid metabolism, and autophagy. Dysregulation of MCSs is closely related to various diseases, such as type 2 diabetes mellitus (T2DM), neurodegenerative diseases, and cancers. Visualization, proteomic mapping and manipulation of MCSs may help the dissection of the physiology and pathology MCSs. Recent technical advances have enabled better understanding of the dynamics and functions of MCSs. Here we present a summary of currently known functions of MCSs, with a focus on optical approaches to visualize and manipulate MCSs, as well as proteomic mapping within MCSs.
Collapse
|
13
|
Moreiras H, Bento-Lopes L, Neto MV, Escrevente C, Cabaço LC, Hall MJ, Ramalho JS, Seabra MC, Barral DC. Melanocore uptake by keratinocytes occurs through phagocytosis and involves Protease-activated receptor-2 internalization. Traffic 2022; 23:331-345. [PMID: 35426185 PMCID: PMC9543991 DOI: 10.1111/tra.12843] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 03/26/2022] [Accepted: 04/02/2022] [Indexed: 12/01/2022]
Abstract
In the skin epidermis, melanin is produced and stored within melanosomes in melanocytes, and then transferred to keratinocytes. Different models have been proposed to explain the melanin transfer mechanism, which differ essentially in how melanin is transferred - either in a membrane-bound melanosome or as a melanosome core, i.e., melanocore. Here, we investigated the endocytic route followed by melanocores and melanosomes during internalization by keratinocytes, by comparing the uptake of melanocores isolated from the supernatant of melanocyte cultures, with melanosomes isolated from melanocytes. We show that inhibition of actin dynamics impairs the uptake of both melanocores and melanosomes. Moreover, depletion of critical proteins involved in actin-dependent uptake mechanisms, namely Rac1, CtBP1/BARS, Cdc42 or RhoA, together with inhibition of Rac1-dependent signaling pathways or macropinocytosis suggest that melanocores are internalized by phagocytosis, whereas melanosomes are internalized by macropinocytosis. Interestingly, we found that Rac1, Cdc42 and RhoA are differently activated by melanocore or melanosome stimulation, supporting the existence of two distinct internalization routes of melanin internalization. Furthermore, we show that melanocore uptake induces Protease-activated receptor-2 (PAR-2) internalization by keratinocytes to a higher extent than melanosomes. Since skin pigmentation was shown to be regulated by PAR-2 activation, our results further support the melanocore-based mechanism of melanin transfer and further refine this model, which can now be described as coupled melanocore exo/phagocytosis.
Collapse
Affiliation(s)
- Hugo Moreiras
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Liliana Bento-Lopes
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Matilde V Neto
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Cristina Escrevente
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Luís C Cabaço
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Michael J Hall
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - José S Ramalho
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Miguel C Seabra
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Duarte C Barral
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
14
|
Mishra S, Ghanim M. Interactions of Liberibacter Species with Their Psyllid Vectors: Molecular, Biological and Behavioural Mechanisms. Int J Mol Sci 2022; 23:ijms23074029. [PMID: 35409386 PMCID: PMC8999863 DOI: 10.3390/ijms23074029] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/02/2022] [Accepted: 04/03/2022] [Indexed: 01/27/2023] Open
Abstract
Liberibacter is a group of plant pathogenic bacteria, transmitted by insect vectors, psyllids (Hemiptera: Psylloidea), and has emerged as one of the most devastating pathogens which have penetrated into many parts of the world over the last 20 years. The pathogens are known to cause plant diseases, such as Huanglongbing (citrus greening disease), Zebra chip disease, and carrot yellowing, etc., threatening some very important agricultural sectors, including citrus, potato and others. Candidatus Liberibacter asiaticus (CLas), the causative agent of citrus greening disease, is one of the most important pathogens of this group. This pathogen has infected most of the citrus trees in the US, Brazil and China, causing tremendous decline in citrus productivity, and, consequently, a severely negative impact on economic and personnel associated with citrus and related industries in these countries. Like other members in this group, CLas is transmitted by the Asian citrus psyllid (ACP, Diaphorina citri) in a persistent circulative manner. An additional important member of this group is Ca. L. solanacearum (CLso), which possesses nine haplotypes and infects a variety of crops, depending on the specific haplotype and the insect vector species. Ongoing pathogen control strategies, that are mainly based on use of chemical pesticides, lack the necessary credentials of being technically feasible, and environmentally safe. For this reason, strategies based on interference with Liberibacter vector transmission have been adopted as alternative strategies for the prevention of infection by these pathogens. A significant amount of research has been conducted during the last 10-15 years to understand the aspects of transmission of these bacterial species by their psyllid vectors. These research efforts span biological, ecological, behavioural and molecular aspects of Liberibacter–psyllid interactions, and will be reviewed in this manuscript. These attempts directed towards devising new means of disease control, endeavoured to explore alternative strategies, instead of relying on using chemicals for reducing the vector populations, which is the sole strategy currently employed and which has profound negative effects on human health, beneficial organisms and the environment.
Collapse
|
15
|
Liu KE, Raymond MH, Ravichandran KS, Kucenas S. Clearing Your Mind: Mechanisms of Debris Clearance After Cell Death During Neural Development. Annu Rev Neurosci 2022; 45:177-198. [PMID: 35226828 PMCID: PMC10157384 DOI: 10.1146/annurev-neuro-110920-022431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neurodevelopment and efferocytosis have fascinated scientists for decades. How an organism builds a nervous system that is precisely tuned for efficient behaviors and survival and how it simultaneously manages constant somatic cell turnover are complex questions that have resulted in distinct fields of study. Although neurodevelopment requires the overproduction of cells that are subsequently pruned back, very few studies marry these fields to elucidate the cellular and molecular mechanisms that drive nervous system development through the lens of cell clearance. In this review, we discuss these fields to highlight exciting areas of future synergy. We first review neurodevelopment from the perspective of overproduction and subsequent refinement and then discuss who clears this developmental debris and the mechanisms that control these events. We then end with how a more deliberate merger of neurodevelopment and efferocytosis could reframe our understanding of homeostasis and disease and discuss areas of future study. Expected final online publication date for the Annual Review of Neuroscience, Volume 45 is July 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Kendra E Liu
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA; .,Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Michael H Raymond
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA; .,Center for Clearance, University of Virginia, Charlottesville, Virginia, USA
| | - Kodi S Ravichandran
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA; .,Center for Clearance, University of Virginia, Charlottesville, Virginia, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA.,VIB-UGent Center for Inflammation Research and the Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sarah Kucenas
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA; .,Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA.,Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
16
|
Ge Y, Huang M, Yao YM. Efferocytosis and Its Role in Inflammatory Disorders. Front Cell Dev Biol 2022; 10:839248. [PMID: 35281078 PMCID: PMC8913510 DOI: 10.3389/fcell.2022.839248] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/10/2022] [Indexed: 12/18/2022] Open
Abstract
Efferocytosis is the effective clearance of apoptotic cells by professional and non-professional phagocytes. The process is mechanically different from other forms of phagocytosis and involves the localization, binding, internalization, and degradation of apoptotic cells. Defective efferocytosis has been demonstrated to associate with the pathogenesis of various inflammatory disorders. In the current review, we summarize recent findings with regard to efferocytosis networks and discuss the relationship between efferocytosis and different immune cell populations, as well as describe how efferocytosis helps resolve inflammatory response and modulate immune balance. Our knowledge so far about efferocytosis suggests that it may be a useful target in the treatment of numerous inflammatory diseases.
Collapse
Affiliation(s)
- Yun Ge
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Man Huang
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yong-ming Yao
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
17
|
Chen R, Zhang N, Zhou Y, Jing J. Optical Sensors and Actuators for Probing Proximity-Dependent Biotinylation in Living Cells. Front Cell Neurosci 2022; 16:801644. [PMID: 35250484 PMCID: PMC8890125 DOI: 10.3389/fncel.2022.801644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/13/2022] [Indexed: 01/09/2023] Open
Abstract
Proximity-dependent biotinylation techniques have been gaining wide applications in the systematic analysis of protein-protein interactions (PPIs) on a proteome-wide scale in living cells. The engineered biotin ligase TurboID is among the most widely adopted given its enhanced biotinylation efficiency, but it faces the background biotinylation complication that might confound proteomic data interpretation. To address this issue, we report herein a set of split TurboID variants that can be reversibly assembled by using light (designated “OptoID”), which enable optogenetic control of biotinylation based proximity labeling in living cells. OptoID could be further coupled with an engineered monomeric streptavidin that permits real-time monitoring of biotinylation with high temporal precision. These optical actuators and sensors will likely find broad applications in precise proximity proteomics and rapid detection of biotinylation in living cells.
