Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.362
Filter
1.
Front Immunol ; 15: 1463971, 2024.
Article in English | MEDLINE | ID: mdl-39351219

ABSTRACT

Introduction: In this study, we report a novel therapeutic approach redirecting antigen-specific CD4+ T cells recognizing a hybrid insulin peptide (BDC2.5 T cell receptor (TCR) transgenic CD4+ T cells) to attract and suppress islet-specific CD8+ T cells T cells in the non-obese diabetic (NOD) mouse model, and prevent the development of autoimmune diabetes. Methods: Purified BDC2.5 CD4+ T cells were induced to differentiate into regulatory T cells (Tregs). The Tregs were then electroporated with mRNA encoding chimeric human ß2 microglobulin (hß2m) covalently linked to insulin B chain amino acids 15-23 (designated INS-eTreg) or islet-specific glucose-6-phosphatase related protein (IGRP) peptide 206-214 (designated IGRP-eTreg). Immunoregulatory functions of these engineered regulatory T cells (eTregs) were tested by in vitro assays and in vivo co-transfer experiments with ß-cell-antigen-specific CD8+ T cells in NOD.Scid mice or by adoptive transfer into young, pre-diabetic NOD mice. Results: These eTregs were phenotyped by flow cytometry, and shown to have high expression of FoxP3, as well as other markers of Treg function, including IL-10. They suppressed polyclonal CD4+ T cells and antigen-specific CD8+ T cells (recognizing insulin or IGRP), decreasing proliferation and increasing exhaustion and regulatory markers in vitro. In vivo, eTregs reduced diabetes development in co-transfer experiments with pathogenic antigen-specific CD8+ T cells (INS-CD8+ or IGRP-CD8+ cells) into NOD.Scid mice. Finally, when the eTreg were injected into young NOD mice, they reduced insulitis and prevented spontaneous diabetes in the recipient mice. Conclusion: Our results suggest a novel therapeutic strategy to protect NOD mice by targeting antigen-specific cytotoxic CD8+ T cells, using redirected antigen-specific CD4+ Treg cells, to suppress autoimmune diabetes. This may suggest an innovative therapy for protection of people at risk of development of type 1 diabetes.


Subject(s)
CD8-Positive T-Lymphocytes , Diabetes Mellitus, Type 1 , Mice, Inbred NOD , T-Lymphocytes, Regulatory , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , CD8-Positive T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Mice , Humans , Female , Mice, SCID , Insulin/immunology , Adoptive Transfer , Mice, Transgenic , Glucose-6-Phosphatase/immunology , Glucose-6-Phosphatase/genetics , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
2.
Science ; 385(6714): eadn1629, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39264994

ABSTRACT

Macrophages maintain hematopoietic stem cell (HSC) quality by assessing cell surface Calreticulin (Calr), an "eat-me" signal induced by reactive oxygen species (ROS). Using zebrafish genetics, we identified Beta-2-microglobulin (B2m) as a crucial "don't eat-me" signal on blood stem cells. A chemical screen revealed inducers of surface Calr that promoted HSC proliferation without triggering ROS or macrophage clearance. Whole-genome CRISPR-Cas9 screening showed that Toll-like receptor 3 (Tlr3) signaling regulated b2m expression. Targeting b2m or tlr3 reduced the HSC clonality. Elevated B2m levels correlated with high expression of repetitive element (RE) transcripts. Overall, our data suggest that RE-associated double-stranded RNA could interact with TLR3 to stimulate surface expression of B2m on hematopoietic stem and progenitor cells. These findings suggest that the balance of Calr and B2m regulates macrophage-HSC interactions and defines hematopoietic clonality.


Subject(s)
Calreticulin , Hematopoietic Stem Cells , Macrophages , Phagocytosis , Toll-Like Receptor 3 , beta 2-Microglobulin , Animals , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism , Calreticulin/metabolism , Calreticulin/genetics , Cell Proliferation , CRISPR-Cas Systems , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Macrophages/metabolism , Reactive Oxygen Species/metabolism , Repetitive Sequences, Nucleic Acid , Signal Transduction , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 3/genetics , Zebrafish , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics
3.
BMC Cancer ; 24(1): 991, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39128984

