Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 882
Filter
1.
Cell Rep Methods ; 4(9): 100857, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39260365

ABSTRACT

We present a TALEN-based workflow to generate and maintain dual-edited (IL-15+/+/TGFßR2-/-) iPSCs that produce enhanced iPSC-derived natural killer (iNK) cells for cancer immunotherapy. It involves using a cell lineage promoter for knocking in (KI) gene(s) to minimize the potential effects of expression of any exogenous genes on iPSCs. As a proof-of-principle, we KI IL-15 under the endogenous B2M promoter and show that it results in high expression of the sIL-15 in iNK cells but minimal expression in iPSCs. Furthermore, given that it is known that knockout (KO) of TGFßR2 in immune cells can enhance resistance to the suppressive TGF-ß signaling in the tumor microenvironment, we develop a customized medium containing Nodal that can maintain the pluripotency of iPSCs with TGFßR2 KO, enabling banking of these iPSC clones. Ultimately, we show that the dual-edited IL-15+/+/TGFßR2-/- iPSCs can be efficiently differentiated into NK cells that show enhanced autonomous growth and are resistant to the suppressive TGF-ß signaling.


Subject(s)
Induced Pluripotent Stem Cells , Interleukin-15 , Killer Cells, Natural , Receptor, Transforming Growth Factor-beta Type II , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Interleukin-15/genetics , Interleukin-15/metabolism , Humans , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Cell Differentiation , Transcription Activator-Like Effector Nucleases/metabolism , Transcription Activator-Like Effector Nucleases/genetics , Gene Editing/methods
2.
Cancer Cell ; 42(8): 1450-1466.e11, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39137729

ABSTRACT

Glioblastoma (GBM) is an aggressive brain cancer with limited therapeutic options. Natural killer (NK) cells are innate immune cells with strong anti-tumor activity and may offer a promising treatment strategy for GBM. We compared the anti-GBM activity of NK cells engineered to express interleukin (IL)-15 or IL-21. Using multiple in vivo models, IL-21 NK cells were superior to IL-15 NK cells both in terms of safety and long-term anti-tumor activity, with locoregionally administered IL-15 NK cells proving toxic and ineffective at tumor control. IL-21 NK cells displayed a unique chromatin accessibility signature, with CCAAT/enhancer-binding proteins (C/EBP), especially CEBPD, serving as key transcription factors regulating their enhanced function. Deletion of CEBPD resulted in loss of IL-21 NK cell potency while its overexpression increased NK cell long-term cytotoxicity and metabolic fitness. These results suggest that IL-21, through C/EBP transcription factors, drives epigenetic reprogramming of NK cells, enhancing their anti-tumor efficacy against GBM.


Subject(s)
Brain Neoplasms , CCAAT-Enhancer-Binding Protein-delta , Glioblastoma , Interleukins , Killer Cells, Natural , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Glioblastoma/immunology , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/therapy , Interleukins/genetics , Interleukins/metabolism , Interleukins/immunology , Humans , Animals , Mice , CCAAT-Enhancer-Binding Protein-delta/metabolism , CCAAT-Enhancer-Binding Protein-delta/genetics , Brain Neoplasms/immunology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Interleukin-15/genetics , Interleukin-15/metabolism , Interleukin-15/immunology , Xenograft Model Antitumor Assays
3.
J Exp Med ; 221(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39167075

ABSTRACT

Changes in mechanosensitive ion channels following radiation have seldom been linked to therapeutic sensitivity or specific factors involved in antitumor immunity. Here, in this study, we found that the mechanical force sensor, Piezo2, was significantly upregulated in tumor cells after radiation, and Piezo2 knockout in tumor cells enhanced tumor growth suppression by radiotherapy. Specifically, loss of Piezo2 in tumor cells induced their IL-15 expression via unleashing JAK2/STAT1/IRF-1 axis after radiation. This increase in IL-15 activates IL-15Rα on tumor-infiltrating CD8+ T cells, thereby leading to their augmented effector and stem cell-like properties, along with reduced terminal exhausted feature. Importantly, Piezo2 expression was negatively correlated with CD8 infiltration, as well as with radiosensitivity of patients with rectum adenocarcinoma receiving radiotherapy treatment. Together, our findings reveal that tumor cell-intrinsic Piezo2 induces radioresistance by dampening the IRF-1/IL-15 axis, thus leading to impaired CD8+ T cell-dependent antitumor responses, providing insights into the further development of combination strategies to treat radioresistant cancers.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-15 , Ion Channels , Radiation Tolerance , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Animals , Humans , Ion Channels/metabolism , Ion Channels/genetics , Radiation Tolerance/genetics , Mice , Interleukin-15/metabolism , Interleukin-15/genetics , Cell Line, Tumor , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Mice, Inbred C57BL , Female , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Male , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Signal Transduction
4.
J Clin Invest ; 134(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39087476

ABSTRACT

NK cells are cytotoxic innate immune cells involved in antitumor immunity, and they provide a treatment option for patients with acute myeloid leukemia (AML). In this issue of the JCI, Cubitt et al. investigated the role of CD8α, a coreceptor present on approximately 40% of human NK cells. IL-15 stimulation of CD8α- NK cells induced CD8α expression via the RUNX3 transcription factor, driving formation of a unique induced CD8α (iCD8α+) population. iCD8α+ NK cells displayed higher proliferation, metabolic activity, and antitumor cytotoxic function compared with preexisting CD8α+ and CD8α- subsets. Therefore, CD8α expression can be used to define a potential dynamic spectrum of NK cell expansion and function. Because these cells exhibit enhanced tumor control, they may be used to improve in NK cell therapies for patients with AML.


