Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31.136
Filter
1.
J Clin Invest ; 134(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39087474

ABSTRACT

Aortic aneurysms, particularly abdominal aortic aneurysms (AAAs), exhibit sex differences, with higher prevalence and severity in males than females, both in humans and experimental mouse models. In fact, male sex has been considered as the most potent nonmodifiable risk factor for AAA. Currently, there are no medications approved for the treatment of aortic aneurysms, despite the high lethality of ruptured aneurysms, which account for nearly 2% of all deaths. Moreover, the underlying molecular mechanisms mediating the sexual dimorphism of aortic aneurysms remain largely unknown. In this issue of the JCI, Mu et al. revealed a mechanism by which androgens, male sex hormones, exacerbate aortic aneurysms by suppressing programmed cell death protein 1 (PD-1) expression in T cells in an aldosterone and high salt-induced aortic aneurysm mouse model.


Subject(s)
Aortic Aneurysm, Abdominal , Programmed Cell Death 1 Receptor , Animals , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Mice , Humans , Female , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Male , Disease Models, Animal , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Androgens/metabolism
2.
Sci Rep ; 14(1): 17917, 2024 08 02.
Article in English | MEDLINE | ID: mdl-39095551

ABSTRACT

Chimeric antigen receptor (CAR) T cells are effective against hematological cancers, but are less effective against solid tumors such as non-small cell lung cancer (NSCLC). One of the reasons is that only a few cell surface targets specific for NSCLC cells have been identified. Here, we report that CD98 heavy chain (hc) protein is overexpressed on the surface of NSCLC cells and is a potential target for CAR T cells against NSCLC. Screening of over 10,000 mAb clones raised against NSCLC cell lines showed that mAb H2A011 bound to NSCLC cells but not normal lung epithelial cells. H2A011 recognized CD98hc. Although CAR T cells derived from H2A011 could not be established presumably due to the high level of H2A011 reactivity in activated T cells, those derived from the anti-CD98hc mAb R8H283, which had been shown to lack reactivity with CD98hc glycoforms expressed on normal hematopoietic cells and some normal tissues, were successfully developed. R8H283 specifically reacted with NSCLC cells in six of 15 patients. R8H283-derived CAR T cells exerted significant anti-tumor effects in a xenograft NSCLC model in vivo. These results suggest that R8H283 CAR T cells may become a new therapeutic tool for NSCLC, although careful testing for off-tumor reactivity should be performed in the future.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Immunotherapy, Adoptive , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Animals , Immunotherapy, Adoptive/methods , Mice , Cell Line, Tumor , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Xenograft Model Antitumor Assays , Antibodies, Monoclonal/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Female
3.
Nat Commun ; 15(1): 6790, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117654

ABSTRACT

Immunochemotherapy has been the mainstay of treatment for newly diagnosed diffuse large B-cell lymphoma (ndDLBCL) yet is inadequate for many patients. In this work, we perform unsupervised clustering on transcriptomic features from a large cohort of ndDLBCL patients and identify seven clusters, one called A7 with poor prognosis, and develop a classifier to identify these clusters in independent ndDLBCL cohorts. This high-risk cluster is enriched for activated B-cell cell-of-origin, low immune infiltration, high MYC expression, and copy number aberrations. We compare and contrast our methodology with recent DLBCL classifiers to contextualize our clusters and show improved prognostic utility. Finally, using pre-clinical models, we demonstrate a mechanistic rationale for IKZF1/3 degraders such as lenalidomide to overcome the low immune infiltration phenotype of A7 by inducing T-cell trafficking into tumors and upregulating MHC I and II on tumor cells, and demonstrate that TCF4 is an important regulator of MYC-related biology in A7.


Subject(s)
Gene Expression Regulation, Neoplastic , Ikaros Transcription Factor , Lenalidomide , Lymphoma, Large B-Cell, Diffuse , Proto-Oncogene Proteins c-myc , Transcription Factor 4 , Transcriptome , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Humans , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Lenalidomide/therapeutic use , Lenalidomide/pharmacology , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Transcription Factor 4/genetics , Transcription Factor 4/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Prognosis , Animals , Cell Line, Tumor , Gene Expression Profiling/methods , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , DNA Copy Number Variations
4.
Nat Commun ; 15(1): 6677, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107283