ABSTRACT

BACKGROUND: Our previous study demonstrated that ß2-microglobulin (ß2M) promoted ER+/HER2- breast cancer survival via the SGK1/Bcl-2 signaling pathway. However, the role of ß2M has not been investigated in ER-/HER2+ breast cancer. Here, we aimed to determine the role of ß2M in ER-/HER2+ breast cancer. METHODS: The interaction between ß2M and HFE was confirmed by co-immunoprecipitation, mass spectrometry, yeast two-hybrid screening, and His pull-down. The knockdown and overexpression of ß2M or HFE were performed in MDA-MB-453 cells, and ERK signaling pathway was subsequently analyzed via western blotting. Apoptotic cells were detected using flow cytometer. ß2M, HFE, and p-ERK1/2 were examined in tumor and paired adjacent tissues via immunohistochemistry. RESULTS: HFE was found to be an interacting protein of ß2M in ER-/HER2+ breast cancer cells MDA-MB-453 by co-immunoprecipitation and mass spectrometry. A yeast two-hybrid system and His-pull down experiments verified that ß2M directly interacted with HFE. ß2M and HFE as a complex were mainly located in the cytoplasm, with some on the cytomembrane of MDA-MB-453 cells. In addition to breast cancer cells BT474, endogenous ß2M directly interacted with HFE in breast cancer cells MDA-MB-453, MDA-MB-231, and MCF-7. ß2M activated the ERK signaling pathway by interacting with HFE and induced apoptosis of MDA-MB-453 cells. The expression of HFE and p-ERK1/2 showed significantly high levels in HER2-overexpressing breast cancer tumor tissue compared with adjacent normal tissue, consistent with the results obtained from the cell experiments. CONCLUSIONS: ß2M induced apoptosis of tumor cells via activation of the ERK signal pathway by directly interacting with HFE in HER2-overexpressing breast cancer.


Subject(s)
Apoptosis , Breast Neoplasms , Hemochromatosis Protein , MAP Kinase Signaling System , Receptor, ErbB-2 , beta 2-Microglobulin , Humans , beta 2-Microglobulin/metabolism , beta 2-Microglobulin/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Female , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Cell Line, Tumor , Hemochromatosis Protein/genetics , Hemochromatosis Protein/metabolism , Protein Binding , Gene Expression Regulation, Neoplastic
4.
Front Immunol ; 15: 1396187, 2024.
Article in English | MEDLINE | ID: mdl-39170623

ABSTRACT

Introduction: Myeloid sarcomas (MS) comprise rare extramedullary manifestations of myeloid neoplasms with poor patients' outcome. While the clinical relevance of the tumor microenvironment (TME) is well established in many malignancies, there exists limited information in MS. Methods: The expression of the human leukocyte antigen class I (HLA-I) antigens, HLA-I antigen processing and presenting machinery (APM) components and the composition of the TME of 45 MS and paired bone marrow (BM) samples from two independent cohorts were assessed by immunohistochemistry, multispectral imaging, and RNA sequencing (RNAseq). Results: A significant downregulation of the HLA-I heavy chain (HC; 67.5%) and ß2-microglobulin (ß2M; 64.8%), but an upregulation of HLA-G was found in MS compared to BM samples, which was confirmed in a publicly available dataset. Moreover, MS tumors showed a predominantly immune cell excluded TME with decreased numbers of tissue infiltrating lymphocytes (TILs) (9.5%) compared to paired BM (22.9%). RNAseq analysis of a subset of 10 MS patients with preserved and reduced HLA-I HC expression revealed 150 differentially expressed genes and a significantly reduced expression of inflammatory response genes was found in samples with preserved HLA-I expression. Furthermore, low HLA-I expression and low TIL numbers in the TME of MS cases were linked to an inferior patients' outcome. Discussion: This study demonstrated a high prevalence of immune escape strategies in the pathogenesis and extramedullary spread of MS, which was also found in patients without evidence of any BM pathology, which yields the rational for the development of novel individually tailored therapies for MS patients.


Subject(s)
Sarcoma, Myeloid , Tumor Microenvironment , Humans , Male , Female , Middle Aged , Tumor Microenvironment/immunology , Sarcoma, Myeloid/immunology , Sarcoma, Myeloid/genetics , Sarcoma, Myeloid/mortality , Adult , Aged , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Tumor Escape , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , beta 2-Microglobulin/genetics , Immune Evasion , Young Adult
5.
Cell Stem Cell ; 31(9): 1376-1386.e8, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38981470

ABSTRACT

Allogeneic cellular immunotherapies hold promise for broad clinical implementation but face limitations due to potential rejection of donor cells by the host immune system. Silencing of beta-2 microglobulin (B2M) expression is commonly employed to evade T cell-mediated rejection by the host, although the absence of B2M is expected to trigger missing-self responses by host natural killer (NK) cells. Here, we demonstrate that genetic deletion of the adhesion ligands CD54 and CD58 in B2M-deficient chimeric antigen receptor (CAR) T cells and multi-edited induced pluripotent stem cell (iPSC)-derived CAR NK cells reduces their susceptibility to rejection by host NK cells in vitro and in vivo. The absence of adhesion ligands limits rejection in a unidirectional manner in B2M-deficient and B2M-sufficient settings without affecting the antitumor functionality of the engineered donor cells. Thus, these data suggest that genetic ablation of adhesion ligands effectively alleviates rejection by host immune cells, facilitating the implementation of universal immunotherapy.


