1
|
Zhang L, Kitzmiller CE, Richard AS, Popli S, Choe H. The ability of human TIM1 to bind phosphatidylethanolamine enhances viral uptake and efferocytosis compared to rhesus and mouse orthologs. J Virol 2024:e0164924. [PMID: 39475278 DOI: 10.1128/jvi.01649-24] [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: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 11/06/2024] Open
Abstract
T-cell immunoglobulin and mucin (TIM) family proteins facilitate the clearance of apoptotic cells, are involved in immune regulation, and promote infection of enveloped viruses. These processes are frequently studied in experimental animals, such as mice or rhesus macaques, but functional differences among the TIM orthologs from these species have not been described. Previously, we reported that while all three human TIM proteins bind phosphatidylserine (PS), only human TIM1 (hTIM1) binds phosphatidylethanolamine (PE), and that this PE-binding ability contributes to both phagocytic clearance of apoptotic cells and viral infection. Here, we show that rhesus macaque TIM1 (rhTIM1) and mouse TIM1 (mTIM1) bind PS but not PE, and that their inability to bind PE makes them less efficient than hTIM1. We also show that alteration of only two residues of mTIM1 or rhTIM1 enables them to bind both PE and PS, and that these PE-binding variants are more efficient at phagocytosis and mediating viral entry. Further, we demonstrate that the mucin domain also contributes to the binding of the virions and apoptotic cells, although it does not directly bind phospholipid. Interestingly, contribution of the hTIM1 mucin domain is more pronounced in the presence of a PE-binding head domain. These results demonstrate that rhTIM1 and mTIM1 are inherently less functional than hTIM1, owing to their inability to bind PE and their less functional mucin domains. They also imply that mouse and macaque models underestimate the activity of hTIM1.IMPORTANCEWe previously reported that human T-cell immunoglobulin and mucin protein 1 (TIM1) binds phosphatidylethanolamine (PE) as well as phosphatidylserine (PS), and that PE is exposed on the apoptotic cells and viral envelopes. Moreover, TIM1 recognition of PE contributes to phagocytic clearance of apoptotic cells and virus uptake. Here, we report that unlike human TIM1, murine and rhesus TIM1 orthologs bind only PS, and as a result, their ability to clear apoptotic cells or promote virus infection is less efficient. These findings are significant because they imply that the activity of TIM1 in humans is greater than what the studies conducted in common animal models would indicate.
Collapse
Affiliation(s)
- Lizhou Zhang
- Division of Infectious Disease, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Research, Jupiter, Florida, USA
| | - Claire E Kitzmiller
- Division of Infectious Disease, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Audrey S Richard
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Research, Jupiter, Florida, USA
| | - Sonam Popli
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Research, Jupiter, Florida, USA
| | - Hyeryun Choe
- Division of Infectious Disease, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Research, Jupiter, Florida, USA
| |
Collapse
|
2
|
Amin N, Jaiswal M, Kannaujiya VK. Effects of temperature on morphology, physiology, and metabolic profile of diazotrophic cyanobacteria inhabiting diverse habitats. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2024; 216:109186. [PMID: 39388751 DOI: 10.1016/j.plaphy.2024.109186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/29/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024]
Abstract
Global population expansion has increased the demand for food supply and agricultural productivity. Abiotic stressors like temperature have significantly restricted agriculture in cropland and jeopardized food security. Cyanobacteria play a crucial role in fostering sustainable agriculture and ensuring global food security. In the present study, we have assessed the effect of temperatures on diazotrophic free living rice-field and hot-spring cyanobacteria. They were treated to a variable range of temperatures to see the changes in cellular morphology, physiology, and biochemical characteristics. The rise of temperatures induces growth (60 %), total protein (54 %) contents of rice-field cyanobacterium until 25 °C, further treatment results in decline (20 %) at 45 °C. However, growth indices were increased till 35 °C (90 %) in hot-spring cyanobacterium and further treatment did not exhibit a significant decline in the same. However, the reactive oxygen species (ROS) generation and lipid peroxidation (LPO) were higher in rice-field (2.8 and 1.7 fold) as compared to hot-spring cyanobacterium (2.2 and 1.6 fold). In response to temperature, enzymatic antioxidant contents were much higher in hot-spring as compared to rice-field cyanobacterium. Similarly, carotenoid and carbohydrate content was also higher in hot spring (2 fold) as compared to rice-field cyanobacterium (1.5 and 1.2 fold). All these data collectively suggest that hot-spring (Nostoc sp. strain VKB02) has a higher thermoprotective capacity with novel defense mechanisms as compared to rice-field cyanobacterium (Anabaena sp. strain VKB01). These findings contributed to a better understanding of the temperature stress, improvement of agricultural productivity and future welfare of green ecosystems.
Collapse
Affiliation(s)
- Nasreen Amin
- Department of Botany, MMV, Banaras Hindu University, Varanasi 221005, India
| | - Megha Jaiswal
- Department of Botany, MMV, Banaras Hindu University, Varanasi 221005, India
| | - Vinod K Kannaujiya
- Department of Botany, MMV, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
3
|
Smith R, Bassand K, Dussol A, Piesse C, Duplus E, El Hadri K. A new model and precious tool to study molecular mechanisms of macrophage aging. Aging (Albany NY) 2024; 16:12697-12725. [PMID: 39373702 PMCID: PMC11501386 DOI: 10.18632/aging.206124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 08/06/2024] [Indexed: 10/08/2024]
Abstract
The accumulation of senescent cells, characterized by a senescence-associated secretory phenotype (SASP), contributes to chronic inflammation and age-related diseases (ARD). During aging, macrophages can adopt a senescent-like phenotype and an altered function, which promotes senescent cell accumulation. In the context of aging and ARD, controlling the resolution of the inflammatory response and preventing chronic inflammation, especially by targeting macrophages, must be a priority. Aging being a dynamic process, we developed a model of in vitro murine peritoneal macrophage aging. Our results show that macrophages cultured for 7 or 14 days exhibit a senescence-like phenotype: proliferation decrease, the levels of cyclin-dependent kinase inhibitors p16INK4A and p21CIP1 and of pro-inflammatory SASP components (MCP-1, IL-6, IL-1β, TNF-α, and MMP-9) increase, phagocytosis capacity decline and glycolytic activity is induced. In our model, chronic treatment with CB3, a thioredoxin-1 mimetic anti-inflammatory peptide, completely prevents p21CIP1 increase and enables day 14 macrophages to maintain proliferative activity.We describe a new model of macrophage aging with a senescence-like phenotype associated with inflammatory, metabolic and functional perturbations. This model is a valuable tool for characterizing macrophage aging mechanisms and developing innovative strategies with promising therapeutical purpose in limiting inflammaging and ARD.
Collapse
Affiliation(s)
- Rémy Smith
- Sorbonne Université, CNRS UMR 8256 Biological Adaptation and Ageing (B2A), INSERM U1164, Institut de Biologie Paris Seine (IBPS), Paris 75005, France
| | - Kévin Bassand
- INSERM U1148, Laboratory for Vascular and Translational Sciences (LVTS), Université Sorbonne Paris Nord, Bobigny 93000, France
| | - Ashok Dussol
- Sorbonne Université, CNRS UMR 8256 Biological Adaptation and Ageing (B2A), INSERM U1164, Institut de Biologie Paris Seine (IBPS), Paris 75005, France
| | - Christophe Piesse
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine (IBPS), Plate-forme Ingénierie des Protéines et Synthèse Peptidique, Paris 75005, France
| | - Eric Duplus
- Sorbonne Université, CNRS UMR 8256 Biological Adaptation and Ageing (B2A), INSERM U1164, Institut de Biologie Paris Seine (IBPS), Paris 75005, France
| | - Khadija El Hadri
- Sorbonne Université, CNRS UMR 8256 Biological Adaptation and Ageing (B2A), INSERM U1164, Institut de Biologie Paris Seine (IBPS), Paris 75005, France
| |
Collapse
|
4
|
Farias HR, Ramos JMO, Griesang CT, Santos L, Junior OVR, Souza DG, Ferreira FS, Somacal S, Martins LAM, de Souza DOG, Moreira JCF, Wyse ATS, Guma FTCR, de Oliveira J. LDL Exposure Disrupts Mitochondrial Function and Dynamics in a Hippocampal Neuronal Cell Line. Mol Neurobiol 2024:10.1007/s12035-024-04476-y. [PMID: 39302616 DOI: 10.1007/s12035-024-04476-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/30/2024] [Indexed: 09/22/2024]
Abstract
Hypercholesterolemia has been associated with cognitive dysfunction and neurodegenerative diseases. Moreover, this metabolic condition disrupts the blood-brain barrier, allowing low-density lipoprotein (LDL) to enter the central nervous system. Thus, we investigated the effects of LDL exposure on mitochondrial function in a mouse hippocampal neuronal cell line (HT-22). HT-22 cells were exposed to human LDL (50 and 300 μg/mL) for 24 h. After this, intracellular lipid droplet (LD) content, cell viability, cell death, and mitochondrial parameters were assessed. We found that the higher LDL concentration increases LD content compared with control. Both concentrations increased the number of Annexin V-positive cells, indicating apoptosis. Moreover, in mitochondrial parameters, the LDL exposure on hippocampal neuronal cell line leads to a decrease in mitochondrial complexes I and II activities in both concentrations tested and a reduction in Mitotracker™ Red fluorescence and Mitotracker™ Red and Mitotracker™ Green ratio in the higher concentration, indicating mitochondrial impairment. The LDL incubation induces mitochondrial superoxide production and decreases superoxide dismutase activity in the lower concentration in HT-22 cells. Finally, LDL exposure increases the expression of genes associated with mitochondrial fusion (OPA1 and mitofusin 2) in the lower concentration. In conclusion, our findings suggest that LDL exposure induces mitochondrial dysfunction and modulates mitochondrial dynamics in the hippocampal neuronal cells.
Collapse
Affiliation(s)
- Hémelin Resende Farias
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Jessica Marques Obelar Ramos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Caroline Tainá Griesang
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Lucas Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Osmar Vieira Ramires Junior
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Debora Guerini Souza
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fernanda Silva Ferreira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Sabrina Somacal
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Leo Anderson Meira Martins
- Programa de Pós-Graduação em Fisiologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Diogo Onofre Gomes de Souza
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - José Cláudio Fonseca Moreira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Angela T S Wyse
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fátima Theresinha Costa Rodrigues Guma
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Jade de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
5
|
Pati A, Das BK, Panda AK. Elevated toll-like receptor 9 is associated with disease severity and kidney involvement in systemic lupus erythematosus. Hum Immunol 2024; 85:111104. [PMID: 39255560 DOI: 10.1016/j.humimm.2024.111104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/22/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024]
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE) is associated with the activation of both innate and adaptive immune system. Infection is a significant environmental factor that is responsible for the development of SLE. Toll-like receptors (TLRs) are responsible for recognizing pathogens, and the expression of TLRs has been found to differ in SLE patients. Additionally, various infections have been reported to influence TLR expression. This study aimed to explore the relationship between TLRs and the onset, severity, and symptoms of SLE in the eastern Indian population. METHODS The study included 70 SLE patients and a control group matched for age and sex. RT-PCR was used to evaluate mRNA expression of TLRs 2, 4, 7, and 9. Statistical analyses were performed using GraphPad Prism software v.10.2.3. RESULTS Patients with SLE expressed significantly higher levels of TLR2 (p < 0.0001) and TLR9 (p = 0.012) than healthy controls. In lupus nephritis, TLR9 expression was higher than in SLE patients without kidney involvement (p = 0.037). Furthermore, a significant relationship was found between TLR9 expression and systemic lupus erythematosus disease activity index (SLEDAI) scores (p < 0.0001, Spearman's r = 0.47), implying the potential role of TLRs in SLE development. However, mRNA expression of TLR4 and TLR7 was not associated with SLE, clinical indices, or disease severity. CONCLUSIONS TLR9 is associated with SLE pathogenesis and clinical severity, making it a promising molecule for targeted therapy in SLE management.
