Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cancer Immunol Res ; 12(5): 592-613, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38393969

RESUMO

Solid tumors are dense three-dimensional (3D) multicellular structures that enable efficient receptor-ligand trans interactions via close cell-cell contact. Immunoglobulin-like transcript (ILT)2 and ILT4 are related immune-suppressive receptors that play a role in the inhibition of myeloid cells within the tumor microenvironment. The relative contribution of ILT2 and ILT4 to immune inhibition in the context of solid tumor tissue has not been fully explored. We present evidence that both ILT2 and ILT4 contribute to myeloid inhibition. We found that although ILT2 inhibits myeloid cell activation in the context of trans-engagement by MHC-I, ILT4 efficiently inhibits myeloid cells in the presence of either cis- or trans-engagement. In a 3D spheroid tumor model, dual ILT2/ILT4 blockade was required for the optimal activation of myeloid cells, including the secretion of CXCL9 and CCL5, upregulation of CD86 on dendritic cells, and downregulation of CD163 on macrophages. Humanized mouse tumor models showed increased immune activation and cytolytic T-cell activity with combined ILT2 and ILT4 blockade, including evidence of the generation of immune niches, which have been shown to correlate with clinical response to immune-checkpoint blockade. In a human tumor explant histoculture system, dual ILT2/ILT4 blockade increased CXCL9 secretion, downregulated CD163 expression, and increased the expression of M1 macrophage, IFNγ, and cytolytic T-cell gene signatures. Thus, we have revealed distinct contributions of ILT2 and ILT4 to myeloid cell biology and provide proof-of-concept data supporting the combined blockade of ILT2 and ILT4 to therapeutically induce optimal myeloid cell reprogramming in the tumor microenvironment.


Assuntos
Antígenos CD , Receptor B1 de Leucócitos Semelhante a Imunoglobulina , Glicoproteínas de Membrana , Células Mieloides , Receptores Imunológicos , Microambiente Tumoral , Receptores Imunológicos/metabolismo , Animais , Humanos , Camundongos , Microambiente Tumoral/imunologia , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Células Mieloides/imunologia , Células Mieloides/metabolismo , Glicoproteínas de Membrana/metabolismo , Linhagem Celular Tumoral , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo
2.
Cell Rep ; 42(12): 113516, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38048225

RESUMO

The immune checkpoint NKG2A/CD94 is a promising target for cancer immunotherapy, and its ligand major histocompatibility complex E (MHC-E) is frequently upregulated in cancer. NKG2A/CD94-mediated inhibition of lymphocytes depends on the presence of specific leader peptides in MHC-E, but when and where they are presented in situ is unknown. We apply a nanobody specific for the Qdm/Qa-1b complex, the NKG2A/CD94 ligand in mouse, and find that presentation of Qdm peptide depends on every member of the endoplasmic reticulum-resident peptide loading complex. With a turnover rate of 30 min, the Qdm peptide reflects antigen processing capacity in real time. Remarkably, Qdm/Qa-1b complexes require inflammatory signals for surface expression in situ, despite the broad presence of Qa-1b molecules in homeostasis. Furthermore, we identify LILRB1 as a functional inhibition receptor for MHC-E in steady state. These data provide a molecular understanding of NKG2A blockade in immunotherapy and assign MHC-E as a convergent ligand for multiple immune checkpoints.


Assuntos
Antígenos de Histocompatibilidade Classe I , Neoplasias , Camundongos , Animais , Antígenos de Histocompatibilidade Classe I/metabolismo , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Células Matadoras Naturais , Ligantes , Peptídeos/metabolismo , Neoplasias/metabolismo , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo
3.
Front Immunol ; 14: 1240275, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37781391

RESUMO

Immune checkpoint blockade is a compelling approach in tumor immunotherapy. Blocking inhibitory pathways in T cells has demonstrated clinical efficacy in different types of cancer and may hold potential to also stimulate innate immune responses. A novel emerging potential target for immune checkpoint therapy is leukocyte immunoglobulin-like receptor subfamily B member 1 (LILRB1). LILRB1 belongs to the superfamily of leukocyte immunoglobulin-like receptors and exerts inhibitory functions. The receptor is expressed by a variety of immune cells including macrophages as well as certain cytotoxic lymphocytes and contributes to the regulation of different immune responses by interaction with classical as well as non-classical human leukocyte antigen (HLA) class I molecules. LILRB1 has gained increasing attention as it has been demonstrated to function as a phagocytosis checkpoint on macrophages by recognizing HLA class I, which represents a 'Don't Eat Me!' signal that impairs phagocytic uptake of cancer cells, similar to CD47. The specific blockade of the HLA class I:LILRB1 axis may provide an option to promote phagocytosis by macrophages and also to enhance cytotoxic functions of T cells and natural killer (NK) cells. Currently, LILRB1 specific antibodies are in different stages of pre-clinical and clinical development. In this review, we introduce LILRB1 and highlight the features that make this immune checkpoint a promising target for cancer immunotherapy.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Macrófagos , Antígenos de Histocompatibilidade Classe I , Células Matadoras Naturais , Imunoglobulinas/metabolismo , Antígenos CD/metabolismo
4.
Front Immunol ; 13: 929339, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36389667