Collapse
Affiliation(s)
- Rui Chen
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ningxia Zhang
- Laboratory of Cancer Biology, Department of Medical Oncology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yubin Zhou
- Department of Translational Medical Sciences, Center for Translational Cancer Research, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, United States
- *Correspondence: Yubin Zhou,
| | - Ji Jing
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, China
- Ji Jing,
| |
Collapse
|
18
|
Wang J, Yu C, Zhuang J, Qi W, Jiang J, Liu X, Zhao W, Cao Y, Wu H, Qi J, Zhao RC. The role of phosphatidylserine on the membrane in immunity and blood coagulation. Biomark Res 2022; 10:4. [PMID: 35033201 PMCID: PMC8760663 DOI: 10.1186/s40364-021-00346-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 12/17/2022] Open
Abstract
The negatively charged aminophospholipid, phosphatidylserine (PtdSer), is located in the inner leaflet of the plasma membrane in normal cells, and may be exposed to the outer leaflet under some immune and blood coagulation processes. Meanwhile, Ptdser exposed to apoptotic cells can be recognized and eliminated by various immune cells, whereas on the surface of activated platelets Ptdser interacts with coagulation factors prompting enhanced production of thrombin which significantly facilitates blood coagulation. In the case where PtdSer fails in exposure or mistakenly occurs, there are occurrences of certain immunological and haematological diseases, such as the Scott syndrome and Systemic lupus erythematosus. Besides, viruses (e.g., Human Immunodeficiency Virus (HIV), Ebola virus (EBOV)) can invade host cells through binding the exposed PtdSer. Most recently, the Corona Virus Disease 2019 (COVID-19) has been similarly linked to PtdSer or its receptors. Therefore, it is essential to comprehensively understand PtdSer and its functional characteristics. Therefore, this review summarizes Ptdser, its eversion mechanism; interaction mechanism, particularly with its immune receptors and coagulation factors; recognition sites; and its function in immune and blood processes. This review illustrates the potential aspects for the underlying pathogenic mechanism of PtdSer-related diseases, and the discovery of new therapeutic strategies as well.
Collapse
Affiliation(s)
- Jiao Wang
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China.
| | - Changxin Yu
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Junyi Zhuang
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Wenxin Qi
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Jiawen Jiang
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Xuanting Liu
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Wanwei Zhao
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Yiyang Cao
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Hao Wu
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Jingxuan Qi
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Robert Chunhua Zhao
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, No. 5 Dongdansantiao, Beijing, 100005, China.
- Centre of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China.
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China.
| |
Collapse
|
19
|
Apoptotic cell-derived metabolites in efferocytosis-mediated resolution of inflammation. Cytokine Growth Factor Rev 2021; 62:42-53. [PMID: 34742632 DOI: 10.1016/j.cytogfr.2021.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 10/13/2021] [Indexed: 12/21/2022]
Abstract
The resolution of inflammation, as part of standard host defense mechanism, is the process to guarantee timely termination of inflammatory responses and eventual restoration of tissue homeostasis . It is mainly achieved via efferocytosis, during which pro-resolving macrophages clear apoptotic neutrophils at the inflammatory site. Unfortunately, impaired resolution can be the leading cause of chronic inflammatory disorders and some autoimmune diseases. Existing studies have provided relatively comprehensive understandings about the recognition and uptake of apoptotic neutrophils by macrophages during early phases of efferocytosis. However, lack of information concerns macrophage metabolism of apoptotic cell-derived metabolites after being released from phagolysosomes or the relationship between such metabolism and efferocytosis. Notwithstanding, three recent studies have revealed macrophage metabolism of cholesterol, fatty acids and arginine, as well as their respective functions in the context of inflammation-resolution. This review provides an overview of the resolution of inflammation, efferocytosis and the key players involved, followed by a focus on the metabolism of apoptotic cell-derived metabolites within efferocytes. Hypotheses of more potential apoptotic cell-derived metabolites and their possible roles in the resolution are also formulated. Understanding the effect of these metabolites further advances the concept that apoptotic cells act as active players to regulate resolution, and also suggests novel therapeutic strategies for diseases driven by defective resolution and even cancer that may be treated through enhanced efferocytosis.
Collapse
|
20
|
Having an Old Friend for Dinner: The Interplay between Apoptotic Cells and Efferocytes. Cells 2021; 10:cells10051265. [PMID: 34065321 PMCID: PMC8161178 DOI: 10.3390/cells10051265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 02/02/2023] Open
Abstract
Apoptosis, the programmed and intentional death of senescent, damaged, or otherwise superfluous cells, is the natural end-point for most cells within multicellular organisms. Apoptotic cells are not inherently damaging, but if left unattended, they can lyse through secondary necrosis. The resulting release of intracellular contents drives inflammation in the surrounding tissue and can lead to autoimmunity. These negative consequences of secondary necrosis are avoided by efferocytosis—the phagocytic clearance of apoptotic cells. Efferocytosis is a product of both apoptotic cells and efferocyte mechanisms, which cooperate to ensure the rapid and complete removal of apoptotic cells. Herein, we review the processes used by apoptotic cells to ensure their timely removal, and the receptors, signaling, and cellular processes used by efferocytes for efferocytosis, with a focus on the receptors and signaling driving this process.
Collapse
|
21
|
Umbilical cord-derived mesenchymal stromal cells immunomodulate and restore actin dynamics and phagocytosis of LPS-activated microglia via PI3K/Akt/Rho GTPase pathway. Cell Death Discov 2021; 7:46. [PMID: 33723246 PMCID: PMC7961004 DOI: 10.1038/s41420-021-00436-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/20/2021] [Accepted: 02/13/2021] [Indexed: 12/27/2022] Open
Abstract
Microglia are the immune cells in the central nervous system surveying environment and reacting to various injuries. Activated microglia may cause impaired synaptic plasticity, therefore modulating and restoring them to neutral phenotype is crucial to counteract a pro-inflammatory, neurotoxic state. In this study, we focused on elucidating whether human umbilical cord (UC) -derived mesenchymal stromal cells (MSCs) can exert immunomodulatory effect and change the phenotype of activated microglia. Primary culture of microglia was activated by lipopolysaccharide (LPS) and was co-cultured with three lots of MSCs. We investigated immunomodulation, actin dynamics and phagocytic capacity of activated microglia, and examined change of Rho GTPase in microglia as the mechanism. MSCs suppressed the expression of IL-1β and pNFκB in LPS-activated microglia, and conversely elevated the expression of IL-1β in resting-surveying microglia with lot-to-lot variation. Morphological and phagocytotic analyses revealed that LPS stimulation significantly increased active Rho GTPase, Rac1, and Cdc42 levels in the microglia, and their morphology changed to amoeboid in which F-actin spread with ruffle formation. The F-actin spreading persisted after removal of LPS stimulation and reduced phagocytosis. On the other hand, MSC co-culture induced bimodal increase in active Rac1 and Cdc42 levels in LPS-activated microglia. Moreover, extended ruffles of F-actin shrinked and concentrated to form an actin ring, thereby restoring phagocytosis. We confirmed inhibition of the PI3K/Akt pathway attenuated F-actin dynamics and phagocytosis restored by MSCs. Overall, we demonstrated that MSCs immunomodulated microglia with lot-to-lot variation, and changed the phenotype of LPS-activated microglia restoring actin dynamics and phagocytosis by increase of active Rho GTPase.
Collapse
|
22
|
Husain I, Luo X. Apoptotic Donor Cells in Transplantation. Front Immunol 2021; 12:626840. [PMID: 33717145 PMCID: PMC7947657 DOI: 10.3389/fimmu.2021.626840] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/18/2021] [Indexed: 12/31/2022] Open
Abstract
Despite significant advances in prevention and treatment of transplant rejection with immunosuppressive medications, we continue to face challenges of long-term graft survival, detrimental medication side effects to both the recipient and transplanted organ together with risks for opportunistic infections. Transplantation tolerance has so far only been achieved through hematopoietic chimerism, which carries with it a serious and life-threatening risk of graft versus host disease, along with variability in persistence of chimerism and uncertainty of sustained tolerance. More recently, numerous in vitro and in vivo studies have explored the therapeutic potential of silent clearance of apoptotic cells which have been well known to aid in maintaining peripheral tolerance to self. Apoptotic cells from a donor not only have the ability of down regulating the immune response, but also are a way of providing donor antigens to recipient antigen-presenting-cells that can then promote donor-specific peripheral tolerance. Herein, we review both laboratory and clinical evidence that support the utility of apoptotic cell-based therapies in prevention and treatment of graft versus host disease and transplant rejection along with induction of donor-specific tolerance in solid organ transplantation. We have highlighted the potential limitations and challenges of this apoptotic donor cell-based therapy together with ongoing advancements and attempts made to overcome them.
