Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Trends Immunol ; 45(6): 442-453, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38782625

ABSTRACT

Activated CD8+ T cells directly kill target cells. Therefore, the regulation of their function is central to avoiding immunopathology. Mechanisms that curb effector functions in CD4+ and CD8+ T cells are mostly shared, yet important differences occur. Here, we focus on the control of CD8+ T cell activity and discuss the importance of a poorly understood aspect of tolerance that directly impairs engagement of target cells: the downregulation of CD8. We contextualize this process and propose that it represents a key element during CD8+ T cell modulation.


Subject(s)
CD8-Positive T-Lymphocytes , Immune Tolerance , Animals , Humans , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/metabolism , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Down-Regulation/immunology , Lymphocyte Activation/immunology
2.
Clin Immunol ; 263: 110225, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642784

ABSTRACT

Systemic lupus erythematosus (SLE) and other autoimmune diseases are thought to develop in genetically predisposed individuals when triggered by environmental factors. This paradigm does not fully explain disease development, as it fails to consider the delay between birth and disease expression. In this review, we discuss observations described in T cells from patients with SLE that are not related to hereditary factors and have therefore been considered secondary to the disease process itself. Here, we contextualize some of those observations and argue that they may represent a pathogenic layer between genetic factors and disease development. Acquired changes in T cell phenotype and function in the setting of SLE may affect the immune system, creating a predisposition towards a more inflammatory and pathogenic system that amplifies autoimmunity and facilitates disease development.


Subject(s)
Lupus Erythematosus, Systemic , T-Lymphocytes , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/genetics , T-Lymphocytes/immunology , Autoimmunity/immunology , Autoimmunity/genetics , Genetic Predisposition to Disease , Animals
3.
JCI Insight ; 9(6)2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38516890

ABSTRACT

Sodium-glucose cotransporter 2 (SGLT2) inhibitor, dapagliflozin (Dapa), exhibited nephroprotective effects in patients with chronic kidney disease (CKD). We assessed the efficacy of short-term Dapa administration following acute kidney injury (AKI) in preventing CKD. Male Wistar rats were randomly assigned to Sham surgery, bilateral ischemia for 30 minutes (abbreviated as IR), and IR + Dapa groups. Daily treatment with Dapa was initiated just 24 hours after IR and maintained for only 10 days. Initially, rats were euthanized at this point to study early renal repair. After severe AKI, Dapa promptly restored creatinine clearance (CrCl) and significantly reduced renal vascular resistance compared with the IR group. Furthermore, Dapa effectively reversed the mitochondrial abnormalities, including increased fission, altered mitophagy, metabolic dysfunction, and proapoptotic signaling. To study this earlier, another set of rats was studied just 5 days after AKI. Despite persistent renal dysfunction, our data reveal a degree of mitochondrial protection. Remarkably, a 10-day treatment with Dapa demonstrated effectiveness in preventing CKD transition in an independent cohort monitored for 5 months after AKI. This was evidenced by improvements in proteinuria, CrCl, glomerulosclerosis, and fibrosis. Our findings underscore the potential of Dapa in preventing maladaptive repair following AKI, emphasizing the crucial role of early intervention in mitigating AKI long-term consequences.


Subject(s)
Acute Kidney Injury , Renal Insufficiency, Chronic , Reperfusion Injury , Animals , Humans , Male , Rats , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Acute Kidney Injury/metabolism , Glucose , Rats, Wistar , Renal Insufficiency, Chronic/drug therapy , Reperfusion Injury/complications , Reperfusion Injury/metabolism , Sodium/metabolism , Sodium-Glucose Transporter 2/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Benzhydryl Compounds/pharmacology , Benzhydryl Compounds/therapeutic use
4.
Arthritis Rheumatol ; 75(6): 961-972, 2023 06.
Article in English | MEDLINE | ID: mdl-36575804