Subject(s)
Killer Cells, Natural , Animals , Mice , Ligands , Killer Cells, Natural/immunology , Induced Pluripotent Stem Cells/metabolism , Mice, Inbred C57BL , Graft Rejection/immunology , Immunotherapy/methods , CD58 Antigens/metabolism , CD58 Antigens/genetics , Humans , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Intercellular Adhesion Molecule-1/metabolism
6.
Mol Genet Genomics ; 299(1): 74, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39085666

ABSTRACT

Derivation of hypoimmunogenic human cells from genetically manipulated pluripotent stem cells holds great promise for future transplantation medicine and adoptive immunotherapy. Disruption of beta-2-microglobulin (B2M) in pluripotent stem cells followed by differentiation into specialized cell types is a promising approach to derive hypoimmunogenic cells. Given the attractive features of CRISPR/Cas9-based gene editing tool and baculoviral delivery system, baculovirus can deliver CRISPR/Cas9 components for site-specific gene editing of B2M. Herein, we report the development of a baculoviral CRISPR/Cas9 vector system for the B2M locus disruption in human cells. When tested in human embryonic stem cells (hESCs), the B2M gene knockdown/out was successfully achieved, leading to the stable down-regulation of human leukocyte antigen class I expression on the cell surface. Fibroblasts derived from the B2M gene-disrupted hESCs were then used as stimulator cells in the co-cultures with human peripheral blood mononuclear cells. These fibroblasts triggered significantly reduced alloimmune responses as assessed by sensitive Elispot assays. The B2M-negative hESCs maintained the pluripotency and the ability to differentiate into three germ lineages in vitro and in vivo. These findings demonstrated the feasibility of using the baculoviral-CRISPR/Cas9 system to establish B2M-disrupted pluripotent stem cells. B2M knockdown/out sufficiently leads to hypoimmunogenic conditions, thereby supporting the potential use of B2M-negative cells as universal donor cells for allogeneic cell therapy.


Subject(s)
Baculoviridae , CRISPR-Cas Systems , Cell Differentiation , Gene Editing , Genetic Vectors , Pluripotent Stem Cells , beta 2-Microglobulin , Humans , beta 2-Microglobulin/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Baculoviridae/genetics , Gene Editing/methods , Genetic Vectors/genetics , Cell Differentiation/genetics , Gene Knockout Techniques/methods , Animals , Fibroblasts/metabolism , Fibroblasts/cytology , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Mice
7.
Gene Ther ; 31(9-10): 467-476, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39013986

ABSTRACT

Gene therapy using siRNA has become a promising strategy to achieve targeted gene knockdown for treatment of cardiovascular pathologies. However, efficient siRNA transfection often relies on cationic delivery vectors such as synthetic cell-penetrating polymers which are susceptible to interference by negatively charged molecules. Anticoagulants such as heparin, which is negatively charged and widely used in cardiovascular applications, may pose a significant barrier to effective siRNA delivery. We therefore conducted in vitro studies utilizing human smooth muscle and endothelial cells transfected with glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and ß2-microglobulin (B2M) siRNA in the presence of heparin, argatroban, and bivalirudin in order to determine which anticoagulant therapy is most compatible for siRNA delivery. We observed that while heparin, at clinical doses, decreases the efficiency of siRNA targeted mRNA knockdown, mRNA knockdown is not inhibited in the presence of either argatroban or bivalirudin. Our data suggests that heparin should be avoided during siRNA therapy with cationic transfection agents, and argatroban and bivalirudin should be used in its stead.


Subject(s)
Arginine , Heparin , RNA, Small Interfering , Transfection , Humans , Heparin/pharmacology , RNA, Small Interfering/pharmacology , RNA, Small Interfering/genetics , Transfection/methods , Arginine/analogs & derivatives , Arginine/pharmacology , Hirudins/pharmacology , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Peptide Fragments/pharmacology , Pipecolic Acids/pharmacology , Sulfonamides/pharmacology , beta 2-Microglobulin/genetics , Antithrombins/pharmacology , Gene Knockdown Techniques/methods , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/antagonists & inhibitors , Cell-Penetrating Peptides/pharmacology , Genetic Therapy/methods , Anticoagulants/pharmacology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects
8.
Sci Transl Med ; 16(756): eadi9548, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39018368

ABSTRACT

Immune rejection remains the major obstacle to long-term survival of allogeneic lung transplants. The expression of major histocompatibility complex molecules and minor histocompatibility antigens triggers allogeneic immune responses that can lead to allograft rejection. Transplant outcomes therefore depend on long-term immunosuppression, which is associated with severe side effects. To address this problem, we investigated the effect of genetically engineered transplants with permanently down-regulated swine leukocyte antigen (SLA) expression to prevent rejection in a porcine allogeneic lung transplantation (LTx) model. Minipig donor lungs with unmodified SLA expression (control group, n = 7) or with modified SLA expression (treatment group, n = 7) were used to evaluate the effects of SLA knockdown on allograft survival and on the nature and strength of immune responses after terminating an initial 4-week period of immunosuppression after LTx. Genetic engineering to down-regulate SLA expression was achieved during ex vivo lung perfusion by lentiviral transduction of short hairpin RNAs targeting mRNAs encoding ß2-microglobulin and class II transactivator. Whereas all grafts in the control group were rejected within 3 months, five of seven animals in the treatment group maintained graft survival without immunosuppression during the 2-year monitoring period. Compared with controls, SLA-silenced lung recipients had lower donor-specific antibodies and proinflammatory cytokine concentrations in the serum. Together, these data demonstrate a survival benefit of SLA-down-regulated lung transplants in the absence of immunosuppression.


