Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 573
Filter
1.
Sci Signal ; 17(837): eadq4734, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38771919

ABSTRACT

Antibody fragments can act as pharmacological tools to modulate the functions of G protein-coupled receptors.


Subject(s)
Receptors, G-Protein-Coupled , Single-Domain Antibodies , Single-Domain Antibodies/immunology , Humans , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/immunology , Animals
2.
J Immunol ; 212(10): 1531-1539, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38506555

ABSTRACT

Immune thrombocytopenia (ITP) is an autoimmune disease characterized by low platelet counts primarily due to antiplatelet autoantibodies. Anti-D is a donor-derived polyclonal Ab against the rhesus D Ag on erythrocytes used to treat ITP. Unfortunately, adverse inflammatory/hypersensitivity reactions and a Food and Drug Administration-issued black box warning have limited its clinical use. This underscores the imperative to understand the inflammatory pathway associated with anti-erythrocyte Ab-based therapies. TER119 is an erythrocyte-specific Ab with anti-D-like therapeutic activity in murine ITP, while also exhibiting a distinct inflammatory signature involving production of CCL2, CCL5, and CXCL9 but not IFN-γ. Therefore, TER119 has been used to elucidate the potential mechanism underlying the adverse inflammatory activity associated with anti-erythrocyte Ab therapy in murine ITP. Prior work has demonstrated that TER119 administration is associated with a dramatic decrease in body temperature and inflammatory cytokine/chemokine production. The work presented in the current study demonstrates that inhibiting the highly inflammatory platelet-activating factor (PAF) pathway with PAF receptor antagonists prevents TER119-driven changes in body temperature and inhibits the production of the CCL2, CCL5, and CXCL9 inflammatory cytokines in CD-1 mice. Phagocytic cells and a functional TER119 Fc region were found to be necessary for TER119-induced body temperature changes and increases in CXCL9 and CCL2. Taken together, this work reveals the novel requirement of the PAF pathway in causing adverse inflammatory activity associated with anti-erythrocyte Ab therapy in a murine model and provides a strategy of mitigating these potential reactions without altering therapeutic activity.


Subject(s)
Chemokine CCL2 , Erythrocytes , Inflammation , Platelet Activating Factor , Platelet Membrane Glycoproteins , Purpura, Thrombocytopenic, Idiopathic , Animals , Mice , Platelet Activating Factor/immunology , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Erythrocytes/immunology , Inflammation/immunology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/immunology , Chemokine CCL2/immunology , Chemokine CCL5/immunology , Chemokine CXCL9/immunology , Receptors, G-Protein-Coupled/immunology , Signal Transduction/immunology , Mice, Inbred C57BL , Autoantibodies/immunology , Disease Models, Animal
4.
Immunity ; 57(1): 28-39, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38198852

ABSTRACT

The discovery of Mas-related G protein-coupled receptors (Mrgprs) has opened a compelling chapter in our understanding of immunity and sensory biology. This family of receptors, with their unique expression and diverse ligands, has emerged as key players in inflammatory states and hold the potential to alleviate human diseases. This review will focus on the members of this receptor family expressed on immune cells and how they govern immune and neuro-immune pathways underlying various physiological and pathological states. Immune cell-specific Mrgprs have been shown to control a variety of manifestations, including adverse drug reactions, inflammatory conditions, bacterial immunity, and the sensing of environmental exposures like allergens and irritants.


Subject(s)
Immunity , Receptors, G-Protein-Coupled , Humans , Environmental Exposure , Receptors, G-Protein-Coupled/immunology
5.
Nat Cancer ; 4(11): 1536-1543, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37653140

ABSTRACT

Bispecific antibodies targeting GPRC5D demonstrated promising efficacy in multiple myeloma, but acquired resistance usually occurs within a few months. Using a single-nucleus multi-omic strategy in three patients from the MYRACLE cohort (ClinicalTrials.gov registration: NCT03807128 ), we identified two resistance mechanisms, by bi-allelic genetic inactivation of GPRC5D or by long-range epigenetic silencing of its promoter and enhancer regions. Molecular profiling of target genes may help to guide the choice of immunotherapy and early detection of resistance in multiple myeloma.


Subject(s)
Antibodies, Bispecific , Multiple Myeloma , Humans , Antibodies, Bispecific/therapeutic use , Epigenesis, Genetic , Immunotherapy/methods , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , T-Lymphocytes
6.
World J Surg Oncol ; 20(1): 382, 2022 Dec 04.
Article in English | MEDLINE | ID: mdl-36464675

ABSTRACT

BACKGROUND: CD97 is the most widely expressed G protein-coupled receptor in the epidermal growth factor seven-span transmembrane family. It plays a vital role in cell adhesion, migration, and cell connection regulation. We explored the role of CD97 in hepatocellular carcinoma (HCC). METHODS: We evaluated CD97 mRNA expression in HCC using TNMplot and the Gene Expression Omnibus database. The clinical prognostic significance of CD97 in HCC patients was evaluated by gene expression profiling interactive analysis, the Kaplan-Meier plotter, and the UALCAN database. The Tumor Immune Estimation Resource (TIMER) and CIBERSORT databases were used to analyze the relationships among CD97, genes positively related with CD97, and tumor-infiltrating immune cells. RESULTS: CD97 was highly expressed in HCC tissues and was associated with an adverse prognosis. CD97 and genes positively related with CD97 were positively correlated with the abundance of tumor-infiltrating immune cells and strongly correlated with tumor-infiltrating macrophages (all r ≥ 0.513, P < 0.001). CD97 was positively correlated with M2 macrophage and tumor-associated macrophage markers (both r ≥ 0.464, P < 0.001). CD97 was found to be an immune-related gene in HCC and positively correlated with the TOX, PD-L1, PD-L2, CTLA4, and PD-1 immune checkpoint genes. CD97 copy number alterations affect the level of immune cell infiltration and mRNA expression. CONCLUSIONS: CD97 can be used as a potential molecular marker of prognosis in HCC, which is associated with immune cell infiltration.


Subject(s)
Biomarkers, Tumor , Carcinoma, Hepatocellular , Liver Neoplasms , Receptors, G-Protein-Coupled , Humans , Biomarkers , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Tumor-Associated Macrophages/immunology , Databases, Factual
7.
N Engl J Med ; 387(24): 2232-2244, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36507686

ABSTRACT

BACKGROUND: G protein-coupled receptor, family C, group 5, member D (GPRC5D) is an orphan receptor expressed in malignant plasma cells. Talquetamab, a bispecific antibody against CD3 and GPRC5D, redirects T cells to mediate killing of GPRC5D-expressing myeloma cells. METHODS: In a phase 1 study, we evaluated talquetamab administered intravenously weekly or every other week (in doses from 0.5 to 180 µg per kilogram of body weight) or subcutaneously weekly, every other week, or monthly (5 to 1600 µg per kilogram) in patients who had heavily pretreated relapsed or refractory multiple myeloma that had progressed with established therapies (a median of six previous lines of therapy) or who could not receive these therapies without unacceptable side effects. The primary end points - the frequency and type of dose-limiting toxic effects (study part 1 only), adverse events, and laboratory abnormalities - were assessed in order to select the recommended doses for a phase 2 study. RESULTS: At the data-cutoff date, 232 patients had received talquetamab (102 intravenously and 130 subcutaneously). At the two subcutaneous doses recommended for a phase 2 study (405 µg per kilogram weekly [30 patients] and 800 µg per kilogram every other week [44 patients]), common adverse events were cytokine release syndrome (in 77% and 80% of the patients, respectively), skin-related events (in 67% and 70%), and dysgeusia (in 63% and 57%); all but one cytokine release syndrome event were of grade 1 or 2. One dose-limiting toxic effect of grade 3 rash was reported in a patient who had received talquetamab at the 800-µg dose level. At median follow-ups of 11.7 months (in patients who had received talquetamab at the 405-µg dose level) and 4.2 months (in those who had received it at the 800-µg dose level), the percentages of patients with a response were 70% (95% confidence interval [CI], 51 to 85) and 64% (95% CI, 48 to 78), respectively. The median duration of response was 10.2 months and 7.8 months, respectively. CONCLUSIONS: Cytokine release syndrome, skin-related events, and dysgeusia were common with talquetamab treatment but were primarily low-grade. Talquetamab induced a substantial response among patients with heavily pretreated relapsed or refractory multiple myeloma. (Funded by Janssen Research and Development; MonumenTAL-1 ClinicalTrials.gov number, NCT03399799.).


Subject(s)
Antibodies, Bispecific , CD3 Complex , Multiple Myeloma , Receptors, G-Protein-Coupled , T-Lymphocytes , Humans , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/immunology , Antibodies, Bispecific/therapeutic use , Cytokine Release Syndrome/chemically induced , Cytokine Release Syndrome/etiology , Dysgeusia/chemically induced , Dysgeusia/etiology , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Neoplasm Recurrence, Local/drug therapy , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , CD3 Complex/antagonists & inhibitors , CD3 Complex/immunology , Administration, Intravenous , Injections, Subcutaneous , Skin Diseases/chemically induced , Skin Diseases/etiology
8.
Mol Med Rep ; 26(3)2022 Sep.
Article in English | MEDLINE | ID: mdl-35856408

ABSTRACT

Sepsis serves as a leading cause of admission to and death of patients in the intensive care unit (ICU) and is described as a systemic inflammatory response syndrome caused by abnormal host response to infection. Adipose­derived mesenchymal stem cells (ADSCs) have exhibited reliable and promising clinical application potential in multiple disorders. However, the function and the mechanism of ADSCs in sepsis remain elusive. In the present study, the crucial inhibitory effect of ADSC­derived hydroxy­carboxylic acid receptor 1 (HCAR1) on sepsis was identified. Reverse transcription quantitative­PCR determined that the mRNA expression of HCAR1 was reduced while the mRNA expression of Toll­like receptor 4 (TLR4), major histocompatibility complex class II (MHC II), NOD­like receptor family pyrin domain containing 3 (NLRP3), and the levels of interleukin­1ß (IL­1ß), tumor necrosis factor­α (TNF­α), interleukin­10 (IL­10), and interleukin­18 (IL­18) were enhanced in the peripheral blood of patients with sepsis. The expression of HCAR1 was negatively correlated with TLR4 (r=­0.666), MHC II (r=­0.587), and NLRP3 (r=­0.621) expression and the expression of TLR4 was positively correlated with NLRP3 (r=0.641), IL­1ß (r=0.666), TNF­α (r=0.606), and IL­18 (r=0.624) levels in the samples. Receiver operating characteristic (ROC) curve analysis revealed that the area under the ROC curve (AUC) of HCAR1, TLR4, MHC II and NLRP3 mRNA expression was 0.830, 0.853, 0.735 and 0.945, respectively, in which NLRP3 exhibited the highest diagnostic value, and the AUC values of IL­1ß, IL­18, TNF­α, and IL­10 were 0.751, 0.841, 0.924 and 0.729, respectively, in which TNF­α exhibited the highest diagnostic value. A sepsis rat model was established by injecting lipopolysaccharide (LPS) and the rats were randomly divided into 5 groups, including a normal control group (NC group; n=6), a sepsis model group (LPS group; n=6), an ADSC transplantation group (L + M group; n=6), a combined HCAR1 receptor agonist group [L + HCAR1 inducer (Gi) + M group; n=6], and a combined HCAR1 receptor inhibitor group [L + HCAR1 blocker (Gk) + M group; n=6]. Hematoxylin and eosin staining determined that ADSCs attenuated the lung injury of septic rats and ADSC­derived HCAR1 enhanced the effect of ADSCs. The expression of HCAR1, TLR4, MHC II, NLRP3, IL­1ß, IL­18 and TNF­α levels were suppressed by ADSCs and the effect was further induced by ADSC­derived HCAR1. However, ADSC­derived HCAR1 induced the levels of anti­inflammatory factor IL­10. The negative correlation of HCAR1 expression with TLR4, MHC II, and NLRP3 expression in the peripheral blood and lung tissues of the rats was then identified. It is thus concluded that ADSC­derived HCAR1 regulates immune response in the attenuation of sepsis. ADSC­derived HCAR1 may be a promising therapeutic strategy for sepsis.


Subject(s)
Adipose Tissue , Mesenchymal Stem Cells , Receptors, G-Protein-Coupled , Sepsis , Adipose Tissue/cytology , Adipose Tissue/immunology , Animals , Immunity , Interleukin-10/immunology , Interleukin-18/immunology , Lipopolysaccharides/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , RNA, Messenger/metabolism , Rats , Receptors, G-Protein-Coupled/immunology , Sepsis/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
9.
Nat Commun ; 13(1): 4046, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35831277

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease affecting synovial joints where different CD4+ T cell subsets may contribute to pathology. Here, we perform single cell sequencing on synovial CD4+ T cells from anti-citrullinated protein antibodies (ACPA)+ and ACPA- RA patients and identify two peripheral helper T cell (TPH) states and a cytotoxic CD4+ T cell subset. We show that the adhesion G-protein coupled receptor 56 (GPR56) delineates synovial CXCL13high TPH CD4+ T cells expressing LAG-3 and the tissue-resident memory receptors CXCR6 and CD69. In ACPA- SF, TPH cells display lower levels of GPR56 and LAG-3. Further, most expanded T cell clones in the joint are within CXCL13high TPH CD4+ T cells. Finally, RNA-velocity analyses suggest a common differentiation pathway between the two TPH clusters and effector CD4+ T cells. Our study provides comprehensive immunoprofiling of the synovial CD4+ T cell subsets in ACPA+ and ACPA- RA.


Subject(s)
Arthritis, Rheumatoid , Receptors, G-Protein-Coupled , T-Lymphocytes, Helper-Inducer , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Humans , Joints/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology
10.
Cancer Discov ; 12(8): OF3, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35748592

ABSTRACT

Patients with refractory or relapsed multiple myeloma usually develop resistance to the two approved BCMA-targeting chimeric antigen receptor (CAR) T cells. A preliminary study of a new CAR T-cell therapy that zeroes in on the GPRC5D protein on multiple myeloma cells suggests that this approach is safe and effective. All 10 patients treated with the GPRC5D-targeting cells showed responses.


Subject(s)
Drug Resistance, Neoplasm , Immunotherapy, Adoptive , Multiple Myeloma , Receptors, Chimeric Antigen , Receptors, G-Protein-Coupled , B-Cell Maturation Antigen/antagonists & inhibitors , B-Cell Maturation Antigen/immunology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Humans , Immunotherapy, Adoptive/methods , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/therapeutic use
11.
EBioMedicine ; 80: 104060, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35588628

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) has complex genetic and environmental aspects, and free fatty acid receptors (FFARs) may bridge genetic and dietary aspects. FFAR4 is highly expressed in the intestine and acts primarily as the receptor of long-chain fatty acids, which are major components of the human diet. It is unclear what role, if any, FFAR4 may play in IBD. METHODS: Mouse and human colitis samples, mice with complete FFAR4 knockout, intestine-specific FFAR4 knockout and FFAR4 overexpression and cell culture were used. RNA-sequencing analysis and flow cytometry were performed to examine the mechanisms. FINDINGS: The results showed that FFAR4 expression was upregulated in colitis tissues and that the loss of intestinal FFAR4 ameliorated colitis, whereas intestinal FFAR4 overexpression exacerbated the disease. We identified intestinal epithelial cell deletion of FFAR4 by upregulating ZBED6, which in turn induced L33 transcription, and L33 elevated Treg cell numbers, ameliorating colitis. INTERPRETATION: FFAR4 deletion attenuates colitis by modulating Treg cells via the ZBED6-IL33 pathway. FUNDING: National Natural Science Foundation of China, Innovation and Application Project of Medical and Public Health Technology of Wuxi Science and Technology, Fundamental Research Funds for the Central Universities and the Fund of Wuxi Healthcare Commission.


Subject(s)
Colitis , Interleukin-33 , Receptors, G-Protein-Coupled , Repressor Proteins , T-Lymphocytes, Regulatory , Animals , Colitis/immunology , Colitis/metabolism , Humans , Interleukin-33/immunology , Interleukin-33/metabolism , Mice , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Repressor Proteins/immunology , Repressor Proteins/metabolism , T-Lymphocytes, Regulatory/immunology
12.
FASEB J ; 36(5): e22322, 2022 05.
Article in English | MEDLINE | ID: mdl-35429062

ABSTRACT

Emerging evidence suggests that signaling through the C3a anaphylatoxin receptor (C3aR) protects against various inflammation-related diseases. However, the role of C3aR in psoriasis remains unknown. The purpose of this study was to investigate the possible protective role of C3aR in psoriasis and to explore the underlying molecular mechanisms. We initially found that the psoriatic epidermis exhibited significantly decreased C3aR expression. C3aR showed protective roles in mouse models of imiquimod (IMQ)- and interleukin-23-induced psoriasis. Furthermore, increased epidermal thickness and keratin 6 (K6), K16, and K17 expression occurred in the ears and backs of C3aR-/- mice. Pharmacological treatment with a C3aR agonist ameliorated IMQ-induced psoriasiform lesions in mice and decreased the expression of K6, K16, and K17. Additionally, the signal transducer and activator of transcription 3 (STAT3) pathway participated in the protective function of C3aR. More importantly, the expression levels of K6, K16, and K17 in keratinocytes were all restored in HaCaT cells transfected with a C3aR-overexpression plasmid after treating them with colivelin (a STAT3 activator). Our findings demonstrate that C3aR protects against the development of psoriasis and suggest that C3aR confers protection by negatively regulating K6, K16, and K17 expression in a STAT3-dependent manner, thus inhibiting keratinocyte proliferation and helping reverse the pathogenesis of psoriasis.


Subject(s)
Keratinocytes , Keratins , Psoriasis , Receptors, G-Protein-Coupled , Anaphylatoxins , Animals , Cell Proliferation , Disease Models, Animal , Keratin-16/immunology , Keratin-17/immunology , Keratin-6/immunology , Keratinocytes/metabolism , Keratinocytes/pathology , Keratins/immunology , Mice , Psoriasis/drug therapy , Psoriasis/immunology , Psoriasis/pathology , Receptors, G-Protein-Coupled/immunology , Skin/metabolism
13.
Nat Commun ; 13(1): 1220, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35264564

ABSTRACT

COVID-19 shares the feature of autoantibody production with systemic autoimmune diseases. In order to understand the role of these immune globulins in the pathogenesis of the disease, it is important to explore the autoantibody spectra. Here we show, by a cross-sectional study of 246 individuals, that autoantibodies targeting G protein-coupled receptors (GPCR) and RAS-related molecules associate with the clinical severity of COVID-19. Patients with moderate and severe disease are characterized by higher autoantibody levels than healthy controls and those with mild COVID-19 disease. Among the anti-GPCR autoantibodies, machine learning classification identifies the chemokine receptor CXCR3 and the RAS-related molecule AGTR1 as targets for antibodies with the strongest association to disease severity. Besides antibody levels, autoantibody network signatures are also changing in patients with intermediate or high disease severity. Although our current and previous studies identify anti-GPCR antibodies as natural components of human biology, their production is deregulated in COVID-19 and their level and pattern alterations might predict COVID-19 disease severity.


Subject(s)
Autoantibodies/immunology , COVID-19/immunology , Receptors, G-Protein-Coupled/immunology , Renin-Angiotensin System/immunology , Autoantibodies/blood , Autoimmunity , Biomarkers/blood , COVID-19/blood , COVID-19/classification , Cross-Sectional Studies , Female , Humans , Machine Learning , Male , Multivariate Analysis , Receptor, Angiotensin, Type 1/immunology , Receptors, CXCR3/immunology , SARS-CoV-2 , Severity of Illness Index
14.
Nat Commun ; 13(1): 97, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013216

ABSTRACT

For many solid tumors, immune checkpoint blockade therapy has become first line treatment, yet a large proportion of patients with immunologically cold tumors do not benefit due to the paucity of tumor infiltrating lymphocytes. Here we show that the orphan G Protein-Coupled Receptor 182 (GPR182) contributes to immunotherapy resistance in cancer via scavenging chemokines that are important for lymphocyte recruitment to tumors. GPR182 is primarily upregulated in melanoma-associated lymphatic endothelial cells (LECs) during tumorigenesis, and this atypical chemokine receptor endocytoses chemokines promiscuously. In GPR182-deficient mice, T cell infiltration into transplanted melanomas increases, leading to enhanced effector T cell function and improved antitumor immunity. Ablation of GPR182 leads to increased intratumoral concentrations of multiple chemokines and thereby sensitizes poorly immunogenic tumors to immune checkpoint blockade and adoptive cellular therapies. CXCR3 blockade reverses the improved antitumor immunity and T cell infiltration characteristic of GPR182-deficient mice. Our study thus identifies GPR182 as an upstream regulator of the CXCL9/CXCL10/CXCR3 axis that limits antitumor immunity and as a potential therapeutic target in immunologically cold tumors.


Subject(s)
Chemokine CXCL10/genetics , Chemokine CXCL9/genetics , Melanoma, Experimental/genetics , Melanoma/genetics , Receptors, CXCR3/genetics , Receptors, G-Protein-Coupled/genetics , Skin Neoplasms/genetics , Animals , Cell Movement , Chemokine CXCL10/immunology , Chemokine CXCL9/immunology , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Melanoma/mortality , Melanoma/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/mortality , Melanoma, Experimental/therapy , Mice , Mice, Knockout , Protein Binding , Receptors, CXCR3/immunology , Receptors, G-Protein-Coupled/immunology , Signal Transduction , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Skin Neoplasms/therapy , Survival Analysis , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation , Tumor Burden , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
15.
J Clin Invest ; 132(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-34784295

ABSTRACT

Bitter taste receptors (taste 2 receptors, TAS2Rs) serve as warning sensors in the lingual system against the ingestion of potentially poisonous food. Here, we investigated the functional role of TAS2Rs in the human gut and focused on their potential to trigger an additional host defense pathway in the intestine. Human jejunal crypts, especially those from individuals with obesity, responded to bitter agonists by inducing the release of antimicrobial peptides (α-defensin 5 and regenerating islet-derived protein 3 α [REG3A]) but also regulated the expression of other innate immune factors (mucins, chemokines) that affected E. coli growth. We found that the effect of aloin on E. coli growth and on the release of the mucus glycoprotein CLCA1, identified via proteomics, was affected by TAS2R43 deletion polymorphisms and thus confirmed a role for TAS2R43. RNA-Seq revealed that denatonium benzoate induced an NRF2-mediated nutrient stress response and an unfolded protein response that increased the expression of the mitokine GDF15 but also ADM2 and LDLR, genes that are involved in anorectic signaling and lipid homeostasis. In conclusion, TAS2Rs in the intestine constitute a promising target for treating diseases that involve disturbances in the innate immune system and body weight control. TAS2R polymorphisms may be valuable genetic markers to predict therapeutic responses.


Subject(s)
Immunity, Innate , Intestinal Mucosa/immunology , Obesity/immunology , Receptors, G-Protein-Coupled/immunology , Growth Differentiation Factor 15/immunology , Humans , Male , Middle Aged , Pancreatitis-Associated Proteins/immunology , Peptide Hormones/immunology , RNA-Seq , Receptors, LDL/immunology
16.
Int Immunopharmacol ; 102: 108389, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34920312

ABSTRACT

The G protein-coupled receptor MrgprX2 in mast cells is known to be a crucial receptor for pseudo-allergic reactions. MrgprX2 activation leads to elevated intracellular calcium levels and mast cell degranulation, but the underlying mechanism remains to be elucidated. Herein, we investigated the role of the phosphatidylinositol 3 kinase (PI3K)/serum-threonine kinase (AKT) signaling pathway and phospholipase C gamma (PLCγ) in mast cell degranulation mediated by MrgprX2 in LAD2 human-derived mast cells. The results showed that phosphorylated AKT (p-AKT) and PLCγ up-regulation were accompanied by an increase in intracellular calcium following activation of MrgprX2 by Compound 48/80, an inducer of mast cell degranulation. In contrast, p-AKT and PLCγ were down-regulated and intracellular calcium levels decreased after MrgprX2 knockdown. Mast cell degranulation was clearly suppressed; however, inhibiting PI3K and PLCγ phosphorylation did not influence MrgprX2 expression. The increase in calcium concentration was suppressed and mast cell degranulation was weakened. Furthermore, by inhibiting PI3K and PLCγ phosphorylation in animals, the allergic symptoms caused by C48/80 were obviously reduced. We deduced that during the mast cell degranulation observed in pseudoallergic reactions, MrgprX2 regulated intracellular calcium levels via the PI3K/AKT and PLCγ pathways.


Subject(s)
Hypersensitivity/immunology , Mast Cells/immunology , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Animals , Calcium/metabolism , Cell Degranulation , Cell Line , Humans , Hypersensitivity/metabolism , Mast Cells/metabolism , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Phosphatidylinositol 3-Kinase/immunology , Phospholipase C gamma/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics , Signal Transduction
17.
Front Immunol ; 12: 688930, 2021.
Article in English | MEDLINE | ID: mdl-34867939

ABSTRACT

Acute anaphylaxis to small molecule drugs is largely considered to be antibody-mediated with immunogloblin E (IgE) and mast cell activation being key. More recently, a role for drug-reactive immunoglobulin G (IgG) with neutrophil activation has also been suggested, at least in reactions to neuromuscular blocking agents (NMBAs). However, the mast cell receptor MRGPRX2 has also been highlighted as a possible triggering mechanism in acute anaphylaxis to many clinically used drugs. Significantly, MRGPRX2 activation is not dependent upon the presence of drug-recognising antibody. Given the reasonable assumption that MRGPRX2 is expressed in all individuals, the corollary of this is that in theory, anybody could respond detrimentally to triggering drugs (recently suggested to be around 20% of a drug-like compound library). But this clearly is not the case, as the incidence of acute drug-induced anaphylaxis is very low. In this mini-review we consider antibody-dependent and -independent mechanisms of mast cell activation by small molecule drugs with a focus on the MRGPRX2 pathway. Moreover, as a juxtaposition to these adverse drug actions, we consider how increased understanding of the role of MRGPRX2 in anaphylaxis is important for future drug development and can complement exploration of this receptor as a drug target in broader clinical settings.


Subject(s)
Anaphylaxis/immunology , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Anaphylaxis/etiology , Anaphylaxis/therapy , Drug Hypersensitivity/etiology , Drug Hypersensitivity/immunology , Drug Hypersensitivity/therapy , Drug-Related Side Effects and Adverse Reactions/etiology , Drug-Related Side Effects and Adverse Reactions/immunology , Drug-Related Side Effects and Adverse Reactions/therapy , Gene Expression , Humans , Mast Cell Activation Disorders/etiology , Mast Cell Activation Disorders/immunology , Mast Cell Activation Disorders/therapy , Mast Cells/drug effects , Mast Cells/immunology , Models, Immunological , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics
18.
Bull Cancer ; 108(10S): S205-S212, 2021 Oct.
Article in French | MEDLINE | ID: mdl-34920804

ABSTRACT

Immunotherapies have recently emerged as potential game changers in the treatment of multiple myeloma (MM). Those include monoclonal antibodies (targeting CD38 or CS1), bispecific antibodies (BsAb, mainly targeting BCMA, GPRC5D or FcRH5), antibody-drug conjugate (mainly targeting BCMA) and CAR-T cells (mainly targeting BCMA). BsAb have the capacity to bind two different antigens, one at the tumor cell surface and one on T cells (CD3), recreating the immune synapse. In this article, we discuss the main clinical data on BsAb in MM, as well as their different constructs and the potential mechanism of resistance.


Subject(s)
Antibodies, Bispecific/therapeutic use , Immunotherapy/methods , Multiple Myeloma/therapy , ADP-ribosyl Cyclase 1/immunology , Antibodies, Bispecific/immunology , Antigens, Neoplasm/immunology , Drug Resistance, Neoplasm/immunology , Humans , Multiple Myeloma/immunology , Receptors, Fc/immunology , Receptors, G-Protein-Coupled/immunology , Signaling Lymphocytic Activation Molecule Family/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...