RESUMO

Antibody-dependent cellular phagocytosis (ADCP) by macrophages, an important effector function of tumor targeting antibodies, is hampered by 'Don´t Eat Me!' signals such as CD47 expressed by cancer cells. Yet, human leukocyte antigen (HLA) class I expression may also impair ADCP by engaging leukocyte immunoglobulin-like receptor subfamily B (LILRB) member 1 (LILRB1) or LILRB2. Analysis of different lymphoma cell lines revealed that the ratio of CD20 to HLA class I cell surface molecules determined the sensitivity to ADCP by the combination of rituximab and an Fc-silent variant of the CD47 antibody magrolimab (CD47-IgGσ). To boost ADCP, Fc-silent antibodies against LILRB1 and LILRB2 were generated (LILRB1-IgGσ and LILRB2-IgGσ, respectively). While LILRB2-IgGσ was not effective, LILRB1-IgGσ significantly enhanced ADCP of lymphoma cell lines when combined with both rituximab and CD47-IgGσ. LILRB1-IgGσ promoted serial engulfment of lymphoma cells and potentiated ADCP by non-polarized M0 as well as polarized M1 and M2 macrophages, but required CD47 co-blockade and the presence of the CD20 antibody. Importantly, complementing rituximab and CD47-IgGσ, LILRB1-IgGσ increased ADCP of chronic lymphocytic leukemia (CLL) or lymphoma cells isolated from patients. Thus, dual checkpoint blockade of CD47 and LILRB1 may be promising to improve antibody therapy of CLL and lymphomas through enhancing ADCP by macrophages.


Assuntos
Antígeno CD47 , Leucemia Linfocítica Crônica de Células B , Humanos , Antígeno CD47/metabolismo , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Rituximab/farmacologia , Rituximab/uso terapêutico , Rituximab/metabolismo , Leucemia Linfocítica Crônica de Células B/metabolismo , Linhagem Celular Tumoral , Fagocitose , Macrófagos , Anticorpos/metabolismo , Antígenos CD/metabolismo
5.
Sci Rep ; 12(1): 15420, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-36104364

RESUMO

Inborn errors of immunity are known to cause not only immunodeficiencies and allergies but also autoimmunity. Leukocyte immunoglobulin-like receptor B1 (LILRB1) is a receptor on leukocytes playing a role in regulating immune responses. No phenotypes have been reported to be caused by germline mutations in LILRB1. We aimed to identify the causative variant in a three-generation family with nine members suffering from one of the three autoimmune diseases-Graves' disease, Hashimoto's thyroiditis, or systemic lupus erythematosus. Whole-genome linkage study revealed a locus on chromosome 19q13.4 with the maximum LOD score of 2.71. Whole-exome sequencing identified a heterozygous missense variant, c.479G > A (p. G160E) in LILRB1, located within the chromosomal-linked region, in all nine affected members. The variant has never been previously reported. Jurkat cells transfected with the mutant LILRB1, compared with those with the wild-type LILRB1, showed decreased phosphorylation of both LILRB1 and its downstream protein, SHP-1. Flow cytometry was used to study immunophenotype and revealed that LILRB1 was significantly lower on the surface of activated regulatory T lymphocytes (Treg) cells of patients. Single-cell RNA sequencing showed substantially increased M1-like monocytes in peripheral blood mononuclear cells of affected individuals. This study, for the first time, implicates LILRB1 as a new disease gene for autoimmunity.


Assuntos
Doença de Graves , Leucócitos Mononucleares , Antígenos CD/genética , Humanos , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Leucócitos Mononucleares/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Sequenciamento do Exoma
6.
Am J Reprod Immunol ; 88(5): e13612, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36004818

RESUMO

PROBLEM: NK cells are important for healthy pregnancy and aberrant phenotypes or effector functions have been associated with RPL. We compared expression of a broad panel of NK cell receptors, including immune checkpoint receptors, and investigated their clinical association with RPL as this might improve patient stratification and prediction of RPL. METHOD OF STUDY: Peripheral blood mononuclear cells were isolated from 52 women with RPL and from 2 women with an uncomplicated pregnancy for flowcytometric analysis and plasma was used to determine anti-CMV IgG antibodies. RESULTS: Between RPL and controls, we observed no difference in frequencies of T-, NKT or NK cells, in CD56dimCD16+ or CD56brightCD16- NK cell subsets or in the expression of KIRs, NKG2A, NKG2C, NKG2D, NKp30, NKp44, NKp46 or DNAM1. NK cells from women with RPL had a higher expression of LILRB1 and TACTILE and this was associated with the number of losses. The immune checkpoint receptors PD1, TIM3 and LAG3 were not expressed on peripheral blood NK cells. In RPL patients, there was a large variation in NKG2C expression and higher levels could be explained by CMV seropositivity. CONCLUSION: Our study identified LILRB1 and TACTILE as NK cell receptors associated with RPL. Moreover, we provide first support for the potential role of CMV in RPL via its impact on the NK cell compartment. Thereby our study could guide future studies to confirm the clinical association of LILRB1, TACTILE and NKG2C with RPL in a larger cohort and to explore their functional relevance in reproductive success.