Subject(s)
Gene Knockdown Techniques , Graft Survival , Histocompatibility Antigens Class I , Immunosuppression Therapy , Lung Transplantation , Animals , Swine , Graft Survival/immunology , Histocompatibility Antigens Class I/metabolism , Graft Rejection/immunology , Swine, Miniature , Histocompatibility Antigens Class II/metabolism , Transplantation, Homologous , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism , Lung/metabolism , Nuclear Proteins , Trans-Activators
9.
Front Immunol ; 15: 1404668, 2024.
Article in English | MEDLINE | ID: mdl-38903492

ABSTRACT

Heart transplantation is associated with major hurdles, including the limited number of available organs for transplantation, the risk of rejection due to genetic discrepancies, and the burden of immunosuppression. In this study, we demonstrated the feasibility of permanent genetic engineering of the heart during ex vivo perfusion. Lentiviral vectors encoding for short hairpin RNAs targeting beta2-microglobulin (shß2m) and class II transactivator (shCIITA) were delivered to the graft during two hours of normothermic EVHP. Highly efficient genetic engineering was indicated by stable reporter gene expression in endothelial cells and cardiomyocytes. Remarkably, swine leucocyte antigen (SLA) class I and SLA class II expression levels were decreased by 66% and 76%, respectively, in the vascular endothelium. Evaluation of lactate, troponin T, and LDH levels in the perfusate and histological analysis showed no additional cell injury or tissue damage caused by lentiviral vectors. Moreover, cytokine secretion profiles (IL-6, IL-8, and TNF-α) of non-transduced and lentiviral vector-transduced hearts were comparable. This study demonstrated the ex vivo generation of genetically engineered hearts without compromising tissue integrity. Downregulation of SLA expression may contribute to reduce the immunogenicity of the heart and support graft survival after allogeneic or xenogeneic transplantation.


Subject(s)
Genetic Vectors , Heart Transplantation , Histocompatibility Antigens Class I , Lentivirus , Animals , Lentivirus/genetics , Heart Transplantation/methods , Genetic Vectors/genetics , Swine , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Perfusion/methods , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/immunology , beta 2-Microglobulin/genetics , Cytokines/metabolism , Genetic Engineering , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/immunology , Humans , RNA, Small Interfering/genetics , Graft Survival/immunology , Graft Survival/genetics , Endothelial Cells/metabolism , Endothelial Cells/immunology , Nuclear Proteins , Trans-Activators
10.
Arterioscler Thromb Vasc Biol ; 44(7): 1570-1583, 2024 07.
Article in English | MEDLINE | ID: mdl-38813697

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) represents an important phenotype in heart failure with preserved ejection fraction (HFpEF). However, management of PH-HFpEF is challenging because mechanisms involved in the regulation of PH-HFpEF remain unclear. METHODS: We used a mass spectrometry-based comparative plasma proteomics approach as a sensitive and comprehensive hypothesis-generating discovery technique to profile proteins in patients with PH-HFpEF and control subjects. We then validated and investigated the role of one of the identified proteins using in vitro cell cultures, in vivo animal models, and independent cohort of human samples. RESULTS: Plasma proteomics identified high protein abundance levels of B2M (ß2-microglobulin) in patients with PH-HFpEF. Interestingly, both circulating and skeletal muscle levels of B2M were increased in mice with skeletal muscle SIRT3 (sirtuin-3) deficiency or high-fat diet-induced PH-HFpEF. Plasma and muscle biopsies from a validation cohort of PH-HFpEF patients were found to have increased B2M levels, which positively correlated with disease severity, especially pulmonary capillary wedge pressure and right atrial pressure at rest. Not only did the administration of exogenous B2M promote migration/proliferation in pulmonary arterial vascular endothelial cells but it also increased PCNA (proliferating cell nuclear antigen) expression and cell proliferation in pulmonary arterial vascular smooth muscle cells. Finally, B2m deletion improved glucose intolerance, reduced pulmonary vascular remodeling, lowered PH, and attenuated RV hypertrophy in mice with high-fat diet-induced PH-HFpEF. CONCLUSIONS: Patients with PH-HFpEF display higher circulating and skeletal muscle expression levels of B2M, the magnitude of which correlates with disease severity. Our findings also reveal a previously unknown pathogenic role of B2M in the regulation of pulmonary vascular proliferative remodeling and PH-HFpEF. These data suggest that circulating and skeletal muscle B2M can be promising targets for the management of PH-HFpEF.