ABSTRACT

OBJECTIVE: Variants in STAT4 are associated with systemic lupus erythematosus (SLE) and other autoimmune diseases. We undertook this study to investigate how disease-associated variants affect STAT4 expression, in particular in CD4+ T cells where STAT4 plays an essential role. METHODS: We compared Th1 differentiation between naive CD4+ T cells from healthy donors homozygous for the risk (R/R) or nonrisk (NR/NR) alleles. We analyzed epigenetic marks in STAT4 and evaluated the relevance of its third intron, assessed the consequences of Stat4 overexpression in vivo in mice, and analyzed the effects of the STAT4 genotype in patients with lupus nephritis. RESULTS: Naive CD4+ T cells from NR/NR healthy donors down-regulated STAT4 in response to interleukin-12 (IL-12). In contrast, cells from R/R healthy donors maintained high levels. R/R cells exhibited a higher abundance of transcriptionally active STAT4 and increased interferon-γ production. Accordingly, R/R healthy donors exhibited a stronger induction of local active enhancer marks. Genetic editing confirmed the presence of a negative regulatory region in the STAT4 third intron, where most of the SLE-associated STAT4 single-nucleotide polymorphisms (SNPs) are located. In vivo forced expression demonstrated that increases in Stat4 levels in T cells enhanced glomerulonephritis in mice. Accordingly, the R/R genotype was associated with suboptimal response to treatment and with worse clinical outcomes in patients with proliferative lupus nephritis. CONCLUSION: The SLE-associated STAT4 haplotype correlates with an abnormal IL-12-mediated STAT4 transcriptional regulation. Carriers of the risk variant exhibit exaggerated CD4+ proinflammatory capacities that, in the context of SLE, contribute to more severe disease. R/R patients may benefit from blockade of the IL-12/STAT4 pathway.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Animals , Mice , CD4-Positive T-Lymphocytes/metabolism , Down-Regulation , Haplotypes , Interferon-gamma/genetics , Interleukin-12 , Lupus Erythematosus, Systemic/genetics , Lupus Nephritis/genetics , Polymorphism, Single Nucleotide , STAT4 Transcription Factor/genetics , Humans
5.
J Exp Med ; 219(5)2022 05 02.
Article in English | MEDLINE | ID: mdl-35404389

ABSTRACT

Monocytes undergo phenotypic and functional changes in response to inflammatory cues, but the molecular signals that drive different monocyte states remain largely undefined. We show that monocytes acquire macrophage markers upon glomerulonephritis and may be derived from CCR2+CX3CR1+ double-positive monocytes, which are preferentially recruited, dwell within glomerular capillaries, and acquire proinflammatory characteristics in the nephritic kidney. Mechanistically, the transition to immature macrophages begins within the vasculature and relies on CCR2 in circulating cells and TNFR2 in parenchymal cells, findings that are recapitulated in vitro with monocytes cocultured with TNF-TNFR2-activated endothelial cells generating CCR2 ligands. Single-cell RNA sequencing of cocultures defines a CCR2-dependent monocyte differentiation path associated with the acquisition of immune effector functions and generation of CCR2 ligands. Immature macrophages are detected in the urine of lupus nephritis patients, and their frequency correlates with clinical disease. In conclusion, CCR2-dependent functional specialization of monocytes into macrophages begins within the TNF-TNFR2-activated vasculature and may establish a CCR2-based autocrine, feed-forward loop that amplifies renal inflammation.