ABSTRACT

Clarification of the cytotoxic function of T cells is crucial for understanding human immune responses and immunotherapy procedures. Here, we report a high-throughput Bessel oblique plane microscopy (HBOPM) platform capable of 3D live imaging and phenotyping of chimeric antigen receptor (CAR)-modified T-cell cytotoxicity against cancer cells. The HBOPM platform has the following characteristics: an isotropic subcellular resolution of 320 nm, large-scale scouting over 400 interacting cell pairs, long-term observation across 5 hours, and quantitative analysis of the Terabyte-scale 3D, multichannel, time-lapse image datasets. Using this advanced microscopy platform, several key subcellular events in CAR-T cells are captured and comprehensively analyzed; these events include the instantaneous formation of immune synapses and the sustained changes in the microtubing morphology. Furthermore, we identify the actin retrograde flow speed, the actin depletion coefficient, the microtubule polarization and the contact area of the CAR-T/target cell conjugates as essential parameters strongly correlated with CAR-T-cell cytotoxic function. Our approach will be useful for establishing criteria for quantifying T-cell function in individual patients for all T-cell-based immunotherapies.


Subject(s)
Imaging, Three-Dimensional , Immunotherapy, Adoptive , Microtubules , Receptors, Chimeric Antigen , T-Lymphocytes , Humans , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Imaging, Three-Dimensional/methods , Immunotherapy, Adoptive/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Microtubules/metabolism , Cell Line, Tumor , Immunological Synapses/immunology , Immunological Synapses/metabolism , Cytotoxicity, Immunologic , Actins/metabolism , Microscopy/methods , Phenotype
5.
Theranostics ; 14(11): 4481-4498, 2024.
Article in English | MEDLINE | ID: mdl-39113807

ABSTRACT

Rationale: Since oncogene expression products often exhibit upregulation or abnormally activated activity, developing a technique to regulate abnormal protein levels represent a viable approach for treating tumors and protein abnormality-related diseases. Methods: We first screened out eMIATAC components with high targeted degradation efficiency and explored the mechanism by which eMIATAC induced target protein degradation, and verified the degradation efficiency of the target protein by protein imprinting and flow cytometry. Next, we recombined eMIATAC with some controllable elements to verify the regulatable degradation performance of the target protein. Subsequently, we constructed eMIATAC that can express targeted degradation of AKT1 and verified its effect on GBM cell development in vitro and in vivo. Finally, we concatenated eMIATAC with CAR sequences to construct CAR-T cells with low BATF protein levels and verified the changes in their anti-tumor efficacy. Results: we developed a system based on the endosome-microautophagy-lysosome pathway for degrading endogenous proteins: endosome-MicroAutophagy TArgeting Chimera (eMIATAC), dependent on Vps4A instead of lysosomal-associated membrane protein 2A (LAMP2A) to bind to the chaperone Hsc70 and the protein of interest (POI). The complex was then transported to the lysosome by late endosomes, where degradation occurred similarly to microautophagy. The eMIATACs demonstrated accuracy, efficiency, reversibility, and controllability in degrading the target protein EGFP. Moreover, eMIATAC exhibited excellent performance in knocking down POI when targeting endogenous proteins in vivo and in vitro. Conclusions: The eMIATACs could not only directly knock down abnormal proteins for glioma treatment but also enhance the therapeutic effect of CAR-T cell therapy for tumors by knocking down T cell exhaustion-related proteins. The newly developed eMIATAC system holds promise as a novel tool for protein knockdown strategies. By enabling direct control over endogenous protein levels, eMIATAC has the potential to revolutionize treatment for cancer and genetic diseases.


Subject(s)
Autophagy , Endosomes , Immunotherapy, Adoptive , Proteolysis , Humans , Animals , Endosomes/metabolism , Cell Line, Tumor , Mice , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/metabolism , Glioblastoma/therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomal-Associated Membrane Protein 2/genetics , Xenograft Model Antitumor Assays , HSC70 Heat-Shock Proteins/metabolism , Lysosomes/metabolism , T-Lymphocytes/metabolism
6.
J Immunother Cancer ; 12(8)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39111833