Subject(s)
Disease Models, Animal , Heart Failure , Hypertension, Pulmonary , Proteomics , Stroke Volume , beta 2-Microglobulin , Adult , Aged , Animals , Humans , Male , Mice , Middle Aged , beta 2-Microglobulin/genetics , beta 2-Microglobulin/blood , beta 2-Microglobulin/metabolism , Biomarkers/blood , Case-Control Studies , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Heart Failure/physiopathology , Heart Failure/metabolism , Heart Failure/blood , Heart Failure/genetics , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/genetics , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Proteomics/methods , Pulmonary Artery/physiopathology , Pulmonary Artery/metabolism , Sirtuin 3/genetics , Sirtuin 3/metabolism , Vascular Remodeling , Ventricular Function, Left
11.
Virology ; 595: 110083, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38696887

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) infection inhibits swine leukocyte antigen class I (SLA-I) expression in pigs, resulting in inefficient antigen presentation and subsequent low levels of cellular PRRSV-specific immunity as well as persistent viremia. We previously observed that the non-structural protein 4 (nsp4) of PRRSV contributed to inhibition of the ß2-microglobulin (ß2M) and SLA-I expression in cells. Here, we constructed a series of nsp4 mutants with different combination of amino acid mutations to attenuate the inhibitory effect of nsp4 on ß2M and SLA-I expression. Almost all nsp4 mutants exogenously expressed in cells showed an attenuated effect on inhibition of ß2M and SLA-I expression, but the recombinant PRRSV harboring these nsp4 mutants failed to be rescued with exception of the rPRRSV-nsp4-mut10 harboring three amino acid mutations. However, infection of rPRRSV-nsp4-mut10 not only enhanced ß2M and SLA-I expression in both cells and pigs but also promoted the DCs to active the CD3+CD8+T lymphocytes more efficiently, as compared with its parental PRRSV (rPRRVS-nsp4-wt). These data suggested that the inhibition of nsp4-mediated ß2M downregulation improved ß2M/SLA-I expression in pigs.


Subject(s)
Down-Regulation , Histocompatibility Antigens Class I , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Viral Nonstructural Proteins , beta 2-Microglobulin , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/physiology , Porcine respiratory and reproductive syndrome virus/immunology , Animals , Swine , Porcine Reproductive and Respiratory Syndrome/virology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/immunology , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/immunology , Cell Line , CD8-Positive T-Lymphocytes/immunology , Mutation
12.
Mol Genet Genomics ; 299(1): 47, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649532

ABSTRACT

Multiple myeloma (MM) is a plasma cell dyscrasia that is characterized by the uncontrolled proliferation of malignant PCs in the bone marrow. Due to immunotherapy, attention has returned to the immune system in MM, and it appears necessary to identify biomarkers in this area. In this study, we created a prognostic model for MM using immune-related gene pairs (IRGPs), with the advantage that it is not affected by technical bias. After retrieving microarray data of MM patients, bioinformatics analyses like COX regression and least absolute shrinkage and selection operator (LASSO) were used to construct the signature. Then its prognostic value is assessed via time-dependent receiver operating characteristic (ROC) and the Kaplan-Meier (KM) analysis. We also used XCELL to examine the status of immune cell infiltration among MM patients. 6-IRGP signatures were developed and proved to predict MM prognosis with a P-value of 0.001 in the KM analysis. Moreover, the risk score was significantly associated with clinicopathological characteristics and was an independent prognostic factor. Of note, the combination of age and ß2-microglobulin with risk score could improve the accuracy of determining patients' prognosis with the values of the area under the curve (AUC) of 0.73 in 5 years ROC curves. Our model was also associated with the distribution of immune cells. This novel signature, either alone or in combination with age and ß2-microglobulin, showed a good prognostic predictive value and might be used to guide the management of MM patients in clinical practice.


Subject(s)
Bone Marrow , Gene Expression Profiling , Multiple Myeloma , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/mortality , Humans , Female , Prognosis , Male , Gene Expression Profiling/methods , Bone Marrow/pathology , Bone Marrow/immunology , Middle Aged , Aged , Gene Expression Regulation, Neoplastic , beta 2-Microglobulin/genetics , Biomarkers, Tumor/genetics , Kaplan-Meier Estimate , ROC Curve , Transcriptome/genetics
13.
Front Immunol ; 15: 1285049, 2024.
Article in English | MEDLINE | ID: mdl-38455061