Collapse
Affiliation(s)
- Irma Husain
- Department of Medicine, Duke University, Durham, NC, United States
| | - Xunrong Luo
- Department of Medicine, Duke University, Durham, NC, United States
| |
Collapse
|
23
|
Abstract
The clearance of apoptotic cells by professional and non-professional phagocytes - a process termed 'efferocytosis' - is essential for the maintenance of tissue homeostasis. Accordingly, defective efferocytosis underlies a growing list of chronic inflammatory diseases. Although much has been learnt about the mechanisms of apoptotic cell recognition and uptake, several key areas remain incompletely understood. This Review focuses on new discoveries related to how phagocytes process the metabolic cargo they receive during apoptotic cell uptake; the links between efferocytosis and the resolution of inflammation in health and disease; and the roles of efferocytosis in host defence. Understanding these aspects of efferocytosis sheds light on key physiological and pathophysiological processes and suggests novel therapeutic strategies for diseases driven by defective efferocytosis and impaired inflammation resolution.
Collapse
|
24
|
Jing J, Liu G, Huang Y, Zhou Y. A molecular toolbox for interrogation of membrane contact sites. J Physiol 2019; 598:1725-1739. [PMID: 31119749 PMCID: PMC7098838 DOI: 10.1113/jp277761] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/17/2019] [Indexed: 01/04/2023] Open
Abstract
Membrane contact sites (MCSs) are specialized subcellular compartments formed by closely apposed membranes from two organelles. The intermembrane gap is separated by a distance ranging from 10 to 35 nm. MCSs are typically maintained through dynamic protein–protein and protein–lipid interactions. These intermembrane contact sites constitute important intracellular signalling hotspots to mediate a plethora of cellular processes, including calcium homeostasis, lipid metabolism, membrane biogenesis and organelle remodelling. In recent years, a series of genetically encoded probes and chemogenetic or optogenetic actuators have been invented to aid the visualization and interrogation of MCSs in both fixed and living cells. These molecular tools have greatly accelerated the pace of mechanistic dissection of membrane contact sites at the molecular level. In this review, we present an overview on the latest progress in this endeavour, and provide a general guide to the selection of methods and molecular tools for probing interorganellar membrane contact sites.
![]()
Collapse
Affiliation(s)
- Ji Jing
- Center for Translational Cancer Research, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, 77030, USA
| | - Gan Liu
- Cockrell School of Engineering, University of Texas, Austin, TX, 78712, USA
| | - Yun Huang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, 77030, USA
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, 77030, USA
| |
Collapse
|
25
|
Bewley MA, Budd RC, Ryan E, Cole J, Collini P, Marshall J, Kolsum U, Beech G, Emes RD, Tcherniaeva I, Berbers GAM, Walmsley SR, Donaldson G, Wedzicha JA, Kilty I, Rumsey W, Sanchez Y, Brightling CE, Donnelly LE, Barnes PJ, Singh D, Whyte MKB, Dockrell DH. Opsonic Phagocytosis in Chronic Obstructive Pulmonary Disease Is Enhanced by Nrf2 Agonists. Am J Respir Crit Care Med 2018; 198:739-750. [PMID: 29547002 PMCID: PMC6222469 DOI: 10.1164/rccm.201705-0903oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 03/14/2018] [Indexed: 01/01/2023] Open
Abstract
RATIONALE Previous studies have identified defects in bacterial phagocytosis by alveolar macrophages (AMs) in patients with chronic obstructive pulmonary disease (COPD), but the mechanisms and clinical consequences remain incompletely defined. OBJECTIVES To examine the effect of COPD on AM phagocytic responses and identify the mechanisms, clinical consequences, and potential for therapeutic manipulation of these defects. METHODS We isolated AMs and monocyte-derived macrophages (MDMs) from a cohort of patients with COPD and control subjects within the Medical Research Council COPDMAP consortium and measured phagocytosis of bacteria in relation to opsonic conditions and clinical features. MEASUREMENTS AND MAIN RESULTS COPD AMs and MDMs have impaired phagocytosis of Streptococcus pneumoniae. COPD AMs have a selective defect in uptake of opsonized bacteria, despite the presence of antipneumococcal antibodies in BAL, not observed in MDMs or healthy donor AMs. AM defects in phagocytosis in COPD are significantly associated with exacerbation frequency, isolation of pathogenic bacteria, and health-related quality-of-life scores. Bacterial binding and initial intracellular killing of opsonized bacteria in COPD AMs was not reduced. COPD AMs have reduced transcriptional responses to opsonized bacteria, such as cellular stress responses that include transcriptional modules involving antioxidant defenses and Nrf2 (nuclear factor erythroid 2-related factor 2)-regulated genes. Agonists of the cytoprotective transcription factor Nrf2 (sulforaphane and compound 7) reverse defects in phagocytosis of S. pneumoniae and nontypeable Haemophilus influenzae by COPD AMs. CONCLUSIONS Patients with COPD have clinically relevant defects in opsonic phagocytosis by AMs, associated with impaired transcriptional responses to cellular stress, which are reversed by therapeutic targeting with Nrf2 agonists.
Collapse
Affiliation(s)
- Martin A Bewley
- 1 Department of Infection, Immunity and Cardiovascular Disease and
- 2 The Florey Institute for Host-Pathogen Interactions, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Richard C Budd
- 1 Department of Infection, Immunity and Cardiovascular Disease and
- 2 The Florey Institute for Host-Pathogen Interactions, University of Sheffield Medical School, Sheffield, United Kingdom
- 3 Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Eilise Ryan
- 4 Department of Respiratory Medicine
- 5 MRC Centre for Inflammation Research, and
| | - Joby Cole
- 1 Department of Infection, Immunity and Cardiovascular Disease and
- 2 The Florey Institute for Host-Pathogen Interactions, University of Sheffield Medical School, Sheffield, United Kingdom
- 3 Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Paul Collini
- 1 Department of Infection, Immunity and Cardiovascular Disease and
- 2 The Florey Institute for Host-Pathogen Interactions, University of Sheffield Medical School, Sheffield, United Kingdom
- 3 Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Jennifer Marshall
- 5 MRC Centre for Inflammation Research, and
- 6 Department of Infection Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Umme Kolsum
- 7 Medicines Evaluation Unit, University of Manchester, Manchester, United Kingdom
- 8 University Hospital of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| | - Gussie Beech
- 7 Medicines Evaluation Unit, University of Manchester, Manchester, United Kingdom
- 8 University Hospital of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| | - Richard D Emes
- 9 School of Veterinary Medicine and Science and
- 10 Advanced Data Analysis Centre, University of Nottingham, United Kingdom
| | - Irina Tcherniaeva
- 11 Centre for Infectious Disease Control (CIb), National Institute for Public Health and the Environment (RIVM), Utrecht, the Netherlands
| | - Guy A M Berbers
- 11 Centre for Infectious Disease Control (CIb), National Institute for Public Health and the Environment (RIVM), Utrecht, the Netherlands
| | - Sarah R Walmsley
- 4 Department of Respiratory Medicine
- 5 MRC Centre for Inflammation Research, and
| | - Gavin Donaldson
- 12 National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jadwiga A Wedzicha
- 12 National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Iain Kilty
- 13 Pfizer Inc., Cambridge, Massachusetts
| | - William Rumsey
- 14 Stress and Repair Discovery Performance Unit, Respiratory Therapy Area, GSK, King of Prussia, Pennsylvania; and
| | - Yolanda Sanchez
- 14 Stress and Repair Discovery Performance Unit, Respiratory Therapy Area, GSK, King of Prussia, Pennsylvania; and
| | | | - Louise E Donnelly
- 12 National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Peter J Barnes
- 12 National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Dave Singh
- 7 Medicines Evaluation Unit, University of Manchester, Manchester, United Kingdom
- 8 University Hospital of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| | - Moira K B Whyte
- 4 Department of Respiratory Medicine
- 5 MRC Centre for Inflammation Research, and
| | - David H Dockrell
- 5 MRC Centre for Inflammation Research, and
- 6 Department of Infection Medicine, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
26
|
Abdolmaleki F, Farahani N, Gheibi Hayat SM, Pirro M, Bianconi V, Barreto GE, Sahebkar A. The Role of Efferocytosis in Autoimmune Diseases. Front Immunol 2018; 9:1645. [PMID: 30083153 PMCID: PMC6064952 DOI: 10.3389/fimmu.2018.01645] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/04/2018] [Indexed: 12/28/2022] Open
Abstract
Apoptosis happens continuously for millions of cells along with the active removal of apoptotic debris in order to maintain tissue homeostasis. In this respect, efferocytosis, i.e., the process of dead cell clearance, is orchestrated through cell exposure of a set of "find me," "eat me," and "tolerate me" signals facilitating the engulfment of dying cells through phagocytosis by macrophages and dendritic cells. The clearance of dead cells via phagocytes is of utmost importance to maintain the immune system tolerance to self-antigens. Accordingly, this biological activity prevents the release of autoantigens by dead cells, thus potentially suppressing the undesirable autoreactivity of immune cells and the appearance of inflammatory autoimmune disorders as systemic lupus erythematous and rheumatoid arthritis. In the present study, the apoptosis pathways and their immune regulation were reviewed. Moreover, efferocytosis process and its impairment in association with some autoimmune diseases were discussed.