Subject(s)
CD8 Antigens , Core Binding Factor Alpha 3 Subunit , Interleukin-15 , Killer Cells, Natural , Leukemia, Myeloid, Acute , Humans , CD8 Antigens/metabolism , CD8 Antigens/immunology , CD8 Antigens/genetics , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Core Binding Factor Alpha 3 Subunit/immunology , Interleukin-15/immunology , Interleukin-15/metabolism , Interleukin-15/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism
5.
Mol Ther ; 32(8): 2728-2740, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38879754

ABSTRACT

Despite the remarkable success of chimeric antigen receptor (CAR) T therapy in hematological malignancies, its efficacy in solid tumors remains limited. Cytokine-engineered CAR T cells offer a promising avenue, yet their clinical translation is hindered by the risks associated with constitutive cytokine expression. In this proof-of-concept study, we leverage the endogenous interferon (IFN)-γ promoter for transgenic interleukin (IL)-15 expression. We demonstrate that IFN-γ expression is tightly regulated by T cell receptor signaling. By introducing an internal ribosome entry site IL15 into the 3' UTR of the IFN-γ gene via homology directed repair-mediated knock-in, we confirm that IL-15 expression can co-express with IFN-γ in an antigen stimulation-dependent manner. Importantly, the insertion of transgenes does not compromise endogenous IFN-γ expression. In vitro and in vivo data demonstrate that IL-15 driven by the IFN-γ promoter dramatically improves CAR T cells' antitumor activity, suggesting the effectiveness of IL-15 expression. Last, as a part of our efforts toward clinical translation, we have developed an innovative two-gene knock-in approach. This approach enables the simultaneous integration of CAR and IL-15 genes into TRAC and IFN-γ gene loci using a single AAV vector. CAR T cells engineered to express IL-15 using this approach demonstrate enhanced antitumor efficacy. Overall, our study underscores the feasibility of utilizing endogenous promoters for transgenic cytokines expression in CAR T cells.


Subject(s)
Immunotherapy, Adoptive , Interferon-gamma , Interleukin-15 , Promoter Regions, Genetic , Receptors, Chimeric Antigen , Interferon-gamma/metabolism , Humans , Animals , Mice , Immunotherapy, Adoptive/methods , Interleukin-15/genetics , Interleukin-15/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Genetic Vectors/genetics , Cell Line, Tumor , Transgenes , Cytokines/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Xenograft Model Antitumor Assays , Gene Expression
6.
Front Immunol ; 15: 1404891, 2024.
Article in English | MEDLINE | ID: mdl-38919611

ABSTRACT

Background: Inflammatory cytokines play key pathogenic roles in liver fibrosis. IL-15 is a proinflammatory cytokine produced by myeloid cells. IL-15 promotes pathogenesis of several chronic inflammatory diseases. However, increased liver fibrosis has been reported in mice lacking IL-15 receptor alpha chain (IL-15Rα), suggesting an anti-fibrogenic role for IL-15. As myeloid cells are key players in liver fibrosis and IL-15 signaling can occur independently of IL-15Rα, we investigated the requirement of IL-15 and IL-15Rα in liver fibrosis. Methods: We induced liver fibrosis in Il15-/- , Il15ra-/- and wildtype C57BL/6 mice by the administration of carbon tetrachloride (CCl4). Liver fibrosis was evaluated by Sirius red and Mason's trichrome staining and α-smooth muscle acting immunostaining of myofibroblasts. Gene expression of collagens, matrix modifying enzymes, cytokines and chemokines was quantified by RT-qPCR. The phenotype and the numbers of intrahepatic lymphoid and myeloid cell subsets were evaluated by flow cytometry. Results: Both Il15-/- and Il15ra-/- mice developed markedly reduced liver fibrosis compared to wildtype control mice, as revealed by reduced collagen deposition and myofibroblast content. Il15ra-/- mice showed further reduction in collagen deposition compared to Il15-/- mice. However, Col1a1 and Col1a3 genes were similarly induced in the fibrotic livers of wildtype, Il15-/- and Il15ra-/- mice, although notable variations were observed in the expression of matrix remodeling enzymes and chemokines. As expected, Il15-/- and Il15ra-/- mice showed markedly reduced numbers of NK cells compared to wildtype mice. They also showed markedly less staining of CD45+ immune cells and CD68+ macrophages, and significantly reduced inflammatory cell infiltration into the liver, with fewer pro-inflammatory and anti-inflammatory monocyte subsets compared to wildtype mice. Conclusion: Our findings indicate that IL-15 exerts its profibrogenic role in the liver by promoting macrophage activation and that this requires trans-presentation of IL-15 by IL-15Rα.


