Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 937
Filter
1.
Proc Natl Acad Sci U S A ; 121(13): e2319856121, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38513098

ABSTRACT

The use of lipid nanoparticles (LNP) to encapsulate and deliver mRNA has become an important therapeutic advance. In addition to vaccines, LNP-mRNA can be used in many other applications. For example, targeting the LNP with anti-CD5 antibodies (CD5/tLNP) can allow for efficient delivery of mRNA payloads to T cells to express protein. As the percentage of protein expressing T cells induced by an intravenous injection of CD5/tLNP is relatively low (4-20%), our goal was to find ways to increase mRNA-induced translation efficiency. We showed that T cell activation using an anti-CD3 antibody improved protein expression after CD5/tLNP transfection in vitro but not in vivo. T cell health and activation can be increased with cytokines, therefore, using mCherry mRNA as a reporter, we found that culturing either mouse or human T cells with the cytokine IL7 significantly improved protein expression of delivered mRNA in both CD4+ and CD8+ T cells in vitro. By pre-treating mice with systemic IL7 followed by tLNP administration, we observed significantly increased mCherry protein expression by T cells in vivo. Transcriptomic analysis of mouse T cells treated with IL7 in vitro revealed enhanced genomic pathways associated with protein translation. Improved translational ability was demonstrated by showing increased levels of protein expression after electroporation with mCherry mRNA in T cells cultured in the presence of IL7, but not with IL2 or IL15. These data show that IL7 selectively increases protein translation in T cells, and this property can be used to improve expression of tLNP-delivered mRNA in vivo.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Interleukin-7 , Liposomes , Nanoparticles , Protein Biosynthesis , RNA, Messenger , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Interleukin-7/pharmacology , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , Mice, Inbred C57BL , Cells, Cultured , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology
2.
Cancer Lett ; 588: 216714, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38369003

ABSTRACT

Glioblastoma, the most prevalent malignant primary brain tumor, presents substantial treatment challenges because of its inherent aggressiveness and limited therapeutic options. Lymphopenia, defined as reduced peripheral blood lymphocyte count, commonly occurs as a consequence of the disease and its treatment. Recent studies have associated lymphopenia with a poor prognosis. Factors that contribute to lymphopenia include radiotherapy, chemotherapy, and the tumor itself. Patients who are female, older, using dexamethasone, or receiving higher doses of radiation therapy are particularly vulnerable to this condition. Several preclinical studies have explored the use of interleukin-7, a crucial cytokine for lymphocyte homeostasis, to restore lymphocyte counts and potentially rebuild the immune system to combat glioblastoma cells. With the development of recombinant interleukin-7 for prolonged activity in the body, various clinical trials are underway to explore this treatment in patients with glioblastoma. Our study provides a comprehensive summary of the incidence of lymphopenia, its potential biological background, and the associated clinical risk factors. Furthermore, we reviewed several clinical trials using IL-7 cytokine therapy in glioblastoma patients. We propose IL-7 as a promising immunotherapeutic strategy for glioblastoma treatment. We are optimistic that our study will enhance understanding of the complex interplay between lymphopenia and glioblastoma and will pave the way for the development of more effective treatment modalities.


Subject(s)
Glioblastoma , Lymphopenia , Humans , Female , Male , Glioblastoma/drug therapy , Glioblastoma/pathology , Interleukin-7/therapeutic use , Interleukin-7/pharmacology , Lymphopenia/etiology , Lymphopenia/pathology , Lymphocytes , Immunotherapy/adverse effects
3.
Br J Cancer ; 130(8): 1388-1401, 2024 May.
Article in English | MEDLINE | ID: mdl-38424167

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors unleash inhibitory signals on T cells conferred by tumors and surrounding stromal cells. Despite the clinical efficacy of checkpoint inhibitors, the lack of target expression and persistence of immunosuppressive cells limit the pervasive effectiveness of the therapy. These limitations may be overcome by alternative approaches that co-stimulate T cells and the immune microenvironment. METHODS: We analyzed single-cell RNA sequencing data from multiple human cancers and a mouse tumor transplant model to discover the pleiotropic expression of the Interleukin 7 (IL-7) receptor on T cells, macrophages, and dendritic cells. RESULTS: Our experiment on the mouse model demonstrated that recombinant IL-7 therapy induces tumor regression, expansion of effector CD8 T cells, and pro-inflammatory activation of macrophages. Moreover, spatial transcriptomic data support immunostimulatory interactions between macrophages and T cells. CONCLUSION: These results indicate that IL-7 therapy induces anti-tumor immunity by activating T cells and pro-inflammatory myeloid cells, which may have diverse therapeutic applicability.


Subject(s)
Interleukin-7 , Neoplasms , Humans , Animals , Mice , Interleukin-7/genetics , Interleukin-7/pharmacology , Immunotherapy , Neoplasms/genetics , Neoplasms/therapy , T-Lymphocytes , Sequence Analysis, RNA , Tumor Microenvironment/genetics , CD8-Positive T-Lymphocytes
4.
Sci Adv ; 9(48): eadh9879, 2023 12.
Article in English | MEDLINE | ID: mdl-38019919

ABSTRACT

Cancer immunotherapy is moving toward combination regimens with agents of complementary mechanisms of action to achieve more frequent and robust efficacy. However, compared with single-agent therapies, combination immunotherapies are associated with increased overall toxicity because the very same mechanisms also work in concert to enhance systemic inflammation and promote off-tumor toxicity. Therefore, rational design of combination regimens that achieve improved antitumor control without exacerbated toxicity is a main objective in combination immunotherapy. Here, we show that the combination of engineered, tumor matrix-binding interleukin-7 (IL-7) and IL-12 achieves remarkable anticancer effects by activating complementary pathways without inducing any additive immunotoxicity. Mechanistically, engineered IL-12 provided effector properties to T cells, while IL-7 prevented their exhaustion and boosted memory formation as assessed by tumor rechallenge experiments. The dual combination also rendered checkpoint inhibitor (CPI)-resistant genetically engineered melanoma model responsive to CPI. Thus, our approach provides a framework of evaluation of rationally designed combinations in immuno-oncology and yields a promising therapy.


Subject(s)
Interleukin-12 , Melanoma , Humans , Interleukin-12/genetics , Interleukin-7/pharmacology , T-Cell Exhaustion , Immunotherapy , Melanoma/pathology
5.
Biomed Pharmacother ; 167: 115599, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37783150

ABSTRACT

B-lymphocytopenia among myelosuppression is the most intractable side effect of chemotherapy. Here, we investigated ways to alleviate 5-fluorouracil-caused stress hematopoietic impairment. We found that intraperitoneally injected ASP (Angelica sinensis polysaccharides) (100 mg/kg per day), one main active ingredient of Angelica sinensis, for consecutive 7 days, significantly recovered mouse bone marrow pro-B and pre-B cells, reversed the capacity of CFU-PreB colony forming, thus alleviating B cell reduction in the spleen and peripheral blood, as well as ameliorating immunoglobin from spleen and serum. The mechanism is related to the protective effects of ASP on IL-7 producing cells, including perivascular Leptin+ and CXCL12+ mesenchymal stem and progenitor cells (MSPCs), thus promoting IL-7 production, and activating IL-7R-mediated STAT5, PI3K-AKT signaling, including survival signals and EBF1, PAX5 transcription factor expression. Additionally, ASP's IL-7 promoting effect was demonstrated to be associated with maintaining osteogenesis/adipogenesis balance of MSPCs via the NRF2 antioxidant pathway. Collectively, our findings indicate that ASP reverse stress B-lymphocytopenia via improving Nrf2 signaling, promoting IL-7 production in MSPCs, and subsequently maintaining survival, proliferation, and differentiation of B cell progenitors, which may represent a promising therapeutic strategy.


Subject(s)
Angelica sinensis , Lymphopenia , Mice , Animals , Interleukin-7/pharmacology , Fluorouracil/pharmacology , NF-E2-Related Factor 2 , Phosphatidylinositol 3-Kinases , Oxidative Stress , Stem Cells , Polysaccharides/pharmacology
6.
PLoS One ; 18(10): e0286834, 2023.
Article in English | MEDLINE | ID: mdl-37874823

ABSTRACT

Interleukin (IL)-7 is broadly active on T-cell populations, and modified versions have been clinically evaluated for a variety of therapeutic applications, including cancer, lymphopenia, and infectious diseases; and found to be relatively well-tolerated and biologically active. Here we describe novel IL-7R agonists that are unrelated in structure to IL-7, bind to the receptor subunits differently from IL-7, but closely emulate IL-7 biology. The small size, low structural complexity, and the natural amino acid composition of the pharmacologically active peptide MDK1472 allows facile incorporation into protein structures, such as the IgG2-Fc fusion MDK-703. This molecule possesses properties potentially better suited to therapeutic applications than native IL-7 or its derivatives. We compared these compounds with IL-7 for immune cell selectivity, induction of IL-7R signaling, receptor-mediated internalization, proliferation, and generation of immune cell phenotypes in human and non-human primate (NHP) peripheral blood cells in vitro; and found them to be similar in biological activity to IL-7. In cynomolgus macaques, MDK-703 exhibits a circulating half-life of 46 hr and produces sustained T-cell expansion characteristic of IL-7 treatment. In the huCD34+-engrafted NSG mouse model of the human immune system, MDK-703 induces an immune cell profile very similar to that generated by IL-7-derived compounds; including the pronounced expansion of memory T-cells, particularly the population of stem-like memory T-cells (Tscm) which may be important for anti-tumor activities reported with IL-7 treatment. Clinical administration of IL-7 and modified variants has been reported to induce anti-drug antibodies (ADAs), including IL-7 neutralizing antibodies. The novel peptide agonist reported here scores very low in predicted immunogenicity, and because the peptide lacks sequence similarity with IL-7, the problematic immunogenic neutralization of endogenous cytokine should not occur. The properties we report here implicate MDK-703 as a candidate for clinical evaluation in oncology, anti-viral and other infectious disease, vaccine enhancement, and treatment of lymphopenia.


Subject(s)
Interleukin-7 , Lymphopenia , Receptors, Interleukin-7 , Animals , Humans , Mice , Cytokines/metabolism , Interleukin-7/pharmacology , Peptides/pharmacology , Receptors, Interleukin-7/agonists
7.
Hum Vaccin Immunother ; 19(2): 2232247, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37417353

ABSTRACT

Following acute stress such as trauma or sepsis, most of critically ill elderly patients become immunosuppressed and susceptible to secondary infections and enhanced mortality. We have developed a virus-based immunotherapy encoding human interleukin-7 (hIL-7) aiming at restoring both innate an adaptative immune homeostasis in these patients. We assessed the impact of this encoded hIL-7 on the ex vivo immune functions of T cells from PBMC of immunosenescent patients with or without hip fracture. T-cell ex vivo phenotyping was characterized in terms of senescence (CD57), IL-7 receptor (CD127) expression, and T cell differentiation profile. Then, post stimulation, activation status, and functionality (STAT5/STAT1 phosphorylation and T cell proliferation assays) were evaluated by flow cytometry. Our data show that T cells from both groups display immunosenescence features, express CD127 and are activated after stimulation by virotherapy-produced hIL-7-Fc. Interestingly, hip fracture patients exhibit a unique functional ability: An important T cell proliferation occurred compared to controls following stimulation with hIL-7-Fc. In addition, stimulation led to an increased naïve T cell as well as a decreased effector memory T cell proportions compared to controls. This preliminary study indicates that the produced hIL-7-Fc is well recognized by T cells and initiates IL-7 signaling through STAT5 and STAT1 phosphorylation. This signaling efficiently leads to T cell proliferation and activation and enables a T cell "rejuvenation." These results are in favor of the clinical development of the hIL-7-Fc expressing virotherapy to restore or induce immune T cell responses in immunosenescent hip fracture patients.


Subject(s)
Immunosenescence , Interleukin-7 , T-Lymphocytes , Aged , Humans , Immunotherapy , Interleukin-7/pharmacology , Leukocytes, Mononuclear/metabolism , STAT5 Transcription Factor/metabolism , T-Lymphocytes/metabolism
8.
Aging Cell ; 22(7): e13864, 2023 07.
Article in English | MEDLINE | ID: mdl-37165998

ABSTRACT

Age-related immune dysfunctions, such as decreased T-cell output, are closely related to pathologies like cancers and lack of vaccine efficacy among the elderly. Engineered fusokine, GIFT-7, a fusion of interleukin 7 (IL-7) and GM-CSF, can reverse aging-related lymphoid organ atrophy. We generated a GIFT-7 fusokine tumor vaccine and employed it in aged syngeneic mouse models of glioblastoma and found that peripheral vaccination with GIFT-7TVax resulted in thymic regeneration and generated durable long-term antitumor immunity specifically in aged mice. Global cytokine analysis showed increased pro-inflammatory cytokines including IL-1ß in the vaccinated group that resulted in hyperactivation of dendritic cells. In addition, GIFT-7 vaccination resulted in increased T-cell trafficking to the brain and robust Th-17 long-term effector memory T-cell formation. TCR-seq analysis showed increased productive frequency among detected rearrangements within the vaccinated group. Overall, our data demonstrate that aging immune system can be therapeutically augmented to generate lasting antitumor immunity.


Subject(s)
Cancer Vaccines , Glioblastoma , Mice , Animals , Cytokines , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-7/pharmacology , Glioblastoma/therapy
9.
Blood Adv ; 6(23): 6093-6107, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36206199

ABSTRACT

Interleukin-7 (IL-7) is an essential cytokine for T-cell homeostatic proliferation and maintenance. Clinical studies have shown the potential benefits of IL-7 therapy in various diseases associated with lymphopenia. However, the kinetics of the T-cell response to a single administration of IL-7 in humans have not been fully elucidated. Here, we investigated the effects of Fc-fused long-acting recombinant human IL-7 (hIL-7-hyFc, efineptakin alfa) on lymphocytes in healthy adults after a single subcutaneous or intramuscular administration. Administration of hIL-7-hyFc increased the CD8+ and CD4+ T-cell numbers up to 2.5-fold, with corresponding upregulation of Ki-67 and Bcl-2 expression, peaking at day 3 or 7. Regulatory T cells (Tregs) did not expand. Among CD8+ and CD4+ T cells, all T-cell subsets (TN, TEM, TCM, TEMRA, and TSCM) increased for 56 days. The T-cell receptor repertoire diversity of naive CD8+ and CD4+ T cells was increased by hIL-7-hyFc, whereas the memory T-cell subsets did not differ between day 56 and day 0. Transcriptomic analysis revealed that hIL-7-hyFc induced robust T-cell expansion without changes in gene expression profiles associated with T-cell functions or genes related to T-cell exhaustion, senescence, and anergy. The effector functions of antigen-specific CD8+ T cells were preserved after hIL-7-hyFc administration. Our results suggest that hIL-7-hyFc administration induced a sustained increase in the numbers of CD8+ and CD4+ T cells, but not Tregs, without qualitative changes. These results support the potential of hIL-7-hyFc as a treatment for patients with compromised T-cell immunity or as a vaccine adjuvant.


Subject(s)
Interleukin-7 , T-Lymphocyte Subsets , Adult , Humans , Interleukin-7/pharmacology , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Proliferation
10.
Cytokine ; 160: 156049, 2022 12.
Article in English | MEDLINE | ID: mdl-36201890

ABSTRACT

OVERVIEW: IL-7 is a member of the family of cytokines with four anti-parallel α helixes that bind Type I cytokine receptors. It is produced by stromal cells and is required for development and homeostatic survival of lymphoid cells. GENOMIC ARCHITECTURE: Interleukin 7 (IL7) human IL7: gene ID: 3574 on ch 8; murine Il7 gene ID: 16,196 on ch 3. PROTEIN: Precursor contains a signal sequence, mature human IL-7 peptide 152aa, predicted 17.4kd peptide, glycosylated resulting in 25kd. Crystal structure: http://www.rcsb.org/structure/3DI2. REGULATION OF IL-7 PRODUCTION: Major producers are stromal cells in thymus, bone marrow and lymphoid organs but also reported in other tissues. Production is primarily constitutive but reported to be affected by IFNγ and other factors. IL-7 RECEPTORS: Two chains IL-7Rα (IL-7R) and γc (IL-2RG). Human IL-7R: gene ID 3575 on ch 5; human IL2RG: gene ID 3561 on ch X; mouse IL-7R: gene ID 16,197 on ch 15; murine Il2rg gene ID 16,186 on ch X. Member of γc family of receptors for cytokines IL-2, -4, -9, -15, and -21. Primarily expressed on lymphocytes but reports of other cell types. Expression in T-cells downregulated by IL-7. Low expression on Tregs, no expression on mature B-cells. Crystal structure: http://www.rcsb.org/structure/3DI2. IL-7 RECEPTOR SIGNAL TRANSDUCTION PATHWAYS: Major signals through JAK1, JAK3 to STAT5 and through non-canonical STAT3, STAT1, PI3K/AKT and MEK/ERK pathways. BIOLOGICAL ACTIVITY OF IL-7: Required for survival of immature thymocytes, naïve T-cells, memory T-cells, pro-B-cells and innate lymphocytes. Pharmacological treatment with IL-7 induces expansion of naïve and memory T-cells and pro-B-cells. ABNORMALITIES OF THE IL-7 PATHWAY IN DISEASE: Deficiencies in the IL-7 pathway in humans and mice result in severe combined immunodeficiency due to lymphopenia. Excessive signaling of the pathway in mice drives autoimmune diseases and in humans is associated with autoimmune syndromes including multiple sclerosis, type 1 diabetes, rheumatoid arthritis, sarcoidosis, atopic dermatitis and asthma. Mutations in the IL-7 receptor pathway drive acute lymphoblastic leukemia. CLINICAL APPLICATIONS: IL-7 has been evaluated in patients with cancer and shown to expand lymphocytes. It accelerated lymphocyte recovery after hematopoietic stem cell transfer, and increased lymphocyte counts in AIDS patients and sepsis patients. Monoclonal antibodies blocking the IL-7 receptor are being evaluated in autoimmune diseases. Cytotoxic monoclonals are being evaluated in acute lymphoblastic leukemia. Drugs blocking the signal transduction pathway are being tested in autoimmunity and acute lymphoblastic leukemia.


Subject(s)
Autoimmune Diseases , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Animals , Antibodies, Monoclonal , Humans , Interleukin-2/metabolism , Interleukin-7/pharmacology , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Sorting Signals , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , STAT5 Transcription Factor/metabolism
11.
Front Immunol ; 13: 922307, 2022.
Article in English | MEDLINE | ID: mdl-35874706

ABSTRACT

Objectives: This study aimed to elucidate the changes and associated mechanisms of circulating CD28- cytotoxic T lymphocytes (CTLs) in patients with IgG4-related disease (IgG4-RD). Methods: Fifty IgG4-RD patients and 15 healthy controls (HCs) were recruited. Peripheral blood mononuclear cells (PBMCs) were isolated, the levels of circulating CD28- CTLs were detected by flow cytometry, and the proportions of CD127lo or GZMB+CD28- CTL subsets were analyzed in the meantime. Mechanistically, PBMCs isolated from IgG4-RD patients were stimulated with IL-7 in the presence or absence of the JAK inhibitor tofacitinib. Flow cytometry was used to analyze the proliferation of CD28- CTLs and the changes in related subpopulations. Results: Circulating CD4+CD28- CTLs and CD8+CD28- CTLs were significantly increased in IgG4-RD patients compared with HCs, accompanied by an elevation of CD127lo or GZMB+ CTL subsets. The ex vivo culture of PBMCs showed that IL-7 could induce the amplification of CD4+CD28- CTLs and CD8+CD28- CTLs in IgG4-RD. Furthermore, IL-7 promotes the proliferation and functional subset changes of these CD28- CTLs in this disease. The selective JAK inhibitor tofacitinib significantly inhibited the effects of IL-7 on CD4+CD28- CTLs and CD8+CD28- CTLs. Conclusion: IL-7 can affect the immune balance of IgG4-RD patients by promoting the expansion and function of CD4+CD28- and CD8+CD28- CTLs in IgG4-RD through the JAK pathway. Blockade of the IL-7 signaling pathway may be a new therapeutic strategy for IgG4-RD.


Subject(s)
Immunoglobulin G4-Related Disease , Janus Kinase Inhibitors , CD28 Antigens , Humans , Interleukin-7/metabolism , Interleukin-7/pharmacology , Janus Kinase Inhibitors/pharmacology , Leukocytes, Mononuclear/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic
12.
Sci Rep ; 12(1): 11208, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35778432

ABSTRACT

Interleukin 7 (IL-7) has been demonstrated regulating lymphangiogenesis, apoptosis, and proliferation. Whether IL-7 induce or inhibit autophagy in non-small cell lung cancer (NSCLC) are unknown. In this study, Western blot was used to detect cytoplasmic and nuclear protein of p53, total protein of AMP-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR) and Light chain 3 (LC3). Quantitative Real-Time PCR (qRT-PCR) was used to detect p53 mRNA level after treated with IL-7. Then using transmission electron microscopy to observe the morphological change of autophagosome. 123 cases of NSCLC were collected for survival analysis, immunohistochemistry staining and cox regression multivariate analysis. We find that IL-7 induce the p53 translocation from nucleus to cytoplasm, then IL-7 down-regulate phosphorylation of AMPK and up-regulate phosphorylation of mTOR. The expression of AMPK and p53 were associated with IL-7/IL-7R and mTOR expression. Clinically, AMPK and p53 were well correlated with stage and survival of lung cancer patients. IL-7R, mTOR and tumor stage were the strongest predictors of survival. In conclusion, IL-7 inhibit autophagy in NSCLC via P53 regulated AMPK/mTOR signaling pathway. AMPK and p53 are correlated with patients' survival. IL-7R, mTOR and tumor stage are the strongest predictor of survival.


Subject(s)
AMP-Activated Protein Kinases , Carcinoma, Non-Small-Cell Lung , Interleukin-7 , Lung Neoplasms , TOR Serine-Threonine Kinases , Tumor Suppressor Protein p53 , AMP-Activated Protein Kinases/metabolism , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Interleukin-7/metabolism , Interleukin-7/pharmacology , Lung Neoplasms/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism
13.
Sci Rep ; 12(1): 10461, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35729189

ABSTRACT

HER-2 targeted therapies, such as monoclonal antibodies (mAbs) and CAR-T cell therapy have been applied in the treatment of various of cancers. However, the anti-HER2 CAR-T cell therapy are limited by its expensive production procedure and fatal side effects such as cytokine storm or "On target, off tumor". The application of anti-HER2 mAbs to the soild tumor are also plagued by the patients resistant with different mechanisms. Thus, the recombinant protein technology can be presented as an attractive methods in advantage its less toxic and lower cost. In this study, we produced a HER-2-targeting recombinant protein, which is the fusion of the anti-HER-2 single chain fragment variable domain, CCL19 and IL7 (HCI fusion protein). Our results showed that the recombinant protein can induce the specific lysis effects of immune cells on HER-2-positive gastric tumor cells and can suppress gastric tumor growth in a xenograft model by chemotactic autoimmune cell infiltration into tumor tissues and activated T cells. Taken together, our results revealed that the HCI fusion protein can be applied as a subsequent clinical drug in treating HER-2 positive gastric tumors.


Subject(s)
Chemokine CCL19 , Interleukin-7 , Receptors, Chimeric Antigen , Recombinant Fusion Proteins , Stomach Neoplasms , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Chemokine CCL19/genetics , Chemokine CCL19/pharmacology , Humans , Interleukin-7/genetics , Interleukin-7/pharmacology , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Xenograft Model Antitumor Assays
14.
Nat Commun ; 13(1): 3296, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35697686

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy is routinely used to treat patients with refractory hematologic malignancies. However, a significant proportion of patients experience suboptimal CAR T cell cytotoxicity and persistence that can permit tumor cell escape and disease relapse. Here we show that a prototype pro-lymphoid growth factor is able to enhance CAR T cell efficacy. We demonstrate that a long-acting form of recombinant human interleukin-7 (IL-7) fused with hybrid Fc (rhIL-7-hyFc) promotes proliferation, persistence and cytotoxicity of human CAR T cells in xenogeneic mouse models, and murine CAR T cells in syngeneic mouse models, resulting in long-term tumor-free survival. Thus, rhIL-7-hyFc represents a tunable clinic-ready adjuvant for improving suboptimal CAR T cell activity.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Animals , Cell Proliferation , Humans , Interleukin-7/pharmacology , Mice , Recombinant Fusion Proteins , T-Lymphocytes
15.
Environ Sci Pollut Res Int ; 29(45): 68990-69007, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35554836

ABSTRACT

Diabetes mellitus (DM) is a metabolic disorder that causes severe complications in several tissues due to redox imbalances, which in turn cause defective angiogenesis in response to ischemia and activate a number of proinflammatory pathways. Our study aimed to investigate the effect of bee gomogenat (BG) dietary supplementation on the architecture of immune organs in a streptozotocin (STZ)-induced type 1 diabetes (T1D) mouse model. Three animal groups were used: the control non-diabetic, diabetic, and BG-treated diabetic groups. STZ-induced diabetes was associated with increased levels of blood glucose, ROS, and IL-6 and decreased levels of IL-2, IL-7, IL-4, and GSH. Moreover, diabetic mice showed alterations in the expression of autophagy markers (LC3, Beclin-1, and P62) and apoptosis markers (Bcl-2 and Bax) in the thymus, spleen, and lymph nodes. Most importantly, the phosphorylation level of AKT (a promoter of cell survival) was significantly decreased, but the expression levels of MCP-1 and HSP-70 (markers of inflammation) were significantly increased in the spleen and lymph nodes in diabetic mice compared to control animals. Interestingly, oral supplementation with BG restored the levels of blood glucose, ROS, IL-6, IL-2, IL-4, IL-7, and GSH in diabetic mice. Treatment with BG significantly abrogated apoptosis and autophagy in lymphoid organs in diabetic mice by restoring the expression levels of LC3, Beclin-1, P62, Bcl-2, and Bax; decreasing inflammatory signals by downregulating the expression of MCP-1 and HSP-70; and promoting cell survival by enhancing the phosphorylation of AKT. Our data were the first to reveal the therapeutic potential of BG on the architecture of lymphoid organs and enhancing the immune system during T1D.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Animals , Apoptosis , Autophagy , Beclin-1/metabolism , Beclin-1/pharmacology , Bees , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/drug therapy , Interleukin-2/metabolism , Interleukin-2/pharmacology , Interleukin-2/therapeutic use , Interleukin-4/metabolism , Interleukin-4/pharmacology , Interleukin-4/therapeutic use , Interleukin-6/metabolism , Interleukin-7/metabolism , Interleukin-7/pharmacology , Interleukin-7/therapeutic use , Mice , Oxidative Stress , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Streptozocin/pharmacology , Streptozocin/therapeutic use , bcl-2-Associated X Protein/metabolism
16.
Cells ; 11(10)2022 05 20.
Article in English | MEDLINE | ID: mdl-35626739

ABSTRACT

The growth of T cells ex vivo for the purpose of T cell therapies is a rate-limiting step in the overall process for cancer patients to achieve remission. Growing T cells is a fiscally-, time-, and resource-intensive process. Cytokines have been shown to accelerate the growth of T cells, specifically IL-2, IL-7, and IL-15. Here a design of experiments was conducted to optimize the growth rate of different naïve and memory T cell subsets using combinations of cytokines. Mathematical models were developed to study the impact of IL-2, IL-7, and IL-15 on the growth of T cells. The results show that CD4+ and CD8+ naïve T cells grew effectively using moderate IL-2 and IL-7 in combination, and IL-7, respectively. CD4+ and CD8+ memory cells favored moderate IL-2 and IL-15 in combination and moderate IL-7 and IL-15 in combination, respectively. A statistically significant interaction was observed between IL-2 and IL-7 in the growth data of CD4+ naïve T cells, while the interaction between IL-7 and IL-15 was found for CD8+ naïve T cells. The important genes and related signaling pathways and metabolic reactions were identified from the RNA sequencing data for each of the four subsets stimulated by each of the three cytokines. This systematic investigation lays the groundwork for studying other T cell subsets.


Subject(s)
Interleukin-15 , Interleukin-7 , Cells, Cultured , Cytokines , Humans , Immunologic Memory , Interleukin-15/pharmacology , Interleukin-2/pharmacology , Interleukin-7/pharmacology , Memory T Cells , Transcriptome
17.
J Surg Res ; 274: 145-152, 2022 06.
Article in English | MEDLINE | ID: mdl-35151957

ABSTRACT

INTRODUCTION: The wound is known as damage to the skin structure by external stimuli, such as cut, bruises, and burn, which typically leaves the internal tissue exposed. The wound repair process when hampered leads to an excessive burden on the healthcare setting. Therefore, there is an urgent need to develop effective agents that can promote the wound healing process. In the present study, we intended to investigate the pharmacological benefit of Casticin (CST), a polymethylflavone against wound. METHODS: The wound in Wistar rats was induced by a surgical procedure. After surgery, the wound was examined for wound size over a period and for the expression of cyclooxygenase-2 and induced collagen III expressions. The effect of CST was examined on tumor necrosis factor α, interferon gamma, interleukin (IL)-10, transforming growth factor beta, and IL-7 by real-time polymerase chain reaction. The expression of matrix metalloproteinases (MMP)-2 and MMP-9 and its natural inhibitor (tissue inhibitors of metalloproteinases, TIMP1), level of macrophages, and lymphocytes were also quantified. The effect of CST was determined also on apoptosis of rats' splenocytes. RESULTS: CST significantly enhances wound healing of rat postsurgery, with maximum activity achieved in the case of a 60 µM treated group. The expression of cyclooxygenase-2 was found reduced together with an increase in collagen III, tumor necrosis factor α, interferon gamma, IL-10, transforming growth factor beta, and IL-7 in the CST group. The levels of MMP-2 and MMP-9 were also found reduced together with an increase in TIMP1 level in CST-treated group. The levels of CD4+, CD8+, and CD11b+ cells at the wound site 24 and 120 h postsurgery was also found reduced in CST-treated group. However, it showed no apoptosis in murine splenocytes. CONCLUSIONS: Collectively, CST can promote the wound healing process by modulation of inflammation and immune response, which induces tissue remodeling.


Subject(s)
Matrix Metalloproteinase 9 , Tumor Necrosis Factor-alpha , Animals , Collagen/metabolism , Cyclooxygenase 2/metabolism , Flavonoids , Interferon-gamma/metabolism , Interleukin-7/metabolism , Interleukin-7/pharmacology , Matrix Metalloproteinase 9/metabolism , Mice , Rats , Rats, Wistar , Skin/pathology , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Wound Healing
18.
Biochem J ; 479(3): 225-243, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35015072

ABSTRACT

The ability of the cellular immune system to discriminate self from foreign antigens depends on the appropriate calibration of the T cell receptor (TCR) signalling threshold. The lymphocyte homeostatic cytokine interleukin 7 (IL-7) is known to affect TCR thresholding, but the molecular mechanism is not fully elucidated. A better understanding of this process is highly relevant in the context of autoimmune disease therapy and cancer immunotherapy. We sought to characterise the early signalling events attributable to IL-7 priming; in particular, the altered phosphorylation of signal transduction proteins and their molecular localisation to the TCR. By integrating high-resolution proximity- phospho-proteomic and imaging approaches using primary T cells, rather than engineered cell lines or an in vitro expanded T cell population, we uncovered transduction events previously not linked to IL-7. We show that IL-7 leads to dephosphorylation of cytohesin interacting protein (CYTIP) at a hitherto undescribed phosphorylation site (pThr280) and alters the co-localisation of cytohesin-1 with the TCR and LFA-1 integrin. These results show that IL-7, acting via CYTIP and cytohesin-1, may impact TCR activation thresholds by enhancing the co-clustering of TCR and LFA-1 integrin.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Interleukin-7/pharmacology , Lymphocyte Function-Associated Antigen-1/metabolism , Proteome/metabolism , Proteomics/methods , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/drug effects , T-Lymphocytes/immunology , Transcription Factors/metabolism , Actin Cytoskeleton/metabolism , Blood Donors , Cells, Cultured , Humans , Lymphocyte Activation/drug effects , Phosphorylation/drug effects , Recombinant Proteins/pharmacology , Threonine/metabolism
19.
Haematologica ; 107(6): 1293-1310, 2022 06 01.
Article in English | MEDLINE | ID: mdl-34670357

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive cancer arising from lymphoblasts of T-cell origin. While TALL accounts for only 15% of childhood and 25% of adult ALL, 30% of patients relapse with a poor outcome. Targeted therapy of resistant and high-risk pediatric T-ALL is therefore urgently needed, together with precision medicine tools allowing the testing of efficacy in patient samples. Furthermore, leukemic cell heterogeneity requires drug response assessment at the single-cell level. Here we used single-cell mass cytometry to study signal transduction pathways such as JAK-STAT, PI3K-AKT-mTOR and MEK-ERK in 16 diagnostic and five relapsed T-ALL primary samples, and investigated the in vitro response of cells to Interleukin-7 (IL-7) and the inhibitor BEZ-235. T-ALL cells showed upregulated activity of the PI3K-AKT-mTOR and MEK-ERK pathways and increased expression of proliferation and translation markers. We found that perturbation induced by the ex vivo administration of either IL-7 or BEZ-235 reveals a high degree of exclusivity with respect to the phospho-protein responsiveness to these agents. Notably, these response signatures were maintained from diagnosis to relapse in individual patients. In conclusion, we demonstrated the power of mass cytometry single-cell profiling of signal transduction pathways in T-ALL. Taking advantage of this advanced approach, we were able to identify distinct clusters with different responsiveness to IL-7 and BEZ-235 that can persist at relapse. Collectively our observations can contribute to a better understanding of the complex signaling network governing T-ALL behavior and its correlation with influence on the response to therapy.


Subject(s)
Interleukin-7 , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Interleukin-7/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Recurrence , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/metabolism
20.
Front Immunol ; 12: 680442, 2021.
Article in English | MEDLINE | ID: mdl-34956167

ABSTRACT

Interleukin-7 (IL-7) is produced by stromal cells, keratinocytes, and epithelial cells in host tissues or tumors and exerts a wide range of immune effects mediated by the IL-7 receptor (IL-7R). IL-7 is primarily involved in regulating the development of B cells, T cells, natural killer cells, and dendritic cells via the JAK-STAT, PI3K-Akt, and MAPK pathways. This cytokine participates in the early generation of lymphocyte subsets and maintain the survival of all lymphocyte subsets; in particular, IL-7 is essential for orchestrating the rearrangement of immunoglobulin genes and T-cell receptor genes in precursor B and T cells, respectively. In addition, IL-7 can aid the activation of immune cells in anti-virus and anti-tumor immunity and plays important roles in the restoration of immune function. These biological functions of IL-7 make it an important molecular adjuvant to improve vaccine efficacy as it can promote and extend systemic immune responses against pathogens by prolonging lymphocyte survival, enhancing effector cell activity, and increasing antigen-specific memory cell production. This review focuses on the biological function and mechanism of IL-7 and summarizes its contribution towards improved vaccine efficacy. We hope to provide a thorough overview of this cytokine and provide strategies for the development of the future vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunogenicity, Vaccine/physiology , Immunomodulation/physiology , Interleukin-7/physiology , Vaccine Development , Animals , Cytokines/physiology , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Immunity, Mucosal , Immunologic Memory , Interleukin-7/administration & dosage , Interleukin-7/deficiency , Interleukin-7/pharmacology , Interleukin-7/therapeutic use , Intraepithelial Lymphocytes/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Mice , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...