Collapse
Affiliation(s)
- Fereshte Abdolmaleki
- Cellular and Molecular Research Center, School of Paramedical Sciences, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Najmeh Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Vanessa Bianconi
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - George E. Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
27
|
Manara MC, Terracciano M, Mancarella C, Sciandra M, Guerzoni C, Pasello M, Grilli A, Zini N, Picci P, Colombo MP, Morrione A, Scotlandi K. CD99 triggering induces methuosis of Ewing sarcoma cells through IGF-1R/RAS/Rac1 signaling. Oncotarget 2018; 7:79925-79942. [PMID: 27835596 PMCID: PMC5346761 DOI: 10.18632/oncotarget.13160] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 10/14/2016] [Indexed: 12/27/2022] Open
Abstract
CD99 is a cell surface molecule that has emerged as a novel target for Ewing sarcoma (EWS), an aggressive pediatric bone cancer. This report provides the first evidence of methuosis in EWS, a non-apoptotic form of cell death induced by an antibody directed against the CD99 molecule. Upon mAb triggering, CD99 induces an IGF-1R/RAS/Rac1 complex, which is internalized into RAB5-positive endocytic vacuoles. This complex is then dissociated, with the IGF-1R recycling to the cell membrane while CD99 and RAS/Rac1 are sorted into immature LAMP-1-positive vacuoles, whose excessive accumulation provokes methuosis. This process, which is not detected in CD99-expressing normal mesenchymal cells, is inhibited by disruption of the IGF-1R signaling, whereas enhanced by IGF-1 stimulation. Induction of IGF-1R/RAS/Rac1 was also observed in the EWS xenografts that respond to anti-CD99 mAb, further supporting the role of the IGF/RAS/Rac1 axis in the hyperstimulation of macropinocytosis and selective death of EWS cells. Thus, we describe a vulnerability of EWS cells, including those resistant to standard chemotherapy, to a treatment with anti-CD99 mAb, which requires IGF-1R/RAS signaling but bypasses the need for their direct targeting. Overall, we propose CD99 targeting as new opportunity to treat EWS patients resistant to canonical apoptosis-inducing agents.
Collapse
Affiliation(s)
- Maria Cristina Manara
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Mario Terracciano
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna 40136, Italy.,Department of Urology and Biology of Prostate Cancer Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Caterina Mancarella
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Marika Sciandra
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna 40136, Italy.,PROMETEO Laboratory, STB, RIT Department, Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Clara Guerzoni
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna 40136, Italy.,PROMETEO Laboratory, STB, RIT Department, Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Michela Pasello
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna 40136, Italy.,PROMETEO Laboratory, STB, RIT Department, Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Andrea Grilli
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Nicoletta Zini
- CNR, National Research Council of Italy, Institute of Molecular Genetics, Bologna 40136, Italy.,SC Laboratory of Musculoskeletal Cell Biology, Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Piero Picci
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna 40136, Italy.,PROMETEO Laboratory, STB, RIT Department, Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Mario P Colombo
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS "Istituto Nazionale dei Tumori," Milan 20133, Italy
| | - Andrea Morrione
- Department of Urology and Biology of Prostate Cancer Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Katia Scotlandi
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna 40136, Italy.,PROMETEO Laboratory, STB, RIT Department, Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| |
Collapse
|
28
|
Pushkarsky I, Tseng P, Black D, France B, Warfe L, Koziol-White CJ, Jester WF, Trinh RK, Lin J, Scumpia PO, Morrison SL, Panettieri RA, Damoiseaux R, Di Carlo D. Elastomeric sensor surfaces for high-throughput single-cell force cytometry. Nat Biomed Eng 2018; 2:124-137. [PMID: 31015629 DOI: 10.1038/s41551-018-0193-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 01/09/2018] [Indexed: 11/09/2022]
Abstract
As cells with aberrant force-generating phenotypes can directly lead to disease, cellular force-generation mechanisms are high-value targets for new therapies. Here, we show that single-cell force sensors embedded in elastomers enable single-cell force measurements with ~100-fold improvement in throughput than was previously possible. The microtechnology is scalable and seamlessly integrates with the multi-well plate format, enabling highly parallelized time-course studies. In this regard, we show that airway smooth muscle cells isolated from fatally asthmatic patients have innately greater and faster force-generation capacity in response to stimulation than healthy control cells. By simultaneously tracing agonist-induced calcium flux and contractility in the same cell, we show that the calcium level is ultimately a poor quantitative predictor of cellular force generation. Finally, by quantifying phagocytic forces in thousands of individual human macrophages, we show that force initiation is a digital response (rather than a proportional one) to the proper immunogen. By combining mechanobiology at the single-cell level with high-throughput capabilities, this microtechnology can support drug-discovery efforts for clinical conditions associated with aberrant cellular force generation.
Collapse
Affiliation(s)
- Ivan Pushkarsky
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peter Tseng
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Electrical Engineering and Computer Science, University of California, Los Angeles, Irvine, CA, USA
| | - Dylan Black
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bryan France
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lyndon Warfe
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Cynthia J Koziol-White
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, NJ, USA
| | - William F Jester
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, NJ, USA
| | - Ryan K Trinh
- Department of Microbiology, Immunology and Molecular Genetics and The Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jonathan Lin
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Philip O Scumpia
- Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sherie L Morrison
- Department of Microbiology, Immunology and Molecular Genetics and The Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, NJ, USA
| | - Robert Damoiseaux
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Molecular and Medicinal Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA. .,California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA. .,Department of Mechanical Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
29
|
Yurdagul A, Doran AC, Cai B, Fredman G, Tabas IA. Mechanisms and Consequences of Defective Efferocytosis in Atherosclerosis. Front Cardiovasc Med 2018; 4:86. [PMID: 29379788 PMCID: PMC5770804 DOI: 10.3389/fcvm.2017.00086] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/11/2017] [Indexed: 12/22/2022] Open
Abstract
Efficient clearance of apoptotic cells, termed efferocytosis, critically regulates normal homeostasis whereas defective uptake of apoptotic cells results in chronic and non-resolving inflammatory diseases, such as advanced atherosclerosis. Monocyte-derived macrophages recruited into developing atherosclerotic lesions initially display efficient efferocytosis and temper inflammatory responses, processes that restrict plaque progression. However, during the course of plaque development, macrophages undergo cellular reprogramming that reduces efferocytic capacity, which results in post-apoptotic necrosis of apoptotic cells and inflammation. Furthermore, defective efferocytosis in advanced atherosclerosis is a major driver of necrotic core formation, which can trigger plaque rupture and acute thrombotic cardiovascular events. In this review, we discuss the molecular and cellular mechanisms that regulate efferocytosis, how efferocytosis promotes the resolution of inflammation, and how defective efferocytosis leads to the formation of clinically dangerous atherosclerotic plaques.
Collapse
Affiliation(s)
- Arif Yurdagul
- Department of Medicine, Columbia University, New York, NY, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Department of Physiology, Columbia University, New York, NY, United States
| | - Amanda C Doran
- Department of Medicine, Columbia University, New York, NY, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Department of Physiology, Columbia University, New York, NY, United States
| | - Bishuang Cai
- Department of Medicine, Columbia University, New York, NY, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Department of Physiology, Columbia University, New York, NY, United States
| | - Gabrielle Fredman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Ira A Tabas
- Department of Medicine, Columbia University, New York, NY, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Department of Physiology, Columbia University, New York, NY, United States
| |
Collapse
|
30
|
Guduru SKR, Arya P. Synthesis and biological evaluation of rapamycin-derived, next generation small molecules. MEDCHEMCOMM 2018; 9:27-43. [PMID: 30108899 PMCID: PMC6072512 DOI: 10.1039/c7md00474e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 11/21/2017] [Indexed: 12/20/2022]
Abstract
Over the years, rapamycin has attracted serious attention due to its remarkable biological properties and as a potent inhibitor of the mammalian target of rapamycin (mTOR) protein through its binding with FKBP-12. Several efficient strategies that utilize synthetic and biosynthetic approaches have been utilized to develop small molecule rapamycin analogs or for synthesizing hybrid compounds containing a partial rapamycin structure to improve pharmacokinetic properties. Herein, we report selected case studies related to the synthesis of rapamycin-derived compounds and hybrid molecules to explore their biological properties.