Subject(s)
Carbon Tetrachloride , Disease Models, Animal , Interleukin-15 Receptor alpha Subunit , Interleukin-15 , Liver Cirrhosis , Mice, Inbred C57BL , Mice, Knockout , Animals , Interleukin-15/metabolism , Interleukin-15/genetics , Mice , Interleukin-15 Receptor alpha Subunit/genetics , Interleukin-15 Receptor alpha Subunit/metabolism , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/chemically induced , Male , Liver/pathology , Liver/metabolism , Liver/immunology , Cytokines/metabolism , Receptors, Interleukin-15
7.
Avian Dis ; 68(2): 117-128, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38885053

ABSTRACT

Cytokines are co-administrated with vaccines or co-expressed in the vaccine virus genome to improve protective efficacy by stimulating immune responses. Using glycosylphosphatidylinositol (GPI) anchoring by attachment to the target cytokine, we constructed recombinant Marek's disease virus (MDV) vaccine strain 301B/1 (v301B/1-rtg-IL-15) that expresses chicken interleukin-15 (IL-15) as the membrane-bound form at the cell surface. We evaluated the vaccine efficacy of v301B/1-rtg-IL-15 given as a bivalent Marek's disease (MD) vaccine in combination with turkey herpesvirus (HVT) against a very virulent plus MDV strain 648A challenge. The efficacy was compared with that of conventional bivalent MD vaccine, as a mixture with HVT plus parental v301B/1 or v301B/1-IL-15, which expresses a natural form of IL-15. The membrane-bound IL-15 expression did not interfere with the virus growth of recombinant v301B/1-rtg-IL-15. However, the MD incidence in birds vaccinated with v301B/1-rtg-IL-15 was higher than that of birds given the conventional bivalent MD vaccine containing parental v301B/1 virus, although the v301B/1-rtg-IL-15 vaccinated group showed increased natural killer cell activation at day 5 postvaccination, the same day as challenge. Overall, the protection of v301B/1-rtg-IL-15 was not improved from that of v301B/1 against very virulent plus MDV challenge.


Eficacia de una vacuna contra el virus de la enfermedad de Marek cepa 301B/1 recombinante que expresa la interleucina-15 de pollo anclada a la membrana. Las citocinas se administran junto con vacunas o se co-expresan en el genoma del virus de la vacuna para mejorar la eficacia protectora mediante la estimulación de respuestas inmunitarias. Utilizando el anclaje de glicosilfosfatidilinositol (GPI) mediante unión a la citoquina objetivo, se construyó una cepa de vacuna recombinante del virus de la enfermedad de Marek (MDV) 301B/1 (v301B/1-rtg-IL-15) que expresa la interleucina-15 de pollo (IL-15) como la forma unida a la membrana en la superficie celular. Se evaluó la eficacia de la vacuna v301B/1-rtg-IL-15 administrada como vacuna bivalente en combinación con el herpesvirus del pavo (HVT) contra el desafío con un virus muy virulento cepa 648A de la enfermedad de Marek (MD). La eficacia se comparó con la de la vacuna bivalente convencional contra la enfermedad de Marek, como una mezcla con HVT más la cepa v301B/1 parental o con el virus recombinante v301B/1-IL-15, que expresa una forma natural de IL-15. La expresión de IL-15 unida a membrana no interfirió con el crecimiento del virus de v301B/1-rtg-IL-15 recombinante. Sin embargo, la incidencia de la enfermedad de Marek en aves vacunadas con v301B/1-rtg-IL-15 fue mayor que la de las aves que recibieron la vacuna de Marek bivalente convencional que contenía el virus v301B/1 parental, aunque el grupo vacunado con v301B/1-rtg-IL-15 mostró una mayor activación de las células asesinas naturales en el día 5 después de la vacunación, que fue el mismo día del desafío. En general, la protección por la vacuna v301B/1-rtg-IL-15 no mejoró con respecto a la conferida por v301B/1 contra un desafío muy virulento de la enfermedad de Marek.


Subject(s)
Chickens , Herpesvirus 2, Gallid , Interleukin-15 , Marek Disease Vaccines , Marek Disease , Vaccines, Synthetic , Animals , Interleukin-15/genetics , Interleukin-15/immunology , Interleukin-15/metabolism , Marek Disease/prevention & control , Marek Disease/immunology , Marek Disease Vaccines/immunology , Marek Disease Vaccines/genetics , Vaccines, Synthetic/immunology , Herpesvirus 2, Gallid/genetics , Herpesvirus 2, Gallid/immunology , Poultry Diseases/prevention & control , Poultry Diseases/virology , Poultry Diseases/immunology , Herpesvirus 1, Meleagrid/immunology , Herpesvirus 1, Meleagrid/genetics , Herpesvirus 1, Meleagrid/metabolism
8.
J Clin Invest ; 134(15)2024 May 28.
Article in English | MEDLINE | ID: mdl-38805302