ABSTRACT

BACKGROUND: High-grade gliomas including glioblastoma (GBM) and diffuse midline gliomas (DMG) represent the most lethal and aggressive brain cancers where current treatment modalities offer limited efficacy. Chimeric antigen receptor (CAR) T cell therapies have emerged as a promising strategy, boasting tumor-specific targeting and the unique ability to penetrate the blood-brain barrier. However, the effective clinical application hinges on the optimal choice of antigen, with a limited number, currently under investigation. METHODS: We employed cell surface proteomic analysis of primary human high-grade glioma samples from both adult and pediatric patients. This led to the identification of Ephrin type-A receptor 3 (EphA3) as a prevalently expressed target. We engineered a second-generation EphA3-targeted CAR T cell and assessed function using in vitro and in vivo models of GBM and DMG. RESULTS: EphA3-targeted CAR T cells demonstrated robust antigen-specific killing of human GBM and DMG cell lines in vitro. In an orthotopic xenograft NSG mouse model, EphA3-targeted CAR T cells not only effectively eradicated tumors but also established a functional T cell population protective on rechallenge. Remarkably, mice rechallenged with a second contralateral orthotopic tumor implantation achieved complete tumor clearance and maintained a sustained complete response 6 months following initial treatment. CONCLUSION: Building on the proven safety profile of EphA3 antibodies in clinical settings, our study provides compelling preclinical evidence supporting the efficacy of EphA3-targeted CAR T cells against high-grade gliomas. These findings underscore the potential for transitioning this innovative therapy into clinical trials, aiming to revolutionize the treatment landscape for patients afflicted with these formidable brain cancers.


Subject(s)
Glioma , Receptor, EphA3 , Receptors, Chimeric Antigen , Humans , Animals , Mice , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Glioma/therapy , Glioma/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Xenograft Model Antitumor Assays , Immunotherapy, Adoptive/methods , Cell Line, Tumor , Female , Immunologic Memory
7.
PLoS One ; 19(8): e0307450, 2024.
Article in English | MEDLINE | ID: mdl-39178184

ABSTRACT

Adenosine to inosine (A-to-I) RNA editing by ADAR1 has been implicated in maintaining self-tolerance, preventing autoimmunity, and mediating antiviral immunity. Foreign viral double-stranded RNA triggers rapid interferon response and activates ADAR1 in the host immune system. Emerging data points to a role of ADAR1 A-to-I editing in the inflammatory response associated with severe COVID-19 disease. We identify A-to-I editing events within human whole transcriptome data from SARS-CoV-2 infected individuals, non-infected individuals, and individuals with other viral illnesses from nasopharyngeal swabs. High levels of RNA editing in host cells are associated with low SARS-CoV-2 viral load (p = 9.27 E-06), suggesting an inhibitory effect of ADAR1 on viral infection. Additionally, we find differentially expressed genes associated with RNA-modifications and interferon response. Single cell RNA-sequencing analysis of SARS-CoV-2 infected nasopharyngeal swabs reveals that cytotoxic CD8 T cells upregulate ADAR1 in COVID-19 positive samples (p = 0.0269). We further reveal ADAR1 expression increases with CD4 and CD8 T cell activation, and knockdown of ADAR1 leads to apoptosis and aberrant IL-2 secretion. Together, our data suggests A-to-I RNA editing is required to maintain healthy homeostasis of activated T cells to combat SARS-CoV-2 infection.


Subject(s)
Adenosine Deaminase , COVID-19 , Homeostasis , RNA Editing , RNA-Binding Proteins , SARS-CoV-2 , Humans , COVID-19/immunology , COVID-19/virology , COVID-19/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , SARS-CoV-2/physiology , SARS-CoV-2/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , CD8-Positive T-Lymphocytes/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Viral Load , Inosine/metabolism , Adenosine/metabolism , Lymphocyte Activation/immunology
8.
Front Immunol ; 15: 1415328, 2024.
Article in English | MEDLINE | ID: mdl-39192973

