Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters

Publication year range
1.
Cell ; 185(26): 4887-4903.e17, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36563662

ABSTRACT

Our bodies turn over billions of cells daily via apoptosis and are in turn cleared by phagocytes via the process of "efferocytosis." Defects in efferocytosis are now linked to various inflammatory diseases. Here, we designed a strategy to boost efferocytosis, denoted "chimeric receptor for efferocytosis" (CHEF). We fused a specific signaling domain within the cytoplasmic adapter protein ELMO1 to the extracellular phosphatidylserine recognition domains of the efferocytic receptors BAI1 or TIM4, generating BELMO and TELMO, respectively. CHEF-expressing phagocytes display a striking increase in efferocytosis. In mouse models of inflammation, BELMO expression attenuates colitis, hepatotoxicity, and nephrotoxicity. In mechanistic studies, BELMO increases ER-resident enzymes and chaperones to overcome protein-folding-associated toxicity, which was further validated in a model of ER-stress-induced renal ischemia-reperfusion injury. Finally, TELMO introduction after onset of kidney injury significantly reduced fibrosis. Collectively, these data advance a concept of chimeric efferocytic receptors to boost efferocytosis and dampen inflammation.


Subject(s)
Macrophages , Phagocytosis , Animals , Mice , Macrophages/metabolism , Inflammation/metabolism , Phagocytes/metabolism , Carrier Proteins/metabolism , Apoptosis , Adaptor Proteins, Signal Transducing/metabolism
2.
Cell ; 180(3): 502-520.e19, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31983537

ABSTRACT

The tumor microenvironment (TME) is critical for tumor progression. However, the establishment and function of the TME remain obscure because of its complex cellular composition. Using a mouse genetic system called mosaic analysis with double markers (MADMs), we delineated TME evolution at single-cell resolution in sonic hedgehog (SHH)-activated medulloblastomas that originate from unipotent granule neuron progenitors in the brain. First, we found that astrocytes within the TME (TuAstrocytes) were trans-differentiated from tumor granule neuron precursors (GNPs), which normally never differentiate into astrocytes. Second, we identified that TME-derived IGF1 promotes tumor progression. Third, we uncovered that insulin-like growth factor 1 (IGF1) is produced by tumor-associated microglia in response to interleukin-4 (IL-4) stimulation. Finally, we found that IL-4 is secreted by TuAstrocytes. Collectively, our studies reveal an evolutionary process that produces a multi-lateral network within the TME of medulloblastoma: a fraction of tumor cells trans-differentiate into TuAstrocytes, which, in turn, produce IL-4 that stimulates microglia to produce IGF1 to promote tumor progression.


Subject(s)
Astrocytes/metabolism , Carcinogenesis/metabolism , Cell Transdifferentiation , Cerebellar Neoplasms/metabolism , Medulloblastoma/metabolism , Paracrine Communication , Animals , Cell Lineage , Cerebellar Neoplasms/pathology , Disease Models, Animal , Female , Hedgehog Proteins/metabolism , Heterografts , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Male , Medulloblastoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Tumor Microenvironment
3.
Immunity ; 57(7): 1629-1647.e8, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38754432

ABSTRACT

The pancreatic islet microenvironment is highly oxidative, rendering ß cells vulnerable to autoinflammatory insults. Here, we examined the role of islet resident macrophages in the autoimmune attack that initiates type 1 diabetes. Islet macrophages highly expressed CXCL16, a chemokine and scavenger receptor for oxidized low-density lipoproteins (OxLDLs), regardless of autoimmune predisposition. Deletion of Cxcl16 in nonobese diabetic (NOD) mice suppressed the development of autoimmune diabetes. Mechanistically, Cxcl16 deficiency impaired clearance of OxLDL by islet macrophages, leading to OxLDL accumulation in pancreatic islets and a substantial reduction in intra-islet transitory (Texint) CD8+ T cells displaying proliferative and effector signatures. Texint cells were vulnerable to oxidative stress and diminished by ferroptosis; PD-1 blockade rescued this population and reversed diabetes resistance in NOD.Cxcl16-/- mice. Thus, OxLDL scavenging in pancreatic islets inadvertently promotes differentiation of pathogenic CD8+ T cells, presenting a paradigm wherein tissue homeostasis processes can facilitate autoimmune pathogenesis in predisposed individuals.