ABSTRACT

The surface receptor CD8α is present on 20%-80% of human (but not mouse) NK cells, yet its function on NK cells remains poorly understood. CD8α expression on donor NK cells was associated with a lack of therapeutic responses in patients with leukemia in prior studies, thus, we hypothesized that CD8α may affect critical NK cell functions. Here, we discovered that CD8α- NK cells had improved control of leukemia in xenograft models compared with CD8α+ NK cells, likely due to an enhanced capacity for proliferation. Unexpectedly, we found that CD8α expression was induced on approximately 30% of previously CD8α- NK cells following IL-15 stimulation. These induced CD8α+ (iCD8α+) NK cells had the greatest proliferation, responses to IL-15 signaling, and metabolic activity compared with those that sustained existing CD8α expression (sustained CD8α+) or those that remained CD8α- (persistent CD8α-). These iCD8α+ cells originated from an IL-15Rßhi NK cell population, with CD8α expression dependent on the transcription factor RUNX3. Moreover, CD8A CRISPR/Cas9 deletion resulted in enhanced responses through the activating receptor NKp30, possibly by modulating KIR inhibitory function. Thus, CD8α status identified human NK cell capacity for IL-15-induced proliferation and metabolism in a time-dependent fashion, and its presence had a suppressive effect on NK cell-activating receptors.


Subject(s)
CD8 Antigens , Cell Proliferation , Interleukin-15 , Killer Cells, Natural , Lymphocyte Activation , Humans , CD8 Antigens/metabolism , CD8 Antigens/immunology , CD8 Antigens/genetics , Interleukin-15/immunology , Interleukin-15/metabolism , Interleukin-15/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism
9.
Front Immunol ; 15: 1368290, 2024.
Article in English | MEDLINE | ID: mdl-38690288

ABSTRACT

Background: NK cells can be genetically engineered to express a transgenic T-cell receptor (TCR). This approach offers an alternative strategy to target heterogenous tumors, as NK:TCR cells can eradicate both tumor cells with high expression of HLA class I and antigen of interest or HLA class I negative tumors. Expansion and survival of NK cells relies on the presence of IL-15. Therefore, autonomous production of IL-15 by NK:TCR cells might improve functional persistence of NK cells. Here we present an optimized NK:TCR product harnessed with a construct encoding for soluble IL-15 (NK:TCR/IL-15), to support their proliferation, persistence and cytotoxic capabilities. Methods: Expression of tumor-specific TCRs in peripheral blood derived NK-cells was achieved following retroviral transduction. NK:TCR/IL-15 cells were compared with NK:TCR cells for autonomous cytokine production, proliferation and survival. NK:BOB1-TCR/IL-15 cells, expressing a HLA-B*07:02-restricted TCR against BOB1, a B-cell lineage specific transcription factor highly expressed in all B-cell malignancies, were compared with control NK:BOB1-TCR and NK:CMV-TCR/IL-15 cells for effector function against TCR antigen positive malignant B-cell lines in vitro and in vivo. Results: Viral incorporation of the interleukin-15 gene into engineered NK:TCR cells was feasible and high expression of the TCR was maintained, resulting in pure NK:TCR/IL-15 cell products generated from peripheral blood of multiple donors. Self-sufficient secretion of IL-15 by NK:TCR cells enables engineered NK cells to proliferate in vitro without addition of extra cytokines. NK:TCR/IL-15 demonstrated a marked enhancement of TCR-mediated cytotoxicity as well as enhanced NK-mediated cytotoxicity resulting in improved persistence and performance of NK:BOB1-TCR/IL-15 cells in an orthotopic multiple myeloma mouse model. However, in contrast to prolonged anti-tumor reactivity by NK:BOB1-TCR/IL-15, we observed in one of the experiments an accumulation of NK:BOB1-TCR/IL-15 cells in several organs of treated mice, leading to unexpected death 30 days post-NK infusion. Conclusion: This study showed that NK:TCR/IL-15 cells secrete low levels of IL-15 and can proliferate in an environment lacking cytokines. Repeated in vitro and in vivo experiments confirmed the effectiveness and target specificity of our product, in which addition of IL-15 supports TCR- and NK-mediated cytotoxicity.


Subject(s)
Interleukin-15 , Killer Cells, Natural , Receptors, Antigen, T-Cell , Interleukin-15/genetics , Interleukin-15/immunology , Interleukin-15/metabolism , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Animals , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Cytotoxicity, Immunologic , Cell Proliferation , Cell Line, Tumor , Immunotherapy, Adoptive/methods , Genetic Engineering
10.
Oncoimmunology ; 13(1): 2358590, 2024.
Article in English | MEDLINE | ID: mdl-38812569

ABSTRACT

Chimeric antigen receptor (CAR) T cells have demonstrated outstanding therapeutic success in hematological malignancies. Yet, their efficacy against solid tumors remains constrained due to inadequate infiltration of cytotoxic T and CAR-T cells in the tumor microenvironment (TME), a factor correlated with poor prognosis in patients with solid tumors. To overcome this limitation, we engineered CAR-T cells to secrete CXCL10 and IL15 (10 × 15 CAR-T), which sustain T cell viability and enhance their recruitment, thereby amplifying the long-term cytotoxic capacity of CAR-T cells in vitro. In a xenograft model employing NUGC4-T21 cells, mice receiving 10 × 15 CAR-T cells showed superior tumor reduction and extended survival rates compared to those treated with second-generation CAR-T cells. Histopathological evaluations indicated a pronounced increase in cytotoxic T cell accumulation in the TME post 10 × 15 CAR-T cell treatment. Therefore, the synergistic secretion of CXCL10 and IL15 in these CAR-T cells enhances T cell recruitment and adaptability within tumor tissues, improving tumor control. This approach may offer a promising strategy for advancing CAR-T therapies in the treatment of solid tumors.