ABSTRACT

Background: The non-viral production of CAR-T cells through electroporation of transposon DNA plasmids is an alternative approach to lentiviral/retroviral methods. This method is particularly suitable for early-phase clinical trials involving novel types of CAR-T cells. The primary disadvantage of non-viral methods is the lower production efficiency compared to viral-based methods, which becomes a limiting factor for CAR-T production, especially in chemotherapy-pretreated lymphopenic patients. Methods: We describe a good manufacturing practice (GMP)-compliant protocol for producing CD19 and CD123-specific CAR-T cells based on the electroporation of transposon vectors. The lymphocytes were purified from the blood of patients undergoing chemotherapy for B-NHL or AML and were electroporated with piggyBac transposon encoding CAR19 or CAR123, respectively. Electroporated cells were then polyclonally activated by anti-CD3/CD28 antibodies and a combination of cytokines (IL-4, IL-7, IL-21). The expansion was carried out in the presence of irradiated allogeneic blood-derived mononuclear cells (i.e., the feeder) for up to 21 days. Results: Expansion in the presence of the feeder enhanced CAR-T production yield (4.5-fold in CAR19 and 9.3-fold in CAR123). Detailed flow-cytometric analysis revealed the persistence of early-memory CAR-T cells and a low vector-copy number after production in the presence of the feeder, with no negative impact on the cytotoxicity of feeder-produced CAR19 and CAR123 T cells. Furthermore, large-scale manufacturing of CAR19 carried out under GMP conditions using PBMCs obtained from B-NHL patients (starting number=200x10e6 cells) enabled the production of >50x10e6 CAR19 in 7 out of 8 cases in the presence of the feeder while only in 2 out of 8 cases without the feeder. Conclusions: The described approach enables GMP-compatible production of sufficient numbers of CAR19 and CAR123 T cells for clinical application and provides the basis for non-viral manufacturing of novel experimental CAR-T cells that can be tested in early-phase clinical trials. This manufacturing approach can complement and advance novel experimental immunotherapeutic strategies against human hematologic malignancies.


Subject(s)
Antigens, CD19 , DNA Transposable Elements , Immunotherapy, Adoptive , Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive/methods , Antigens, CD19/immunology , Antigens, CD19/genetics , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/genetics , Feeder Cells , Lymphoma, B-Cell/therapy , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Electroporation , Allogeneic Cells/immunology
9.
Cancer Res Commun ; 4(8): 2255-2266, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39099200

ABSTRACT

Recent insights have identified adrenergic (ADRN) and mesenchymal (MES) cell lineages as distinct biologic cell types and T-cell inflammation as a prognostic marker in neuroblastoma. We hypothesized that elucidating unique and overlapping aspects of these biologic features could serve as novel biomarkers for informing ongoing efforts to improve therapeutic approaches for children with high-risk neuroblastoma. We identified lineage-specific, single-stranded super-enhancers to define ADRN and MES specific genes. Publicly available RNA-seq of diagnostic tumor biopsies was used in Discovery and Validation cohorts. Each tumor was assigned a relative MES score and T-cell inflammation (TCI) score. Survival was assessed using the Kaplan-Meier method, and differences were assessed by the log-rank test. Inflammation scores were correlated with MES scores and anticorrelated with MYCN-amplification in both cohorts. Among patients with high-risk, ADRN tumors, those with TCI tumors had superior overall survival to those with non-inflamed tumors. A similar, but nonsignificant, trend was observed in the Validation cohort. Conversely, there was no difference according to TCI status in the MES cohort in either the Discover or Validation cohorts. High-inflammation scores were correlated with improved survival in some patients with high-risk, ADRN but not MES neuroblastoma. Our findings bolster support for further developing T-cell-based and immunotherapy-based approaches for children with high-risk neuroblastoma of varying MES and ADRN expression. SIGNIFICANCE: Adrenergic (ADRN) and mesenchymal (MES) lineages are distinct biologic cell types in neuroblastoma. We defined ADRN and MES specific genes and found that high-risk, ADRN tumors harboring elevated T-cell inflammation signatures had superior overall survival. Our findings bolster support for further developing immunotherapy-based approaches for children with high-risk neuroblastoma.


Subject(s)
Inflammation , Neuroblastoma , T-Lymphocytes , Humans , Neuroblastoma/mortality , Neuroblastoma/pathology , Neuroblastoma/immunology , Neuroblastoma/genetics , Inflammation/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Prognosis , Male , Female , Child, Preschool , Biomarkers, Tumor/genetics , Infant , Child , Gene Expression Regulation, Neoplastic
10.
Sci Adv ; 10(34): eadl3975, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39167656

ABSTRACT

Genetic TNFAIP3 (A20) inactivation is a classical somatic lymphoma lesion and the genomic trait in haploinsufficiency of A20 (HA20). In a cohort of 34 patients with HA20, we show that heterozygous TNFAIP3 loss skews immune repertoires toward lymphocytes with classical self-reactive antigen receptors typically found in B and T cell lymphomas. This skewing was mediated by a feed-forward tumor necrosis factor (TNF)/A20/nuclear factor κB (NF-κB) loop that shaped pre-lymphoma transcriptome signatures in clonally expanded B (CD81, BACH2, and NEAT1) or T (GATA3, TOX, and PDCD1) cells. The skewing was reversed by anti-TNF treatment but could also progress to overt lymphoma. Analysis of conditional TNFAIP3 knock-out mice reproduced the wiring of the TNF/A20/NF-κB signaling axis with permissive antigen receptors and suggested a distinct regulation in B and T cells. Together, patients with the genetic disorder HA20 provide an exceptional window into A20/TNF/NF-κB-mediated control of immune homeostasis and early steps of lymphomagenesis that remain clinically unrecognized.