Assuntos
Aborto Habitual , Antígenos CD , Receptor B1 de Leucócitos Semelhante a Imunoglobulina , Leucócitos Mononucleares , Feminino , Humanos , Gravidez , Antígenos CD/metabolismo , Células Matadoras Naturais , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Receptores de Células Matadoras Naturais
7.
World J Surg Oncol ; 20(1): 92, 2022 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-35321724

RESUMO

BACKGROUND: Leukocyte immunoglobulin-like receptor subfamily B (LILRB), including 5 subtypes, is a group of inhibitory receptors in the immune system. The LILRB family is known to be involved in the tumor progression of various cancer types, especially liver cancer. However, the expression patterns and prognostic values of LILRB family members in liver cancer tissues remain unclear. METHODS: We used the Oncomine database, GEPIA database, Kaplan-Meier Plotter, Timer, and TISIDB to assess the expression and prognostic value of the LILRB family in liver cancer patients. We also verified the expression of the LILRB family in tumor tissues and tumor-free liver tissues at the protein level by using immunohistochemistry. The STRING website was used to explore the interaction between the LILRB family and their related genes. The DAVID database was used to perform the gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Flow cytometry was used to assess the infiltrated NK cells in liver cancer tissues. RESULTS: Our study revealed that the mRNA expression of LILRB1, LILRB2, LILRB3, and LILRB5 was downregulated, while compared with normal tissues, the mRNA expression of LILRB4 was upregulated in liver cancer tissues. Survival analysis revealed that LILRB2 and LILRB5 mRNA expression levels were significantly positively associated with overall survival (OS) and disease-free survival (DSS) and that the mRNA expression of all LILRB family members was significantly positively correlated with recurrence-free survival (RFS) and progression-free survival (PFS). Next, we further found that the mRNA expression of all LILRB family members was significantly associated with the infiltration of B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells in liver cancer. Finally, GO and KEGG analyses found that the LILRB family and its related genes were involved in antigen processing and presentation and natural killer cell-mediated cytotoxicity pathways. CONCLUSIONS: Our study suggested that LILRB family expression was associated with the prognosis of liver cancer patients and infiltrated immune cells. The LILRB family might be involved in antigen processing and presentation and natural killer cell-mediated cytotoxicity pathways.


Assuntos
Antígenos CD , Linfócitos T CD8-Positivos , Neoplasias Hepáticas , Receptores Imunológicos , Antígenos CD/genética , Antígenos CD/metabolismo , Humanos , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/genética , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/imunologia , Glicoproteínas de Membrana/genética , Prognóstico , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo
8.
Ann Hematol ; 101(4): 739-748, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35041051

RESUMO

Acquired aplastic anemia (AA) is an autoimmune disease characterized by hematopoietic stem and progenitor cell destruction in bone marrow. The non-classic human leukocyte class I antigen (HLA-) G interacts with multiple cell subsets, such as T cells and B cells. HLA-G exerts powerful immune suppression by binding with its receptors, immunoglobulin-like transcripts (ILTs). Here, we compared 46 AA patients and 28 healthy controls. Soluble HLA-G levels in bone marrow supernatants from AA patients were higher than controls. The proportion of bone marrow B cells was decreased and the ILT2-expressing cells among CD19+ cells were increased in AA patients. In addition, the percentage of mature B cells among marrow B cells was increased in AA patient, while the percentage of pro-B plus pre-B cells was decreased. More immature B cells and pro-B plus pre-B cells expressed ILT2 in AA patients than in controls, while mature B cells expressing ILT2 did not differ significantly. Functional studies demonstrated that high-level soluble HLA-G inhibited bone marrow B cell proliferation by interacting with ILT2 in AA, and was blocked by anti-HLA-G and anti-ILT2 monoclonal antibodies. Together, these results suggest that the abnormal decrease of pro-B plus pre-B cells in AA patients was related to the enhanced suppression by the excess HLA-G and ILT2 proteins. Therapeutic blockade of the HLA-G-ILT2 interaction may help to normalize bone marrow B cell proliferation.


Assuntos
Anemia Aplástica , Antígenos CD/metabolismo , Antígenos HLA-G , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Medula Óssea/metabolismo , Proliferação de Células , Humanos
9.
J Am Soc Nephrol ; 33(1): 121-137, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34725108