Collapse
Affiliation(s)
- Shiva Krishna Reddy Guduru
- Center for Drug Discovery , Department of Pharmacology and Chemical Biology , Baylor College of Medicine , One Baylor Plaza , Houston , Texas 77030 , USA . ; ; Tel: +1 713 798 8794
- Department of Pharmacology and Chemical Biology , Baylor College of Medicine , One Baylor Plaza , Houston , Texas 77030 , USA
| | - Prabhat Arya
- Chemistry and Chemical Biology , Dr. Reddy's Institute of Life Sciences (DRILS) , University of Hyderabad Campus , Hyderabad 500046 , India
| |
Collapse
|
31
|
Böttcher RT, Veelders M, Rombaut P, Faix J, Theodosiou M, Stradal TE, Rottner K, Zent R, Herzog F, Fässler R. Kindlin-2 recruits paxillin and Arp2/3 to promote membrane protrusions during initial cell spreading. J Cell Biol 2017; 216:3785-3798. [PMID: 28912124 PMCID: PMC5674885 DOI: 10.1083/jcb.201701176] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 07/14/2017] [Accepted: 08/08/2017] [Indexed: 12/18/2022] Open
Abstract
Cell spreading requires the coupling of actin-driven membrane protrusion and integrin-mediated adhesion to the extracellular matrix. The integrin-activating adaptor protein kindlin-2 plays a central role for cell adhesion and membrane protrusion by directly binding and recruiting paxillin to nascent adhesions. Here, we report that kindlin-2 has a dual role during initial cell spreading: it binds paxillin via the pleckstrin homology and F0 domains to activate Rac1, and it directly associates with the Arp2/3 complex to induce Rac1-mediated membrane protrusions. Consistently, abrogation of kindlin-2 binding to Arp2/3 impairs lamellipodia formation and cell spreading. Our findings identify kindlin-2 as a key protein that couples cell adhesion by activating integrins and the induction of membrane protrusions by activating Rac1 and supplying Rac1 with the Arp2/3 complex.
Collapse
Affiliation(s)
- Ralph T Böttcher
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Maik Veelders
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Pascaline Rombaut
- Gene Center Munich, Ludwig Maximilians University Munich, Munich, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Marina Theodosiou
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Klemens Rottner
- Helmholtz Centre for Infection Research, Braunschweig, Germany
- Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Roy Zent
- Division of Nephrology, Department of Medicine, Vanderbilt University, Nashville, TN
- Department of Medicine, Veterans Affairs Medical Center, Nashville, TN
| | - Franz Herzog
- Gene Center Munich, Ludwig Maximilians University Munich, Munich, Germany
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
32
|
Akhtar N, Li W, Mironov A, Streuli CH. Rac1 Controls Both the Secretory Function of the Mammary Gland and Its Remodeling for Successive Gestations. Dev Cell 2017; 38:522-35. [PMID: 27623383 PMCID: PMC5022528 DOI: 10.1016/j.devcel.2016.08.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 06/30/2016] [Accepted: 08/12/2016] [Indexed: 12/27/2022]
Abstract
An important feature of the mammary gland is its ability to undergo repeated morphological changes during each reproductive cycle with profound tissue expansion in pregnancy and regression in involution. However, the mechanisms that determine the tissue's cyclic regenerative capacity remain elusive. We have now discovered that Cre-Lox ablation of Rac1 in mammary epithelia causes gross enlargement of the epithelial tree and defective alveolar regeneration in a second pregnancy. Architectural defects arise because loss of Rac1 disrupts clearance in involution following the first lactation. We show that Rac1 is crucial for mammary alveolar epithelia to switch from secretion to a phagocytic mode and rapidly remove dying neighbors. Moreover, Rac1 restricts the extrusion of dying cells into the lumen, thus promoting their eradication by live phagocytic neighbors while within the epithelium. Without Rac1, residual milk and cell corpses flood the ductal network, causing gross dilation, chronic inflammation, and defective future regeneration. Rac1 is required for full secretory differentiation of the mammary gland Rac1 restricts apoptotic cell shedding into the lumen to limit inflammation Rac1 contributes to post-lactational tissue remodeling during involution Defective clearance of milk and dead cells in Rac1-null glands causes ductal bloating
Collapse
Affiliation(s)
- Nasreen Akhtar
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, and Manchester Breast Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK; Department of Oncology and Metabolism, The Bateson Centre, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK.
| | - Weiping Li
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, and Manchester Breast Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Aleksander Mironov
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, and Manchester Breast Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Charles H Streuli
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, and Manchester Breast Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
33
|
Abstract
Mammary epithelial phagocytosis is critical for removal of apoptotic cells during involution, but the mechanisms governing this process are largely unknown. In this issue of Developmental Cell, Akhtar et al. (2016) provide insight into mechanisms regulating involution, demonstrating that Rac1 drives the switch from differentiation to phagocytosis in mammary epithelium.
Collapse
Affiliation(s)
- Matthew J Naylor
- School of Medical Sciences and Bosch Institute, Sydney Medical School, The University of Sydney, New South Wales 2006, Australia.
| |
Collapse
|
34
|
Ikeda Y, Kawai K, Ikawa A, Kawamoto K, Egami Y, Araki N. Rac1 switching at the right time and location is essential for Fcγ receptor-mediated phagosome formation. J Cell Sci 2017; 130:2530-2540. [DOI: 10.1242/jcs.201749] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 06/04/2017] [Indexed: 12/28/2022] Open
Abstract
Lamellipodia are sheet-like cell protrusions driven by actin polymerization mainly through Rac1, a GTPase molecular switch. In Fcγ receptor-mediated phagocytosis of IgG-opsonized erythrocytes (IgG-Es), Rac1 activation is required for lamellipodial extension along the surface of IgG-Es. However, the significance of Rac1 deactivation in phagosome formation is poorly understood. Our live-cell imaging and electron microscopy revealed that RAW264 macrophages expressing a constitutively active Rac1 mutant showed defects in phagocytic cup formation, while lamellipodia were formed around IgG-Es. Because the activated Rac1 reduced the phosphorylation levels of myosin light chain, failure of the cup formation were probably due to inhibition of actin/myosin II contractility. Reversible photo-manipulation of the Rac1 switch in macrophages fed with IgG-Es could phenocopy two lamellipodial motilities: outward-extension and cup-constriction by Rac1 ON and OFF, respectively. In conjunction with FRET imaging of Rac1 activity, we provide a novel mechanistic model of phagosome formation spatiotemporally controlled by Rac1 switching within a phagocytic cup.
Collapse
Affiliation(s)
- Yuka Ikeda
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Katsuhisa Kawai
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Akira Ikawa
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Kyoko Kawamoto
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Youhei Egami
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Nobukazu Araki
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Miki, Kagawa 761-0793, Japan
| |
Collapse
|
35
|
Ross B, Mehta S, Zhang J. Molecular tools for acute spatiotemporal manipulation of signal transduction. Curr Opin Chem Biol 2016; 34:135-142. [PMID: 27639090 DOI: 10.1016/j.cbpa.2016.08.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 08/15/2016] [Accepted: 08/17/2016] [Indexed: 01/14/2023]
Abstract
The biochemical activities involved in signal transduction in cells are under tight spatiotemporal regulation. To study the effects of the spatial patterning and temporal dynamics of biochemical activities on downstream signaling, researchers require methods to manipulate signaling pathways acutely and rapidly. In this review, we summarize recent developments in the design of three broad classes of molecular tools for perturbing signal transduction, classified by their type of input signal: chemically induced, optically induced, and magnetically induced.
Collapse
Affiliation(s)
- Brian Ross
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sohum Mehta
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Jin Zhang
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
36
|
Gordon P, Okai B, Hoare JI, Erwig LP, Wilson HM. SOCS3 is a modulator of human macrophage phagocytosis. J Leukoc Biol 2016; 100:771-780. [PMID: 27106674 DOI: 10.1189/jlb.3a1215-554rr] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 04/03/2016] [Indexed: 01/20/2023] Open
Abstract
Suppressor of cytokine signaling (SOCS) proteins are recognized as key feedback inhibitors modulating the inflammatory activities of macrophages, but comparatively little is known about whether and how they affect phagocytosis. Here, we evaluated the role of SOCS3 in driving the inflammatory phenotype and phagocytic uptake of apoptotic cells by human macrophages and the signaling pathways that are necessary for efficient phagocytosis. In M1-activated human monocyte-derived macrophages, SOCS3 silencing, using short interfering RNA technology, resulted in a decreased expression of proinflammatory markers and an increased expression of M2 macrophage markers. Strikingly, we demonstrated for the first time that SOCS3 knockdown significantly enhances the phagocytic capacity of M1 macrophages for carboxylate-modified beads and apoptotic neutrophils. With the use of live-cell video microscopy, we showed that SOCS3 knockdown radically affects the temporal dynamics of particle engulfment, enabling more rapid uptake of a second target and delaying postengulfment processing, as evidenced by deferred acquisition of phagosome maturation markers. SOCS3 knockdown impacts on phagocytosis through increased PI3K and Ras-related C3 botulinum toxin substrate 1 (Rac1) activity, pathways essential for engulfment and clearance of apoptotic cells. Enhanced phagocytosis in SOCS3-silenced cells was reversed by pharmacological PI3K inhibition. Furthermore, we revealed that actin polymerization, downstream of PI3K/Rac1 activation, was significantly altered in SOCS3-silenced cells, providing a mechanism for their greater phagocytic activity. The findings support a new model, whereby SOCS3 not only plays an important role in driving macrophage inflammatory responses but modulates key signaling pathways organizing the actin cytoskeleton to regulate the efficiency of phagocytic processes.