Subject(s)
Endothelial Cells , Monocytes , Receptors, CCR2 , Receptors, Tumor Necrosis Factor, Type II , Humans , Ligands , Macrophages , Receptors, CCR2/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics
6.
Int J Mol Sci ; 23(5)2022 Feb 25.
Article in English | MEDLINE | ID: mdl-35269715

ABSTRACT

Acute kidney injury (AKI) is a public health problem worldwide. Sirtuins are a family of seven NAD+-dependent deacylases, Overexpression of Sirtuin 1, 3, and 5 protect against AKI. However, the role of Sirtuin 7 (Sirt7) in AKI is not known. Here, we analyzed how Sirt7 deficient mice (KO-Sirt7) were affected by AKI. As expected, wild-type and Sirt7 heterozygotes mice that underwent renal ischemia/reperfusion (IR) exhibited the characteristic hallmarks of AKI: renal dysfunction, tubular damage, albuminuria, increased oxidative stress, and renal inflammation. In contrast, the KO-Sirt7+IR mice were protected from AKI, exhibiting lesser albuminuria and reduction in urinary biomarkers of tubular damage, despite similar renal dysfunction. The renoprotection in the Sirt7-KO+IR group was associated with reduced kidney weight, minor expression of inflammatory cytokines and less renal infiltration of inflammatory cells. This anti-inflammatory effect was related to diminished p65 expression and in its active phosphorylation, as well as by a reduction in p65 nuclear translocation. Sirt7 deficient mice are protected from AKI, suggesting that this histone deacetylase promotes tubular damage and renal inflammation. Therefore, our findings indicate that Sirt7 inhibitors may be an attractive therapeutic target to reduce NFκB signaling.


Subject(s)
Acute Kidney Injury , Reperfusion Injury , Sirtuins/metabolism , Acute Kidney Injury/metabolism , Albuminuria , Animals , Inflammation/metabolism , Kidney/metabolism , Mice , Mice, Inbred C57BL , Reperfusion Injury/metabolism , Sirtuins/genetics
7.
Nat Rev Rheumatol ; 18(4): 232-244, 2022 04.
Article in English | MEDLINE | ID: mdl-35075294

ABSTRACT

Adaptive immune responses rely on the proliferation of T lymphocytes able to recognize and eliminate pathogens. The magnitude and duration of the expansion of activated T cell clones are finely regulated to minimize immunopathology and avoid autoimmunity. In patients with rheumatic autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis, activated lymphocytes survive and exert effector functions for prolonged periods, defying the mechanisms that normally curb their capacities during acute and chronic infections. Here, we review the molecular mechanisms that limit the duration of immune responses in health and discuss the factors that alter such regulation in the setting of systemic lupus erythematosus and rheumatoid arthritis. We highlight defects that could contribute to the development and progression of autoimmune disease and describe how chronic inflammation can alter the regulation of activated lymphocyte survival, promoting its perpetuation. These concepts might contribute to the understanding of the mechanisms that underlie the chronicity of inflammation in the context of autoimmunity.


Subject(s)
Arthritis, Rheumatoid , Autoimmune Diseases , Lupus Erythematosus, Systemic , Rheumatic Diseases , Autoimmunity , Cell Survival , Humans , Inflammation , T-Lymphocytes
8.
Rev Invest Clin ; 73(5): 297-301, 2021.
Article in English | MEDLINE | ID: mdl-34609362

ABSTRACT

Systemic autoimmune diseases are complex clinical conditions that arise in genetically predisposed individuals as a result of the interplay between their immune system and their environment. In this perspective, we briefly discuss our current understanding of the pathogenesis of autoimmunity and indicate four research avenues whose exploration will bring us closer to resolving fundamental questions that remain unanswered in this enigmatic field.


Subject(s)
Autoimmune Diseases , Autoimmunity , Genetic Predisposition to Disease , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmunity/genetics , Humans
9.
Nat Commun ; 12(1): 4791, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34373452