Subject(s)
Chemokine CXCL10 , Immunotherapy, Adoptive , Interleukin-15 , Receptors, Chimeric Antigen , Stomach Neoplasms , Tumor Microenvironment , Xenograft Model Antitumor Assays , Animals , Chemokine CXCL10/metabolism , Chemokine CXCL10/genetics , Stomach Neoplasms/therapy , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/genetics , Humans , Mice , Interleukin-15/genetics , Interleukin-15/metabolism , Immunotherapy, Adoptive/methods , Tumor Microenvironment/immunology , Cell Line, Tumor , T-Lymphocytes, Cytotoxic/immunology , Cell Survival , Female
11.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 10-17, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814240

ABSTRACT

The present study investigated the relationship between single nucleotide polymorphisms in the interleukin (IL)-15 gene located (exon 8) on the chromosomal location 4q31.21 and acute lymphoblastic leukaemia (ALL) risk in Iraqi patients. A total of 78 (49 male -29 female) primary ALL (62B-cell, 16 T-cells lineages cases and 30 healthy control subjects (median age 11, age range were 4-21.5), were enrolled at the Nanakaly Hospital of Erbil Province between February 2021 and January 2022. The genotype analysis was performed using polymerase chain reaction (PCR) and Sanger DNA sequencing. The IL15 homozygous rs10833 (100%) and rs2291596 (63.6%) genotypes indicated high frequencies and were associated with a risk of developing ALL, while the remaining 16 novel mutations indicated in low frequency (9.1%) except for the 97270G>GT genotype (18.2%). High expression levels were noted for different clusters of differentiation (CD) biomarkers between both subtypes of ALL, including, CD10, CD19, CD22, CD79a, CD99,  terminal deoxynucleotidyl transferase (TdT), and human leukocyte antigen DR (HLA-DR) isotype in B-cells lineages, while, CD2, CD3, CD5, CD7, CD13, CD117 and TdT are more specific to T-cells lineages. On the other hand,  significant changes were noted in certain hematological parameters including red blood cells (RBCs), haemoglobin (g/dl), haematocrite (HCt %), red blood cell distribution width (RDW %), and platelet counts (PLT- 109/L) compared with those of healthy subjects. Finally, it was concluded that various novel mutations were recorded with different subtypes of ALL diseases, and mild anemia was observed among patients. Future studies will be towered to associate these mutations with prognosis and therapeutic response of diseases.


Subject(s)
Genetic Predisposition to Disease , Interleukin-15 , Mutation , Polymorphism, Single Nucleotide , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adolescent , Child , Child, Preschool , Female , Humans , Male , Young Adult , Case-Control Studies , Gene Frequency/genetics , Genotype , Interleukin-15/genetics , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Risk Factors
12.
Clin Exp Med ; 24(1): 102, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758367

ABSTRACT

Immunotherapy is widely used in cancer treatment; however, only a subset of patients responds well to it. Significant efforts have been made to identify patients who will benefit from immunotherapy. Successful anti-tumor immunity depends on an intact cancer-immunity cycle, especially long-lasting CD8+ T-cell responses. Interferon (IFN)-α/ß/IFN-γ/interleukin (IL)-15 pathways have been reported to be involved in the development of CD8+ T cells. And these pathways may predict responses to immunotherapy. Herein, we aimed to analyze multiple public databases to investigate whether IFN-α/ß/IFN-γ/IL-15 pathways could be used to predict the response to immunotherapy. Results showed that IFN-α/ß/IFN-γ/IL-15 pathways could efficiently predict immunotherapy response, and guanylate-binding protein 1 (GBP1) could represent the IFN-α/ß/IFN-γ/IL-15 pathways. In public and private cohorts, we further demonstrated that GBP1 could efficiently predict the response to immunotherapy. Functionally, GBP1 was mainly expressed in macrophages and strongly correlated with chemokines involved in T-cell migration. Therefore, our study comprehensively investigated the potential role of GBP1 in immunotherapy, which could serve as a novel biomarker for immunotherapy and a target for drug development.