ABSTRACT

Background: Downregulation of MHC class I expression and/or defects in the antigen presentation pathways are commonly reported in human cancers. Numerous studies previously have explored extensively the molecular mechanisms that underlie HLA-class I and Beta2-Microglobulin (B2M) downregulation. However, the techniques presently available to detect expression of MHC class I proteins lack the robustness, specificity and sensitivity needed for systematic integration and analysis in clinical trials. Furthermore, the dynamics of HLA-class I and B2M expression have not been comprehensively studied as a potential biomarker for immunotherapy. Methods: Using novel, validated, immunohistochemistry (IHC)-based methods for quantifying B2M and HLA-A in tumor samples from diverse cancer types, we have determined loss of B2M and HLA-A proteins in 336 archived, primary specimens and 329 biopsies from metastatic patients collected during Roche-sponsored Phase 1 clinical trials investigating novel immunotherapy candidates as monotherapy or in combination with CPI. Results: Up to 56% of cases with B2M or HLA-A loss were noted in the investigated tumor types. The frequency of loss was dependent on indication and stage of disease and revealed heterogeneous expression patterns across patients. B2M and HLA-A loss was increased in metastatic lesions compared to primary tumors, indicating selection of MHC class I low clones in metastatic and refractory tumor cells. High on-treatment B2M expression correlated with successful clinical outcome (RECIST), while high baseline B2M did not. A treatment-induced increase of B2M expression was noted in most of the patients with low B2M levels at baseline. The triple biomarker combination of B2M, CD8 and PDL1 strongly improved response prediction to cancer immunotherapy. Conclusion: Our results indicate that B2M and HLA-A loss occurs frequently in tumors and is reversed in most instances following immunotherapy which supports the conclusion that MHC class I loss is not the dominant resistance mechanism to CPI treatment. This investigation reveals a highly dynamic expression of HLA-A and B2M in tumors affected by indication, metastatic status, immunophenotype and immunotherapy treatment. Baseline expression levels of B2M on tumors may be of utility as a constituent of a biomarker panel used for selecting patients for immunotherapy clinical trials.


Subject(s)
Neoplasms , beta 2-Microglobulin , Humans , beta 2-Microglobulin/genetics , Histocompatibility Antigens Class I/genetics , Immunotherapy , HLA-A Antigens
14.
Vet Microbiol ; 292: 110036, 2024 May.
Article in English | MEDLINE | ID: mdl-38458048

ABSTRACT

Group A Rotavirus (RVA) is a major cause of diarrhea in infants and piglets. ß2-microglobulin (ß2 M), encoded by the B2M gene, serves as a crucial subunit of the major histocompatibility complex class I (MHC-I) molecules. ß2 M is indispensable for the transport of MHC-I to the cell membrane. MHC-I, also known as swine leukocyte antigen class I (SLA-I) in pigs, presents viral antigens to the cell surface. In this study, RVA infection down-regulated ß2 M expression in both porcine intestinal epithelial cells-J2 (IPEC-J2) and MA-104 cells. RVA infection did not down-regulate the mRNA level of the B2M gene, indicating that the down-regulation of ß2 M occurred on the protein level. Mechanismly, RVA infection triggered ß2 M aggregation in the endoplasmic reticulum (ER) and enhanced the Lys48 (K48)-linked ubiquitination of ß2 M, leading to the degradation of ß2 M through ERAD-proteasome pathway. Furthermore, we found that RVA infection significantly impeded the level of SLA-I on the surface, and the overexpression of ß2 M could recover its expression. In this study, our study demonstrated that RVA infection degrades ß2 M via ERAD-proteasome pathway, consequently hampering SLA-I expression on the cell surface. This study would enhance the understanding of the mechanism of how RVA infection induces immune escape.


Subject(s)
Rotavirus Infections , Swine Diseases , Animals , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism , Cell Membrane , Endoplasmic Reticulum-Associated Degradation , Histocompatibility Antigens Class I/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Rotavirus Infections/veterinary , Swine , Swine Diseases/metabolism
15.
Nature ; 613(7945): 743-750, 2023 01.
Article in English | MEDLINE | ID: mdl-36631610

ABSTRACT

DNA mismatch repair-deficient (MMR-d) cancers present an abundance of neoantigens that is thought to explain their exceptional responsiveness to immune checkpoint blockade (ICB)1,2. Here, in contrast to other cancer types3-5, we observed that 20 out of 21 (95%) MMR-d cancers with genomic inactivation of ß2-microglobulin (encoded by B2M) retained responsiveness to ICB, suggesting the involvement of immune effector cells other than CD8+ T cells in this context. We next identified a strong association between B2M inactivation and increased infiltration by γδ T cells in MMR-d cancers. These γδ T cells mainly comprised the Vδ1 and Vδ3 subsets, and expressed high levels of PD-1, other activation markers, including cytotoxic molecules, and a broad repertoire of killer-cell immunoglobulin-like receptors. In vitro, PD-1+ γδ T cells that were isolated from MMR-d colon cancers exhibited enhanced reactivity to human leukocyte antigen (HLA)-class-I-negative MMR-d colon cancer cell lines and B2M-knockout patient-derived tumour organoids compared with antigen-presentation-proficient cells. By comparing paired tumour samples from patients with MMR-d colon cancer that were obtained before and after dual PD-1 and CTLA-4 blockade, we found that immune checkpoint blockade substantially increased the frequency of γδ T cells in B2M-deficient cancers. Taken together, these data indicate that γδ T cells contribute to the response to immune checkpoint blockade in patients with HLA-class-I-negative MMR-d colon cancers, and underline the potential of γδ T cells in cancer immunotherapy.


Subject(s)
Colonic Neoplasms , Genes, MHC Class I , Histocompatibility Antigens Class I , Immune Checkpoint Inhibitors , Immunotherapy , Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocytes , Humans , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics , DNA Mismatch Repair/genetics , Receptors, KIR , Cell Line, Tumor , Organoids , Antigen Presentation , Genes, MHC Class I/genetics
16.
Otol Neurotol ; 43(9): e1049-e1055, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36006779

ABSTRACT

BACKGROUND AND OBJECTIVES: Vestibular schwannoma (VS), the most common intercranial schwannoma, originates from the sheath of the vestibular nerve. The growth rate of VS varies greatly, with the tumor enlarging gradually, which can compress the peripheral nerve tissue and reveal corresponding symptoms. This study was aimed to elucidate the growth mechanism of VS by analyzing cellular changes at protein, messenger ribonucleic acid (mRNA), and other molecular levels. METHODS: We determined mRNA and protein levels of ß 2 -microglobulin (ß 2 -M) and nuclear factor κB (NF-κB) in tumors of different sizes using the real-time polymerase chain reaction and Western blotting, respectively. The relationship between these factors was verified in VS primary cells cultured in vitro, and the potential role of ß 2 -M and NF-κB in VS growth was elucidated. RESULTS: In the secretions of freshly isolated tumor tissue cultured for 72 h, the concentration of ß 2 -M was positively correlated with the tumor diameter. Furthermore, tumors with larger diameter showed higher expressions of ß 2 -M and NF-κB at protein and mRNA level. ß 2 -M treatment resulted in elevated protein expression of NF-κB and also its phosphorylated form in vitro. CONCLUSION: ß 2 -M may participate in VS growth by regulating NF-κB and act as a key regulatory molecule in VS tumor growth.


Subject(s)
NF-kappa B , Neuroma, Acoustic , beta 2-Microglobulin , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Neuroma, Acoustic/genetics , RNA, Messenger/metabolism , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism
17.
Proc Natl Acad Sci U S A ; 119(22): e2200468119, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35613051

ABSTRACT

Aggregation of initially stably structured proteins is involved in more than 20 human amyloid diseases. Despite intense research, however, how this class of proteins assembles into amyloid fibrils remains poorly understood, principally because of the complex effects of amino acid substitutions on protein stability, solubility, and aggregation propensity. We address this question using ß2-microglobulin (ß2m) as a model system, focusing on D76N-ß2m that is involved in hereditary amyloidosis. This amino acid substitution causes the aggregation-resilient wild-type protein to become highly aggregation prone in vitro, although the mechanism by which this occurs remained elusive. Here, we identify the residues key to protecting ß2m from aggregation by coupling aggregation with antibiotic resistance in E. coli using a tripartite ß-lactamase assay (TPBLA). By performing saturation mutagenesis at three different sites (D53X-, D76X-, and D98X-ß2m) we show that residue 76 has a unique ability to drive ß2m aggregation in vivo and in vitro. Using a randomly mutated D76N-ß2m variant library, we show that all of the mutations found to improve protein behavior involve residues in a single aggregation-prone region (APR) (residues 60 to 66). Surprisingly, no correlation was found between protein stability and protein aggregation rate or yield, with several mutations in the APR decreasing aggregation without affecting stability. Together, the results demonstrate the power of the TPBLA to develop proteins that are resilient to aggregation and suggest a model for D76N-ß2m aggregation involving the formation of long-range couplings between the APR and Asn76 in a nonnative state.


Subject(s)
Amyloidosis , Protein Aggregation, Pathological , beta 2-Microglobulin , Amino Acid Substitution , Amyloidogenic Proteins/genetics , Amyloidosis/genetics , Enzyme Assays , Escherichia coli , Humans , Point Mutation , Protein Aggregation, Pathological/genetics , Protein Folding , beta 2-Microglobulin/chemistry , beta 2-Microglobulin/genetics , beta-Lactamases
18.
J Immunother ; 45(4): 187-193, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35343934