Collapse
Affiliation(s)
- Peter Gordon
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, United Kingdom
| | - Blessing Okai
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, United Kingdom
| | - Joseph I Hoare
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, United Kingdom
| | - Lars P Erwig
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, United Kingdom
| | - Heather M Wilson
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, United Kingdom
| |
Collapse
|
37
|
The clearance of dying cells: table for two. Cell Death Differ 2016; 23:915-26. [PMID: 26990661 PMCID: PMC4987729 DOI: 10.1038/cdd.2015.172] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 12/19/2022] Open
Abstract
Phagocytic cells of the immune system must constantly survey for, recognize, and efficiently clear the billions of cellular corpses that arise as a result of development, stress, infection, or normal homeostasis. This process, termed efferocytosis, is critical for the prevention of autoimmune and inflammatory disorders, and persistence of dead cells in tissue is characteristic of many human autoimmune diseases, notably systemic lupus erythematosus. The most notable characteristic of the efferocytosis of apoptotic cells is its ‘immunologically silent' response. Although the mechanisms by which phagocytes facilitate engulfment of dead cells has been a well-studied area, the pathways that coordinate to process the ingested corpse and direct the subsequent immune response is an area of growing interest. The recently described pathway of LC3 (microtubule-associated protein 1A/1B-light chain 3)-associated phagocytosis (LAP) has shed some light on this issue. LAP is triggered when an extracellular particle, such as a dead cell, engages an extracellular receptor during phagocytosis, induces the translocation of autophagy machinery, and ultimately LC3 to the cargo-containing phagosome, termed the LAPosome. In this review, we will examine efferocytosis and the impact of LAP on efferocytosis, allowing us to reimagine the impact of the autophagy machinery on innate host defense mechanisms.
Collapse
|
38
|
Molecular imaging analysis of Rab GTPases in the regulation of phagocytosis and macropinocytosis. Anat Sci Int 2015; 91:35-42. [DOI: 10.1007/s12565-015-0313-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/26/2015] [Indexed: 12/22/2022]
|
39
|
Schlam D, Bagshaw RD, Freeman SA, Collins RF, Pawson T, Fairn GD, Grinstein S. Phosphoinositide 3-kinase enables phagocytosis of large particles by terminating actin assembly through Rac/Cdc42 GTPase-activating proteins. Nat Commun 2015; 6:8623. [PMID: 26465210 PMCID: PMC4634337 DOI: 10.1038/ncomms9623] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 09/11/2015] [Indexed: 02/06/2023] Open
Abstract
Phagocytosis is responsible for the elimination of particles of widely disparate sizes, from large fungi or effete cells to small bacteria. Though superficially similar, the molecular mechanisms involved differ: engulfment of large targets requires phosphoinositide 3-kinase (PI3K), while that of small ones does not. Here, we report that inactivation of Rac and Cdc42 at phagocytic cups is essential to complete internalization of large particles. Through a screen of 62 RhoGAP-family members, we demonstrate that ARHGAP12, ARHGAP25 and SH3BP1 are responsible for GTPase inactivation. Silencing these RhoGAPs impairs phagocytosis of large targets. The GAPs are recruited to large—but not small—phagocytic cups by products of PI3K, where they synergistically inactivate Rac and Cdc42. Remarkably, the prominent accumulation of phosphatidylinositol 3,4,5-trisphosphate characteristic of large-phagosome formation is less evident during phagocytosis of small targets, accounting for the contrasting RhoGAP distribution and the differential requirement for PI3K during phagocytosis of dissimilarly sized particles. Phagocytosis of large (but not small) particles requires PI 3-kinase activity. Here, Schlam et al. show that Rho GTPase-activating proteins are recruited to the phagocytic cup by products of PI 3-kinase, resulting in the local inactivation of Rac and Cdc42 and allowing for the completion of internalization of large particles.
Collapse
Affiliation(s)
- Daniel Schlam
- Division of Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada M5G1X8.,Institute of Medical Science, University of Toronto, Faculty of Medicine, 1 King's College Circle, Toronto, Ontario, Canada M5S1A8
| | - Richard D Bagshaw
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, Canada M5G1X5
| | - Spencer A Freeman
- Division of Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada M5G1X8
| | - Richard F Collins
- Division of Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada M5G1X8
| | - Tony Pawson
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, Canada M5G1X5
| | - Gregory D Fairn
- Institute of Medical Science, University of Toronto, Faculty of Medicine, 1 King's College Circle, Toronto, Ontario, Canada M5S1A8.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada M5B1T8
| | - Sergio Grinstein
- Division of Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada M5G1X8.,Institute of Medical Science, University of Toronto, Faculty of Medicine, 1 King's College Circle, Toronto, Ontario, Canada M5S1A8.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada M5B1T8
| |
Collapse
|
40
|
Tao H, Yancey PG, Babaev VR, Blakemore JL, Zhang Y, Ding L, Fazio S, Linton MF. Macrophage SR-BI mediates efferocytosis via Src/PI3K/Rac1 signaling and reduces atherosclerotic lesion necrosis. J Lipid Res 2015; 56:1449-60. [PMID: 26059978 PMCID: PMC4513986 DOI: 10.1194/jlr.m056689] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Indexed: 12/13/2022] Open
Abstract
Macrophage apoptosis and efferocytosis are key determinants of atherosclerotic plaque inflammation and necrosis. Bone marrow transplantation studies in ApoE- and LDLR-deficient mice revealed that hematopoietic scavenger receptor class B type I (SR-BI) deficiency results in severely defective efferocytosis in mouse atherosclerotic lesions, resulting in a 17-fold higher ratio of free to macrophage-associated dead cells in lesions containing SR-BI−/− cells, 5-fold more necrosis, 65.2% less lesional collagen content, nearly 7-fold higher dead cell accumulation, and 2-fold larger lesion area. Hematopoietic SR-BI deletion elicited a maladaptive inflammatory response [higher interleukin (IL)-1β, IL-6, and TNF-α lower IL-10 and transforming growth factor β]. Efferocytosis of apoptotic thymocytes was reduced by 64% in SR-BI−/− versus WT macrophages, both in vitro and in vivo. In response to apoptotic cells, macrophage SR-BI bound with phosphatidylserine and induced Src phosphorylation and cell membrane recruitment, which led to downstream activation of phosphoinositide 3-kinase (PI3K) and Ras-related C3 botulinum toxin substrate 1 (Rac1) for engulfment and clearance of apoptotic cells, as inhibition of Src decreased PI3K, Rac1-GTP, and efferocytosis in WT cells. Pharmacological inhibition of Rac1 reduced macrophage efferocytosis in a SR-BI-dependent fashion, and activation of Rac1 corrected the defective efferocytosis in SR-BI−/− macrophages. Thus, deficiency of macrophage SR-BI promotes defective efferocytosis signaling via the Src/PI3K/Rac1 pathway, resulting in increased plaque size, necrosis, and inflammation.
Collapse
Affiliation(s)
- Huan Tao
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Patricia G Yancey
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Vladimir R Babaev
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - John L Blakemore
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Youmin Zhang
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Lei Ding
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Sergio Fazio
- Department of Medicine, Physiology, and Pharmacology, Center of Preventive Cardiology, Oregon Health and Science University, Portland, OR 97239
| | - MacRae F Linton
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232 Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
41
|
Mai P, Tian L, Yang L, Wang L, Yang L, Li L. Cannabinoid receptor 1 but not 2 mediates macrophage phagocytosis by G(α)i/o /RhoA/ROCK signaling pathway. J Cell Physiol 2015; 230:1640-50. [PMID: 25545473 DOI: 10.1002/jcp.24911] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 12/18/2014] [Indexed: 12/24/2022]
Abstract
Phagocytosis is critical to macrophages linking innate and adaptive immune reaction. Cannabinoid receptor 1 (CB1) and 2 (CB2) mediate immune modulation. However, the role of cannabinoid receptors in macrophage phagocytosis is undefined. In this study, we found that two murine macrophage lines (J774A.1 and RAW264.7) and peripheral blood macrophages all expressed CB1 and CB2 by immunofluorescence-staining, real time RT-PCR and Western blot. Macrophage phagocytic activity was determined by quantifying fluorescent intensity of the engulfed BioParticles or fluorescence-activated cell sorting. mAEA (CB1 agonist) enhanced phagocytosis of macrophages, but JWH133 (CB2 agonist) had no influence. Pharmacological or genetic ablation of CB1 inhibited mAEA-enhanced phagocytosis, while CB2 had no such effects. Meanwhile, activation of CB1 increased GTP-bounding active form of small GTPase RhoA, but not Rac1 or Cdc42. AM281 (CB1 antagonist) and pertussis toxin (PTX, G((α)i/o) protein inhibitor) decreased GTP-bound RhoA protein level with mAEA. In addition, PTX, C3 Transferase (RhoA inhibitor) or Y27632 (Rho-associated kinase ROCK inhibitor) attenuated CB1-mediated phagocytosis. These results confirm that activation of CB1 regulates macrophage phagocytosis through G((α)i/o)/RhoA/ROCK signaling pathway. Moreover, activation of CB1 induced significant up-regulation of CB1 expression by real time RT-PCR and Western blot analysis, but not CB2. It indicated the existence of a positive feedback between CB1 activation and CB1 expression. The up-regulation of CB1 was RhoA-independent but it may contribute to maintaining high phagocytic activity of macrophages for a longer time. In conclusion, CB1 mediates macrophage phagocytosis by G((α)i/o)/RhoA/ROCK signal axis. These data further underline the role of CB1 in macrophage phagocytic process.