RESUMO

BACKGROUND: The reported association of mTOR-inhibitor (mTORi) treatment with a lower incidence of cytomegalovirus (CMV) infection in kidney transplant recipients (KTR) who are CMV seropositive (R+) remains unexplained. METHODS: The incidence of CMV infection and T-cell profile was compared between KTRs treated with mTORis and mycophenolic acid (MPA), and in vitro mTORi effects on T-cell phenotype and functions were analyzed. RESULTS: In KTRs who were R+ and treated with MPA, both αß and γδ T cells displayed a more dysfunctional phenotype (PD-1+, CD85j+) at day 0 of transplantation in the 16 KTRs with severe CMV infection, as compared with the 17 KTRs without or with spontaneously resolving CMV infection. In patients treated with mTORis (n=27), the proportion of PD-1+ and CD85j+ αß and γδ T cells decreased, when compared with patients treated with MPA (n=44), as did the frequency and severity of CMV infections. mTORi treatment also led to higher proportions of late-differentiated and cytotoxic γδ T cells and IFNγ-producing and cytotoxic αß T cells. In vitro, mTORis increased proliferation, viability, and CMV-induced IFNγ production of T cells and decreased PD-1 and CD85j expression in T cells, which shifted the T cells to a more efficient EOMESlow Hobithigh profile. In γδ T cells, the mTORi effect was related to increased TCR signaling. CONCLUSION: Severe CMV replication is associated with a dysfunctional T-cell profile and mTORis improve T-cell fitness along with better control of CMV. A dysfunctional T-cell phenotype could serve as a new biomarker to predict post-transplantation infection and to stratify patients who should benefit from mTORi treatment. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER: Proportion of CMV Seropositive Kidney Transplant Recipients Who Will Develop a CMV Infection When Treated With an Immunosuppressive Regimen Including Everolimus and Reduced Dose of Cyclosporine Versus an Immunosuppressive Regimen With Mycophenolic Acid and Standard Dose of Cyclosporine A (EVERCMV), NCT02328963.


Assuntos
Infecções por Citomegalovirus/prevenção & controle , Transplante de Rim/efeitos adversos , Inibidores de MTOR/uso terapêutico , Subpopulações de Linfócitos T/efeitos dos fármacos , Idoso , Antibacterianos/uso terapêutico , Antígenos CD/metabolismo , Técnicas de Cultura de Células , Infecções por Citomegalovirus/epidemiologia , Infecções por Citomegalovirus/patologia , Feminino , Humanos , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Masculino , Pessoa de Meia-Idade , Ácido Micofenólico/uso terapêutico , Receptor de Morte Celular Programada 1/metabolismo , Subpopulações de Linfócitos T/metabolismo
10.
Front Immunol ; 12: 728685, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34659215

RESUMO

Mucosal-associated invariant T (MAIT) cells are an innate-like population of T cells that display a TCR Vα7.2+ CD161+ phenotype and are restricted by the nonclassical MHC-related molecule 1 (MR1). Although B cells control MAIT cell development and function, little is known about the mechanisms underlying their interaction(s). Here, we report, for the first time, that during Salmonella enterica serovar Typhi (S. Typhi) infection, HLA-G expression on B cells downregulates IFN-γ production by MAIT cells. In contrast, blocking HLA-G expression on S. Typhi-infected B cells increases IFN-γ production by MAIT cells. After interacting with MAIT cells, kinetic studies show that B cells upregulate HLA-G expression and downregulate the inhibitory HLA-G receptor CD85j on MAIT cells resulting in their loss. These results provide a new role for HLA-G as a negative feedback loop by which B cells control MAIT cell responses to antigens.


Assuntos
Antígenos CD/metabolismo , Linfócitos B/metabolismo , Antígenos HLA-G/metabolismo , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Células T Invariantes Associadas à Mucosa/metabolismo , Salmonella typhi/patogenicidade , Febre Tifoide/metabolismo , Adulto , Antígenos CD/genética , Linfócitos B/imunologia , Linfócitos B/microbiologia , Células Cultivadas , Técnicas de Cocultura , Feminino , Interações Hospedeiro-Patógeno , Humanos , Interferon gama/genética , Interferon gama/metabolismo , Cinética , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/genética , Masculino , Pessoa de Meia-Idade , Células T Invariantes Associadas à Mucosa/imunologia , Células T Invariantes Associadas à Mucosa/microbiologia , Fenótipo , Salmonella typhi/imunologia , Transdução de Sinais , Febre Tifoide/genética , Febre Tifoide/imunologia , Febre Tifoide/microbiologia , Adulto Jovem
11.
Mol Immunol ; 136: 82-97, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34098344

RESUMO

BACKGROUND: Leukocyte immunoglobulin-like receptor subfamily B (LILRB) is a group of inhibitory receptors involved in innate immune mainly expressed on lymphoid and myelomonocytic cells. LILRB is proposed to serve as immune checkpoint like PD-1 and CTLA-4 for tumor treatment. We recently reported that the expression of LILRB2 in CD1c+ mDC from tumor tissue might suppress immune for HCC patients. However, the expression of all the LILRB family on other immune cells in peripheral blood and tumor microenvironment of HCC patients has not been systematically studied. METHODS: The expression of LILRB family (LILRB1, LILRB2, LILRB3, LILRB4 and LILRB5) on immune cells, including granulocytes, NK cells, NKT cells, monocyte subsets, TAMs, B cells, γδ T cells, CD4+ T cells, CD8+ T cells and MDSC subsets, was analyzed by flow cytometry in the peripheral blood of 20 HCC patients and 20 healthy donors as well as in the tumor and tumor free tissues of 10 HCC patients. RESULTS: LILRB1, LILRB2 and LILRB3 in granulocytes from peripheral blood were expressed increased in HCC patients compared with healthy donors. The expression of LILRB5 in NK cells and NKT cells from HCC blood were higher compared with healthy donors` blood. CD14+CD16+ monocyte subsets in blood of HCC patients expressed increased LILRB1 and LILRB4 than that in healthy donors. CD14+CD16- monocyte subsets in blood of HCC patients expressed increased LILRB3 than that in healthy donors. Compared to corresponding TFL, LILRB3, LILRB4 and LILRB5 were expressed enhanced in TAMs from HCC tumors. LILRB1 expressed on the B cells both in the blood and tumor had significantly increased compared with healthy donors or corresponding TFL. Different from peripheral blood, in the HCC microenvironment, CD4+ T cells expressed lower LILRB2, LILRB3 and LILRB4 than that from TFL and CD8+ T cells expressed decreased LILRB2. And γδ T cells expressed LILRB1 in HCC blood and microenvironment. Surprisingly, the percentage of LILRB1 expressed on MDSC from HCC peripheral blood and tumors was lower than that from healthy donors and corresponding TFL. CONCLUSIONS: This is the first systemically examination of the LILRB family expression on a variety of immune cells from both peripheral blood and microenvironment in HCC patients. The specific increasing expression of LILRB on immune cells may regulate innate and adaptive immune and impact on HCC progression. Our findings justify further investigation of LILRB function in HCC.