Collapse
Affiliation(s)
- Abhijit Pati
- ImmGen EvSys Laboratory, Department of Biotechnology, Berhampur University, Odisha 767007, India
| | - Bidyut K Das
- Department of Clinical Immunology and Rheumatology, SCB Medical College Cuttack, Odisha 753007, India
| | - Aditya K Panda
- ImmGen EvSys Laboratory, Department of Biotechnology, Berhampur University, Odisha 767007, India; Centre of Excellence on Bioprospecting of "Ethnopharmaceuticals of Southern Odisha" (CoE-BESO), Berhampur University, Bhanja Bihar, Berhampur, Odisha 760007, India.
| |
Collapse
|
6
|
Zhang L, Kitzmiller CE, Richard AS, Popli S, Choe H. The ability of human TIM1 to bind phosphatidylethanolamine enhances viral uptake and efferocytosis compared to rhesus and mouse orthologs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605603. [PMID: 39131348 PMCID: PMC11312472 DOI: 10.1101/2024.07.29.605603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
T-cell Immunoglobulin and Mucin (TIM)-family proteins facilitate the clearance of apoptotic cells, are involved in immune regulation, and promote infection of enveloped viruses. These processes are frequently studied in experimental animals such as mice or rhesus macaques, but functional differences among the TIM orthologs from these species have not been described. Previously, we reported that while all three human TIM proteins bind phosphatidylserine (PS), only human TIM1 (hTIM1) binds phosphatidylethanolamine (PE), and that this PE-binding ability contributes to both phagocytic clearance of apoptotic cells and virus infection. Here we show that rhesus macaque TIM1 (rhTIM1) and mouse TIM1 (mTIM1) bind PS but not PE and that their inability to bind PE makes them less efficient than hTIM1. We also show that alteration of only two residues of mTIM1 or rhTIM1 enables them to bind both PE and PS, and that these PE-binding variants are more efficient at phagocytosis and mediating viral entry. Further, we demonstrate that the mucin domain also contributes to the binding of the virions and apoptotic cells, although it does not directly bind phospholipid. Interestingly, contribution of the hTIM1 mucin domain is more pronounced in the presence of a PE-binding head domain. These results demonstrate that rhTIM1 and mTIM1 are inherently less functional than hTIM1, owing to their inability to bind PE and their less functional mucin domains. They also imply that mouse and macaque models underestimate the activity of hTIM1.
Collapse
Affiliation(s)
- Lizhou Zhang
- Division of Infectious Disease, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Research, Jupiter, FL 33458, USA
| | - Claire E. Kitzmiller
- Division of Infectious Disease, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Audrey S. Richard
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Research, Jupiter, FL 33458, USA
| | - Sonam Popli
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Research, Jupiter, FL 33458, USA
| | - Hyeryun Choe
- Division of Infectious Disease, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Research, Jupiter, FL 33458, USA
| |
Collapse
|
7
|
Dingwall CB, Sasaki Y, Strickland A, Summers DW, Bloom AJ, DiAntonio A, Milbrandt J. Suppressing phagocyte activation by overexpressing the phosphatidylserine lipase ABHD12 preserves sarmopathic nerves. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599919. [PMID: 38979309 PMCID: PMC11230269 DOI: 10.1101/2024.06.20.599919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Programmed axon degeneration (AxD) is a key feature of many neurodegenerative diseases. In healthy axons, the axon survival factor NMNAT2 inhibits SARM1, the central executioner of AxD, preventing it from initiating the rapid local NAD+ depletion and metabolic catastrophe that precipitates axon destruction. Because these components of the AxD pathway act within neurons, it was also assumed that the timetable of AxD was set strictly by a cell-intrinsic mechanism independent of neuron-extrinsic processes later activated by axon fragmentation. However, using a rare human disease model of neuropathy caused by hypomorphic NMNAT2 mutations and chronic SARM1 activation (sarmopathy), we demonstrated that neuronal SARM1 can initiate macrophage-mediated axon elimination long before stressed-but-viable axons would otherwise succumb to cell-intrinsic metabolic failure. Investigating potential SARM1-dependent signals that mediate macrophage recognition and/or engulfment of stressed-but-viable axons, we found that chronic SARM1 activation triggers axonal blebbing and dysregulation of phosphatidylserine (PS), a potent phagocyte immunomodulatory molecule. Neuronal expression of the phosphatidylserine lipase ABDH12 suppresses nerve macrophage activation, preserves motor axon integrity, and rescues motor function in this chronic sarmopathy model. We conclude that PS dysregulation is an early SARM1-dependent axonal stress signal, and that blockade of phagocytic recognition and engulfment of stressed-but-viable axons could be an attractive therapeutic target for management of neurological disorders involving SARM1 activation.
Collapse
|
8
|
Guillet S, Lazarov T, Jordan N, Boisson B, Tello M, Craddock B, Zhou T, Nishi C, Bareja R, Yang H, Rieux-Laucat F, Lorenzo RIF, Dyall SD, Isenberg D, D’Cruz D, Lachmann N, Elemento O, Viale A, Socci ND, Abel L, Nagata S, Huse M, Miller WT, Casanova JL, Geissmann F. ACK1 and BRK non-receptor tyrosine kinase deficiencies are associated with familial systemic lupus and involved in efferocytosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.15.24302255. [PMID: 38883731 PMCID: PMC11177913 DOI: 10.1101/2024.02.15.24302255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Systemic Lupus Erythematosus (SLE) is an autoimmune disease, the pathophysiology and genetic basis of which are incompletely understood. Using a forward genetic screen in multiplex families with systemic lupus erythematosus (SLE) we identified an association between SLE and compound heterozygous deleterious variants in the non-receptor tyrosine kinases (NRTKs) ACK1 and BRK. Experimental blockade of ACK1 or BRK increased circulating autoantibodies in vivo in mice and exacerbated glomerular IgG deposits in an SLE mouse model. Mechanistically, non-receptor tyrosine kinases (NRTKs) regulate activation, migration, and proliferation of immune cells. We found that the patients' ACK1 and BRK variants impair efferocytosis, the MERTK-mediated anti-inflammatory response to apoptotic cells, in human induced Pluripotent Stem Cell (hiPSC)-derived macrophages, which may contribute to SLE pathogenesis. Overall, our data suggest that ACK1 and BRK deficiencies are associated with human SLE and impair efferocytosis in macrophages.
Collapse
Affiliation(s)
- Stephanie Guillet
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Ecole doctorale Bio Sorbonne Paris Cité, Université Paris Descartes-Sorbonne Paris Cité.Paris, France
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of MedicalSciences, New York, New York 10065, USA
| | - Natasha Jordan
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ Hospitals, London SE1 1UL, UK
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
- University of Paris Cité, Imagine Institute, Paris, France
| | - Maria Tello
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Barbara Craddock
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Ting Zhou
- SKI Stem Cell Research Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Chihiro Nishi
- Laboratory of Biochemistry & Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871
| | - Rohan Bareja
- Cary and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Meyer Cancer Center Weill Cornell Medical College, New York, New York 10065, USA
| | - Hairu Yang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | | | | | - Sabrina D. Dyall
- Department of Biosciences and Ocean Studies, Faculty of Science, University of Mauritius, Reduit, Mauritius
| | - David Isenberg
- Centre for Rheumatology, Division of Medicine, University College London, The Rayne Building, University College London
| | - David D’Cruz
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ Hospitals, London SE1 1UL, UK
| | - Nico Lachmann
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover 30625, Germany
| | - Olivier Elemento
- Cary and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Meyer Cancer Center Weill Cornell Medical College, New York, New York 10065, USA
| | - Agnes Viale
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Nicholas D. Socci
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Bioinformatics Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
- University of Paris Cité, Imagine Institute, Paris, France
| | - Shigekazu Nagata
- Laboratory of Biochemistry & Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871
| | - Morgan Huse
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
- University of Paris Cité, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, 10065 NY, USA
- Lab of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick Children, Paris, France, EU
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France, EU
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of MedicalSciences, New York, New York 10065, USA
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ Hospitals, London SE1 1UL, UK
| |
Collapse
|
9
|
Wang J, Li J, Yin L, Wang X, Dong Y, Zhao G, Shen S, Hou Y. MSCs promote the efferocytosis of large peritoneal macrophages to eliminate ferroptotic monocytes/macrophages in the injured endometria. Stem Cell Res Ther 2024; 15:127. [PMID: 38693589 PMCID: PMC11064342 DOI: 10.1186/s13287-024-03742-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Endometria are one of the important components of the uterus, which is located in the peritoneal cavity. Endometrial injury usually leads to intrauterine adhesions (IUA), accompanied by inflammation and cell death. We previously reported that both the endometrial ferroptosis was increased and monocytes/macrophages were involved in endometrial injury of IUA. Large peritoneal macrophages (LPMs) are recently reported to migrate into the injured tissues and phagocytose dead cells to repair the tissues. We previously demonstrated that mesenchymal stromal cells (MSCs) had made excellent progress in the repair of endometrial injury. However, it is unclear whether MSCs regulate the LPM efferocytosis against ferroptotic monocytes/macrophages in the injured endometria. METHODS Here, endometrial injury in IUA mouse model was conducted by uterine curettage and LPS injection surgery and the samples were collected at different times to detect the changes of LPMs and ferroptotic monocytes/macrophages. We conducted LPMs depletion assay in vivo and LPMs and Erastin-induced ferroptotic THP-1 cells coculture systems in vitro to detect the LPM efferocytosis against ferroptotic monocytes/macrophages. The IUA model was treated with MSCs, and their effects on LPMs and endometrial repair were analyzed. Flow cytometry, western blotting, quantitative real-time PCR, immunohistochemical analysis, ELISA, and RNA-sequencing were performed. RESULTS We found that LPMs migrated to the injured uteri in response to the damage in early phase (3 h), and sustained to a later stage (7 days). Astonishingly, we found that ferroptotic monocytes/macrophages were significantly increased in the injured uteri since 12 h after injury. Moreover, LPMs cocultured with Erastin-induced ferroptotic THP-1 cells in vitro, efferocytosis of LPMs against ferroptotic monocytes/macrophages was emerged. The mRNA expression profiles revealed that LPM efferocytosis against ferroptotic monocytes/macrophages was an induction of glycolysis program and depended on the PPARγ-HK2 pathway. Importantly, we validated that MSCs promoted the efferocytic capability and migration of LPMs to the injured uteri via secreting stanniocalcin-1 (STC-1). CONCLUSION The data collectively demonstrated first the roles of LPMs via removal of ferroptotic monocytes/macrophages and provided a novel mechanism of MSCs in repairing the endometrial injury.
Collapse
Affiliation(s)
- Jiali Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China
| | - Jingman Li
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China
| | - Lijie Yin
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China
| | - Xiuzhu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China
| | - Yue Dong
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China
| | - Guangfeng Zhao
- Department of Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Sunan Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, No. 22 Hankou Rd., Gulou District, Nanjing, Jiangsu, 210093, People's Republic of China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
10
|
Mann V, Sundaresan A, Shishodia S. Overnutrition and Lipotoxicity: Impaired Efferocytosis and Chronic Inflammation as Precursors to Multifaceted Disease Pathogenesis. BIOLOGY 2024; 13:241. [PMID: 38666853 PMCID: PMC11048223 DOI: 10.3390/biology13040241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/25/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
Overnutrition, driven by the consumption of high-fat, high-sugar diets, has reached epidemic proportions and poses a significant global health challenge. Prolonged overnutrition leads to the deposition of excessive lipids in adipose and non-adipose tissues, a condition known as lipotoxicity. The intricate interplay between overnutrition-induced lipotoxicity and the immune system plays a pivotal role in the pathogenesis of various diseases. This review aims to elucidate the consequences of impaired efferocytosis, caused by lipotoxicity-poisoned macrophages, leading to chronic inflammation and the subsequent development of severe infectious diseases, autoimmunity, and cancer, as well as chronic pulmonary and cardiovascular diseases. Chronic overnutrition promotes adipose tissue expansion which induces cellular stress and inflammatory responses, contributing to insulin resistance, dyslipidemia, and metabolic syndrome. Moreover, sustained exposure to lipotoxicity impairs the efferocytic capacity of macrophages, compromising their ability to efficiently engulf and remove dead cells. The unresolved chronic inflammation perpetuates a pro-inflammatory microenvironment, exacerbating tissue damage and promoting the development of various diseases. The interaction between overnutrition, lipotoxicity, and impaired efferocytosis highlights a critical pathway through which chronic inflammation emerges, facilitating the development of severe infectious diseases, autoimmunity, cancer, and chronic pulmonary and cardiovascular diseases. Understanding these intricate connections sheds light on potential therapeutic avenues to mitigate the detrimental effects of overnutrition and lipotoxicity on immune function and tissue homeostasis, thereby paving the way for novel interventions aimed at reducing the burden of these multifaceted diseases on global health.