Collapse
Affiliation(s)
- Ping Mai
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | | | | | | | | | | |
Collapse
|
42
|
Genua M, D'Alessio S, Cibella J, Gandelli A, Sala E, Correale C, Spinelli A, Arena V, Malesci A, Rutella S, Ploplis VA, Vetrano S, Danese S. The urokinase plasminogen activator receptor (uPAR) controls macrophage phagocytosis in intestinal inflammation. Gut 2015; 64:589-600. [PMID: 24848264 DOI: 10.1136/gutjnl-2013-305933] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Inflammation plays crucial roles in the pathogenesis of several chronic inflammatory disorders, including Crohn's disease (CD) and UC, the two major forms of IBD. The urokinase plasminogen activator receptor (uPAR) exerts pleiotropic functions over the course of both physiological and pathological processes. uPAR not only has a key role in fibrinolysis but also modulates the development of protective immunity. Additionally, uPAR supports extracellular matrix degradation and regulates cell migration, adhesion and proliferation, thus influencing the development of inflammatory and immune responses. This study aimed to evaluate the role of uPAR in the pathogenesis of IBD. DESIGN The functional role of uPAR was assessed in established experimental models of colitis. uPAR deficiency effects on cytokine release, polarisation and bacterial phagocytosis were analysed in colonic macrophages. uPAR expression was analysed in surgical specimens collected from normal subjects and patients with IBD. RESULTS In mice, uPAR expression is positively regulated as colitis progresses. uPAR-KO mice displayed severe inflammation compared with wild-type littermates, as indicated by clinical assessment, endoscopy and colon histology. The absence of uPAR led to an increased production of inflammatory cytokines by macrophages that showed an M1 polarisation and impaired phagocytosis. In human IBD, CD68(+) macrophages derived from the inflamed mucosa expressed low levels of uPAR. CONCLUSIONS These findings point to uPAR as an essential component of intestinal macrophage functions and unravel a new potential target to control mucosal inflammation in IBD.
Collapse
Affiliation(s)
- Marco Genua
- IBD Center, Humanitas Clinical and Research Center, Rozzano, Italy Department of Translational Medicine, University of Milan, Milan, Italy
| | - Silvia D'Alessio
- IBD Center, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Javier Cibella
- IBD Center, Humanitas Clinical and Research Center, Rozzano, Italy
| | | | - Emanuela Sala
- IBD Center, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Carmen Correale
- IBD Center, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Antonino Spinelli
- Department of Translational Medicine, University of Milan, Milan, Italy Department of Surgery-IBD Surgery Unit, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Vincenzo Arena
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Alberto Malesci
- IBD Center, Humanitas Clinical and Research Center, Rozzano, Italy Department of Translational Medicine, University of Milan, Milan, Italy
| | - Sergio Rutella
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Victoria A Ploplis
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana, USA
| | - Stefania Vetrano
- IBD Center, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Silvio Danese
- IBD Center, Humanitas Clinical and Research Center, Rozzano, Italy
| |
Collapse
|
43
|
Abstract
During development, stress, infection, or normal homeostasis, billions of cells die on a daily basis, and the responsibility of clearing these cellular corpses lies with the phagocytes of innate immune system. This process, termed efferocytosis , is critical for the prevention of inflammation and autoimmunity , as well as modulation of the adaptive immune response. Defective clearance of dead cells is characteristic of many human autoimmune or autoinflammatory disorders, such as systemic lupus erythematosus (SLE), atherosclerosis, and diabetes. The mechanisms that phagocytes employ to sense, engulf, and process dead cells for an appropriate immune response have been an area of great interest. However, insight into novel mechanisms of programmed cell death , such as necroptosis, has shed light on the fact that while the diner (or phagocyte) is important, the meal itself (the type of dead cell) can play a crucial role in shaping the pursuant immune response.
Collapse
Affiliation(s)
- Jennifer Martinez
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
44
|
Fisher TW, Vyas M, He R, Nelson W, Cicero JM, Willer M, Kim R, Kramer R, May GA, Crow JA, Soderlund CA, Gang DR, Brown JK. Comparison of potato and asian citrus psyllid adult and nymph transcriptomes identified vector transcripts with potential involvement in circulative, propagative liberibacter transmission. Pathogens 2014; 3:875-907. [PMID: 25436509 PMCID: PMC4282890 DOI: 10.3390/pathogens3040875] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/18/2014] [Accepted: 10/20/2014] [Indexed: 01/01/2023] Open
Abstract
The potato psyllid (PoP) Bactericera cockerelli (Sulc) and Asian citrus psyllid (ACP) Diaphorina citri Kuwayama are the insect vectors of the fastidious plant pathogen, Candidatus Liberibacter solanacearum (CLso) and Ca. L. asiaticus (CLas), respectively. CLso causes Zebra chip disease of potato and vein-greening in solanaceous species, whereas, CLas causes citrus greening disease. The reliance on insecticides for vector management to reduce pathogen transmission has increased interest in alternative approaches, including RNA interference to abate expression of genes essential for psyllid-mediated Ca. Liberibacter transmission. To identify genes with significantly altered expression at different life stages and conditions of CLso/CLas infection, cDNA libraries were constructed for CLso-infected and -uninfected PoP adults and nymphal instars. Illumina sequencing produced 199,081,451 reads that were assembled into 82,224 unique transcripts. PoP and the analogous transcripts from ACP adult and nymphs reported elsewhere were annotated, organized into functional gene groups using the Gene Ontology classification system, and analyzed for differential in silico expression. Expression profiles revealed vector life stage differences and differential gene expression associated with Liberibacter infection of the psyllid host, including invasion, immune system modulation, nutrition, and development.
Collapse
Affiliation(s)
- Tonja W Fisher
- School of Plant Sciences, The University of Arizona, Tucson, AZ 85721, USA.
| | - Meenal Vyas
- School of Plant Sciences, The University of Arizona, Tucson, AZ 85721, USA.
| | - Ruifeng He
- Institute of Biological Chemistry, Washington State University, Pullman, WA 99164, USA.
| | | | - Joseph M Cicero
- School of Plant Sciences, The University of Arizona, Tucson, AZ 85721, USA.
| | - Mark Willer
- BIO5, The University of Arizona, Tucson, AZ 85721, USA.
| | - Ryan Kim
- National Center for Genome Resources, 2935 Rodeo Park Drive East, Santa Fe, NM 87505, USA.
| | - Robin Kramer
- National Center for Genome Resources, 2935 Rodeo Park Drive East, Santa Fe, NM 87505, USA.
| | - Greg A May
- National Center for Genome Resources, 2935 Rodeo Park Drive East, Santa Fe, NM 87505, USA.
| | - John A Crow
- National Center for Genome Resources, 2935 Rodeo Park Drive East, Santa Fe, NM 87505, USA.
| | | | - David R Gang
- Institute of Biological Chemistry, Washington State University, Pullman, WA 99164, USA.
| | - Judith K Brown
- School of Plant Sciences, The University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
45
|
Adjobo-Hermans MJW. Fast reversibility of dimeriser system enables quantification of signal molecule turnover. Chembiochem 2014; 15:2037-9. [PMID: 25145328 DOI: 10.1002/cbic.201402294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Indexed: 11/10/2022]
Abstract
The design of a brake: Chemical induced dimerisation systems have revolutionised signal transduction research by allowing fast activation of specific proteins. A recent report describes the design of tools that enable the rapid switching off of the induced signal, thereby enabling quantification of signal molecule turnover.