Subject(s)
GTP-Binding Proteins , Immunotherapy , Neoplasms , Humans , CD8-Positive T-Lymphocytes/immunology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Immunotherapy/methods , Interferon-alpha/metabolism , Interferon-beta/metabolism , Interferon-gamma/metabolism , Interleukin-15/genetics , Neoplasms/immunology , Neoplasms/therapy , Signal Transduction
13.
Biosci Trends ; 18(2): 198-200, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38616129

ABSTRACT

Alopecia areata (AA) is an autoimmune disease characterized by damage to hair follicles and hair loss. Cell-free DNA (cfDNA) has recently received attention as a biomarker of various disorders including inflammatory skin diseases. In this study, we aimed to investigate the clinical significance of cfDNA and the circulating DNAs of disease-associated cytokines in AA patients. Serum samples were obtained from 63 patients with AA and 32 healthy controls (HC). Using droplet digital polymerase chain reaction, circulating C-X-C motif chemokine ligand (CXCL) 9, CXCL10, CXCL11, C-X-C motif chemokine receptor 3, interferon (IFN)-γ, interleukin (IL) -7, IL-15, and Janus kinase (JAK) 2 were detectable in both HC and AA patients. Among the detectable DNAs, copies of circulating CXCL9, CXCL11, IL-15, IFN-γ, and JAK2 were significantly higher in AA patients than in HC. These results suggest that increased circulating DNA levels may reflect damage to hair follicles in AA patients.


Subject(s)
Alopecia Areata , Cell-Free Nucleic Acids , Cytokines , Humans , Alopecia Areata/blood , Alopecia Areata/genetics , Cell-Free Nucleic Acids/blood , Male , Female , Adult , Cytokines/blood , Case-Control Studies , Biomarkers/blood , Middle Aged , Young Adult , Janus Kinase 2/genetics , Janus Kinase 2/blood , Chemokine CXCL9/blood , Chemokine CXCL9/genetics , Chemokine CXCL11/blood , Chemokine CXCL11/genetics , Interferon-gamma/blood , Hair Follicle , Chemokine CXCL10/blood , Adolescent , Interleukin-15/blood , Interleukin-15/genetics
14.
Transplant Proc ; 56(3): 678-685, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38433025

ABSTRACT

BACKGROUND: Abdominal aortic calcification (AAC) is associated with cardiovascular-related mortality, along with an elevated risk of coronary, cerebrovascular, and cardiovascular events. Notably, AAC is strongly associated with poor overall and recurrence free survival posthepatectomy for hepatocellular carcinoma. Despite the acknowledged significance of atherosclerosis in systemic inflammation, its response to ischemia/reperfusion injury (IRI) remains poorly elucidated. In this study, we aimed to clarify the impact of atherosclerosis on the liver immune system using a warm IRI mouse model. METHODS: Injury was induced in an atherosclerotic mouse model (ApoE-/-) or C57BL/6J wild-type (WT) mice through 70% clamping for 1 hour and analyzed after 6 hours of reperfusion. RESULTS: Elevated serum levels of aspartate and alanine aminotransferase, along with histological assessment, indicated considerable damage in the livers of ApoE-/- mice than that in WT mice. This indicates a substantial contribution of atherosclerosis to IRI. Furthermore, T and natural killer (NK) cells in ApoE-/- mouse livers displayed a more inflammatory phenotype than those in WT mouse livers. Reverse transcription-polymerase chain reaction analysis revealed a significant upregulation of interleukin (IL)-15 and its transcriptional regulator, interferon regulatory factor-1 (IRF-1) in ApoE-/- mouse livers compared with that in WT mouse livers. CONCLUSIONS: These findings suggest that in an atherosclerotic mouse model, atherosclerosis can mirror intrahepatic immunity, particularly activating liver NK and T cells through IL-15 production, thereby exacerbating hepatic damage. The upregulation of IL-15 expression is associated with IRF-1 overexpression.


Subject(s)
Atherosclerosis , Disease Models, Animal , Interferon Regulatory Factor-1 , Liver , Mice, Inbred C57BL , Reperfusion Injury , Animals , Reperfusion Injury/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Mice , Liver/pathology , Liver/metabolism , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Male , Killer Cells, Natural/immunology , Interleukin-15/genetics
15.
Int J Pharm ; 655: 124027, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38554742

ABSTRACT

Cancer immunotherapy has emerged as a promising clinical treatment strategy in recent years. Unfortunately, the satisfactory antitumor therapeutic efficacy of immunotherapy is limited by intricate immunosuppressive tumor microenvironment (ITM). To remodel the ITM and alleviate the immune evasion, we constructed FA-PEG-modified liposomes to deliver plasmid IL-15 (pIL-15) and gemcitabine (GEM) (FPCL@pIL-15 + FPGL), respectively. The FPCL@pIL-15 (150 nm) and FPGL (120 nm) exhibited symmetrically spherical structures as well as desirable penetration and accumulation on tumor tissue depending on folic acid (FA) specialized targeting function. The transfected expression of IL-15 efficiently fosters the proliferation and co-activation of Natural killer (NK) cells and CD8+T cells through binding to IL-15R. FPGL upregulated the expression of Natural killer group 2 member D ligands (NKG2DLs) and reinforced recognition by NK cells to alleviate the immune evasion, and simultaneously promoted activation of CD8+T cells through immunogenic cell death (ICD) effects. More importantly, the combinatorial administration achieved intended anti-tumor efficacy in the subcutaneous 4T1 tumor model. In essence, we demonstrated that combining FPCL@pIL-15 with FPGL synergistically stimulates and mobilizes the immune system to reverse the ITM and trigger an anti-tumor immune response, indicating a tremendous potential for application in immunotherapy.


Subject(s)
Gemcitabine , Neoplasms , Cell Line, Tumor , Immunotherapy , Interleukin-15/genetics , Plasmids , Tumor Microenvironment
16.
J Transl Med ; 22(1): 171, 2024 02 18.
Article in English | MEDLINE | ID: mdl-38368374

ABSTRACT

BACKGROUND: Chimeric antigen receptor (CAR) T cell therapy has shown remarkable responses in hematological malignancies with several approved products, but not in solid tumors. Patients suffer from limited response and tumor relapse due to low efficacy of CAR-T cells in the complicated and immunosuppressive tumor microenvironment. This clinical challenge has called for better CAR designs and combined strategies to improve CAR-T cell therapy against tumor changes. METHODS: In this study, IL-15/IL-15Rα was inserted into the extracellular region of CAR targeting mesothelin. In-vitro cytotoxicity and cytokine production were detected by bioluminescence-based killing and ELISA respectively. In-vivo xenograft mice model was used to evaluate the anti-tumor effect of CAR-T cells. RNA-sequencing and online database analysis were used to identify new targets in residual gastric cancer cells after cytotoxicity assay. CAR-T cell functions were detected in vitro and in vivo after GLI Pathogenesis Related 1 (GLIPR1) knockdown in gastric cancer cells. Cell proliferation and migration of gastric cancer cells were detected by CCK-8 and scratch assay respectively after GLIPR1 were overexpressed or down-regulated. RESULTS: CAR-T cells constructed with IL-15/IL-15Rα (CAR-ss-T) showed significantly improved CAR-T cell expansion, cytokine production and cytotoxicity, and resulted in superior tumor control compared to conventional CAR-T cells in gastric cancer. GLIPR1 was up-regulated after CAR-T treatment and survival was decreased in gastric cancer patients with high GLIPR1 expression. Overexpression of GLIPR1 inhibited cytotoxicity of conventional CAR-T but not CAR-ss-T cells. CAR-T treatment combined with GLIPR1 knockdown increased anti-tumor efficacy in vitro and in vivo. CONCLUSIONS: Our data demonstrated for the first time that this CAR structure design combined with GLIPR1 knockdown in gastric cancer improved CAR-T cell-mediated anti-tumor response.


Subject(s)
Receptors, Chimeric Antigen , Stomach Neoplasms , Humans , Animals , Mice , Stomach Neoplasms/genetics , Stomach Neoplasms/therapy , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Interleukin-15/genetics , Interleukin-15/metabolism , Cell Line, Tumor , Neoplasm Recurrence, Local/metabolism , Immunotherapy, Adoptive/methods , T-Lymphocytes , Xenograft Model Antitumor Assays , Tumor Microenvironment , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism
17.
J Control Release ; 367: 45-60, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38246204

ABSTRACT

PD-1/PD-L1-based immune checkpoint blockade therapy has shown limited benefits in tumor patients, partially attributed to the inadequate infiltration of immune effector cells within tumors. Here, we established a nanoplatform named DPPA/IL-15 NPs to target PD-L1 for the tumor delivery of IL-15 messenger RNA (mRNA). DPPA/IL-15 NPs were endowed with ultrasound responsiveness and contrast-enhanced ultrasound (CEUS) imaging performance. They effectively protected IL-15 mRNA from degradation and specifically transfected it into tumor cells through the utilization of ultrasound-targeted microbubble destruction (UTMD). This resulted in the activation of IL-15-related immune effector cells while blocking the PD-1/PD-L1 pathway. In addition, UTMD could generate reactive oxygen species (ROS) that induce endoplasmic reticulum (ER) stress-driven immunogenic cell death (ICD), initiating anti-tumor immunity. In vitro and in vivo studies revealed that this combination therapy could induce a robust systemic immune response and enhance anti-tumor efficacy. Thus, this combination therapy has the potential for clinical translation through enhanced immunotherapy and provides real-time ultrasound imaging guidance.


Subject(s)
B7-H1 Antigen , Neoplasms , Humans , Microbubbles , Programmed Cell Death 1 Receptor/metabolism , Interleukin-15/genetics , Neoplasms/therapy , Immunotherapy/methods , Tumor Microenvironment , Cell Line, Tumor
18.
Signal Transduct Target Ther ; 9(1): 16, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38212320

ABSTRACT

Multiple myeloma (MM) remains a challenging hematologic malignancy despite advancements in chimeric antigen receptor T-cell (CAR-T) therapy. Current targets of CAR-T cells used in MM immunotherapy have limitations, with a subset of patients experiencing antigen loss resulting in relapse. Therefore, novel targets for enhancing CAR-T cell therapy in MM remain needed. Fc receptor-like 5 (FCRL5) is a protein marker with considerably upregulated expression in MM and has emerged as a promising target for CAR-T cell therapeutic interventions, offering an alternative treatment for MM. To further explore this option, we designed FCRL5-directed CAR-T cells and assessed their cytotoxicity in vitro using a co-culture system and in vivo using MM cell-derived xenograft models, specifically focusing on MM with gain of chromosome 1q21. Given the challenges in CAR-T therapies arising from limited T cell persistence, our approach incorporates interleukin-15 (IL-15), which enhances the functionality of central memory T (TCM) cells, into the design of FCRL5-directed CAR-T cells, to improve cytotoxicity and reduce T-cell dysfunction, thereby promoting greater CAR-T cell survival and efficacy. Both in vitro and xenograft models displayed that FCRL5 CAR-T cells incorporating IL-15 exhibited potent antitumor efficacy, effectively inhibiting the proliferation of MM cells and leading to remarkable tumor suppression. Our results highlight the capacity of FCRL5-specific CAR-T cells with the integration of IL-15 to improve the therapeutic potency, suggesting a potential novel immunotherapeutic strategy for MM treatment.


Subject(s)
Multiple Myeloma , Receptors, Chimeric Antigen , Humans , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Receptors, Chimeric Antigen/genetics , Interleukin-15/genetics , Interleukin-15/metabolism , Cell Line, Tumor , T-Lymphocytes , Receptors, Fc/metabolism
19.
J Clin Invest ; 134(6)2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38271093

ABSTRACT

Virus-induced memory T cells often express functional cross-reactivity, or heterologous immunity, to other viruses and to allogeneic MHC molecules that is an important component of pathogenic responses to allogeneic transplants. During immune responses, antigen-reactive naive and central memory T cells proliferate in secondary lymphoid organs to achieve sufficient cell numbers to effectively respond, whereas effector memory T cell proliferation occurs directly within the peripheral inflammatory microenvironment. Mechanisms driving heterologous memory T cell proliferation and effector function expression within peripheral tissues remain poorly understood. Here, we dissected proliferation of heterologous donor-reactive memory CD8+ T cells and their effector functions following infiltration into heart allografts with low or high intensities of ischemic inflammation. Proliferation within both ischemic conditions required p40 homodimer-induced IL-15 transpresentation by graft DCs, but expression of effector functions mediating acute allograft injury occurred only in high-ischemic allografts. Transcriptional responses of heterologous donor-reactive memory CD8+ T cells were distinct from donor antigen-primed memory CD8+ T cells during early activation in allografts and at graft rejection. Overall, the results provide insights into mechanisms driving heterologous effector memory CD8+ T cell proliferation and the separation between proliferation and effector function that is dependent on the intensity of inflammation within the tissue microenvironment.


Subject(s)
Heart Transplantation , Interleukin-15 , Animals , Mice , CD8-Positive T-Lymphocytes , Graft Rejection , Immunologic Memory , Interleukin-15/genetics , Mice, Inbred C57BL , Transplantation, Homologous , Interleukin-9/metabolism
20.
Am J Physiol Endocrinol Metab ; 326(3): E326-E340, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38294696

ABSTRACT

This study aimed to evaluate the role of skeletal muscle-derived interleukin (IL)-15 in the regulation of skeletal muscle autophagy using IL-15 knockout (KO) and transgenic (TG) mice. Male C57BL/6 wild-type (WT), IL-15 KO, and IL-15 TG mice were used in this study. Changes in muscle mass, forelimb grip strength, succinate dehydrogenase (SDH) activity, gene and protein expression levels of major regulators and indicators of autophagy, comprehensive gene expression, and DNA methylation in the gastrocnemius muscle were analyzed. Enrichment pathway analyses revealed that the pathology of IL-15 gene deficiency was related to the autophagosome pathway. Moreover, although IL-15 KO mice maintained gastrocnemius muscle mass, they exhibited a decrease in autophagy induction. IL-15 TG mice exhibited a decrease in gastrocnemius muscle mass and an increase in forelimb grip strength and SDH activity in skeletal muscle. In the gastrocnemius muscle, the ratio of phosphorylated adenosine monophosphate-activated protein kinase α (AMPKα) to total AMPKα and unc-51-like autophagy activating kinase 1 and Beclin1 protein expression were higher in the IL-15 TG group than in the WT group. IL-15 gene deficiency induces a decrease in autophagy induction. In contrast, IL-15 overexpression could improve muscle quality by activating autophagy induction while decreasing muscle mass. The regulation of IL-15 in autophagy in skeletal muscles may lead to the development of therapies for the autophagy-induced regulation of skeletal muscle mass and cellular quality control.NEW & NOTEWORTHY IL-15 gene deficiency can decrease autophagy induction. However, although IL-15 overexpression induced a decrease in muscle mass, it led to an improvement in muscle quality. Based on these results, understanding the role of IL-15 in regulating autophagy pathways within skeletal muscle may lead to the development of therapies for the autophagy-induced regulation of skeletal muscle mass and cellular quality control.


Subject(s)
Interleukin-15 , Muscle, Skeletal , Mice , Male , Animals , Interleukin-15/genetics , Interleukin-15/metabolism , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Mice, Transgenic , Mice, Knockout , AMP-Activated Protein Kinases/metabolism , Autophagy
SELECTION OF CITATIONS
SEARCH DETAIL