Collapse
Affiliation(s)
| | | | - Shishir Shishodia
- Department of Biology, Texas Southern University, Houston, TX 77004, USA; (V.M.); (A.S.)
| |
Collapse
|
11
|
Zhan J, Wang J, Liang Y, Wang L, Huang L, Liu S, Zeng X, Zeng E, Wang H. Apoptosis dysfunction: unravelling the interplay between ZBP1 activation and viral invasion in innate immune responses. Cell Commun Signal 2024; 22:149. [PMID: 38402193 PMCID: PMC10893743 DOI: 10.1186/s12964-024-01531-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/13/2024] [Indexed: 02/26/2024] Open
Abstract
Apoptosis plays a pivotal role in pathogen elimination and maintaining homeostasis. However, viruses have evolved strategies to evade apoptosis, enabling their persistence within the host. Z-DNA binding protein 1 (ZBP1) is a potent innate immune sensor that detects cytoplasmic nucleic acids and activates the innate immune response to clear pathogens. When apoptosis is inhibited by viral invasion, ZBP1 can be activated to compensate for the effect of apoptosis by triggering an innate immune response. This review examined the mechanisms of apoptosis inhibition and ZBP1 activation during viral invasion. The authors outlined the mechanisms of ZBP1-induced type I interferon, pyroptosis and necroptosis, as well as the crosstalk between ZBP1 and the cGAS-STING signalling pathway. Furthermore, ZBP1 can reverse the suppression of apoptotic signals induced by viruses. Intriguingly, a positive feedback loop exists in the ZBP1 signalling pathway, which intensifies the innate immune response while triggering a cytokine storm, leading to tissue and organ damage. The prudent use of ZBP1, which is a double-edged sword, has significant clinical implications for treating infections and inflammation.
Collapse
Affiliation(s)
- Jianhao Zhan
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- HuanKui Academy, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Jisheng Wang
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Yuqing Liang
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Lisha Wang
- HuanKui Academy, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Le Huang
- HuanKui Academy, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Shanshan Liu
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Xiaoping Zeng
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Medical College, Jinhua Polytechnic, Jinhua, Zhejiang Province, 321017, China
| | - Erming Zeng
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China.
| | - Hongmei Wang
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China.
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi Province, 330006, China.
- Medical College, Jinhua Polytechnic, Jinhua, Zhejiang Province, 321017, China.
| |
Collapse
|
12
|
Yarychkivska O, Sharmin R, Elkhalil A, Ghose P. Apoptosis and beyond: A new era for programmed cell death in Caenorhabditis elegans. Semin Cell Dev Biol 2024; 154:14-22. [PMID: 36792437 DOI: 10.1016/j.semcdb.2023.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 01/27/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023]
Abstract
Programmed cell death (PCD) is crucial for normal development and homeostasis. Our first insights into the genetic regulation of apoptotic cell death came from in vivo studies in the powerful genetic model system of C. elegans. More recently, novel developmental cell death programs occurring both embryonically and post-embryonically, and sex-specifically, have been elucidated. Recent studies in the apoptotic setting have also shed new light on the intricacies of phagocytosis in particular. This review provides a brief historical perspective of the origins of PCD studies in C. elegans, followed by a more detailed description of non-canonical apoptotic and non-apoptotic death programs. We conclude by posing open questions and commenting on our outlook on the future of PCD studies in C. elegans, highlighting the importance of advanced imaging tools and the continued leveraging of C. elegans genetics both with classical and modern cutting-edge approaches.
Collapse
Affiliation(s)
| | | | | | - Piya Ghose
- The University of Texas at Arlington, USA.
| |
Collapse
|
13
|
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
|
14
|
Hou G, Wang X, Wang A, Yuan L, Zheng Q, Xiao H, Wang H. The role of secreted proteins in efferocytosis. Front Cell Dev Biol 2024; 11:1332482. [PMID: 38259511 PMCID: PMC10800375 DOI: 10.3389/fcell.2023.1332482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
The clearance of apoptotic cells known as efferocytosis is the final stage of apoptosis, and includes the recognition, phagocytosis, and degradation of apoptotic cells. The maintenance of tissue homeostasis requires the daily elimination of billions of apoptotic cells from the human body via the process of efferocytosis. Accordingly, aberrations in efferocytosis underlie a growing list of diseases, including atherosclerosis, cancer, and infections. During the initial phase of apoptosis, "Eat-Me" signals are exposed and recognized by phagocytes either directly through phagocyte receptors or indirectly through secreted proteins that function as bridge molecules that cross-link dying cells to phagocytes. Here, we set out to provide a comprehensive review of the molecular mechanisms and biological significance of secreted proteins in apoptotic cell clearance. Specifically, it focuses on how these secreted proteins act as bridging molecules to facilitate the clearance process.
Collapse
Affiliation(s)
| | | | | | | | | | - Hui Xiao
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Hui Wang
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| |
Collapse
|
15
|
Zhao X, Ting SM, Sun G, Bautista Garrido J, Obertas L, Aronowski J. Clearance of Neutrophils From ICH-Affected Brain by Macrophages Is Beneficial and Is Assisted by Lactoferrin and CD91. Stroke 2024; 55:166-176. [PMID: 38063014 PMCID: PMC10842928 DOI: 10.1161/strokeaha.123.045194] [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: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Within hours after intracerebral hemorrhage (ICH) onset, masses of polymorphonuclear neutrophils (PMNs) infiltrate the ICH-affected brain. After degranulation involving controlled release of many toxic antimicrobial molecules, the PMNs undergo rapid apoptosis and then are removed by phagocytic microglia/macrophages (MΦ) through a process called efferocytosis. Effective removal of PMNs may limit secondary brain damage and inflammation; however, the molecular mechanisms governing these cleanup activities are not well understood. We propose that scavenger receptor CD91 on myeloid phagocytes especially in presence of CD91 ligand, LTF (lactoferrin, protein abundant in PMNs), plays an important role in clearance of dead apoptotic PMNs (ANs). METHODS Mice/rats were subjected to an autologous blood injection model of ICH. Primary cultured microglia were used to assess phagocytosis of ANs. Immunohistochemistry was employed to assess CD91 expression and PMN infiltration. CD91 knockout mice selectively in myeloid phagocytes (Mac-CD91-KO) were used to establish the CD91/LTF function in phagocytosis and in reducing ICH-induced injury, as assessed using behavioral tests, hematoma resolution, and oxidative stress. RESULTS Masses of PMNs are found in ICH-affected brain, and they contain LTF. MΦ at the outer border of hematoma are densely packed, expressing CD91 and phagocytosing ANs. Microglia deficient in CD91 demonstrate defective phagocytosis of ANs, and mice deficient in CD91 (Mac-CD91-KO) subjected to ICH injury have increased neurological dysfunction that is associated with impaired hematoma resolution (hemoglobin and iron clearance) and elevated oxidative stress. LTF that normally ameliorates ICH injury in CD91-proficient control mice shows reduced therapeutic effects in Mac-CD91-KO mice. CONCLUSIONS Our study suggests that CD91 plays a beneficial role in improving ANs phagocytosis and ultimately post-ICH outcome and that the beneficial effect of LTF in ICH is in part dependent on presence of CD91 on MΦ.
Collapse
Affiliation(s)
- Xiurong Zhao
- Department of Neurology, University of Texas HSC, McGovern Medical School, Houston
| | - Shun-Ming Ting
- Department of Neurology, University of Texas HSC, McGovern Medical School, Houston
| | - Guanghua Sun
- Department of Neurology, University of Texas HSC, McGovern Medical School, Houston
| | | | - Lidiya Obertas
- Department of Neurology, University of Texas HSC, McGovern Medical School, Houston
| | - Jaroslaw Aronowski
- Department of Neurology, University of Texas HSC, McGovern Medical School, Houston
| |
Collapse
|
16
|
Linder S, Barcelona B. Get a grip: Podosomes as potential players in phagocytosis. Eur J Cell Biol 2023; 102:151356. [PMID: 37625234 DOI: 10.1016/j.ejcb.2023.151356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/27/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023] Open
Abstract
Podosomes have been known for several decades as micron-sized, F-actin-rich structures that play a pivotal role in cell migration and invasion, as they are able to mediate both cell-matrix attachment as well as extracellular matrix degradation. Particularly in monocytic cells, podosomes have been shown to fulfill a variety of additional functions such as sensing of substrate rigidity and topography, or cell-cell fusion. Increasing evidence now points to the involvement of podosome-like structures also during phagocytosis by immune cells such as macrophages, dendritic cells, and neutrophils. Here, we compare the different cell models and experimental set ups where "phagocytic podosomes" have been described. We also discuss the composition and architecture of these structures, their potential involvement in mechanosensing and particle disruption, as well as the pros and cons for addressing them as bona fide podosomes.
Collapse
Affiliation(s)
- Stefan Linder
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany.
| | - Bryan Barcelona
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
17
|
Hajibabaie F, Abedpoor N, Mohamadynejad P. Types of Cell Death from a Molecular Perspective. BIOLOGY 2023; 12:1426. [PMID: 37998025 PMCID: PMC10669395 DOI: 10.3390/biology12111426] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023]
Abstract
The former conventional belief was that cell death resulted from either apoptosis or necrosis; however, in recent years, different pathways through which a cell can undergo cell death have been discovered. Various types of cell death are distinguished by specific morphological alterations in the cell's structure, coupled with numerous biological activation processes. Various diseases, such as cancers, can occur due to the accumulation of damaged cells in the body caused by the dysregulation and failure of cell death. Thus, comprehending these cell death pathways is crucial for formulating effective therapeutic strategies. We focused on providing a comprehensive overview of the existing literature pertaining to various forms of cell death, encompassing apoptosis, anoikis, pyroptosis, NETosis, ferroptosis, autophagy, entosis, methuosis, paraptosis, mitoptosis, parthanatos, necroptosis, and necrosis.
Collapse
Affiliation(s)
- Fatemeh Hajibabaie
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord 88137-33395, Iran;
- Department of Physiology, Medicinal Plants Research Center, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan 81551-39998, Iran
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord 88137-33395, Iran
| | - Navid Abedpoor
- Department of Physiology, Medicinal Plants Research Center, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan 81551-39998, Iran
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan 81551-39998, Iran
| | - Parisa Mohamadynejad
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord 88137-33395, Iran;
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord 88137-33395, Iran
| |
Collapse
|
18
|
Duszyc K, von Pein JB, Ramnath D, Currin-Ross D, Verma S, Lim F, Sweet MJ, Schroder K, Yap AS. Apical extrusion prevents apoptosis from activating an acute inflammatory program in epithelia. Dev Cell 2023; 58:2235-2248.e6. [PMID: 37647898 DOI: 10.1016/j.devcel.2023.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/20/2023] [Accepted: 08/07/2023] [Indexed: 09/01/2023]
Abstract
Apoptosis is traditionally considered to be an immunologically silent form of cell death. Multiple mechanisms exist to ensure that apoptosis does not stimulate the immune system to cause inflammation or autoimmunity. Against this expectation, we now report that epithelia are programmed to provoke, rather than suppress, inflammation in response to apoptosis. We found that an acute inflammatory response led by neutrophils occurs in zebrafish and cell culture when apoptotic epithelial cells cannot be expelled from the monolayer by apical extrusion. This reflects an intrinsic circuit where ATP released from apoptotic cells stimulates epithelial cells in the immediate vicinity to produce interleukin-8 (IL-8). Apical extrusion therefore prevents inappropriate epithelial inflammation by physically eliminating apoptotic cells before they can activate this pro-inflammatory circuit. This carries the implication that epithelia may be predisposed to inflammation, elicited by sporadic or induced apoptosis, if apical extrusion is compromised.