Subject(s)
Haploinsufficiency , Homeostasis , NF-kappa B , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Animals , Humans , Mice , NF-kappa B/metabolism , Mice, Knockout , Female , Male , Signal Transduction , Middle Aged , Lymphocytes/immunology , Lymphocytes/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Adult , Tumor Necrosis Factor-alpha/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/pathology
11.
Nat Commun ; 15(1): 7265, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39179539

ABSTRACT

Rosacea patients show facial hypersensitivity to stimulus factors (such as heat and capsaicin); however, the underlying mechanism of this hyperresponsiveness remains poorly defined. Here, we show capsaicin stimulation in mice induces exacerbated rosacea-like dermatitis but has no apparent effect on normal skin. Nociceptor ablation substantially reduces the hyperresponsiveness of rosacea-like dermatitis. Subsequently, we find that γδ T cells express Ramp1, the receptor of the neuropeptide CGRP, and are in close contact with these nociceptors in the skin. γδ T cells are significantly increased in rosacea skin lesions and can be further recruited and activated by neuron-secreted CGRP. Rosacea-like dermatitis is reduced in T cell receptor δ-deficient (Tcrd-/-) mice, and the nociceptor-mediated aggravation of rosacea-like dermatitis is also reduced in these mice. In vitro experiments show that CGRP induces IL17A secretion from γδ T cells by regulating inflammation-related and metabolism-related pathways. Finally, rimegepant, a CGRP receptor antagonist, shows efficacy in the treatment of rosacea-like dermatitis. In conclusion, our findings demonstrate a neuron-CGRP-γδT cell axis that contributes to the hyperresponsiveness of rosacea, thereby showing that targeting CGRP is a potentially effective therapeutic strategy for rosacea.


Subject(s)
Calcitonin Gene-Related Peptide , Capsaicin , Receptors, Antigen, T-Cell, gamma-delta , Rosacea , Sensory Receptor Cells , Animals , Rosacea/immunology , Mice , Calcitonin Gene-Related Peptide/metabolism , Sensory Receptor Cells/metabolism , Capsaicin/pharmacology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/genetics , Skin/pathology , Skin/immunology , Skin/metabolism , Interleukin-17/metabolism , Interleukin-17/immunology , Mice, Knockout , Mice, Inbred C57BL , Dermatitis/immunology , Dermatitis/metabolism , Dermatitis/pathology , Disease Models, Animal , Male , Nociceptors/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Humans , Receptors, Calcitonin Gene-Related Peptide/metabolism
12.
Front Immunol ; 15: 1377955, 2024.
Article in English | MEDLINE | ID: mdl-39165363

ABSTRACT

Ataxia telangiectasia (AT) is a rare autosomal-recessive disorder characterized by profound neurodegeneration, combined immunodeficiency, and an increased risk for malignant diseases. Treatment options for AT are limited, and the long-term survival prognosis for patients remains grim, primarily due to the emergence of chronic respiratory pathologies, malignancies, and neurological complications. Understanding the dysregulation of the immune system in AT is fundamental for the development of novel treatment strategies. In this context, we performed a retrospective longitudinal immunemonitoring of lymphocyte subset distribution in a cohort of AT patients (n = 65). Furthermore, we performed FACS analyses of peripheral blood mononuclear cells from a subgroup of 12 AT patients to examine NK and T cells for the expression of activating and functional markers. We observed reduced levels of peripheral blood CD3+CD8+ cytotoxic T cells, CD3+CD4+ T helper cells, and CD19+ B cells, whereas the amount of CD3--CD56+ NK cells and CD3+CD56+ NKT-like cells was similar compared with age-matched controls. Notably, there was no association between the age-dependent kinetic of T-, B-, or NK-cell counts and the occurrence of malignancy in AT patients. Additionally, our results indicate an altered NK- and T-cell response to cytokine stimulation in AT with increased levels of TRAIL, FasL, and CD16 expression in NK cells, as well as an elevated activation level of T cells in AT with notably higher expression levels of IFN-γ, CD107a, TRAIL, and FasL. Together, these findings imply function alterations in AT lymphocytes, specifically in T and NK cells, shedding light on potential pathways for innovative therapies.


Subject(s)
Ataxia Telangiectasia , Killer Cells, Natural , Humans , Ataxia Telangiectasia/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Female , Child , Adolescent , Adult , Retrospective Studies , Child, Preschool , Young Adult , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Immunophenotyping
13.
Curr Protoc ; 4(8): e1107, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39166803

ABSTRACT

CAR-T cell therapy has emerged as a potent and effective tool in the immunotherapy of refractory cancers. However, challenges exist in their clinical application, necessitating extensive preclinical research to optimize their function. Various preclinical in vitro and in vivo models have been proposed for such purpose; among which immunocompetent mouse models serve as an invaluable tool in studying host immune interactions within a more realistic simulation of the tumor milieu. We hereby describe a standardized protocol for the generation of high-titer γ-retroviral vectors through transfection of the HEK293T packaging cell line. The virus-containing supernatant is further concentrated using an inhouse concentrator solution, titrated, and applied to mouse T cells purified via a convenient and rapid method by nylon-wool columns. Using the method presented here, we were able to achieve high titer γ-retrovirus and highly pure mouse T cells with desirable CAR transduction efficiency. The mouse CAR T cells produced through this protocol demonstrate favorable CAR expression and viability, thus making them suitable for further in vitro/in vivo assays. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Production of γ-retroviral vectors from retrovirus-backbone plasmids Basic Protocol 2: Concentration of γ-retrovirus-containing supernatants Basic Protocol 3: Titration of concentrated γ-retrovirus Basic Protocol 4: Isolation and activation of mouse T cells Basic Protocol 5: Transduction of activated mouse T cells, assessment of CAR expression, and expansion of CAR T cells for further in vitro/in vivo studies Support Protocol: Surface staining of cells for flow cytometric assessment of CAR expression.


Subject(s)
Receptors, Chimeric Antigen , T-Lymphocytes , Animals , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Mice , Humans , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , HEK293 Cells , Immunotherapy, Adoptive/methods , Disease Models, Animal , Retroviridae/genetics , Neoplasms/immunology , Neoplasms/therapy , Genetic Vectors
14.
J Exp Med ; 221(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39167073

ABSTRACT

The rate at which cells enter the T cell pathway depends not only on the immigration of hematopoietic precursors into the strong Notch signaling environment of the thymus but also on the kinetics with which each individual precursor cell reaches T-lineage commitment once it arrives. Notch triggers a complex, multistep gene regulatory network in the cells in which the steps are stereotyped but the transition speeds between steps are variable. Progenitor-associated transcription factors delay T-lineage differentiation even while Notch-induced transcription factors within the same cells push differentiation forward. Progress depends on regulator cross-repression, on breaching chromatin barriers, and on shifting, competitive collaborations between stage-specific and stably expressed transcription factors, as reviewed here.


Subject(s)
Cell Differentiation , Gene Regulatory Networks , Receptors, Notch , T-Lymphocytes , T-Lymphocytes/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Cell Differentiation/genetics , Humans , Receptors, Notch/metabolism , Receptors, Notch/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Signal Transduction , Cell Lineage/genetics , Transcription, Genetic
15.
ACS Nano ; 18(34): 23741-23756, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39158207

ABSTRACT

Cell backpacks present significant potential in both therapeutic and diagnostic applications, making it essential to further explore their interactions with host cells. Current evidence indicates that backpacks can induce sustained immune responses. Our original objective was to incorporate a model antigen into the backpacks to promote dendritic cell maturation and facilitate antigen presentation, thereby inducing immune responses. However, we unexpectedly discovered that both antigen-loaded backpacks and empty backpacks demonstrated comparable abilities to induce dendritic cell maturation, resulting in nearly identical potency in T-cell proliferation. Our mechanistic studies suggest that the attachment of backpacks induces mechanical forces on dendritic cells via opening the PIEZO1 mechanical ion channel. This interaction leads to the remodeling of the intracellular cytoskeleton and facilitates the production of type I interferons by dendritic cells. Consequently, the mechano-immune-driven dendritic cell backpacks, when combined with radiotherapy, induce a robust antitumor effect. This research presents an avenue for leveraging mechanotransduction to enhance combination immunotherapeutic strategies, potentially leading to groundbreaking advancements in the field.