Subject(s)
Autoimmunity , CD8-Positive T-Lymphocytes , Cell Differentiation , Chemokine CXCL16 , Diabetes Mellitus, Type 1 , Islets of Langerhans , Lipoproteins, LDL , Macrophages , Mice, Inbred NOD , Mice, Knockout , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Chemokine CXCL16/metabolism , Macrophages/immunology , Macrophages/metabolism , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Mice, Inbred C57BL
4.
Nat Immunol ; 20(2): 141-151, 2019 02.
Article in English | MEDLINE | ID: mdl-30643265

ABSTRACT

Rheumatoid arthritis is characterized by progressive joint inflammation and affects ~1% of the human population. We noted single-nucleotide polymorphisms (SNPs) in the apoptotic cell-engulfment genes ELMO1, DOCK2, and RAC1 linked to rheumatoid arthritis. As ELMO1 promotes cytoskeletal reorganization during engulfment, we hypothesized that ELMO1 loss would worsen inflammatory arthritis. Surprisingly, Elmo1-deficient mice showed reduced joint inflammation in acute and chronic arthritis models. Genetic and cell-biology studies revealed that ELMO1 associates with receptors linked to neutrophil function in arthritis and regulates activation and early neutrophil recruitment to the joints, without general inhibition of inflammatory responses. Further, neutrophils from the peripheral blood of human donors that carry the SNP in ELMO1 associated with arthritis display increased migratory capacity, whereas ELMO1 knockdown reduces human neutrophil migration to chemokines linked to arthritis. These data identify 'noncanonical' roles for ELMO1 as an important cytoplasmic regulator of specific neutrophil receptors and promoter of arthritis.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Neutrophils/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis/immunology , Arthritis, Experimental/diagnosis , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Chemotaxis/genetics , Chemotaxis/immunology , Collagen/immunology , Complement C5a/immunology , Complement C5a/metabolism , Cytoplasm/immunology , Cytoplasm/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Healthy Volunteers , Humans , Intravital Microscopy , Joints/cytology , Joints/immunology , Leukotriene B4/immunology , Leukotriene B4/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Polymorphism, Single Nucleotide , Proteomics , Severity of Illness Index , Signal Transduction/immunology , Time-Lapse Imaging
5.
Annu Rev Neurosci ; 45: 177-198, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35226828

ABSTRACT

Neurodevelopment and efferocytosis have fascinated scientists for decades. How an organism builds a nervous system that is precisely tuned for efficient behaviors and survival and how it simultaneously manages constant somatic cell turnover are complex questions that have resulted in distinct fields of study. Although neurodevelopment requires the overproduction of cells that are subsequently pruned back, very few studies marry these fields to elucidate the cellular and molecular mechanisms that drive nervous system development through the lens of cell clearance. In this review, we discuss these fields to highlight exciting areas of future synergy. We first review neurodevelopment from the perspective of overproduction and subsequent refinement and then discuss who clears this developmental debris and the mechanisms that control these events. We then end with how a more deliberate merger ofneurodevelopment and efferocytosis could reframe our understanding of homeostasis and disease and discuss areas of future study.


Subject(s)
Apoptosis , Phagocytes , Apoptosis/physiology , Cell Death , Homeostasis , Phagocytes/metabolism , Phagocytosis/physiology
6.
Immunity ; 54(8): 1715-1727.e7, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34283971

ABSTRACT

Allergic airway inflammation is driven by type-2 CD4+ T cell inflammatory responses. We uncover an immunoregulatory role for the nucleotide release channel, Panx1, in T cell crosstalk during airway disease. Inverse correlations between Panx1 and asthmatics and our mouse models revealed the necessity, specificity, and sufficiency of Panx1 in T cells to restrict inflammation. Global Panx1-/- mice experienced exacerbated airway inflammation, and T-cell-specific deletion phenocopied Panx1-/- mice. A transgenic designed to re-express Panx1 in T cells reversed disease severity in global Panx1-/- mice. Panx1 activation occurred in pro-inflammatory T effector (Teff) and inhibitory T regulatory (Treg) cells and mediated the extracellular-nucleotide-based Treg-Teff crosstalk required for suppression of Teff cell proliferation. Mechanistic studies identified a Salt-inducible kinase-dependent phosphorylation of Panx1 serine 205 important for channel activation. A genetically targeted mouse expressing non-phosphorylatable Panx1S205A phenocopied the exacerbated inflammation in Panx1-/- mice. These data identify Panx1-dependent Treg:Teff cell communication in restricting airway disease.