Assuntos
Carcinoma Hepatocelular/metabolismo , Granulócitos/metabolismo , Neoplasias Hepáticas/metabolismo , Linfócitos/metabolismo , Receptores Imunológicos/metabolismo , Idoso , Antígenos CD/metabolismo , Feminino , Citometria de Fluxo , Humanos , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Pessoa de Meia-Idade
12.
Front Immunol ; 12: 679090, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34054869

RESUMO

Immune checkpoint inhibitors (ICIs) have become a promising area of research for cancer treatment. In addition to the well-known ICIs targeting PD-1/PD-L1, HLA-G/ILT-2/-4 is relatively new immune checkpoint that has been evaluated in early clinical trials in patients with advanced solid tumors. In this study, the expression of HLA-G (n=157), ILT-2/4 (n=82), and PD-L1 (n=70) in epithelial cell adhesion molecule (EpCAM)-positive colorectal cancer (CRC) cells was analyzed by multicolor flow cytometry, and the prognostic significance of these molecules was evaluated. In EpCAM+ CRC cells, the median percentages of HLA-G, ILT-2, ILT-4, and PD-L1 were 14.90%, 67.70%, 8.55% and 80.30%, respectively. In addition, a positive correlation was observed between them (all p<0.001). Higher levels of these immune checkpoint proteins are associated with lymph node metastasis. In addition to the AJCC stage (p=0.001), Kaplan-Meier survival analysis showed that higher levels of HLA-G (p=0.041), ILT-2 (p=0.060), ILT-4 (p<0.001), PD-L1 (p=0.012), HLA-GILT4 (p<0.001) and ILT-2ILT-4 (p<0.001) were significantly associated with shorter survival of CRC patients. When CRC patients were stratified by early and advanced AJCC stages, HLA-G levels were only related to the survival among CRC patients with early disease stage (p=0.024), while ILT-4 levels were significant for both CRC patients with early (p=0.001) and advanced (p=0.020) disease stages. Multivariate cox regression analysis revealed that advanced AJCC stage (HR=2.435; p=0.005) and higher ILT-4 levels (HR=2.198; p=0.063) were independent risk factors for poor outcomes in patients with CRC. In summary, among the immune checkpoints, HLA-G/ILT-2/4 and PD-L1, ILT-4 is the most significant prognostic indicator of CRC. This finding indicated that a combination of immunotherapy strategies, such as ILT-4 blockade, could improve the clinical outcomes in patients with cancer. Moreover, multicolor flow cytometry can be employed as a reliable and efficient, alternative to immunohistochemistry, for evaluating the immune checkpoint proteins expressed in tumor lesions.


Assuntos
Antígenos CD/metabolismo , Antígeno B7-H1/metabolismo , Biomarcadores Tumorais , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/metabolismo , Antígenos HLA-G/imunologia , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos CD/genética , Antígeno B7-H1/genética , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/mortalidade , Suscetibilidade a Doenças , Feminino , Expressão Gênica , Antígenos HLA-G/genética , Humanos , Proteínas de Checkpoint Imunológico/genética , Proteínas de Checkpoint Imunológico/metabolismo , Imunofenotipagem , Estimativa de Kaplan-Meier , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/genética , Masculino , Glicoproteínas de Membrana/genética , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Receptores Imunológicos/genética
13.
Biomed Res Int ; 2021: 6673427, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33997038

RESUMO

RESULTS: KIR2DL1 and ILT-2 expression on idNK cells was higher in healthy women than in RPL patients. Sildenafil enhanced NKG2A expression in RPL patients. VEGF concentration was higher in fertile woman idNK cell cultures. idNK cells were more sensitive for necrosis in RPL than in fertile women. SC did not influence VEGF production or idNK cell apoptosis. CONCLUSIONS: A combination of hypoxia, IL-15, and AZA promotes the conversion of pbNK into idNK cells CD56+CD16--expressing KIR receptors and produces VEGF. Alterations in KIR2DL1 and ILT-2 expression as well as impaired VEGF production were associated with RPL. SC affects NKG2A expression on RPL idNK cells. SC had no effect on VEGF release or idNK cell apoptosis.