Collapse
Affiliation(s)
- Kinga Duszyc
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia.
| | - Jessica B von Pein
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Divya Ramnath
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Denni Currin-Ross
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Suzie Verma
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Fayth Lim
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Matthew J Sweet
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Kate Schroder
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Alpha S Yap
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia.
| |
Collapse
|
19
|
Wang R, Li H, Ling C, Zhang X, Lu J, Luan W, Zhang J, Shi L. A novel phenotype of B cells associated with enhanced phagocytic capability and chemotactic function after ischemic stroke. Neural Regen Res 2023; 18:2413-2423. [PMID: 37282471 DOI: 10.4103/1673-5374.371365] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
Accumulating evidence has demonstrated the involvement of B cells in neuroinflammation and neuroregeneration. However, the role of B cells in ischemic stroke remains unclear. In this study, we identified a novel phenotype of macrophage-like B cells in brain-infiltrating immune cells expressing a high level of CD45. Macrophage-like B cells characterized by co-expression of B-cell and macrophage markers, showed stronger phagocytic and chemotactic functions compared with other B cells and showed upregulated expression of phagocytosis-related genes. Gene Ontology analysis found that the expression of genes associated with phagocytosis, including phagosome- and lysosome-related genes, was upregulated in macrophage-like B cells. The phagocytic activity of macrophage-like B cells was verified by immunostaining and three-dimensional reconstruction, in which TREM2-labeled macrophage-like B cells enwrapped and internalized myelin debris after cerebral ischemia. Cell-cell interaction analysis revealed that macrophage-like B cells released multiple chemokines to recruit peripheral immune cells mainly via CCL pathways. Single-cell RNA sequencing showed that the transdifferentiation to macrophage-like B cells may be induced by specific upregulation of the transcription factor CEBP family to the myeloid lineage and/or by downregulation of the transcription factor Pax5 to the lymphoid lineage. Furthermore, this distinct B cell phenotype was detected in brain tissues from mice or patients with traumatic brain injury, Alzheimer's disease, and glioblastoma. Overall, these results provide a new perspective on the phagocytic capability and chemotactic function of B cells in the ischemic brain. These cells may serve as an immunotherapeutic target for regulating the immune response of ischemic stroke.
Collapse
Affiliation(s)
- Rui Wang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Huaming Li
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Chenhan Ling
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Xiaotao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jianan Lu
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Weimin Luan
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Brain Research Institute, Zhejiang University; Stroke Research Center for Diagnostic and Therapeutic Technologies of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Ligen Shi
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, Zhejiang Province, China
| |
Collapse
|
20
|
Roselli D, Bonifacio MA, Barbuti G, Rossiello MR, Ranieri P, Mariggiò MA. Anti-Phosphatidylserine, Anti-Prothrombin, and Anti-Annexin V Autoantibodies in Antiphospholipid Syndrome: A Real-Life Study. Diagnostics (Basel) 2023; 13:2507. [PMID: 37568869 PMCID: PMC10416833 DOI: 10.3390/diagnostics13152507] [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: 06/23/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The antiphospholipid antibodies (aPL) increase the risk of developing thrombotic events and may coexist with a variety of autoimmune diseases. They can be detected chronically or temporarily in patients with infectious diseases, during drug therapy, or in cases of cancer. A thrombotic event with aPL detection is known as antiphospholipid syndrome (APS) and the diagnostic criteria include the presence of lupus anticoagulant (LA), anticardiolipin (aCL) and β2-glycoprotein-1(aβ2GPI) antibodies. Other autoantigens recognized in APS are phosphatidylserine (aPS), prothrombin (aPT) and Annexin-5 (aA5). This real life study aimed to explore the connections between laboratory criteria and the prevalence of "non-criteria aPL" in APS. This study followed 300 patients with thrombosis and employed two phospholipid sensitivity assays for LA detection, chemiluminescence assays for aCL and aβ2GPI and enzyme-linked immunoassays for aPS, aPT and aA5. A significant association was found between aPS and aCL (r = 0.76) as well as aβ2GPI (r = 0.77), while the association with LA was less significant (r = 0.33). The results of the aPT and aA5 test did not correlate with criteria-antiphospholipid antibodies (r < 0.30). Since the risk of thrombotic complications increases with the intensity and the number of positive autoantibodies, measuring aPT and aA5 autoantibodies may be useful, particularly in aCL/aβ2GPI-negative patients or in cases of isolated LA positivity.
Collapse
Affiliation(s)
- Daniele Roselli
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro Medical School, 70124 Bari, Italy; (D.R.); (M.A.B.); (G.B.); (M.R.R.)
| | - Maria Addolorata Bonifacio
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro Medical School, 70124 Bari, Italy; (D.R.); (M.A.B.); (G.B.); (M.R.R.)
| | - Giovanna Barbuti
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro Medical School, 70124 Bari, Italy; (D.R.); (M.A.B.); (G.B.); (M.R.R.)
| | - Maria Rosaria Rossiello
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro Medical School, 70124 Bari, Italy; (D.R.); (M.A.B.); (G.B.); (M.R.R.)
| | - Prudenza Ranieri
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Experimental and Clinical Pathology, Azienda Ospedaliero-Universitaria Consorziale Policlinico di Bari, 70124 Bari, Italy
| | - Maria Addolorata Mariggiò
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro Medical School, 70124 Bari, Italy; (D.R.); (M.A.B.); (G.B.); (M.R.R.)
| |
Collapse
|
21
|
Whitelaw BS, Stoessel MB, Majewska AK. Movers and shakers: Microglial dynamics and modulation of neural networks. Glia 2023; 71:1575-1591. [PMID: 36533844 PMCID: PMC10729610 DOI: 10.1002/glia.24323] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Microglia are multifaceted cells that act as immune sentinels, with important roles in pathological events, but also as integral contributors to the normal development and function of neural circuits. In the last decade, our understanding of the contributions these cells make to synaptic health and dysfunction has expanded at a dizzying pace. Here we review the known mechanisms that govern the dynamics of microglia allowing these motile cells to interact with synapses, and recruit microglia to specific sites on neurons. We then review the molecular signals that may underlie the function of microglia in synaptic remodeling. The emerging picture from the literature suggests that microglia are highly sensitive cells, reacting to neuronal signals with dynamic and specific actions tuned to the need of specific synapses and networks.
Collapse
Affiliation(s)
- Brendan Steven Whitelaw
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, New York, USA
| | - Mark Blohm Stoessel
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, New York, USA
| | - Ania Katarzyna Majewska
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, New York, USA
| |
Collapse
|
22
|
Harden S, Tan TY, Ku CW, Zhou J, Chen Q, Chan JKY, Brosens J, Lee YH. Peritoneal autoantibody profiling identifies p53 as an autoantibody target in endometriosis. Fertil Steril 2023; 120:176-187. [PMID: 36828054 DOI: 10.1016/j.fertnstert.2023.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023]
Abstract
OBJECTIVE To map the peritoneal autoantibody (AAb) landscape in women with endometriosis. DESIGN Case-control laboratory study. SETTING Academic medical and research units. PATIENT(S) Women who presented with or without endometriosis. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Using native-conformation and citrullinated modified protein arrays, proteome-wide analysis of AAbs against 1,623 proteins were profiled in peritoneal fluids (PFs) of 25 women with endometriosis and 25 women without endometriosis. RESULT(S) In women with endometriosis, the median number of AAbs detected was 4, including AAbs that targeted autoantigens involved in implantation, B-cell activation/development, and aberrant migration and mitogenicity. Forty-six percent of women with endometriosis have ≥5 peritoneal AAbs. Conversely, in women without endometriosis, the median number of detected AAbs was 1. Autoantibodies recognizing tumor suppressor protein p53 were the most commonly detected AAbs, being present in 35% of women with endometriosis, and p53 AAb was associated with a monocyte/macrophage-like PF cytokine signature. Further investigation of the global reactivity of AAbs against citrullinated PF antigens by peptidylarginine deiminase enzymes 1, 2, and 6 revealed anticitrullinated p53 as the only AAb target elevated and citrullinated by all 3 peptidylarginine deiminase isotypes. Furthermore, unsupervised hierarchical clustering and integrative pathway analysis revealed that 60% of women with endometriosis-associated infertility were positive for AAbs, which are involved in platelet-derived growth factor, transforming growth factor-β, RAC1/PAK1/p38/MMP2 signaling, LAT2/NTAL/LAB-mediated calcium mobilization, and integrin-mediated cell adhesion. CONCLUSION(S) Together, our data identify peritoneal autoimmunity in a significant subset of women with endometriosis, with implications on infertility and disease pathophysiology. In these patients, p53 was identified as the most frequent PF AAb target, which was present in both the native and citrullinated forms.
Collapse
Affiliation(s)
- Sarah Harden
- Critical Analytics for Manufacturing Precision Medicine, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore; Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Tse Yeun Tan
- Department of Reproductive Medicine, KKH, Singapore, Singapore; OBGYN-Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Chee Wai Ku
- Department of Reproductive Medicine, KKH, Singapore, Singapore; OBGYN-Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Jieliang Zhou
- KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KKH, Singapore, Singapore; OBGYN-Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Jan Brosens
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom; Tommy's National Centre for Miscarriage Research, University Hospitals Coventry & Warwickshire, Coventry, United Kingdom; Centre for Early Life, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Yie Hou Lee
- Critical Analytics for Manufacturing Precision Medicine, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore; OBGYN-Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore; KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore.
| |
Collapse
|
23
|
Harwood JL. Polyunsaturated Fatty Acids: Conversion to Lipid Mediators, Roles in Inflammatory Diseases and Dietary Sources. Int J Mol Sci 2023; 24:ijms24108838. [PMID: 37240183 DOI: 10.3390/ijms24108838] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are important components of the diet of mammals. Their role was first established when the essential fatty acids (EFAs) linoleic acid and α-linolenic acid were discovered nearly a century ago. However, most of the biochemical and physiological actions of PUFAs rely on their conversion to 20C or 22C acids and subsequent metabolism to lipid mediators. As a generalisation, lipid mediators formed from n-6 PUFAs are pro-inflammatory while those from n-3 PUFAs are anti-inflammatory or neutral. Apart from the actions of the classic eicosanoids or docosanoids, many newly discovered compounds are described as Specialised Pro-resolving Mediators (SPMs) which have been proposed to have a role in resolving inflammatory conditions such as infections and preventing them from becoming chronic. In addition, a large group of molecules, termed isoprostanes, can be generated by free radical reactions and these too have powerful properties towards inflammation. The ultimate source of n-3 and n-6 PUFAs are photosynthetic organisms which contain Δ-12 and Δ-15 desaturases, which are almost exclusively absent from animals. Moreover, the EFAs consumed from plant food are in competition with each other for conversion to lipid mediators. Thus, the relative amounts of n-3 and n-6 PUFAs in the diet are important. Furthermore, the conversion of the EFAs to 20C and 22C PUFAs in mammals is rather poor. Thus, there has been much interest recently in the use of algae, many of which make substantial quantities of long-chain PUFAs or in manipulating oil crops to make such acids. This is especially important because fish oils, which are their main source in human diets, are becoming limited. In this review, the metabolic conversion of PUFAs into different lipid mediators is described. Then, the biological roles and molecular mechanisms of such mediators in inflammatory diseases are outlined. Finally, natural sources of PUFAs (including 20 or 22 carbon compounds) are detailed, as well as recent efforts to increase their production.
Collapse
Affiliation(s)
- John L Harwood
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, Wales, UK
| |
Collapse
|
24
|
Jian Y, Zhou X, Shan W, Chen C, Ge W, Cui J, Yi W, Sun Y. Crosstalk between macrophages and cardiac cells after myocardial infarction. Cell Commun Signal 2023; 21:109. [PMID: 37170235 PMCID: PMC10173491 DOI: 10.1186/s12964-023-01105-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 03/18/2023] [Indexed: 05/13/2023] Open
Abstract
Cardiovascular diseases, such as myocardial infarction (MI), are a leading cause of death worldwide. Acute MI (AMI) inflicts massive injury to the coronary microcirculation, causing large-scale cardiomyocyte death due to ischemia and hypoxia. Inflammatory cells such as monocytes and macrophages migrate to the damaged area to clear away dead cells post-MI. Macrophages are pleiotropic cells of the innate immune system, which play an essential role in the initial inflammatory response that occurs following MI, inducing subsequent damage and facilitating recovery. Besides their recognized role within the immune response, macrophages participate in crosstalk with other cells (including cardiomyocytes, fibroblasts, immune cells, and vascular endothelial cells) to coordinate post-MI processes within cardiac tissue. Macrophage-secreted exosomes have recently attracted increasing attention, which has led to a more elaborate understanding of macrophage function. Currently, the functional roles of macrophages in the microenvironment of the infarcted heart, particularly with regard to their interaction with surrounding cells, remain unclear. Understanding the specific mechanisms that mediate this crosstalk is essential in treating MI. In this review, we discuss the origin of macrophages, changes in their distribution post-MI, phenotypic and functional plasticity, as well as the specific signaling pathways involved, with a focus on the crosstalk with other cells in the heart. Thus, we provide a new perspective on the treatment of MI. Further in-depth research is required to elucidate the mechanisms underlying crosstalk between macrophages and other cells within cardiac tissue for the identification of potential therapeutic targets. Video Abstract.
Collapse
Affiliation(s)
- Yuhong Jian
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiao Zhou
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenju Shan
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Cheng Chen
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wei Ge
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jun Cui
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Yang Sun
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
25
|
Li J, Chen Q, Zhang R, Liu Z, Cheng Y. The phagocytic role of macrophage following myocardial infarction. Heart Fail Rev 2023:10.1007/s10741-023-10314-5. [PMID: 37160618 DOI: 10.1007/s10741-023-10314-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/12/2023] [Indexed: 05/11/2023]
Abstract
Myocardial infarction (MI) is one of the cardiovascular diseases with high morbidity and mortality. MI causes large amounts of apoptotic and necrotic cells that need to be efficiently and instantly engulfed by macrophage to avoid second necrosis. Phagocytic macrophages can dampen or resolve inflammation to protect infarcted heart. Phagocytosis of macrophages is modulated by various factors including proteins, receptors, lncRNA and cytokines. A better understanding of mechanisms in phagocytosis will be beneficial to regulate macrophage phagocytosis capability towards a desired direction in cardioprotection after MI. In this review, we describe the phagocytosis effect of macrophages and summarize the latest reported signals regulating phagocytosis after MI, which will provide a new thinking about phagocytosis-dependent cardiac protection after MI.
Collapse
Affiliation(s)
- Jiahua Li
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
- Guangdong-Hong Kong-Macau Joint Lab On Chinese Medicine and Immune Disease Research, Guangzhou Univ Chinese Med, Guangzhou, Guangdong, 510006, China
| | - Qi Chen
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Rong Zhang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
- Guangdong-Hong Kong-Macau Joint Lab On Chinese Medicine and Immune Disease Research, Guangzhou Univ Chinese Med, Guangzhou, Guangdong, 510006, China
| | - Zhongqiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
- Guangdong-Hong Kong-Macau Joint Lab On Chinese Medicine and Immune Disease Research, Guangzhou Univ Chinese Med, Guangzhou, Guangdong, 510006, China.
| | - Yuanyuan Cheng
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
- Guangdong-Hong Kong-Macau Joint Lab On Chinese Medicine and Immune Disease Research, Guangzhou Univ Chinese Med, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
26
|
McElhinney K, Irnaten M, O’Brien C. p53 and Myofibroblast Apoptosis in Organ Fibrosis. Int J Mol Sci 2023; 24:ijms24076737. [PMID: 37047710 PMCID: PMC10095465 DOI: 10.3390/ijms24076737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/07/2023] Open
Abstract
Organ fibrosis represents a dysregulated, maladaptive wound repair response that results in progressive disruption of normal tissue architecture leading to detrimental deterioration in physiological function, and significant morbidity/mortality. Fibrosis is thought to contribute to nearly 50% of all deaths in the Western world with current treatment modalities effective in slowing disease progression but not effective in restoring organ function or reversing fibrotic changes. When physiological wound repair is complete, myofibroblasts are programmed to undergo cell death and self-clearance, however, in fibrosis there is a characteristic absence of myofibroblast apoptosis. It has been shown that in fibrosis, myofibroblasts adopt an apoptotic-resistant, highly proliferative phenotype leading to persistent myofibroblast activation and perpetuation of the fibrotic disease process. Recently, this pathological adaptation has been linked to dysregulated expression of tumour suppressor gene p53. In this review, we discuss p53 dysregulation and apoptotic failure in myofibroblasts and demonstrate its consistent link to fibrotic disease development in all types of organ fibrosis. An enhanced understanding of the role of p53 dysregulation and myofibroblast apoptosis may aid in future novel therapeutic and/or diagnostic strategies in organ fibrosis.
Collapse
Affiliation(s)
- Kealan McElhinney
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| | - Mustapha Irnaten
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| | - Colm O’Brien
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| |
Collapse
|
27
|
Ardavín C, Alvarez‐Ladrón N, Ferriz M, Gutiérrez‐González A, Vega‐Pérez A. Mouse Tissue-Resident Peritoneal Macrophages in Homeostasis, Repair, Infection, and Tumor Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206617. [PMID: 36658699 PMCID: PMC10104642 DOI: 10.1002/advs.202206617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/21/2022] [Indexed: 06/17/2023]
Abstract
Large peritoneal macrophages (LPMs) are long-lived, tissue-resident macrophages, formed during embryonic life, developmentally and functionally confined to the peritoneal cavity. LPMs provide the first line of defense against life-threatening pathologies of the peritoneal cavity, such as abdominal sepsis, peritoneal metastatic tumor growth, or peritoneal injuries caused by trauma, or abdominal surgery. Apart from their primary phagocytic function, reminiscent of primitive defense mechanisms sustained by coelomocytes in the coelomic cavity of invertebrates, LPMs fulfill an essential homeostatic function by achieving an efficient clearance of apoptotic, that is crucial for the maintenance of self-tolerance. Research performed over the last few years, in mice, has unveiled the mechanisms by which LPMs fulfill a crucial role in repairing peritoneal injuries and controlling microbial and parasitic infections, reflecting that the GATA6-driven LPM transcriptional program can be modulated by extracellular signals associated with pathological conditions. In contrast, recent experimental evidence supports that peritoneal tumors can subvert LPM metabolism and function, leading to the acquisition of a tumor-promoting potential. The remarkable functional plasticity of LPMs can be nevertheless exploited to revert tumor-induced LPM protumor potential, providing the basis for the development of novel immunotherapeutic approaches against peritoneal tumor metastasis based on macrophage reprogramming.
Collapse
Affiliation(s)
- Carlos Ardavín
- Departamento de Inmunología y OncologíaCentro Nacional de Biotecnología/CSICDarwin 3Madrid28049Spain
| | - Natalia Alvarez‐Ladrón
- Departamento de Inmunología y OncologíaCentro Nacional de Biotecnología/CSICDarwin 3Madrid28049Spain
| | - Margarita Ferriz
- Departamento de Inmunología y OncologíaCentro Nacional de Biotecnología/CSICDarwin 3Madrid28049Spain
| | | | - Adrián Vega‐Pérez
- Departamento de Inmunología y OncologíaCentro Nacional de Biotecnología/CSICDarwin 3Madrid28049Spain
- Present address:
Sandra and Edward Meyer Cancer CenterWeill Cornell Medicine1300 York AvenueNew YorkNY10065USA
| |
Collapse
|
28
|
García-Rodríguez FJ, Buchrieser C, Escoll P. Legionella and mitochondria, an intriguing relationship. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 374:37-81. [PMID: 36858656 DOI: 10.1016/bs.ircmb.2022.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Legionella pneumophila is the causative agent of Legionnaires' disease, a severe pneumonia. L. pneumophila injects via a type-IV-secretion-system (T4SS) more than 300 bacterial proteins into macrophages, its main host cell in humans. Certain of these bacterial effectors target organelles in the infected cell and hijack multiple processes to facilitate all steps of the intracellular life cycle of this pathogen. In this review, we discuss the interplay between L. pneumophila, an intracellular bacterium fully armed with virulence tools, and mitochondria, the extraordinary eukaryotic organelles playing prominent roles in cellular bioenergetics, cell-autonomous immunity and cell death. We present and discuss key findings concerning the multiple interactions of L. pneumophila with mitochondria during infection and the mechanisms employed by T4SS effectors that target mitochondrial functions to subvert infected cells.
Collapse
Affiliation(s)
| | - Carmen Buchrieser
- Institut Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires and CNRS UMR 6047, Paris, France.
| | - Pedro Escoll
- Institut Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires and CNRS UMR 6047, Paris, France.
| |
Collapse
|
29
|
Zhou S, Li Y, Zhang Z, Yuan Y. An insight into the TAM system in Alzheimer's disease. Int Immunopharmacol 2023; 116:109791. [PMID: 36738678 DOI: 10.1016/j.intimp.2023.109791] [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: 12/02/2022] [Revised: 01/13/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
The TAM receptors may help delay the progression of Alzheimer's disease (AD). AD is the most common neurodegenerative disease associated with human aging. The TAM receptors, derived from the first letter of its three constituents -Tyro3, Axl, and Mertk, are associated with immune responses, cellular differentiation and migration, and clearance of apoptotic cells and debris, with the two canonical ligands, Growth Arrest Specific 6 (Gas6) and ProS1. Several kinds of research have indicated the participation of the TAM system in AD pathology. Also, the TAMs regulate multiple features of microglia, the significant sensors of disorder in the central nervous system (CNS). In this review, we describe the biology of the TAM receptors and ligands in the CNS. Then, we discuss the relationship between the TAM system and AD, specially focusing on its functional expression in the microglia. Finally, we also summarize some agents that could interfere with the TAM signaling pathways and discuss potential difficulties and strategies for drug development.
Collapse
Affiliation(s)
- Shiqi Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yanyan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yuhe Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
30
|
Akama-Garren EH, Miller P, Carroll TM, Tellier M, Sutendra G, Buti L, Zaborowska J, Goldin RD, Slee E, Szele FG, Murphy S, Lu X. Regulation of immunological tolerance by the p53-inhibitor iASPP. Cell Death Dis 2023; 14:84. [PMID: 36746936 PMCID: PMC9902554 DOI: 10.1038/s41419-023-05567-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/23/2022] [Accepted: 01/06/2023] [Indexed: 02/08/2023]
Abstract
Maintenance of immunological homeostasis between tolerance and autoimmunity is essential for the prevention of human diseases ranging from autoimmune disease to cancer. Accumulating evidence suggests that p53 can mitigate phagocytosis-induced adjuvanticity thereby promoting immunological tolerance following programmed cell death. Here we identify Inhibitor of Apoptosis Stimulating p53 Protein (iASPP), a negative regulator of p53 transcriptional activity, as a regulator of immunological tolerance. iASPP-deficiency promoted lung adenocarcinoma and pancreatic cancer tumorigenesis, while iASPP-deficient mice were less susceptible to autoimmune disease. Immune responses to iASPP-deficient tumors exhibited hallmarks of immunosuppression, including activated regulatory T cells and exhausted CD8+ T cells. Interestingly, iASPP-deficient tumor cells and tumor-infiltrating myeloid cells, CD4+, and γδ T cells expressed elevated levels of PD-1H, a recently identified transcriptional target of p53 that promotes tolerogenic phagocytosis. Identification of an iASPP/p53 axis of immune homeostasis provides a therapeutic opportunity for both autoimmune disease and cancer.
Collapse
Affiliation(s)
- Elliot H Akama-Garren
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, UK.
- Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Paul Miller
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, UK
| | - Thomas M Carroll
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, UK
| | - Michael Tellier
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Gopinath Sutendra
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, UK
- Department of Medicine, University of Alberta, Edmonton, AB, T6G 2B7, Canada
| | - Ludovico Buti
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, UK
- Charles River Laboratories, Leiden, Netherlands
| | - Justyna Zaborowska
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Robert D Goldin
- Centre for Pathology, St. Mary's Hospital, Imperial College, London, W2 1NY, UK
| | - Elizabeth Slee
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, UK
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Shona Murphy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Xin Lu
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
31
|
Phosphatidylserine in the Nervous System: Cytoplasmic Regulator of the AKT and PKC Signaling Pathways and Extracellular "Eat-Me" Signal in Microglial Phagocytosis. Mol Neurobiol 2023; 60:1050-1066. [PMID: 36401705 DOI: 10.1007/s12035-022-03133-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/08/2022] [Indexed: 11/21/2022]
Abstract
Phosphatidylserine (PtdSer) is an important anionic phospholipid found in eukaryotic cells and has been proven to serve as a beneficial factor in the treatment of neurodegenerative diseases. PtdSer resides in the inner leaflet of the plasma membrane, where it is involved in regulating the AKT and PKC signaling pathways; however, it becomes exposed to the extracellular leaflet during neurodevelopmental processes and neurodegenerative diseases, participating in microglia-mediated synaptic and neuronal phagocytosis. In this paper, we review several characteristics of PtdSer, including the synthesis and translocation of PtdSer, the functions of cytoplasmic and exposed PtdSer, and different PtdSer-detection materials used to further understand the role of PtdSer in the nervous system.
Collapse
|
32
|
Häcker G, Haimovici A. Sub-lethal signals in the mitochondrial apoptosis apparatus: pernicious by-product or physiological event? Cell Death Differ 2023; 30:250-257. [PMID: 36131076 PMCID: PMC9490730 DOI: 10.1038/s41418-022-01058-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/09/2022] [Accepted: 08/18/2022] [Indexed: 11/09/2022] Open
Abstract
One of the tasks of mitochondria is the rule over life and death: when the outer membrane is permeabilized, the release of intermembrane space proteins causes cell death by apoptosis. For a long time, this mitochondrial outer membrane permeabilization (MOMP) has been accepted as the famous step from which no cell returns. Recent results have however shown that this quite plainly does not have to be the case. A cell can also undergo only a little MOMP, and it can efficiently repair damage it has incurred in the process. There is no doubt now that such low-scale permeabilization occurs. A major unclarified issue is the biological relevance. Is small-scale mitochondrial permeabilization an accident, a leakiness of the apoptosis apparatus, perhaps during restructuring of the mitochondrial network? Is it attempted suicide, where cell death by apoptosis is the real goal but the stimulus failed to reach the threshold? Or, more boldly, is there a true biological meaning behind the event of the release of low amounts of mitochondrial components? We will here explore this last possibility, which we believe is on one hand appealing, on the other hand plausible and supported by some evidence. Recent data are consistent with the view that sub-lethal signals in the mitochondrial apoptosis pathway can drive inflammation, the first step of an immune reaction. The apoptosis apparatus is almost notoriously easy to trigger. Sub-lethal signals may be even easier to set off. We suggest that the apoptosis apparatus is used in this way to sound the call when the first human cell is infected by a pathogen.
Collapse
Affiliation(s)
- Georg Häcker
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany.
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| | - Aladin Haimovici
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| |
Collapse
|
33
|
The Protective Role of Interleukin 17A in Acinetobacter baumannii Pneumonia Is Associated with Candida albicans in the Airway. Infect Immun 2023; 91:e0037822. [PMID: 36602381 PMCID: PMC9872622 DOI: 10.1128/iai.00378-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Recent studies have found that the coexistence of fungi and bacteria in the airway may increase the risk of infection, contribute to the development of pneumonia, and increase the severity of disease. Interleukin 17A (IL-17A) plays important roles in host resistance to bacterial and fungal infections. The objective of this study was to determine the effects of IL-17A on Acinetobacter baumannii-infected rats with a previous Candida albicans airway inoculation. The incidence of A. baumannii pneumonia was higher in rats with C. albicans in the airway than in noninoculated rats, and it decreased when amphotericin B was used to clear C. albicans, which influenced IL-17A levels. IL-17A had a protective effect in A. baumannii pneumonia associated with C. albicans in the airway. Compared with A. baumannii-infected rats with C. albicans in the airway that did not receive IL-17A, recombinant IL-17A (rIL-17A) supplementation decreased the incidence of A. baumannii pneumonia (10/15 versus 5/17; P = 0.013) and the proportion of neutrophils in the lung (84 ± 3.5 versus 74 ± 4.3%; P = 0.033), reduced tissue destruction and inflammation, and decreased levels of myeloperoxidase (MPO) (1.267 ± 0.15 versus 0.233 ± 0.06 U/g; P = 0.0004), reactive oxygen species (ROS) (132,333 ± 7,505 versus 64,667 ± 10,115 AU; P = 0.0007) and lactate dehydrogenase (LDH) (2.736 ± 0.05 versus 2.1816 ± 0.29 U/g; P = 0.0313). In vitro experiments revealed that IL-17A had no significant effect on the direct migration ability and bactericidal capability of neutrophils. However, IL-17A restrained lysis cell death and increased apoptosis of neutrophils (2.9 ± 1.14 versus 7 ± 0.5%; P = 0.0048). Taken together, our results suggest that C. albicans can depress IL-17A levels, which when supplemented may have a regulatory function that limits the accumulation of neutrophils in inflammatory areas, providing inflammatory response homeostasis.
Collapse
|
34
|
Li T, Yan Z, Fan Y, Fan X, Li A, Qi Z, Zhang J. Cardiac repair after myocardial infarction: A two-sided role of inflammation-mediated. Front Cardiovasc Med 2023; 9:1077290. [PMID: 36698953 PMCID: PMC9868426 DOI: 10.3389/fcvm.2022.1077290] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Myocardial infarction is the leading cause of death and disability worldwide, and the development of new treatments can help reduce the size of myocardial infarction and prevent adverse cardiovascular events. Cardiac repair after myocardial infarction can effectively remove necrotic tissue, induce neovascularization, and ultimately replace granulation tissue. Cardiac inflammation is the primary determinant of whether beneficial cardiac repair occurs after myocardial infarction. Immune cells mediate inflammatory responses and play a dual role in injury and protection during cardiac repair. After myocardial infarction, genetic ablation or blocking of anti-inflammatory pathways is often harmful. However, enhancing endogenous anti-inflammatory pathways or blocking endogenous pro-inflammatory pathways may improve cardiac repair after myocardial infarction. A deficiency of neutrophils or monocytes does not improve overall cardiac function after myocardial infarction but worsens it and aggravates cardiac fibrosis. Several factors are critical in regulating inflammatory genes and immune cells' phenotypes, including DNA methylation, histone modifications, and non-coding RNAs. Therefore, strict control and timely suppression of the inflammatory response, finding a balance between inflammatory cells, preventing excessive tissue degradation, and avoiding infarct expansion can effectively reduce the occurrence of adverse cardiovascular events after myocardial infarction. This article reviews the involvement of neutrophils, monocytes, macrophages, and regulatory T cells in cardiac repair after myocardial infarction. After myocardial infarction, neutrophils are the first to be recruited to the damaged site to engulf necrotic cell debris and secrete chemokines that enhance monocyte recruitment. Monocytes then infiltrate the infarct site and differentiate into macrophages and they release proteases and cytokines that are harmful to surviving myocardial cells in the pre-infarct period. As time progresses, apoptotic neutrophils are cleared, the recruitment of anti-inflammatory monocyte subsets, the polarization of macrophages toward the repair phenotype, and infiltration of regulatory T cells, which secrete anti-inflammatory factors that stimulate angiogenesis and granulation tissue formation for cardiac repair. We also explored how epigenetic modifications regulate the phenotype of inflammatory genes and immune cells to promote cardiac repair after myocardial infarction. This paper also elucidates the roles of alarmin S100A8/A9, secreted frizzled-related protein 1, and podoplanin in the inflammatory response and cardiac repair after myocardial infarction.
Collapse
Affiliation(s)
- Tingting Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhipeng Yan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yajie Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinbiao Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Aolin Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhongwen Qi
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Zhongwen Qi,
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China,Junping Zhang,
| |
Collapse
|
35
|
Wood SJ, Goldufsky JW, Seu MY, Dorafshar AH, Shafikhani SH. Pseudomonas aeruginosa Cytotoxins: Mechanisms of Cytotoxicity and Impact on Inflammatory Responses. Cells 2023; 12:cells12010195. [PMID: 36611990 PMCID: PMC9818787 DOI: 10.3390/cells12010195] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 01/05/2023] Open
Abstract
Pseudomonas aeruginosa is one of the most virulent opportunistic Gram-negative bacterial pathogens in humans. It causes many acute and chronic infections with morbidity and mortality rates as high as 40%. P. aeruginosa owes its pathogenic versatility to a large arsenal of cell-associated and secreted virulence factors which enable this pathogen to colonize various niches within hosts and protect it from host innate immune defenses. Induction of cytotoxicity in target host cells is a major virulence strategy for P. aeruginosa during the course of infection. P. aeruginosa has invested heavily in this strategy, as manifested by a plethora of cytotoxins that can induce various forms of cell death in target host cells. In this review, we provide an in-depth review of P. aeruginosa cytotoxins based on their mechanisms of cytotoxicity and the possible consequences of their cytotoxicity on host immune responses.
Collapse
Affiliation(s)
- Stephen J. Wood
- Department of Medicine, Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Josef W. Goldufsky
- Department of Medicine, Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Michelle Y. Seu
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Amir H. Dorafshar
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sasha H. Shafikhani
- Department of Medicine, Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
- Cancer Center, Rush University Medical Center, Chicago, IL 60612, USA
- Correspondence:
| |
Collapse
|
36
|
Yang Z, Liu R, Qiu M, Mei H, Hao J, Song T, Zhao K, Zou D, Wang H, Gao M. The roles of ERIANIN in tumor and innate immunity and its' perspectives in immunotherapy. Front Immunol 2023; 14:1170754. [PMID: 37187758 PMCID: PMC10175588 DOI: 10.3389/fimmu.2023.1170754] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Traditional Chinese medicine has been used in China for thousands of years. In 2022, the 14th Five-Year Plan for the Development of Traditional Chinese Medicine was released, aiming to enhance traditional Chinese medicine health services and improve policies and systems for high-quality traditional Chinese medicinal development by 2025. ERIANIN, the main component of the traditional Chinese medicine Dendrobium, plays an important role in anti-inflammatory, antiviral, antitumor, antiangiogenic, and other pharmacological effects. ERIANIN has broad-spectrum antitumor effects, and its tumor-suppressive effects have been confirmed in the study of various diseases, such as precancerous lesions of the stomach, gastric cancer, liver cancer, lung cancer, prostate cancer, bladder cancer, breast cancer, cervical cancer, osteosarcoma, colorectal cancer, leukaemia, nasopharyngeal cancer and melanoma through the multiple signaling pathways. Thus, the aim of this review was to systematically summarise the research on ERIANIN with the aim of serving as a reference for future research on this compound and briefly discuss some future perspectives development of ERIANIN in combined immunotherapy.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Oncology, Tianjin Union Medical Center of Nankai University, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China
| | - Ruxue Liu
- College of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Minghan Qiu
- Department of Oncology, Tianjin Union Medical Center of Nankai University, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China
| | - Hanwei Mei
- College of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jie Hao
- Department of Thyroid and Breast Surgery, Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, China
| | - Teng Song
- Department of Oncology, Tianjin Union Medical Center of Nankai University, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China
| | - Ke Zhao
- Department of Oncology, Tianjin Union Medical Center of Nankai University, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China
| | - Dandan Zou
- Department of Oncology, Tianjin Union Medical Center of Nankai University, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China
| | - Huaqing Wang
- Department of Oncology, Tianjin Union Medical Center of Nankai University, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China
- College of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Huaqing Wang, ; Ming Gao,
| | - Ming Gao
- Department of Thyroid and Breast Surgery, Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, China
- *Correspondence: Huaqing Wang, ; Ming Gao,
| |
Collapse
|
37
|
Naylor G, Julian L, Watson-Bryce S, Mullin M, Nibbs RJ, Olson MF. Immunogenic Death of Hepatocellular Carcinoma Cells in Mice Expressing Caspase-Resistant ROCK1 Is Not Replicated by ROCK Inhibitors. Cancers (Basel) 2022; 14:cancers14235943. [PMID: 36497425 PMCID: PMC9740421 DOI: 10.3390/cancers14235943] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022] Open
Abstract
The morphological changes during apoptosis help facilitate "immunologically silent" cell death. Caspase cleavage of the ROCK1 kinase results in its activation, which drives the forceful contraction of apoptotic cells. We previously showed that when ROCK1 was mutated to render it caspase-resistant, there was greater liver damage and neutrophil recruitment after treatment with the hepatotoxin diethylnitrosamine (DEN). We now show that acute DEN-induced liver damage induced higher levels of pro-inflammatory cytokines/chemokines, indicative of immunogenic cell death (ICD), in mice expressing non-cleavable ROCK1 (ROCK1nc). Hepatocellular carcinoma (HCC) tumours in ROCK1nc mice had more neutrophils and CD8+ T cells relative to mice expressing wild-type ROCK1, indicating that spontaneous tumour cell death also was more immunogenic. Since ICD induction has been proposed to be tumour-suppressive, the effects of two distinct ROCK inhibitors on HCC tumours was examined. Both fasudil and AT13148 significantly decreased tumour numbers, areas and volumes, but neither resulted in greater numbers of neutrophils or CD8+ T cells to be recruited. In the context of acute DEN-induced liver damage, AT13148 inhibited the recruitment of dendritic, natural killer and CD8+ T cells to livers. These observations indicate that there is an important role for ROCK1 cleavage to limit immunogenic cell death, which was not replicated by systemic ROCK inhibitor administration. As a result, concomitant administration of ROCK inhibitors with cancer therapeutics would be unlikely to result in therapeutic benefit by inducing ICD to increase anti-tumour immune responses.
Collapse
Affiliation(s)
- Gregory Naylor
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Linda Julian
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Steven Watson-Bryce
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Margaret Mullin
- Electron Microscopy Facility, School of Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Robert J. Nibbs
- Institute of Infection, Immunity and Inflammation, School of Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Michael F. Olson
- Department of Chemistry and Biology, Toronto Metropolitan University, 661 University Avenue Suite 1105, Toronto, ON M5G 1M1, Canada
- Correspondence:
| |
Collapse
|
38
|
Lang C, Roy S, Wang Y, Graves D, Xu Y, Serezani CH, Korrer M, Kim YJ. Efferocytosis drives myeloid NLRP3 dependent inflammasome signaling secretion of IL-1β to promote tumor growth. Front Immunol 2022; 13:993771. [PMID: 36439171 PMCID: PMC9681818 DOI: 10.3389/fimmu.2022.993771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/19/2022] [Indexed: 11/10/2022] Open
Abstract
Caspase-1 signaling in myeloid suppressor cells can promote T-cell independent cancer progression, but the regulation of inflammasome signaling within the highly heterogeneous myeloid population in the tumor milieu remains elusive. To resolve this complexity, single cell transcriptomic profile of Head and Neck Squamous Cell Carcinoma (HNSCC) identified distinct inflammasome-associated genes within specific clusters of tumor-infiltrating myeloid cells. Among these myeloid cells, the sensor protein, NLRP3, and downstream effector IL-1β transcripts were enriched in discreet monocytic and macrophage subtypes in the TME. We showed that deletion of NLRP3, but not AIM2, phenocopied caspase-1/IL-1β dependent tumor progression in vivo. Paradoxically, we found myeloid-intrinsic caspase-1 signaling increased myeloid survival contrary to what would be predicted from the canonical pyroptotic function of caspase-1. This myeloid NLRP3/IL-1β signaling axis promotion of tumor growth was found to be gasdermin D independent. Mechanistically, we found that phagocyte-mediated efferocytosis of dying tumor cells in the TME directly activated NLRP3-dependent inflammasome signaling to drive IL-1β secretion. Subsequently we showed that NLRP3-mediated IL-1β production drives tumor growth in vivo. Dynamic RNA velocity analysis showed a robust directional flow from efferocytosis gene-set high macrophages to an inflammasome gene-set high macrophage population. We provide a novel efferocytosis-dependent inflammasome signaling pathway which mediates homeostatic tumor cell apoptosis that characterizes chronic inflammation-induced malignancy.
Collapse
Affiliation(s)
- Cara Lang
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, TN, United States
| | - Sohini Roy
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Yu Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, United States
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Diana Graves
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, TN, United States
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, United States
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, United States
| | - C. Henrique Serezani
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Michael Korrer
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Young J. Kim
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, United States
- *Correspondence: Young J. Kim,
| |
Collapse
|
39
|
Qi S, Wang Q, Zhang J, Liu Q, Li C. Pyroptosis and Its Role in the Modulation of Cancer Progression and Antitumor Immunity. Int J Mol Sci 2022; 23:ijms231810494. [PMID: 36142404 PMCID: PMC9501080 DOI: 10.3390/ijms231810494] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Pyroptosis is a type of programmed cell death (PCD) accompanied by an inflammatory reaction and the rupture of a membrane. Pyroptosis is divided into a canonical pathway triggered by caspase-1, and a non-canonical pathway independent of caspase-1. More and more pyroptosis-related participants, pathways, and regulatory mechanisms have been exploited in recent years. Pyroptosis plays crucial roles in the initiation, progression, and metastasis of cancer and it affects the immunotherapeutic outcome by influencing immune cell infiltration as well. Extensive studies are required to elucidate the molecular mechanisms between pyroptosis and cancer. In this review, we introduce the discovery history of pyroptosis, delineate the signaling pathways of pyroptosis, and then make comparisons between pyroptosis and other types of PCD. Finally, we provide an overview of pyroptosis in different cancer types. With the progression in the field of pyroptosis, new therapeutic targets and strategies can be explored to combat cancer.
Collapse
Affiliation(s)
- Sihan Qi
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Qilin Wang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Junyou Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Qian Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Chunyan Li
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing 100191, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China
- Correspondence:
| |
Collapse
|
40
|
Tada R, Nagao K, Tanaka R, Yamada S, Watanabe A, Negishi Y. Involvement of splenic marginal zone macrophages in the recognition of systemically administered phosphatidylserine-coated liposomes in mice. Int Immunopharmacol 2022; 112:109209. [PMID: 36084540 DOI: 10.1016/j.intimp.2022.109209] [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: 07/05/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 11/05/2022]
Abstract
Autoimmune diseases present a significant clinical problem, highlighting the need for the development of novel or improved therapeutic methods. One of the factors that causes autoimmune diseases is a defect in the clearance of apoptotic cells by phagocytes. Thus, improved apoptotic cell processing has been considered as a strategy to treat autoimmune diseases. However, therapeutic strategies focusing on apoptotic cell clearance have not been approved till date. We have reported that liposomes composed of phosphatidylserine (PS liposomes) exhibit anti-inflammatory or immunosuppressive effects in macrophages. A PS liposome display PS on its surface, which plays a crucial role in the phagocytosis of apoptotic cells by marginal zone macrophages (MZMs), a key player in the clearance of apoptotic cells, by recognizing PS exposed on the surface of apoptotic cells. Therefore, we hypothesized that PS liposomes could be used as "antigen delivery vesicles" to act as a substitute for apoptotic cells in the treatment of autoimmune diseases. In this study, we showed that systemically administered PS liposomes accumulated in the marginal zone of the spleen due to recognition of surface-displayed PS by MZMs because it was observed that liposomes without PS did not accumulate in the marginal zone. In conclusion, PS liposomes may be useful vehicles to function as active agents and/or antigens against autoimmune diseases.
Collapse
Affiliation(s)
- Rui Tada
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, Japan.
| | - Koichiro Nagao
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, Japan
| | - Riki Tanaka
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, Japan
| | - Sumire Yamada
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, Japan
| | - Ayano Watanabe
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, Japan
| | - Yoichi Negishi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, Japan
| |
Collapse
|
41
|
Tsai TL, Zhou TA, Hsieh YT, Wang JC, Cheng HK, Huang CH, Tsai PY, Fan HH, Feng HK, Huang YC, Lin CC, Lin CH, Lin CY, Dzhagalov IL, Hsu CL. Multiomics reveal the central role of pentose phosphate pathway in resident thymic macrophages to cope with efferocytosis-associated stress. Cell Rep 2022; 40:111065. [PMID: 35830797 DOI: 10.1016/j.celrep.2022.111065] [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: 12/23/2021] [Revised: 05/07/2022] [Accepted: 06/16/2022] [Indexed: 11/29/2022] Open
Abstract
Tissue-resident macrophages (TRMs) are heterogeneous cell populations found throughout the body. Depending on their location, they perform diverse functions maintaining tissue homeostasis and providing immune surveillance. To survive and function within, TRMs adapt metabolically to the distinct microenvironments. However, little is known about the metabolic signatures of TRMs. The thymus provides a nurturing milieu for developing thymocytes yet efficiently removes those that fail the selection, relying on the resident thymic macrophages (TMφs). This study harnesses multiomics analyses to characterize TMφs and unveils their metabolic features. We find that the pentose phosphate pathway (PPP) is preferentially activated in TMφs, responding to the reduction-oxidation demands associated with the efferocytosis of dying thymocytes. The blockade of PPP in Mφs leads to decreased efferocytosis, which can be rescued by reactive oxygen species (ROS) scavengers. Our study reveals the key role of the PPP in TMφs and underscores the importance of metabolic adaptation in supporting Mφ efferocytosis.
Collapse
Affiliation(s)
- Tsung-Lin Tsai
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Tyng-An Zhou
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Ting Hsieh
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ju-Chu Wang
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Hui-Kuei Cheng
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chen-Hua Huang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Pei-Yuan Tsai
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Hsiu-Han Fan
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Hsing-Kai Feng
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Chia Huang
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chen-Ching Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chao-Hsiung Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chih-Yu Lin
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 112, Taiwan
| | - Ivan L Dzhagalov
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chia-Lin Hsu
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
42
|
Li H, Li J, Shi H, Li C, Huang W, Zhang M, Luo Y, Song L, Yu R, Zhu J. Structural characterization and immunoregulatory activity of a novel acidic polysaccharide from Scapharca subcrenata. Int J Biol Macromol 2022; 210:439-454. [PMID: 35504419 DOI: 10.1016/j.ijbiomac.2022.04.204] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/01/2022] [Accepted: 04/27/2022] [Indexed: 01/01/2023]
Abstract
A novel acidic polysaccharide named SSPA50-1 was isolated from Scapharca subcrenata using a simulated gastric fluid extraction method. SSPA50-1 is a heteropolysaccharide with an average molecular weight of 44.7 kDa that is composed of galacturonic acid, glucose, galactose, mannose, ribose, rhamnose, fucose, xylose and arabinose at a molar ratio of 1.00:5.40:9.04:3.10:1.59:4.01:2.10:2.21:2.28. The structural characterization based on the methylation and 1D/2D NMR analyses indicated that SSPA50-1 is composed of →3)-β-L-Rhap-(1→, →3)-β-L-2-O-Me-Fucp-(1→, →2)-α-D-Xylp-(1→, →5)-α-L-Araf-(1→, →3)-β-D-Galp-(1→, →6)-α-D-Glcp-(1→, →3,4)-β-D-Manp-(1→, →3,4)-β-D-Galp-(1→, β-D-Ribf-(1→, α-D-Glcp-(1→, and α-D-GalAp6Me-(1→. Furthermore, SSPA50-1 possessed potent immunoregulatory activity by enhancing the phagocytosis and NO, iNOS, TNF-α and IL-6 secretion capacity of RAW264.7 cells. Otherwise, SSPA50-1 significantly promoted the proliferation of splenic lymphocytes and RAW264.7 macrophages. These results indicated that SSPA50-1 could be developed as a potential ingredient for immunostimulatory agents.
Collapse
Affiliation(s)
- Hang Li
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China
| | - Jianhuan Li
- Department of Pharmacology, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Hui Shi
- Department of Natural Product Chemistry, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China; Shandong Academy of Pharmaceutical Sciences, Jinan 250101, China
| | - Chunlei Li
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China
| | - Weijuan Huang
- Department of Pharmacology, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Man Zhang
- Department of Natural Product Chemistry, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Yuanyuan Luo
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China
| | - Liyan Song
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China; Department of Pharmacology, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China.
| | - Rongmin Yu
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China; Department of Natural Product Chemistry, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China; Shandong Academy of Pharmaceutical Sciences, Jinan 250101, China.
| | - Jianhua Zhu
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China; Shandong Academy of Pharmaceutical Sciences, Jinan 250101, China.
| |
Collapse
|
43
|
Shiu FH, Wong JC, Yamamoto T, Lala T, Purcell RH, Owino S, Zhu D, Van Meir EG, Hall RA, Escayg A. Mice lacking full length Adgrb1 (Bai1) exhibit social deficits, increased seizure susceptibility, and altered brain development. Exp Neurol 2022; 351:113994. [PMID: 35114205 PMCID: PMC9817291 DOI: 10.1016/j.expneurol.2022.113994] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/20/2021] [Accepted: 01/24/2022] [Indexed: 01/11/2023]
Abstract
The adhesion G protein-coupled receptor BAI1/ADGRB1 plays an important role in suppressing angiogenesis, mediating phagocytosis, and acting as a brain tumor suppressor. BAI1 is also a critical regulator of dendritic spine and excitatory synapse development and interacts with several autism-relevant proteins. However, little is known about the relationship between altered BAI1 function and clinically relevant phenotypes. Therefore, we studied the effect of reduced expression of full length Bai1 on behavior, seizure susceptibility, and brain morphology in Adgrb1 mutant mice. We compared homozygous (Adgrb1-/-), heterozygous (Adgrb1+/-), and wild-type (WT) littermates using a battery of tests to assess social behavior, anxiety, repetitive behavior, locomotor function, and seizure susceptibility. We found that Adgrb1-/- mice showed significant social behavior deficits and increased vulnerability to seizures. Adgrb1-/- mice also showed delayed growth and reduced brain weight. Furthermore, reduced neuron density and increased apoptosis during brain development were observed in the hippocampus of Adgrb1-/- mice, while levels of astrogliosis and microgliosis were comparable to WT littermates. These results show that reduced levels of full length Bai1 is associated with a broader range of clinically relevant phenotypes than previously reported.
Collapse
Affiliation(s)
- Fu Hung Shiu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA; Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - Jennifer C Wong
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Takahiro Yamamoto
- Department of Neurosurgery, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Trisha Lala
- Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA; Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ryan H Purcell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sharon Owino
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Dan Zhu
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Erwin G Van Meir
- Department of Neurosurgery, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Randy A Hall
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrew Escayg
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
44
|
Jia D, Chen S, Bai P, Luo C, Liu J, Sun A, Ge J. Cardiac Resident Macrophage-derived Legumain Improves Cardiac Repair via Promoting Clearance and Degradation of Apoptotic Cardiomyocytes after Myocardial Infarction. Circulation 2022; 145:1542-1556. [PMID: 35430895 DOI: 10.1161/circulationaha.121.057549] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Cardiac resident macrophages are self-maintaining that originate from embryonic hematopoiesis. After myocardial infarction (MI), cardiac resident macrophages are responsible for the efficient clearance and degradation of apoptotic cardiomyocytes (efferocytosis). This process is required for inflammation resolution and tissue repair; however, the underlying molecular mechanisms remain unknown. Therefore, we aimed to identify the mechanisms of the continued clearance and degradation of phagolysosomal cargo by cardiac resident macrophages during MI. Methods: Multiple transgenic mice such us Lgmn-/-, Lgmn F/F; LysMCre, LgmnF/F; Cx3cr1CreER, LgmnF/F; LyveCre, and cardiac macrophage Lgmn overexpression by adenovirus gene transfer were used to determine the functional significance of Lgmn in MI. Immune cell filtration and inflammation were examined by flow cytometry and quantitative real-time polymerase chain reaction (qPCR). Moreover, Lgmn expression was analyzed by immunohistochemistry and qPCR in the cardiac tissues of patients with ischemic cardiomyopathy and healthy controls. Results: We identified legumain (Lgmn) as a gene specifically expressed by cardiac resident macrophages. Lgmn deficiency resulted in a considerable exacerbation in cardiac function, accompanied with the accumulation of apoptotic cardiomyocytes and a reduced index of in vivo efferocytosis in the border area. It also led to decreased cytosolic calcium due to defective intracellular calcium mobilization. Furthermore, the formation of LC3-II-dependent phagosome around secondary-encountered apoptotic cardiomyocytes was disabled. In addition, Lgmn deficiency increased infiltration of MHC-IIhigh CCR2+ macrophages and the enhanced recruitment of MHC-IIlow CCR2+ monocytes with downregulation of anti-inflammatory mediators, IL-10 and TGF-β; and upregulation proinflammatory mediators, IL-1β, TNF-α, IL-6, and IFN-γ. Conclusions: Our results directly link efferocytosis to wound healing in the heart and identify Lgmn as a significant link between acute inflammation resolution and organ function.
Collapse
Affiliation(s)
- Daile Jia
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Siqin Chen
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Peiyuan Bai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Chentao Luo
- Department of Cardiovascular Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jin Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
45
|
Mechanisms and Consequences of Noncanonical Inflammasome-Mediated Pyroptosis. J Mol Biol 2022; 434:167245. [PMID: 34537239 PMCID: PMC8844060 DOI: 10.1016/j.jmb.2021.167245] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/07/2021] [Accepted: 09/07/2021] [Indexed: 12/13/2022]
Abstract
The noncanonical inflammasome, comprising inflammatory caspases 4, 5, or 11, monitors the cytosol for bacterial lipopolysaccharide (LPS). Intracellular LPS-elicited autoproteolysis of these inflammatory caspases leads to the cleavage of the pore-forming protein gasdermin D (GSDMD). GSDMD pore formation induces a lytic form of cell death known as pyroptosis and the release of inflammatory cytokines and DAMPs, thereby promoting inflammation. The noncanonical inflammasome-dependent innate sensing of cytosolic LPS plays important roles in bacterial infections and sepsis pathogenesis. Exciting studies in the recent past have significantly furthered our understanding of the biochemical and structural basis of the caspase-4/11 activation of GSDMD, caspase-4/11's substrate specificity, and the biological consequences of noncanonical inflammasome activation of GSDMD. This review will discuss these recent advances and highlight the remaining gaps in our understanding of the noncanonical inflammasome and pyroptosis.
Collapse
|
46
|
Bonfim-Melo A, Duszyc K, Gomez GA, Yap AS. Regulating life after death: how mechanical communication mediates the epithelial response to apoptosis. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2022; 45:9. [PMID: 35076820 PMCID: PMC8789724 DOI: 10.1140/epje/s10189-022-00163-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/10/2022] [Indexed: 06/14/2023]
Abstract
It is increasingly evident that cells in tissues and organs can communicate with one another using mechanical forces. Such mechanical signalling can serve as a basis for the assembly of cellular communities. For this to occur, there must be local instabilities in tissue mechanics that are the source of the signals, and mechanisms for changes in mechanical force to be transmitted and detected within tissues. In this review, we discuss these principles using the example of cell death by apoptosis, when it occurs in epithelia. This elicits the phenomenon of apical extrusion, which can rapidly eliminate apoptotic cells by expelling them from the epithelium. Apoptotic extrusion requires that epithelial cells detect the presence of nearby apoptotic cells, something which can be elicited by the mechanotransduction of tensile instabilities caused by the apoptotic cell. We discuss the central role that adherens junctions can play in the transmission and detection of mechanical signals from apoptotic cells.
Collapse
Affiliation(s)
- Alexis Bonfim-Melo
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Kinga Duszyc
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Guillermo A Gomez
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, 5000, Australia
| | - Alpha S Yap
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
47
|
Zhang Y, Wang Y, Ding J, Liu P. Efferocytosis in multisystem diseases (Review). Mol Med Rep 2022; 25:13. [PMID: 34779503 PMCID: PMC8600411 DOI: 10.3892/mmr.2021.12529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/15/2021] [Indexed: 01/22/2023] Open
Abstract
Efferocytosis, the phagocytosis of apoptotic cells performed by both specialized phagocytes (such as macrophages) and non‑specialized phagocytes (such as epithelial cells), is involved in tissue repair and homeostasis. Effective efferocytosis prevents secondary necrosis, terminates inflammatory responses, promotes self‑tolerance and activates pro‑resolving pathways to maintain homeostasis. When efferocytosis is impaired, apoptotic cells that could not be cleared in time aggregate, resulting in the necrosis of apoptotic cells and release of pro‑inflammatory factors. In addition, defective efferocytosis inhibits the intracellular cholesterol reverse transportation pathways, which may lead to atherosclerosis, lung damage, non‑alcoholic fatty liver disease and neurodegenerative diseases. The uncleared apoptotic cells can also release autoantigens, which can cause autoimmune diseases. Cancer cells escape from phagocytosis via efferocytosis. Therefore, new treatment strategies for diseases related to defective efferocytosis are proposed. This review illustrated the mechanisms of efferocytosis in multisystem diseases and organismal homeostasis and the pathophysiological consequences of defective efferocytosis. Several drugs and treatments available to enhance efferocytosis are also mentioned in the review, serving as new evidence for clinical application.
Collapse
Affiliation(s)
- Yifan Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yiru Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Jie Ding
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Ping Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| |
Collapse
|
48
|
Zaidan I, Tavares LP, Sugimoto MA, Lima KM, Negreiros-Lima GL, Teixeira LC, Miranda TC, Valiate BV, Cramer A, Vago JP, Campolina-Silva GH, Souza JA, Grossi LC, Pinho V, Campagnole-Santos MJ, Santos RAS, Teixeira MM, Galvão I, Sousa LP. Angiotensin-(1-7)/MasR axis promotes migration of monocytes/macrophages with a regulatory phenotype to perform phagocytosis and efferocytosis. JCI Insight 2021; 7:147819. [PMID: 34874920 PMCID: PMC8765051 DOI: 10.1172/jci.insight.147819] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
Nonphlogistic migration of macrophages contributes to the clearance of pathogens and apoptotic cells, a critical step for the resolution of inflammation and return to homeostasis. Angiotensin-(1-7) [Ang-(1-7)] is a heptapeptide of the renin-angiotensin system that acts through Mas receptor (MasR). Ang-(1-7) has recently emerged as a novel proresolving mediator, yet Ang-(1-7) resolution mechanisms are not fully determined. Herein, Ang-(1-7) stimulated migration of human and murine monocytes/macrophages in a MasR-, CCR2-, and MEK/ERK1/2–dependent manner. Pleural injection of Ang-(1-7) promoted nonphlogistic mononuclear cell influx alongside increased levels of CCL2, IL-10, and macrophage polarization toward a regulatory phenotype. Ang-(1-7) induction of CCL2 and mononuclear cell migration was also dependent on MasR and MEK/ERK. Of note, MasR was upregulated during the resolution phase of inflammation, and its pharmacological inhibition or genetic deficiency impaired mononuclear cell recruitment during self-resolving models of LPS pleurisy and E. coli peritonitis. Inhibition/absence of MasR was associated with reduced CCL2 levels, impaired phagocytosis of bacteria, efferocytosis, and delayed resolution of inflammation. In summary, we have uncovered a potentially novel proresolving feature of Ang-(1-7), namely the recruitment of mononuclear cells favoring efferocytosis, phagocytosis, and resolution of inflammation. Mechanistically, cell migration was dependent on MasR, CCR2, and the MEK/ERK pathway.
Collapse
Affiliation(s)
- Isabella Zaidan
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana P Tavares
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michelle A Sugimoto
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kátia M Lima
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Graziele L Negreiros-Lima
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lívia Cr Teixeira
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thais C Miranda
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bruno Vs Valiate
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Allysson Cramer
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Priscila Vago
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Jéssica Am Souza
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laís C Grossi
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Robson A S Santos
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabela Galvão
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Department of Clinical and Toxicological Analysis from the School of Pharma, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
49
|
Shi D, Jiang P. A Different Facet of p53 Function: Regulation of Immunity and Inflammation During Tumor Development. Front Cell Dev Biol 2021; 9:762651. [PMID: 34733856 PMCID: PMC8558413 DOI: 10.3389/fcell.2021.762651] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
As a key transcription factor, the evolutionarily conserved tumor suppressor p53 (encoded by TP53) plays a central role in response to various cellular stresses. A variety of biological processes are regulated by p53 such as cell cycle arrest, apoptosis, senescence and metabolism. Besides these well-known roles of p53, accumulating evidence show that p53 also regulates innate immune and adaptive immune responses. p53 influences the innate immune system by secreted factors that modulate macrophage function to suppress tumourigenesis. Dysfunction of p53 in cancer affects the activity and recruitment of T and myeloid cells, resulting in immune evasion. p53 can also activate key regulators in immune signaling pathways which support or impede tumor development. Hence, it seems that the tumor suppressor p53 exerts its tumor suppressive effect to a considerable extent by modulating the immune response. In this review, we concisely discuss the emerging connections between p53 and immune responses, and their impact on tumor progression. Understanding the role of p53 in regulation of immunity will help to developing more effective anti-tumor immunotherapies for patients with TP53 mutation or depletion.
Collapse
Affiliation(s)
- Di Shi
- School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Peng Jiang
- School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
50
|
Zhou S, Shang Q, Ji J, Luan Y. A Nanoplatform to Amplify Apoptosis-to-Pyroptosis Immunotherapy via Immunomodulation of Myeloid-Derived Suppressor Cells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:47407-47417. [PMID: 34597015 DOI: 10.1021/acsami.1c16154] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Pyroptosis is a programmed cell death to enhance immunogenicity of tumor cells, but pyroptosis-based immunotherapy is limited due to the immune escape involving myeloid-derived suppressor cells (MDSCs). Therefore, designing a nanoplatform to not only trigger apoptosis-pyroptosis transformation but also combat the MDSC-based immune escape is of great significance. As a proof-of-concept study, here, we designed a metal organic framework (MOF)-based nanoplatform to tailor the pyroptosis immunotherapy through disrupting the MDSC-mediated immunosuppression. By pH-responsive zeolitic imidazolate framework-8 (ZIF-8) modified with hyaluronic acid (HA), the chemotherapeutic drug mitoxantrone (MIT) and DNA demethylating agent hydralazine (HYD) were successfully co-encapsulated into ZIF-8 for achieving (M+H)@ZIF/HA nanoparticles. This nanoplatform demonstrated a powerful apoptosis-to-pyroptosis transformation with a potent disruption of MDSC-mediated T cell paralysis via reducing immunosuppressive methylglyoxal by HYD. Overall, our two-pronged nanoplatform (M+H)@ZIF/HA can switch the cold tumor into an arsenal of antigens that stimulate robust immunological responses, while suppressing immune escape, collectively triggering vigorous cytotoxic T cell responses with remarkable tumor elimination and building a long-term immune memory response against metastasis.
Collapse
Affiliation(s)
- Shiyao Zhou
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qi Shang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yuxia Luan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|