Subject(s)
Asthma/immunology , Cell Communication/immunology , Connexins/metabolism , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line , Cell Proliferation/physiology , Connexins/genetics , Disease Models, Animal , HEK293 Cells , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Respiratory System/immunology
7.
Nature ; 628(8007): 408-415, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38480883

ABSTRACT

During development, inflammation or tissue injury, macrophages may successively engulf and process multiple apoptotic corpses via efferocytosis to achieve tissue homeostasis1. How macrophages may rapidly adapt their transcription to achieve continuous corpse uptake is incompletely understood. Transcriptional pause/release is an evolutionarily conserved mechanism, in which RNA polymerase (Pol) II initiates transcription for 20-60 nucleotides, is paused for minutes to hours and is then released to make full-length mRNA2. Here we show that macrophages, within minutes of corpse encounter, use transcriptional pause/release to unleash a rapid transcriptional response. For human and mouse macrophages, the Pol II pause/release was required for continuous efferocytosis in vitro and in vivo. Interestingly, blocking Pol II pause/release did not impede Fc receptor-mediated phagocytosis, yeast uptake or bacterial phagocytosis. Integration of data from three genomic approaches-precision nuclear run-on sequencing, RNA sequencing, and assay for transposase-accessible chromatin using sequencing (ATAC-seq)-on efferocytic macrophages at different time points revealed that Pol II pause/release controls expression of select transcription factors and downstream target genes. Mechanistic studies on transcription factor EGR3, prominently regulated by pause/release, uncovered EGR3-related reprogramming of other macrophage genes involved in cytoskeleton and corpse processing. Using lysosomal probes and a new genetic fluorescent reporter, we identify a role for pause/release in phagosome acidification during efferocytosis. Furthermore, microglia from egr3-deficient zebrafish embryos displayed reduced phagocytosis of apoptotic neurons and fewer maturing phagosomes, supporting defective corpse processing. Collectively, these data indicate that macrophages use Pol II pause/release as a mechanism to rapidly alter their transcriptional programs for efficient processing of the ingested apoptotic corpses and for successive efferocytosis.


Subject(s)
Efferocytosis , Macrophages , RNA Polymerase II , Transcription Elongation, Genetic , Animals , Humans , Male , Mice , Apoptosis , Cytoskeleton/metabolism , Early Growth Response Protein 3/deficiency , Early Growth Response Protein 3/genetics , Efferocytosis/genetics , Hydrogen-Ion Concentration , Macrophages/immunology , Macrophages/metabolism , Neurons/metabolism , Phagosomes/metabolism , RNA Polymerase II/metabolism , Transcription Factors/genetics , Zebrafish/embryology , Zebrafish/genetics , Time Factors
8.
Nature ; 631(8019): 207-215, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38926576