Subject(s)
Dendritic Cells , Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Mice , Mechanotransduction, Cellular/immunology , Mice, Inbred C57BL , Humans , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/pathology , Neoplasms/radiotherapy , Cell Proliferation/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
16.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(7): 614-622, 2024 Jul.
Article in Chinese | MEDLINE | ID: mdl-39179404

ABSTRACT

Objective To explore the potential of the cell surface receptor c-Met as an effective target for chimeric antigen receptor T-cell (CAR-T) therapy in colorectal cancer. Methods The bioinformatics was used to analyze the specific expression of c-Met in colorectal adenocarcinoma (COAD) and its clinical significance. c-Met protein expression was detected by immunohistochemistry in tumor tissues obtained from colorectal cancer patients. Flow cytometry was utilized to assess the expression of c-Met in the HCT116 human colorectal cancer cell line. Additionally, primary T cells isolated from human peripheral blood mononuclear cells (PBMCs) were transduced with a lentivirus to generate second-generation CAR-T cells targeting c-Met, followed by an observation of the inhibitory effects of these c-Met-targeted CAR-T cells on HCT116 cells. Results Immunohistochemistry and bioinformatics data both demonstrated that c-Met was over-expressed in COAD, with patients exhibiting relatively lower expression showing better prognosis. In normal colonic tissue, c-Met was either expressed at low levels or not expressed. Flow cytometry revealed high expression of c-Met in HCT116 cells as well. The c-Met-targeted CAR-T cells were capable of specifically recognizing and targeting antigen-expressing tumor cells. CAR-T cells proliferated specifically under antigenic stimulation, exerting cytotoxic effects on cancer cells and releasing cytokines interleukin 2 (IL-2) and interferon-gamma (IFN-γ), thereby demonstrating the biological functions. Conclusion c-Met may be a promising therapeutic target in COAD; c-Met-targeted CAR-T cells demonstrate inhibitory effects on colorectal cancer cells in vitro.


Subject(s)
Colorectal Neoplasms , Computational Biology , Proto-Oncogene Proteins c-met , Receptors, Chimeric Antigen , Humans , Colorectal Neoplasms/therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/genetics , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , HCT116 Cells , Epithelial-Mesenchymal Transition , Immunotherapy, Adoptive/methods , Cell Line, Tumor , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
Cell Transplant ; 33: 9636897241273689, 2024.
Article in English | MEDLINE | ID: mdl-39180383

ABSTRACT

Immune rejection presents a significant challenge in xenogenic meniscal transplantation. Pigs are widely regarded as an advantageous tissue source for such transplants, with porcine GGTA1, CMAH, and B4GALNT2 being among the most common xenoreactive antigen (Ag) genes. While some studies have suggested that allogeneic meniscus (AM) transplants may exhibit immunoprivileged properties, our study observed slight immunological rejection has been observed following contact between human meniscal cells (HMCs) and human peripheral blood mononuclear cells (PBMCs). Given the limited systematic research on immune responses following xenograft meniscus transplantation, we established porcine meniscus transplantation (PMT) models to comprehensively assess the immunogenicity of porcine meniscus (PM) from both innate and adaptive immune perspectives. Our investigations confirmed that PMT beneath the epidermis led to innate cell infiltration into the xenografts and T-cell activation in local lymph nodes. T-cell activation upregulated the interleukin (IL)-17 signaling pathway, disrupting collagen organization and metabolic processes, thereby hindering PM regeneration. Using freeze-thaw treatment on PM alleviated T-cell activation post-transplantation by eliminating xenogenic DNA. In vitro findings demonstrated that gene editing in porcine meniscal cells (PMCs) suppressed human T-cell activation by downregulating the expression of xenoreactive Ag genes. These results suggest that GGTA1/CMAH/B4GALNT2 knockout (KO) pigs hold significant promise for advancing the field of meniscal transplantation.