ABSTRACT

Classical dendritic cells (cDC) are professional antigen-presenting cells (APC) that regulate immunity and tolerance. Neutrophil-derived cells with properties of DCs (nAPC) are observed in human diseases and after culture of neutrophils with cytokines. Here we show that FcγR-mediated endocytosis of antibody-antigen complexes or an anti-FcγRIIIB-antigen conjugate converts neutrophils into nAPCs that, in contrast to those generated with cytokines alone, activate T cells to levels observed with cDCs and elicit CD8+ T cell-dependent anti-tumor immunity in mice. Single cell transcript analyses and validation studies implicate the transcription factor PU.1 in neutrophil to nAPC conversion. In humans, blood nAPC frequency in lupus patients correlates with disease. Moreover, anti-FcγRIIIB-antigen conjugate treatment induces nAPCs that can activate autologous T cells when using neutrophils from individuals with myeloid neoplasms that harbor neoantigens or those vaccinated against bacterial toxins. Thus, anti-FcγRIIIB-antigen conjugate-induced conversion of neutrophils to immunogenic nAPCs may represent a possible immunotherapy for cancer and infectious diseases.


Subject(s)
Antigens, Neoplasm/immunology , Neoplasms/immunology , Neutrophils/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Antigen Presentation/immunology , Antigen-Antibody Complex , Bone Marrow , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Movement , Cell Proliferation , Cytokines/immunology , Dendritic Cells/immunology , Endocytosis , Humans , Immunity, Innate , Immunotherapy , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Reactive Oxygen Species , Transcriptome
10.
Front Immunol ; 12: 635862, 2021.
Article in English | MEDLINE | ID: mdl-33841416

ABSTRACT

Activation of self-reactive CD8+ T cells induces a peripheral tolerance mechanism that involves loss of CD8 expression. Because genetic deficiency of Fas and Fasl causes the accumulation of double-negative (DN; CD3+ TCR-αß+ CD4- CD8-) T cells that have been proposed to derive from CD8+ cells, we decided to explore the role of Fas and FasL in self-antigen-induced CD8 downregulation. To this end, we quantified Fas and FasL induction by different stimuli and analyzed the effects of Fas/FasL deficiency during a protective immune response and after exposure to self-antigens. Our data describes how Fas and FasL upregulation differs depending on the setting of CD8 T cell activation and demonstrates that Fas/FasL signaling maintains CD8 expression during repetitive antigen stimulation and following self-antigen encounter. Together, our results reveal an unexpected role of Fas/FasL signaling and offer a new insight into the role of these molecules in the regulation of immune tolerance.


Subject(s)
Autoantigens/metabolism , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Fas Ligand Protein/metabolism , Immune Tolerance , Lymphocyte Activation , fas Receptor/metabolism , Adoptive Transfer , Animals , Autoantigens/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cells, Cultured , Down-Regulation , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Kinetics , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Signal Transduction , fas Receptor/genetics , fas Receptor/immunology
11.
J Clin Invest ; 130(11): 5989-6004, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32750040

ABSTRACT

How T cells integrate environmental cues into signals that limit the magnitude and length of immune responses is poorly understood. Here, we provide data that demonstrate that B55ß, a regulatory subunit of protein phosphatase 2A, represents a molecular link between cytokine concentration and apoptosis in activated CD8+ T cells. Through the modulation of AKT, B55ß induced the expression of the proapoptotic molecule Hrk in response to cytokine withdrawal. Accordingly, B55ß and Hrk were both required for in vivo and in vitro contraction of activated CD8+ lymphocytes. We show that this process plays a role during clonal contraction, establishment of immune memory, and preservation of peripheral tolerance. This regulatory pathway may represent an unexplored opportunity to end unwanted immune responses or to promote immune memory.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Protein Phosphatase 2/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Mice , Mice, Transgenic , Neuropeptides/genetics , Neuropeptides/immunology , Protein Phosphatase 2/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology
12.
J Leukoc Biol ; 108(3): 851-857, 2020 09.
Article in English | MEDLINE | ID: mdl-32052478

ABSTRACT

The cellular origin of CD4- CD8- (double negative, DNT) TCR-α/ß+ T cells remains unknown. Available evidence indicates that they may derive from CD8+ T cells, but most published data have been obtained using cells that bear an invariant transgenic T cell receptor that recognizes an Ag that is not present in normal mice. Here, we have used complementary fate mapping and adoptive transfer experiments to identify the cellular lineage of origin of DNT cells in wild-type mice with a polyclonal T cell repertoire. We show that TCR-α/ß+ DNT cells can be traced back to CD8+ and CD4+ CD8+ double positive cells in the thymus. We also demonstrate that polyclonal DNT cells generated in secondary lymphoid organs proliferate upon adoptive transfer and can regain CD8 expression in lymphopenic environment. These results demonstrate the cellular origin of DNT cells and provide a conceptual framework to understand their presence in pathological circumstances.


Subject(s)
CD4 Antigens/analysis , CD8 Antigens/analysis , CD8-Positive T-Lymphocytes/cytology , Lymphopoiesis , Receptors, Antigen, T-Cell, alpha-beta/analysis , T-Lymphocyte Subsets/cytology , Adoptive Transfer , Animals , CD8 Antigens/biosynthesis , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/classification , Cell Lineage , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Specific Pathogen-Free Organisms , Up-Regulation
13.
Clin Immunol ; 212: 108240, 2020 03.
Article in English | MEDLINE | ID: mdl-31299381

ABSTRACT

CD8 T cells can kill malignant cells in an antigen-specific manner. However, anti-tumoral responses are usually limited by suppressive factors that curb the effector responses of tumor-infiltrating CD8 T cells. Therapeutic strategies to overcome intra-tumoral T cell suppression, for example immune checkpoint inhibition, have been clinically effective in patients with cancer. Here, we provide data that demonstrates that GK-1, a peptide derived from the parasite Taenia crassiceps, promotes an anti-melanoma CD8 T cell response with heightened effector characteristics that leads to an increased amount of tumor-infiltrating CD44+ IFN-γ-producing CD8 T cells. The response induced by GK-1 was associated with a reduction in the expression of PD-1 and PD-L1 on tumor-infiltrating CD8 and dendritic cells, respectively, effects that led to a dramatic decrease in tumor burden. Our results suggest that the immunomodulatory properties of GK-1 may promote a CD8 T cell response that may be therapeutically useful in the setting of cancer.


Subject(s)
B7-H1 Antigen/drug effects , CD8-Positive T-Lymphocytes/drug effects , Dendritic Cells/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma, Experimental/immunology , Peptides, Cyclic/pharmacology , Programmed Cell Death 1 Receptor/drug effects , Skin Neoplasms/immunology , Tumor Microenvironment/drug effects , Adoptive Transfer , Animals , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Down-Regulation , Hyaluronan Receptors/immunology , Interferon-gamma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/transplantation , Taenia , Tumor Microenvironment/immunology
14.
Sci Rep ; 9(1): 15502, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31664045

ABSTRACT

Systemic lupus erythematosus (SLE) is a heterogeneous disease with unpredictable patterns of activity. Patients with similar activity levels may have different prognosis and molecular abnormalities. In this study, we aimed to measure the main differences in drug-induced gene expression signatures across SLE patients and to evaluate the potential for clinical data to build a machine learning classifier able to predict the SLE subset for individual patients. SLE transcriptomic data from two cohorts were compared with drug-induced gene signatures from the CLUE database to compute a connectivity score that reflects the capability of a drug to revert the patient signatures. Patient stratification based on drug connectivity scores revealed robust clusters of SLE patients identical to the clusters previously obtained through longitudinal gene expression data, implying that differential treatment depends on the cluster to which patients belongs. The best drug candidates found, mTOR inhibitors or those reducing oxidative stress, showed stronger cluster specificity. We report that drug patterns for reverting disease gene expression follow the cell-specificity of the disease clusters. We used 2 cohorts to train and test a logistic regression model that we employed to classify patients from 3 independent cohorts into the SLE subsets and provide a clinically useful model to predict subset assignment and drug efficacy.


Subject(s)
Lupus Erythematosus, Systemic/genetics , Transcriptome/drug effects , Case-Control Studies , Cluster Analysis , Cohort Studies , Female , Humans , Longitudinal Studies , Lupus Erythematosus, Systemic/classification , Lupus Erythematosus, Systemic/drug therapy , Male , Severity of Illness Index
15.
JCI Insight ; 52019 07 23.
Article in English | MEDLINE | ID: mdl-31335320

ABSTRACT

Chronic inflammation causes target organ damage in patients with systemic autoimmune diseases. The factors that allow this protracted response are poorly understood. We analyzed the transcriptional regulation of PPP2R2B (B55ß), a molecule necessary for the termination of the immune response, in patients with autoimmune diseases. Altered expression of B55ß conditioned resistance to cytokine withdrawal-induced death (CWID) in patients with autoimmune diseases. The impaired upregulation of B55ß was caused by inflammation-driven hypermethylation of specific cytosines located within a regulatory element of PPP2R2B preventing CTCF binding. This phenotype could be induced in healthy T cells by exposure to TNF-α. Our results reveal a gene whose expression is affected by an acquired defect, through an epigenetic mechanism, in the setting of systemic autoimmunity. Because failure to remove activated T cells through CWID could contribute to autoimmune pathology, this mechanism illustrates a vicious cycle through which autoimmune inflammation contributes to its own perpetuation.


Subject(s)
Apoptosis/drug effects , Autoimmune Diseases/metabolism , DNA Methylation , Nerve Tissue Proteins/metabolism , Protein Phosphatase 2/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , CCCTC-Binding Factor/metabolism , Cytokines/metabolism , Cytosine/metabolism , DNA Methylation/drug effects , Gene Expression Regulation , Humans , Inflammation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Protein Phosphatase 2/genetics , Protein Phosphatase 2/pharmacology , T-Lymphocytes , Up-Regulation
16.
Curr Opin Rheumatol ; 31(2): 185-192, 2019 03.
Article in English | MEDLINE | ID: mdl-30672908

ABSTRACT

PURPOSE OF REVIEW: The aim of this review is to discuss recent developments in our understanding of how systemic lupus erythematosus (SLE)-associated genes contribute to autoimmunity. RECENT FINDINGS: Gene-function studies have revealed mechanisms through which SLE-associated alleles of IFIH1, TNFAIP3, IRF5, and PRDM1 likely contribute to the development of autoimmunity. Novel research has identified Mac-1 (encoded by ITGAM), CaMK4, and iRhom2 as plausible therapeutic targets in lupus nephritis. SUMMARY: The work discussed in this review has broad implications for our understanding of the pathogenesis of SLE and for the development of novel therapeutic strategies.


Subject(s)
CD11b Antigen/genetics , Gene Expression Regulation , Genetic Predisposition to Disease , Genetic Therapy/methods , Lupus Erythematosus, Systemic/genetics , RNA/genetics , CD11b Antigen/biosynthesis , Humans , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/therapy
17.
Nat Commun ; 9(1): 5058, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30498196

ABSTRACT

Vascular-deposited IgG immune complexes promote neutrophil recruitment, but how this process is regulated is still unclear. Here we show that the CD18 integrin Mac-1, in its bent state, interacts with the IgG receptor FcγRIIA in cis to reduce the affinity of FcγRIIA for IgG and inhibit FcγRIIA-mediated neutrophil recruitment under flow. The Mac-1 rs1143679 lupus-risk variant reverses Mac-1 inhibition of FcγRIIA, as does a Mac-1 ligand and a mutation in Mac-1's ligand binding αI-domain. Sialylated complex glycans on FcγRIIA interact with the αI-domain via divalent cations, and this interaction is required for FcγRIIA inhibition by Mac-1. Human neutrophils deficient in CD18 integrins exhibit augmented FcγRIIA-dependent recruitment to IgG-coated endothelium. In mice, CD18 integrins on neutrophils dampen IgG-mediated neutrophil accumulation in the kidney. In summary, cis interaction between sialylated FcγRIIA and the αI-domain of Mac-1 alters the threshold for IgG-mediated neutrophil recruitment. A disruption of this interaction may increase neutrophil influx in autoimmune diseases.


Subject(s)
Macrophage-1 Antigen/metabolism , Neutrophils/metabolism , Receptors, IgG/metabolism , Animals , Basement Membrane/metabolism , Endothelium/metabolism , Glycosylation , HEK293 Cells , Humans , Immunoglobulin G/metabolism , Jurkat Cells/metabolism , Macrophage-1 Antigen/chemistry , Male , Mice , Nephritis/metabolism , Protein Structure, Secondary , Receptors, IgG/chemistry
18.
Trends Mol Med ; 24(4): 364-378, 2018 04.
Article in English | MEDLINE | ID: mdl-29526595

ABSTRACT

Disease heterogeneity remains a major challenge for the understanding of systemic lupus erythematosus (SLE). Recent work has revealed the important role of nonimmune factors in the development of end-organ damage involvement, shifting the current paradigm that views SLE as a disease inflicted by a disturbed immune system on passive target organs. Here, we discuss the pathogenesis of lupus nephritis in a comprehensive manner, by incorporating the role that target organs play by withstanding and modulating the local inflammatory response. Moreover, we consider the effects that genetic variants exert on immune and nonimmune cells in order to shape the phenotype of the disease in each affected individual.


Subject(s)
Lupus Nephritis/immunology , Lupus Nephritis/pathology , Animals , Genetic Variation/immunology , Humans , Immune System/immunology , Immune System/pathology , Inflammation/immunology , Inflammation/pathology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology
20.
J Clin Invest ; 127(10): 3810-3826, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28891817

ABSTRACT

The kidney glomerular capillaries are frequent sites of immune complex deposition and subsequent neutrophil accumulation in post-infectious and rapidly progressive glomerulonephritis. However, the mechanisms of neutrophil recruitment remain enigmatic, and there is no targeted therapeutic to avert this proximal event in glomerular inflammation. The uniquely human activating Fc receptor FcγRIIA promotes glomerular neutrophil accumulation and damage in anti-glomerular basement membrane-induced (anti-GBM-induced) glomerulonephritis when expressed on murine neutrophils. Here, we found that neutrophils are directly captured by immobilized IgG antibodies under physiological flow conditions in vitro through FcγRIIA-dependent, Abl/Src tyrosine kinase-mediated F-actin polymerization. Biophysical measurements showed that the lifetime of FcγRIIA-IgG bonds increased under mechanical force in an F-actin-dependent manner, which could enable the capture of neutrophils under physiological flow. Kidney intravital microscopy revealed that circulating neutrophils, which were similar in diameter to glomerular capillaries, abruptly arrested following anti-GBM antibody deposition via neutrophil FcγRIIA and Abl/Src kinases. Accordingly, inhibition of Abl/Src with bosutinib reduced FcγRIIA-mediated glomerular neutrophil accumulation and renal injury in experimental, crescentic anti-GBM nephritis. These data identify a pathway of neutrophil recruitment within glomerular capillaries following IgG deposition that may be targeted by bosutinib to avert glomerular injury.


Subject(s)
Aniline Compounds/pharmacology , Glomerulonephritis/immunology , Immunoglobulin G/immunology , Kidney Glomerulus/immunology , Neutrophils/immunology , Nitriles/pharmacology , Quinolines/pharmacology , Receptors, IgG/immunology , Animals , Capillaries/immunology , Capillaries/pathology , Glomerulonephritis/genetics , Glomerulonephritis/pathology , HL-60 Cells , Humans , Kidney Glomerulus/pathology , Mice , Mice, Knockout , Neutrophils/pathology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/immunology , Receptors, IgG/genetics , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...