Collapse
Affiliation(s)
- Merel J W Adjobo-Hermans
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Geert Grooteplein 28, 6525 GA, Nijmegen (The Netherlands).
| |
Collapse
|
46
|
Onuma H, Komatsu T, Arita M, Hanaoka K, Ueno T, Terai T, Nagano T, Inoue T. Rapidly rendering cells phagocytic through a cell surface display technique and concurrent Rac activation. Sci Signal 2014; 7:rs4. [PMID: 25028719 DOI: 10.1126/scisignal.2005123] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell surfaces represent a platform through which extracellular signals that determine diverse cellular processes, including migration, division, adhesion, and phagocytosis, are transduced. Techniques to rapidly reconfigure the surface properties of living cells should thus offer the ability to harness these cellular functions. Although the molecular mechanism of phagocytosis is well characterized, the minimal molecular players that are sufficient to activate this elaborate process remain elusive. We developed and implemented a technique to present a molecule of interest at the cell surface in an inducible manner on a time scale of minutes. We simultaneously induced the cell surface display of the C2 domain of milk fat globule epidermal growth factor factor 8 (MFG-E8) and activated the intracellular small guanosine triphosphatase Rac, which stimulates actin polymerization at the cell periphery. The C2 domain binds to phosphatidylserine, a lipid exposed on the surface of apoptotic cells. By integrating the stimulation of these two processes, we converted HeLa cells into a phagocytic cell line that bound to and engulfed apoptotic human Jurkat cells. Inducing either the cell surface display of the C2 domain or activating Rac alone was not sufficient to stimulate phagocytosis, which suggests that attachment to the target cell and actin reorganization together constitute the minimal molecular events that are needed to induce phagocytosis. This cell surface display technique might be useful as part of a targeted, cell-based therapy in which unwanted cells with characteristic surface molecules could be rapidly consumed by engineered cells.
Collapse
Affiliation(s)
- Hiroki Onuma
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Toru Komatsu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan. Precursory Research for Embryonic Science and Technology Investigator, Japan Science and Technology Agency, 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan.
| | - Makoto Arita
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Kenjiro Hanaoka
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Tasuku Ueno
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Takuya Terai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Tetsuo Nagano
- Open Innovation Center for Drug Discovery, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takanari Inoue
- Precursory Research for Embryonic Science and Technology Investigator, Japan Science and Technology Agency, 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan. Department of Cell Biology, School of Medicine, Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA. Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
47
|
Zimmermann M, Cal R, Janett E, Hoffmann V, Bochet CG, Constable E, Beaufils F, Wymann MP. Cell-permeant and photocleavable chemical inducer of dimerization. Angew Chem Int Ed Engl 2014; 53:4717-20. [PMID: 24677313 PMCID: PMC4499241 DOI: 10.1002/anie.201310969] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Indexed: 11/30/2022]
Abstract
Chemical inducers of dimerization (CIDs) have been developed to orchestrate protein dimerization and translocation. Here we present a novel photocleavable HaloTag- and SNAP-tag-reactive CID (MeNV-HaXS) with excellent selectivity and intracellular reactivity. Excitation at 360 nm cleaves the methyl-6-nitroveratryl core of MeNV-HaXS. MeNV-HaXS covalently links HaloTag- and SNAP-tag fusion proteins, and enables targeting of selected membranes and intracellular organelles. MeNV-HaXS-mediated translocation has been validated for plasma membrane, late endosomes, lysosomes, Golgi, mitochondria, and the actin cytoskeleton. Photocleavage of MeNV-HaXS liberates target proteins and provides access to optical manipulation of protein relocation with high spatiotemporal and subcellular precision. MeNV-HaXS supports kinetic studies of protein dynamics and the manipulation of subcellular enzyme activities, which is exemplified for Golgi-targeted cargo and the assessment of nuclear import kinetics.
Collapse
Affiliation(s)
- Mirjam Zimmermann
- University of Basel, Department of BiomedicineMattenstrasse 28, Basel (Switzerland)
| | - Ruben Cal
- University of Basel, Department of BiomedicineMattenstrasse 28, Basel (Switzerland)
| | - Elia Janett
- University of Fribourg, Department of ChemistryChemin du Musée 9, Fribourg (Switzerland)
| | - Viktor Hoffmann
- University of Basel, Department of BiomedicineMattenstrasse 28, Basel (Switzerland)
| | - Christian G Bochet
- University of Fribourg, Department of ChemistryChemin du Musée 9, Fribourg (Switzerland)
| | - Edwin Constable
- University of Basel, Department of ChemistrySpitalstrasse 51, Basel (Switzerland)
| | - Florent Beaufils
- University of Basel, Department of BiomedicineMattenstrasse 28, Basel (Switzerland)
| | - Matthias P Wymann
- University of Basel, Department of BiomedicineMattenstrasse 28, Basel (Switzerland)
| |
Collapse
|
48
|
Zimmermann M, Cal R, Janett E, Hoffmann V, Bochet CG, Constable E, Beaufils F, Wymann MP. Ein zellpermeables und photospaltbares Reagens für die selektive intrazelluläre Protein-Protein-Dimerisierung. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201310969] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
49
|
Cabello J, Sämann J, Gómez-Orte E, Erazo T, Coppa A, Pujol A, Büssing I, Schulze B, Lizcano JM, Ferrer I, Baumeister R, Dalfo E. PDR-1/hParkin negatively regulates the phagocytosis of apoptotic cell corpses in Caenorhabditis elegans. Cell Death Dis 2014; 5:e1120. [PMID: 24625979 PMCID: PMC3973248 DOI: 10.1038/cddis.2014.57] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/23/2013] [Accepted: 01/22/2014] [Indexed: 12/28/2022]
Abstract
Apoptotic cell death is an integral part of cell turnover in many tissues, and proper corpse clearance is vital to maintaining tissue homeostasis in all multicellular organisms. Even in tissues with high cellular turnover, apoptotic cells are rarely seen because of efficient clearance mechanisms in healthy individuals. In Caenorhabditis elegans, two parallel and partly redundant conserved pathways act in cell corpse engulfment. The pathway for cytoskeletal rearrangement requires the small GTPase CED-10 Rac1 acting for an efficient surround of the dead cell. The CED-10 Rac pathway is also required for the proper migration of the distal tip cells (DTCs) during the development of the C. elegans gonad. Parkin, the mammalian homolog of the C. elegans PDR-1, interacts with Rac1 in aged human brain and it is also implicated with actin dynamics and cytoskeletal rearrangements in Parkinsons's disease, suggesting that it might act on engulfment. Our genetic and biochemical studies indicate that PDR-1 inhibits apoptotic cell engulfment and DTC migration by ubiquitylating CED-10 for degradation.
Collapse
Affiliation(s)
- J Cabello
- CIBIR (Centre for Biomedical Research of La Rioja), C/Piqueras 98, Logroño 26006, Spain
| | - J Sämann
- Bioinformatics and Molecular Genetics (Faculty of Biology), Center for Biochemistry and Molecular Cell Research (Faculty of Medicine), Schänzlestrasse 1, Freiburg 79104, Germany
| | - E Gómez-Orte
- CIBIR (Centre for Biomedical Research of La Rioja), C/Piqueras 98, Logroño 26006, Spain
| | - T Erazo
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, Barcelona 08193, Spain
| | - A Coppa
- Neurometabolic Diseases Laboratory, Institut D'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona 08907, Spain
| | - A Pujol
- 1] Neurometabolic Diseases Laboratory, Institut D'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona 08907, Spain [2] ICREA (Institució Catalana de Recerca i Estudis avançats), Barcelona, Spain [3] Institute of Neuropathology, University Hospitall Bellvitge - University of Barcelona - IDIBELL, L'Hospitalet de Llobregat, Ciberned, Spain [4] CIBERER (Centro de Investigación Biomédica en Enfermedades Raras), C/ Álvaro de Bazán, 10 Bajo, Valencia 46010, Spain
| | - I Büssing
- Bioinformatics and Molecular Genetics (Faculty of Biology), Center for Biochemistry and Molecular Cell Research (Faculty of Medicine), Schänzlestrasse 1, Freiburg 79104, Germany
| | - B Schulze
- Bioinformatics and Molecular Genetics (Faculty of Biology), Center for Biochemistry and Molecular Cell Research (Faculty of Medicine), Schänzlestrasse 1, Freiburg 79104, Germany
| | - J M Lizcano
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, Barcelona 08193, Spain
| | - I Ferrer
- Institute of Neuropathology, University Hospitall Bellvitge - University of Barcelona - IDIBELL, L'Hospitalet de Llobregat, Ciberned, Spain
| | - R Baumeister
- 1] Bioinformatics and Molecular Genetics (Faculty of Biology), Center for Biochemistry and Molecular Cell Research (Faculty of Medicine), Schänzlestrasse 1, Freiburg 79104, Germany [2] Centre for Biological Signaling Studies (bioss), University of Freiburg, Freiburg 79104, Germany [3] FRIAS Freiburg Institute for Advanced Studies, Section Life Sciences (LIFENET), University of Freiburg, Schaenzlestrasse 1, Freiburg 79104, Germany
| | - E Dalfo
- 1] Bioinformatics and Molecular Genetics (Faculty of Biology), Center for Biochemistry and Molecular Cell Research (Faculty of Medicine), Schänzlestrasse 1, Freiburg 79104, Germany [2] Neurometabolic Diseases Laboratory, Institut D'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona 08907, Spain [3] Institute of Neuropathology, University Hospitall Bellvitge - University of Barcelona - IDIBELL, L'Hospitalet de Llobregat, Ciberned, Spain [4] CIBERER (Centro de Investigación Biomédica en Enfermedades Raras), C/ Álvaro de Bazán, 10 Bajo, Valencia 46010, Spain
| |
Collapse
|
50
|
|