ABSTRACT

ß2-microglobulin (B2M) and Janus kinases 1 and 2 (JAK1/2) mutations have been suggested as genetic mechanisms of immune evasion for anti-programmed cell death protein 1 (PD-1) therapy. Whether B2M and JAK1/2 lose-of-function mutation can cause primary resistance to anti-PD-1 therapy in colorectal carcinoma (CRC) patients remains controversial. Here, we sought to compare the efficacy of anti-PD-1 therapy in DNA mismatch repair deficient/microsatellite instability-high CRC patients with or without B2M or JAK1/2 mutations. Thirty-Five CRC patients who received anti-PD-1 therapy were enrolled in this study. All tumor samples underwent next-generation sequencing. The clinical and molecular data from 110 CRC patients sequenced with the Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) assay and accessed through cBioportal were also analyzed in this study. Of the 35 CRC patients from our center, 10 (28.6%) had a B2M loss-of-function mutation, and 8 (22.9%) had a JAK1/2 loss-of-function mutation. Compared with B2M wild-type CRCs, B2M-mutated CRCs did not show a higher frequency of resistance to anti-PD-1 therapy (P=0.71). There was even better response to anti-PD-1 therapy in patients with JAK1/2 mutation than in those without (P=0.015). Of the 110 CRC patients in the MSK-IMPACT datasets, 13 (11.8%) had a B2M mutation, and 15 (13.6%) had a JAK1/2 mutation. After analyzing the response to anti-PD-1 therapy in these 110 patients, we found similar results (P=0.438 and 0.071, respectively). Moreover, patients with B2M or JAK1/2 mutation had a lower tumor mutational burden score compared with those without. B2M and JAK1/2 loss-of-function mutations occur frequently in microsatellite instability-high CRC. Our study demonstrated that patients with CRC harboring B2M or JAK1/2 mutations should not be excluded from anti-PD-1 therapy.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Immune Checkpoint Inhibitors , Neoplastic Syndromes, Hereditary , Programmed Cell Death 1 Receptor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Janus Kinase 1/genetics , Janus Kinase 2/genetics , Microsatellite Instability , Mutation , Programmed Cell Death 1 Receptor/antagonists & inhibitors , beta 2-Microglobulin/genetics
19.
Acta Biochim Biophys Sin (Shanghai) ; 54(2): 187-198, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35130623

ABSTRACT

The ΔN6 truncation is the main posttranslational modification of ß-microglobulin (ßM) found in dialysis-related amyloid. Investigation of the interaction of wild-type (WT) ßM with N-terminally truncated variants is therefore of medical relevance. However, it is unclear which residues among the six residues at the N-terminus are crucial to the interactions and the modulation of amyloid fibril propagation of ßM. We herein analyzed homo- and heterotypic seeding of amyloid fibrils of WT human ßM and its N-terminally-truncated variants ΔN1 to ΔN6, lacking up to six residues at the N-terminus. At acidic pH 2.5, we produced amyloid fibrils from recombinant, WT ßM and its six truncated variants, and found that ΔN6 ßM fibrils exhibit a significantly lower conformational stability than WT ßM fibrils. Importantly, under more physiological conditions (pH 6.2), we assembled amyloid fibrils only from recombinant, ΔN4, ΔN5, and ΔN6 ßM but not from WT ßM and its three truncated variants ΔN1 to ΔN3. Notably, the removal of the six, five or four residues at the N-terminus leads to enhanced fibril formation, and homo- and heterotypic seeding of ΔN6 fibrils strongly promotes amyloid fibril formation of WT ßM and its six truncated variants, including at more physiological pH 6.2. Collectively, these results demonstrated that the residues 4 to 6 at the N-terminus particularly modulate amyloid fibril propagation of ßM and the interactions of WT ßM with N-terminally truncated variants, potentially indicating the direct relevance to the involvement of the protein's aggregation in dialysis-related amyloidosis.


Subject(s)
Amyloid , beta 2-Microglobulin , Amyloid/chemistry , Amyloid/genetics , Humans , Hydrogen-Ion Concentration , beta 2-Microglobulin/chemistry , beta 2-Microglobulin/genetics
20.
Nat Commun ; 13(1): 182, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013322

ABSTRACT

Combining immune checkpoint therapy (ICT) and targeted therapy holds great promises for broad and long-lasting anti-cancer therapies. However, combining ICT with anti-PI3K inhibitors have been challenging because the multifaceted effects of PI3K on both cancer cells and immune cells within the tumor microenvironment. Here we find that intermittent but not daily dosing of a PI3Kα/ß/δ inhibitor, BAY1082439, on Pten-null prostate cancer models could overcome ICT resistance and unleash CD8+ T cell-dependent anti-tumor immunity in vivo. Mechanistically, BAY1082439 converts cancer cell-intrinsic immune-suppression to immune-stimulation by promoting IFNα/IFNγ pathway activation, ß2-microglubin expression and CXCL10/CCL5 secretion. With its preferential regulatory T cell inhibition activity, BAY1082439 promotes clonal expansion of tumor-associated CD8+ T cells, most likely via tertiary lymphoid structures. Once primed, tumors remain T cell-inflamed, become responsive to anti-PD-1 therapy and have durable therapeutic effect. Our data suggest that intermittent PI3K inhibition can alleviate Pten-null cancer cell-intrinsic immunosuppressive activity and turn "cold" tumors into T cell-inflamed ones, paving the way for successful ICT.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Class I Phosphatidylinositol 3-Kinases/genetics , Immune Checkpoint Inhibitors/pharmacology , PTEN Phosphohydrolase/genetics , Programmed Cell Death 1 Receptor/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Cell Movement/drug effects , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Class I Phosphatidylinositol 3-Kinases/immunology , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Gene Expression Regulation, Neoplastic , Humans , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Male , Mice , Mice, Knockout , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Signal Transduction , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
SELECTION OF CITATIONS
SEARCH DETAIL