Assuntos
Aborto Habitual , Antígenos CD/análise , Células Matadoras Naturais , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/análise , Receptores KIR2DL1/análise , Fator A de Crescimento do Endotélio Vascular/análise , Aborto Habitual/sangue , Aborto Habitual/metabolismo , Adulto , Antígenos CD/metabolismo , Apoptose , Células Cultivadas , Feminino , Humanos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Receptores KIR2DL1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
J Immunother Cancer ; 9(3)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33737343

RESUMO

BACKGROUND: CAR-T cells immunotherapy is a breakthrough in the treatment of hematological malignancies such as acute lymphoblastic leukemia (ALL) and B-cell malignancies. However, CAR-T therapies face major hurdles such as the lack of tumor-specific antigen (TSA), and immunosuppressive tumor microenvironment sometimes caused by the tumorous expression of immune checkpoints (ICPs) such as HLA-G. Indeed, HLA-G is remarkable because it is both a potent ICP and a TSA. HLA-G tumor expression causes immune escape by impairing innate and adaptive immune responses and by inducing a suppressive microenvironment. Yet, to date, no immunotherapy targets it. METHODS: We have developed two anti-HLA-G third-generation CARs based on new anti-HLA-G monoclonal antibodies. RESULTS: Anti-HLA-G CAR-T cells were specific for immunosuppressive HLA-G isoforms. HLA-G-activated CAR-T cells polarized toward T helper 1, and became cytotoxic against HLA-G+ tumor cells. In vivo, anti-HLA-G CAR-T cells were able to control and eliminate HLA-G+ tumor cells. The interaction of tumor-HLA-G with interleukin (IL)T2-expressing T cells is known to result in effector T cell functional inhibition, but anti-HLA-G CAR-T cells were insensitive to this inhibition and still exerted their function even when expressing ILT2. Lastly, we show that anti-HLA-G CAR-T cells differentiated into long-term memory effector cells, and seemed not to lose function even after repeated stimulation by HLA-G-expressing tumor cells. CONCLUSION: We report for the first time that HLA-G, which is both a TSA and an ICP, constitutes a valid target for CAR-T cell therapy to specifically target and eliminate both tumor cells and HLA-G+ suppressive cells.


Assuntos
Anticorpos Monoclonais/metabolismo , Antígenos HLA-G/metabolismo , Imunoterapia Adotiva , Leucemia Eritroblástica Aguda/terapia , Células T de Memória/transplante , Receptores de Antígenos Quiméricos/genética , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Antígenos CD/metabolismo , Diferenciação Celular , Técnicas de Cocultura , Citotoxicidade Imunológica , Antígenos HLA-G/imunologia , Humanos , Memória Imunológica , Células K562 , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/imunologia , Leucemia Eritroblástica Aguda/metabolismo , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Células T de Memória/imunologia , Células T de Memória/metabolismo , Camundongos Endogâmicos NOD , Camundongos SCID , Fenótipo , Receptores de Antígenos Quiméricos/metabolismo , Fatores de Tempo , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Inflammation ; 44(3): 1108-1118, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33538932

RESUMO

Angiopoietin-like proteins (ANGPTLs) are circulating proteins that are expressed in various cells and tissues and are thought to be involved in the repair and remodeling of damaged tissues; however, ANGPTL2 hyperfunction has been shown to cause chronic inflammation, leading to the progression of various diseases. ANGPTL2 is known to exert cellular effects via receptors such as integrin α5ß1 and leukocyte immunoglobulin-like receptor subfamily B member 2 (LILRB2); however, their roles in ANGPTL2-induced inflammation remain unclear. In this study, we investigated the mechanisms underlying ANGPTL2-induced inflammation involving LILRB2 and various signaling pathways in human fibroblast-like synoviocytes (HFLS). The effects of ANGPTL2 and an anti-LILRB2 antibody on the gene expression of various inflammation-related factors were examined using real-time RT-PCR, while their effects on MAPK, NF-κB, and Akt phosphorylation were analyzed by western blotting. We found that the addition of ANGPTL2 enhanced the gene expression of inflammatory factors, whereas pretreatment with the anti-LILRB2 antibody for 12 h decreased the expression of these factors. Similarly, ANGPTL2 addition activated the phosphorylation of ERK, p38, JNK, NF-κB, and Akt in HFLS; however, this effect was significantly inhibited by pretreatment with the anti-LILRB2 antibody. Together, the findings of this study demonstrate that ANGPTL2 induces the expression of inflammatory factors via LILRB2 in synovial cells. Therefore, LILRB2 could be a potential therapeutic agent for treating matrix degradation in osteoarthritis.