Subject(s)
Galactosyltransferases , Graft Rejection , Meniscus , T-Lymphocytes , Animals , Swine , Humans , Graft Rejection/immunology , Galactosyltransferases/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Down-Regulation , Antigens, Heterophile/immunology , Transplantation, Heterologous , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , Freezing , Mixed Function Oxygenases
18.
Stem Cell Res Ther ; 15(1): 257, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39135206

ABSTRACT

BACKGROUND: Using natural killer (NK) cells to treat hematopoietic and solid tumors has great promise. Despite their availability from peripheral blood and cord blood, stem cell-derived NK cells provide an "off-the-shelf" solution. METHODS: In this study, we developed two CAR-NK cells targeting PD-L1 derived from lentiviral transduction of human umbilical cord blood (UCB)-CD34+ cells and UCB-CD34+-derived NK cells. The transduction efficiencies and in vitro cytotoxic functions including degranulation, cytokine production, and cancer cell necrosis of both resultants PD-L1 CAR-NK cells were tested in vitro on two different PD-L1 low and high-expressing solid tumor cell lines. RESULTS: Differentiated CAR­modified UCB-CD34+ cells exhibited enhanced transduction efficiency. The expression of anti-PD-L1 CAR significantly (P < 0.05) enhanced the cytotoxicity of differentiated CAR­modified UCB-CD34+ cells and CAR-modified UCB-CD34+-derived NK cells against PD-L1 high-expressing tumor cell line. In addition, CAR-modified UCB-CD34+-derived NK cells significantly (P < 0.05) restored the tumor-killing ability of exhausted PD-1 high T cells. CONCLUSION: Considering the more efficient transduction in stem cells and the possibility of producing CAR-NK cell products with higher yields, this approach is recommended for studies in the field of CAR-NK cells. Also, a pre-clinical study is now necessary to evaluate the safety and efficacy of these two CAR-NK cells individually and in combination with other therapeutic approaches.


Subject(s)
Antigens, CD34 , B7-H1 Antigen , Fetal Blood , Killer Cells, Natural , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Fetal Blood/cytology , Antigens, CD34/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Differentiation , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Programmed Cell Death 1 Receptor/metabolism , Cell Line, Tumor , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/pathology
19.
Proc Natl Acad Sci U S A ; 121(33): e2318190121, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39106307

ABSTRACT

We developed a highly sensitive assay for detecting protein-protein interaction using chimeric receptors comprising two molecules of interest in the extracellular domain and interferon alpha and beta receptor subunit 1 or 2 (IFNAR1/2) in the intracellular domain. This intracellular IFNAR1/2 reconstitution system (IFNARRS) proved markedly more sensitive than the NanoBiT system, currently considered one of the best detection systems for protein interaction. Employing chimeric receptors with extracellular domains from the IFNγ or IL-2 receptor and the intracellular domains of IFNAR1/2, the IFNARRS system effectively identifies low IFNγ or IL-2 levels. Cells stably expressing these chimeric receptors responded to IFNγ secreted by activated T cells following various stimuli, including a specific peptide-antigen. The activation signals were further enhanced by the expression of relevant genes, such as costimulators, via IFN-stimulated response elements in the promoters. Besides IFNγ or IL-2, the IFNARRS system demonstrated the capability to detect other cytokines by using the corresponding extracellular domains from these target cytokine receptors.


Subject(s)
Interferon-gamma , Interleukin-2 , Receptor, Interferon alpha-beta , T-Lymphocytes , Humans , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Interleukin-2/metabolism , Interferon-gamma/metabolism , Receptors, Interleukin-2/metabolism , Receptors, Interleukin-2/genetics , Protein Binding , Lymphocyte Activation , HEK293 Cells
20.
Cell Rep Methods ; 4(8): 100841, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39127046

ABSTRACT

Cell-type-specific domains are the anatomical domains in spatially resolved transcriptome (SRT) tissues where particular cell types are enriched coincidentally. It is challenging to use existing computational methods to detect specific domains with low-proportion cell types, which are partly overlapped with or even inside other cell-type-specific domains. Here, we propose De-spot, which synthesizes segmentation and deconvolution as an ensemble to generate cell-type patterns, detect low-proportion cell-type-specific domains, and display these domains intuitively. Experimental evaluation showed that De-spot enabled us to discover the co-localizations between cancer-associated fibroblasts and immune-related cells that indicate potential tumor microenvironment (TME) domains in given slices, which were obscured by previous computational methods. We further elucidated the identified domains and found that Srgn may be a critical TME marker in SRT slices. By deciphering T cell-specific domains in breast cancer tissues, De-spot also revealed that the proportions of exhausted T cells were significantly increased in invasive vs. ductal carcinoma.


Subject(s)
Breast Neoplasms , Transcriptome , Tumor Microenvironment , Tumor Microenvironment/immunology , Humans , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/immunology , Female , Gene Expression Profiling/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Mice , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology
SELECTION OF CITATIONS
SEARCH DETAIL