ABSTRACT

Pyroptosis is a lytic cell death mode that helps limit the spread of infections and is also linked to pathology in sterile inflammatory diseases and autoimmune diseases1-4. During pyroptosis, inflammasome activation and the engagement of caspase-1 lead to cell death, along with the maturation and secretion of the inflammatory cytokine interleukin-1ß (IL-1ß). The dominant effect of IL-1ß in promoting tissue inflammation has clouded the potential influence of other factors released from pyroptotic cells. Here, using a system in which macrophages are induced to undergo pyroptosis without IL-1ß or IL-1α release (denoted Pyro-1), we identify unexpected beneficial effects of the Pyro-1 secretome. First, we noted that the Pyro-1 supernatants upregulated gene signatures linked to migration, cellular proliferation and wound healing. Consistent with this gene signature, Pyro-1 supernatants boosted migration of primary fibroblasts and macrophages, and promoted faster wound closure in vitro and improved tissue repair in vivo. In mechanistic studies, lipidomics and metabolomics of the Pyro-1 supernatants identified the presence of both oxylipins and metabolites, linking them to pro-wound-healing effects. Focusing specifically on the oxylipin prostaglandin E2 (PGE2), we find that its synthesis is induced de novo during pyroptosis, downstream of caspase-1 activation and cyclooxygenase-2 activity; further, PGE2 synthesis occurs late in pyroptosis, with its release dependent on gasdermin D pores opened during pyroptosis. As for the pyroptotic metabolites, they link to immune cell infiltration into the wounds, and polarization to CD301+ macrophages. Collectively, these data advance the concept that the pyroptotic secretome possesses oxylipins and metabolites with tissue repair properties that may be harnessed therapeutically.


Subject(s)
Macrophages , Oxylipins , Pyroptosis , Secretome , Wound Healing , Animals , Female , Humans , Mice , Caspase 1/metabolism , Cell Movement , Cell Proliferation , Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Dinoprostone/metabolism , Fibroblasts/metabolism , Fibroblasts/cytology , Gasdermins/metabolism , Inflammasomes/metabolism , Interleukin-1beta , Lipidomics , Macrophages/metabolism , Macrophages/cytology , Mice, Inbred C57BL , Oxylipins/metabolism , Phosphate-Binding Proteins/metabolism , Secretome/metabolism , Wound Healing/physiology
9.
Immunity ; 50(5): 1149-1162, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31117011

ABSTRACT

Nearly every tissue in the body undergoes routine turnover of cells as part of normal healthy living. The majority of these cells undergoing turnover die via apoptosis, and then are rapidly removed by phagocytes by the process of efferocytosis that is anti-inflammatory. However, a number of pathologies have recently been linked to defective clearance of apoptotic cells. Perturbed clearance arises for many reasons, including overwhelming of the clearance machinery, disruptions at different stages of efferocytosis, and responses of phagocytes during efferocytosis, all of which can alter the homeostatic tissue environment. This review covers linkages of molecules involved in the different phases of efferocytosis to disease pathologies that can arise due to their loss or altered function.


Subject(s)
Apoptosis/physiology , Erythrocytes/physiology , Phagocytes/metabolism , Phagocytosis/physiology , Homeostasis , Humans
10.
Nature ; 606(7915): 776-784, 2022 06.
Article in English | MEDLINE | ID: mdl-35614212

ABSTRACT

Chronic non-healing wounds are a major complication of diabetes, which affects 1 in 10 people worldwide. Dying cells in the wound perpetuate the inflammation and contribute to dysregulated tissue repair1-3. Here we reveal that the membrane transporter SLC7A11 acts as a molecular brake on efferocytosis, the process by which dying cells are removed, and that inhibiting SLC7A11 function can accelerate wound healing. Transcriptomics of efferocytic dendritic cells in mouse identified upregulation of several SLC7 gene family members. In further analyses, pharmacological inhibition of SLC7A11, or deletion or knockdown of Slc7a11 using small interfering RNA enhanced efferocytosis in dendritic cells. Slc7a11 was highly expressed in dendritic cells in skin, and single-cell RNA sequencing of inflamed skin showed that Slc7a11 was upregulated in innate immune cells. In a mouse model of excisional skin wounding, inhibition or loss of SLC7A11 expression accelerated healing dynamics and reduced the apoptotic cell load in the wound. Mechanistic studies revealed a link between SLC7A11, glucose homeostasis and diabetes. SLC7A11-deficient dendritic cells were dependent on aerobic glycolysis using glucose derived from glycogen stores for increased efferocytosis; also, transcriptomics of efferocytic SLC7A11-deficient dendritic cells identified increased expression of genes linked to gluconeogenesis and diabetes. Further, Slc7a11 expression was higher in the wounds of diabetes-prone db/db mice, and targeting SLC7A11 accelerated their wound healing. The faster healing was also linked to the release of the TGFß family member GDF15 from efferocytic dendritic cells. In sum, SLC7A11 is a negative regulator of efferocytosis, and removing this brake improves wound healing, with important implications for wound management in diabetes.


Subject(s)
Amino Acid Transport System y+ , Dendritic Cells , Diabetes Mellitus , Phagocytosis , Wound Healing , Amino Acid Transport System y+/antagonists & inhibitors , Animals , Dendritic Cells/cytology , Dendritic Cells/immunology , Diabetes Mellitus/immunology , Gluconeogenesis , Glucose , Glycolysis , Growth Differentiation Factor 15 , Mice
11.
Annu Rev Pharmacol Toxicol ; 64: 339-357, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-37585658

ABSTRACT

Rapid removal of apoptotic cells by phagocytes, a process known as efferocytosis, is key for the maintenance of tissue homeostasis, the resolution of inflammation, and tissue repair. However, impaired efferocytosis can result in the accumulation of apoptotic cells, subsequently triggering sterile inflammation through the release of endogenous factors such as DNA and nuclear proteins from membrane permeabilized dying cells. Here, we review the molecular basis of the three key phases of efferocytosis, that is, the detection, uptake, and degradation of apoptotic materials by phagocytes. We also discuss how defects in efferocytosis due to the alteration of phagocytes and dying cells can contribute to the low-grade chronic inflammation that occurs during aging, described as inflammaging. Lastly, we explore opportunities in targeting and harnessing the efferocytic machinery to limit aging-associated inflammatory diseases.


Subject(s)
Aging , Efferocytosis , Humans , Biological Transport , Inflammation/drug therapy , Nuclear Proteins
12.
Nat Immunol ; 16(9): 907-17, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26287597

ABSTRACT

Human bodies collectively turn over about 200 billion to 300 billion cells every day. Such turnover is an integral part of embryonic and postnatal development, as well as routine tissue homeostasis. This process involves the induction of programmed cell death in specific cells within the tissues and the specific recognition and removal of dying cells by a clearance 'crew' composed of professional, non-professional and specialized phagocytes. In the past few years, considerable progress has been made in identifying many features of apoptotic cell clearance. Some of these new observations challenge the way dying cells themselves are viewed, as well as how healthy cells interact with and respond to dying cells. Here we focus on the homeostatic removal of apoptotic cells in tissues.


Subject(s)
Apoptosis/physiology , Homeostasis/physiology , Phagocytes/physiology , Phagocytosis/physiology , Humans
13.
Nature ; 596(7871): 262-267, 2021 08.
Article in English | MEDLINE | ID: mdl-34349263

ABSTRACT

Regulated cell death is an integral part of life, and has broad effects on organism development and homeostasis1. Malfunctions within the regulated cell death process, including the clearance of dying cells, can manifest in diverse pathologies throughout various tissues including the gastrointestinal tract2. A long appreciated, yet elusively defined relationship exists between cell death and gastrointestinal pathologies with an underlying microbial component3-6, but the direct effect of dying mammalian cells on bacterial growth is unclear. Here we advance a concept that several Enterobacteriaceae, including patient-derived clinical isolates, have an efficient growth strategy to exploit soluble factors that are released from dying gut epithelial cells. Mammalian nutrients released after caspase-3/7-dependent apoptosis boosts the growth of multiple Enterobacteriaceae and is observed using primary mouse colonic tissue, mouse and human cell lines, several apoptotic triggers, and in conventional as well as germ-free mice in vivo. The mammalian cell death nutrients induce a core transcriptional response in pathogenic Salmonella, and we identify the pyruvate formate-lyase-encoding pflB gene as a key driver of bacterial colonization in three contexts: a foodborne infection model, a TNF- and A20-dependent cell death model, and a chemotherapy-induced mucositis model. These findings introduce a new layer to the complex host-pathogen interaction, in which death-induced nutrient release acts as a source of fuel for intestinal bacteria, with implications for gut inflammation and cytotoxic chemotherapy treatment.


Subject(s)
Apoptosis , Enterobacteriaceae/growth & development , Enterobacteriaceae/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Intestines/cytology , Intestines/microbiology , Acetyltransferases/genetics , Acetyltransferases/metabolism , Animals , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Disease Models, Animal , Epithelial Cells/pathology , Female , Foodborne Diseases/microbiology , Germ-Free Life , Host-Pathogen Interactions , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Male , Mice , Mucositis/chemically induced , Salmonella/enzymology , Salmonella/genetics , Salmonella/growth & development , Salmonella/metabolism , Transcriptome , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Tumor Necrosis Factor-alpha/metabolism
14.
Cell ; 147(7): 1442-5, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22196723

ABSTRACT

Billions of cells die via apoptosis every day and are swiftly removed. When a phagocyte engulfs an apoptotic cell, it essentially doubles its cellular contents, raising the question of how a phagocyte may manage the excess metabolic load. This Minireview discusses phagocyte cellular metabolism, the digestion of the ingested apoptotic cell, and the impact of these processes on engulfment.


Subject(s)
Apoptosis , Phagocytes/metabolism , Phagocytosis , Animals , Cell Physiological Phenomena , Glucose/metabolism , Humans , Lipid Metabolism
15.
Nature ; 580(7801): 130-135, 2020 04.
Article in English | MEDLINE | ID: mdl-32238926

ABSTRACT

Caspase-dependent apoptosis accounts for approximately 90% of homeostatic cell turnover in the body1, and regulates inflammation, cell proliferation, and tissue regeneration2-4. How apoptotic cells mediate such diverse effects is not fully understood. Here we profiled the apoptotic metabolite secretome and determined its effects on the tissue neighbourhood. We show that apoptotic lymphocytes and macrophages release specific metabolites, while retaining their membrane integrity. A subset of these metabolites is also shared across different primary cells and cell lines after the induction of apoptosis by different stimuli. Mechanistically, the apoptotic metabolite secretome is not simply due to passive emptying of cellular contents and instead is a regulated process. Caspase-mediated opening of pannexin 1 channels at the plasma membrane facilitated the release of a select subset of metabolites. In addition, certain metabolic pathways continued to remain active during apoptosis, with the release of only select metabolites from a given pathway. Functionally, the apoptotic metabolite secretome induced specific gene programs in healthy neighbouring cells, including suppression of inflammation, cell proliferation, and wound healing. Furthermore, a cocktail of apoptotic metabolites reduced disease severity in mouse models of inflammatory arthritis and lung-graft rejection. These data advance the concept that apoptotic cells are not inert cells waiting for removal, but instead release metabolites as 'good-bye' signals to actively modulate outcomes in tissues.


Subject(s)
Apoptosis/physiology , Cellular Microenvironment , Second Messenger Systems/physiology , Animals , Arthritis , Caspases/metabolism , Cell Line , Cell Proliferation/genetics , Cell Survival/genetics , Connexins/metabolism , Disease Models, Animal , Graft Rejection , Humans , Inflammation/genetics , Lung Transplantation , Lymphocytes/enzymology , Lymphocytes/metabolism , Macrophages/enzymology , Macrophages/metabolism , Mice , Nerve Tissue Proteins/metabolism , Phagocytes/metabolism , Wound Healing/genetics
16.
Immunity ; 44(4): 807-20, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27037190

ABSTRACT

Few apoptotic corpses are seen even in tissues with high cellular turnover, leading to the notion that the capacity for engulfment in vivo is vast. Whether corpse clearance can be enhanced in vivo for potential benefit is not known. In a colonic inflammation model, we noted that the expression of the phagocytic receptor Bai1 was progressively downmodulated. Consistent with this, BAI1-deficient mice had more pronounced colitis and lower survival, with many uncleared apoptotic corpses and inflammatory cytokines within the colonic epithelium. When we engineered and tested transgenic mice overexpressing BAI1, these had fewer apoptotic cells, reduced inflammation, and attenuated disease. Boosting BAI1-mediated uptake by intestinal epithelial cells (rather than myeloid cells) was important in attenuating inflammation. A signaling-deficient BAI1 transgene could not provide a similar benefit. Collectively, these complementary genetic approaches showed that cell clearance could be boosted in vivo, with potential to regulate tissue inflammation in specific contexts.


Subject(s)
Angiogenic Proteins/genetics , Apoptosis/immunology , Colitis/immunology , Epithelial Cells/immunology , Intestinal Mucosa/immunology , Animals , Cell Line, Tumor , Colitis/chemically induced , Colon/immunology , Colon/pathology , Cytokines/immunology , Dextran Sulfate , HCT116 Cells , Humans , Inflammation/immunology , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/immunology
17.
Nature ; 563(7733): 714-718, 2018 11.
Article in English | MEDLINE | ID: mdl-30464343

ABSTRACT

Development and routine tissue homeostasis require a high turnover of apoptotic cells. These cells are removed by professional and non-professional phagocytes via efferocytosis1. How a phagocyte maintains its homeostasis while coordinating corpse uptake, processing ingested materials and secreting anti-inflammatory mediators is incompletely understood1,2. Here, using RNA sequencing to characterize the transcriptional program of phagocytes actively engulfing apoptotic cells, we identify a genetic signature involving 33 members of the solute carrier (SLC) family of membrane transport proteins, in which expression is specifically modulated during efferocytosis, but not during antibody-mediated phagocytosis. We assessed the functional relevance of these SLCs in efferocytic phagocytes and observed a robust induction of an aerobic glycolysis program, initiated by SLC2A1-mediated glucose uptake, with concurrent suppression of the oxidative phosphorylation program. The different steps of phagocytosis2-that is, 'smell' ('find-me' signals or sensing factors released by apoptotic cells), 'taste' (phagocyte-apoptotic cell contact) and 'ingestion' (corpse internalization)-activated distinct and overlapping sets of genes, including several SLC genes, to promote glycolysis. SLC16A1 was upregulated after corpse uptake, increasing the release of lactate, a natural by-product of aerobic glycolysis3. Whereas glycolysis within phagocytes contributed to actin polymerization and the continued uptake of corpses, lactate released via SLC16A1 promoted the establishment of an anti-inflammatory tissue environment. Collectively, these data reveal a SLC program that is activated during efferocytosis, identify a previously unknown reliance on aerobic glycolysis during apoptotic cell uptake and show that glycolytic by-products of efferocytosis can influence surrounding cells.


Subject(s)
Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose/metabolism , Lactic Acid/metabolism , Phagocytes/metabolism , Phagocytosis/genetics , Transcriptome/genetics , Aerobiosis , Animals , Apoptosis , Cell Line , Glycolysis , Humans , Inflammation/genetics , Inflammation/prevention & control , Jurkat Cells , Phagocytes/cytology , Sequence Analysis, RNA , Transcription, Genetic , Zebrafish
18.
EMBO Rep ; 22(6): e52564, 2021 06 04.
Article in English | MEDLINE | ID: mdl-34041845

ABSTRACT

Timely removal of dying or pathogenic cells by phagocytes is essential to maintaining host homeostasis. Phagocytes execute the clearance process with high fidelity while sparing healthy neighboring cells, and this process is at least partially regulated by the balance of "eat-me" and "don't-eat-me" signals expressed on the surface of host cells. Upon contact, eat-me signals activate "pro-phagocytic" receptors expressed on the phagocyte membrane and signal to promote phagocytosis. Conversely, don't-eat-me signals engage "anti-phagocytic" receptors to suppress phagocytosis. We review the current knowledge of don't-eat-me signaling in normal physiology and disease contexts where aberrant don't-eat-me signaling contributes to pathology.


Subject(s)
Biological Phenomena , Phagocytosis , Apoptosis , Phagocytes , Signal Transduction
19.
Cell ; 133(3): 393-5, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18455977

ABSTRACT

During embryonic development, large numbers of apoptotic cells are rapidly cleared by phagocytes. In this issue, Kurant et al. (2008) describe a new phagocytic receptor, called six-microns-under (SIMU), that promotes engulfment of apoptotic neurons by glial cells in the developing nervous system of Drosophila.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/embryology , Membrane Proteins/metabolism , Phagocytosis , Animals , Apoptosis , Central Nervous System , Drosophila/cytology , Neuroglia/metabolism , Neurons/cytology
SELECTION OF CITATIONS
SEARCH DETAIL