Assuntos
Proteína 2 Semelhante a Angiopoietina/toxicidade , Antígenos CD/metabolismo , Fibroblastos/efeitos dos fármacos , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Sinoviócitos/efeitos dos fármacos , Sinovite/induzido quimicamente , Antígenos CD/genética , Células Cultivadas , Citocinas/metabolismo , Fibroblastos/metabolismo , Humanos , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Sinoviócitos/metabolismo , Sinovite/metabolismo
16.
Trends Cancer ; 7(5): 389-392, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33563576

RESUMO

Despite some success, many patients do not benefit from immunotherapy. New strategies to improve clinical efficacy include identification of novel immune-checkpoint (IC) targets or a combination of immunotherapy with antiangiogenic treatments. Here, we propose the therapeutic use of IC, HLA-G/LILRB, and explore its enhanced synergistic antitumor activity when combined with antiangiogenic therapies.


Assuntos
Antígenos CD/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Antígenos HLA-G/metabolismo , Imunoterapia Adotiva/métodos , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Glicoproteínas de Membrana/metabolismo , Neoplasias/terapia , Receptores Imunológicos/metabolismo , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Sinergismo Farmacológico , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Proteínas de Checkpoint Imunológico/metabolismo , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/antagonistas & inibidores , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Glicoproteínas de Membrana/antagonistas & inibidores , Neoplasias/imunologia , Receptores de Antígenos Quiméricos/imunologia , Receptores Imunológicos/antagonistas & inibidores , Evasão Tumoral , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
17.
Front Immunol ; 11: 2046, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32973812

RESUMO

Tumor immune escape is associated with both, the expression of immune checkpoint molecules on peripheral immune cells and soluble forms of the human leukocyte antigen-G (HLA-G) in the blood, which are consequently discussed as clinical biomarker for disease status and outcome of cancer patients. HLA-G preferentially interacts with the inhibitory receptor immunoglobulin-like transcript (ILT) receptor-2 in the blood and can be secreted as free soluble molecules (sHLA-G) or via extracellular vesicles (EV). To investigate the contribution of these two forms to the expression of checkpoint molecules in peripheral blood, we primed peripheral blood mononuclear cells with purified soluble sHLA-G1 protein, or EV preparations derived from SUM149 cells transfected with membrane-bound HLA-G1 or control vector prior to anti-CD3/CD28 T cell activation. Our study demonstrated that priming of PBMC with sHLA-G1 protein prior to 48 h activation resulted in enhanced frequencies of ILT-2 expressing CD8+ T cells, and in an upregulation of immune checkpoint molecules CTLA-4, PD-1, TIM-3, and CD95 exclusively on ILT-2 positive CD8+ T cells. In contrast, when PBMC were primed with EV (containing HLA-G1 or not) upregulation of CTLA-4, PD-1, TIM-3, and CD95 occurred exclusively on ILT-2 negative CD8+ T cells. Taken together, our data suggest that priming with sHLA-G forms induces a pronounced immunosuppressive/exhausted phenotype and that priming with sHLA-G1 protein or EV derived from HLA-G1 positive or negative SUM149 cells affects CD8+ T cells complementary by targeting either the ILT-2 positive or negative subpopulation, respectively, after T cell activation.


Assuntos
Antígenos CD/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vesículas Extracelulares/metabolismo , Antígenos HLA-G/imunologia , Antígenos HLA-G/metabolismo , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Antígenos CD/genética , Transporte Biológico , Biomarcadores , Linhagem Celular , Meios de Cultivo Condicionados/metabolismo , Expressão Gênica , Antígenos HLA-G/sangue , Humanos , Proteínas de Checkpoint Imunológico/metabolismo , Imunomodulação , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/genética , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Fenótipo
18.
Nature ; 587(7833): 309-312, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32650338

RESUMO

The Plasmodium species that cause malaria are obligate intracellular parasites, and disease symptoms occur when these parasites replicate in human blood. Despite the risk of immune detection, the parasite delivers proteins that bind to host receptors on the cell surfaces of infected erythrocytes. In the causative parasite of the most deadly form of malaria in humans, Plasmodium falciparum, RIFINs form the largest family of surface proteins displayed by erythrocytes1. Some RIFINs can bind to inhibitory immune receptors, and these RIFINs act as targets for unusual antibodies that contain a LAIR1 ectodomain2-4 or as ligands for LILRB15. RIFINs stimulate the activation of and signalling by LILRB15, which could potentially lead to the dampening of human immune responses. Here, to understand how RIFINs activate LILRB1-mediated signalling, we determine the structure of a RIFIN bound to LILRB1. We show that this RIFIN mimics the natural activating ligand of LILRB1, MHC class I, in its LILRB1-binding mode. A single mutation in the RIFIN disrupts the complex, blocks LILRB1 binding of all tested RIFINs and abolishes signalling in a reporter assay. In a supported lipid bilayer system, which mimics the activation of natural killer (NK) cells by antibody-dependent cell-mediated cytotoxicity, both RIFIN and MHC are recruited to the immunological synapse of NK cells and reduce the activation of NK cells, as measured by the mobilization of perforin. Therefore, LILRB1-binding RIFINs mimic the binding mode of the natural ligand of LILRB1 and suppress the function of NK cells.


Assuntos
Receptor B1 de Leucócitos Semelhante a Imunoglobulina/química , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/imunologia , Malária Falciparum/imunologia , Proteínas de Membrana/química , Proteínas de Membrana/imunologia , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Sequência de Aminoácidos , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Sítios de Ligação/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Ligantes , Bicamadas Lipídicas , Ativação Linfocitária , Malária Falciparum/parasitologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Moleculares , Mimetismo Molecular/imunologia , Mutação , Perforina/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Transdução de Sinais
19.
Cancer Biomark ; 28(3): 309-319, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32390601

RESUMO

BACKGROUND: Immunosuppressive receptor LILRB1 regulates tumors progression by transducing immune inhibitory signals via intracellular immunoreceptor tyrosine-based inhibitory motifs. However, its role in Hepatocellular Carcinoma (HCC) remains vague. OBJECTIVE: This study is aimed to disclose the association between LILRB1 and HCC. METHODS: Immunoblotting and qRT-PCR were employed to evaluate the level of LILRB1 in hepatocarcinoma cells. LILRB1-positive cells in tissue array were measured using immunohistochemistry staining. The relation among LILRB1, SHP1 and SHP2 and survival rates were analyzed using Gene Expression Profiling Interactive Analysis (GEPIA) and Oncomine database. RESULTS: LILRB1 was robustly reduced in hepatocarcinoma cells compared to normal cells. Clinically, LILRB1 was significantly higher in 49 of 75 (65%) paired paracarcinoma tissues than that in paired HCC samples. 48 of 75 (64%) HCC subjects in tissue microarray showed low level of LILRB1, compared to 25 of 75 (33%) in paired-adjacent tissues. Oncomine database and GEPIA analysis confirmed that LILRB1 was lower in HCC than normal tissues. Additionally, lowLILRB1 had a significant association with clinicopathological characteristics and Disease Free Survival, but no association with Overall Survival in HCC patients. Mechanismly, positive correlation between LILRB1 and SHP1, but not SHP2 was observed in HCC. CONCLUSIONS: LILRB1 possibly plays an antitumor effect in hepatocarcinoma cells by integrating SHP1, providing evidence that LILRB1 might be involved in the pathologic progression of HCC.


Assuntos
Antígenos CD/metabolismo , Carcinoma Hepatocelular/patologia , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Neoplasias Hepáticas/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/mortalidade , Linhagem Celular Tumoral , Proliferação de Células/genética , Progressão da Doença , Intervalo Livre de Doença , Regulação para Baixo/imunologia , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Fígado/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/mortalidade , Masculino , Pessoa de Meia-Idade , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/imunologia , Análise Serial de Tecidos
20.
Mol Med Rep ; 22(2): 986-996, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32468016

RESUMO

Rheumatoid arthritis (RA), which normally manifests as a multi­joint inflammatory reaction, is a common immunological disease in clinical practice. However, the pathogenesis of RA has not yet been fully elucidated. Rituximab (RTX) is an effective drug in the treatment of RA, however its therapeutic efficacy and mechanism of action require further investigation. Thus, the present study aimed to screen the candidate key regulatory genes and explain the potential mechanisms of RA. Gene chips of RA and normal joint tissues were analyzed and, gene chips of RTX before and after treatment were investigated. In the present study, strong evidence supporting the pathogenesis of RA and mechanism of action of RTX were also revealed. Differentially expressed genes (DEGs) were analyzed using the limma package of RStudio software. A total of 1,150 DEGs were detected in RA compared with normal joint tissues. The upregulated genes were enriched in 'interleukin­12 production', 'I­κB kinase/NF­κB signaling', 'regulation of cytokine production involved in immune response' and 'cytokine metabolic process'. Functional enrichment analysis showed that RTX was primarily involved in the inhibition of 'adaptive immune response', 'B cell activation involved in immune response' and 'immune effector process'. Subsequently, leukocyte immunoglobulin­like receptor subfamily B member 1 (LILRB1), a hub gene with high connectivity degree, was selected, and traditional Chinese medicine libraries were molecularly screened according to the structure of the LILRB1 protein. The results indicated that kaempferol 3­O­ß­D­glucosyl­(1→2)­ß­D­glucoside exhibited the highest docking score. In the present study, the DEGs and their biological functions in RA and the pharmacological mechanism of RTX action were determined. Taken together, the results suggested that LILRB1 may be used as a molecular target for RA treatment, and kaempferol 3­O­ß­D­glucosyl­(1→2)­ß­D­glucoside may inhibit the pathological process of RA.


Assuntos
Artrite Reumatoide/etiologia , Artrite Reumatoide/genética , Avaliação Pré-Clínica de Medicamentos/métodos , Antígenos CD/genética , Antígenos CD/metabolismo , Antirreumáticos/farmacologia , Antirreumáticos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Biologia Computacional/métodos , Bases de Dados Genéticas , Bases de Dados de Produtos Farmacêuticos , Bases de Dados de Proteínas , Perfilação da Expressão Gênica/métodos , Ontologia Genética , Humanos , Quempferóis/química , Quempferóis/farmacologia , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/antagonistas & inibidores , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/genética , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Medicina Tradicional Chinesa , Análise de Sequência com Séries de Oligonucleotídeos , Mapas de Interação de Proteínas , Rituximab/genética , Rituximab/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Membrana Sinovial/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA