Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 9.240
Filtrer
1.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38999946

RÉSUMÉ

The tumor cells reprogram their metabolism to cover their high bioenergetic demands for maintaining uncontrolled growth. This response can be mediated by cytokines such as IL-2, which binds to its receptor and activates the JAK/STAT pathway. Some reports show a correlation between the JAK/STAT pathway and cellular metabolism, since the constitutive activation of STAT proteins promotes glycolysis through the transcriptional activation of genes related to energetic metabolism. However, the role of STAT proteins in the metabolic switch induced by cytokines in cervical cancer remains poorly understood. In this study, we analyzed the effect of IL-2 on the metabolic switch and the role of STAT5 in this response. Our results show that IL-2 induces cervical cancer cell proliferation and the tyrosine phosphorylation of STAT5. Also, it induces an increase in lactate secretion and the ratio of NAD+/NADH, which suggest a metabolic reprogramming of their metabolism. When STAT5 was silenced, the lactate secretion and the NAD+/NADH ratio decreased. Also, the expression of HIF1α and GLUT1 decreased. These results indicate that STAT5 regulates IL-2-induced cell proliferation and the metabolic shift to aerobic glycolysis by regulating genes related to energy metabolism. Our results suggest that STAT proteins modulate the metabolic switch in cervical cancer cells to attend to their high demand of energy required for cell growth and proliferation.


Sujet(s)
Prolifération cellulaire , Interleukine-2 , Facteur de transcription STAT-5 , Tumeurs du col de l'utérus , Humains , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/génétique , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Interleukine-2/métabolisme , Interleukine-2/pharmacologie , Glycolyse/effets des médicaments et des substances chimiques , Métabolisme énergétique/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , Transporteur de glucose de type 1/métabolisme , Transporteur de glucose de type 1/génétique , NAD/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Acide lactique/métabolisme
2.
J Obstet Gynaecol ; 44(1): 2372682, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39034630

RÉSUMÉ

BACKGROUND: Interleukin (IL)-2 is a key cytokine capable of modulating the immune response by activating natural killer (NK) cells. This study was recruited to explore the therapeutic potential of IL-2-activated NK-92 cells in endometriosis in vitro. METHODS: Ectopic endometrial stromal cells (EESCs) were isolated and co-cultured with IL-2-activated NK-92 cells at varying effector-to-target (E:T) ratios (1:0 [Control], 1:1, 1:3, and 1:9). The viability, cytotoxicity, and cell surface antigen expression of IL-2-activated NK-92 cells were assessed. The viability, apoptosis, invasion, and migration ability of EESCs co-cultured with NK-92 cells at different ratios were evaluated. The apoptosis-related proteins, invasion and migration-related proteins as well as MEK/ERK pathway were examined via western blot. Each experiment was repeated three times. RESULTS: IL-2 activation enhanced NK-92 cytotoxicity in a concentration-dependent manner. Co-culturing EESCs with IL-2-activated NK-92 cells at E:T ratios of 1:1, 1:3, and 1:9 reduced EESC viability by 20%, 45%, and 70%, respectively, compared to the control group. Apoptosis rates in EESCs increased in correlation with the NK-92 cell proportion, with the highest rate observed at a 1:9 ratio. Moreover, EESC invasion and migration were significantly inhibited by IL-2-activated NK-92 cells, with a 60% reduction in invasion and a 50% decrease in migration at the 1:9 ratio. Besides, the MEK/ERK signalling pathway was down-regulated in EESCs by IL-2-activated NK-92 cells. CONCLUSION: IL-2-activated NK-92 cells exhibit potent cytotoxic effects against EESCs. They promote EESC apoptosis and inhibit viability, invasion, and migration through modulating the MEK/ERK signalling pathway.


Endometriosis is a common chronic systemic disease affecting approximately 190 million women worldwide. However, clinical treatments for endometriosis remain challenging due to the scarcity of high-quality scientific evidence and conflicting available guidelines. This research was designed to explore whether interleukin (IL)-2 affected the progression of endometriosis by modulating endometrial stromal cell apoptosis and natural killer (NK) cell-mediated cytotoxicity, thereby providing new therapeutic methods for endometriosis.


Sujet(s)
Apoptose , Techniques de coculture , Endométriose , Interleukine-2 , Cellules tueuses naturelles , Humains , Endométriose/anatomopathologie , Endométriose/immunologie , Femelle , Interleukine-2/pharmacologie , Interleukine-2/métabolisme , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Cellules tueuses naturelles/immunologie , Apoptose/effets des médicaments et des substances chimiques , Adulte , Endomètre/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cellules stromales/effets des médicaments et des substances chimiques , Évolution de la maladie , Survie cellulaire/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Cellules cultivées
3.
Signal Transduct Target Ther ; 9(1): 150, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38902241

RÉSUMÉ

This study aimed to develop a pan-genotypic and multifunctional small interfering RNA (siRNA) against hepatitis B virus (HBV) with an efficient delivery system for treating chronic hepatitis B (CHB), and explore combined RNA interference (RNAi) and immune modulatory modalities for better viral control. Twenty synthetic siRNAs targeting consensus motifs distributed across the whole HBV genome were designed and evaluated. The lipid nanoparticle (LNP) formulation was optimized by adopting HO-PEG2000-DMG lipid and modifying the molar ratio of traditional polyethylene glycol (PEG) lipid in LNP prescriptions. The efficacy and safety of this formulation in delivering siHBV (tLNP/siHBV) along with the mouse IL-2 (mIL-2) mRNA (tLNP/siHBVIL2) were evaluated in the rAAV-HBV1.3 mouse model. A siRNA combination (terms "siHBV") with a genotypic coverage of 98.55% was selected, chemically modified, and encapsulated within an optimized LNP (tLNP) of high efficacy and security to fabricate a therapeutic formulation for CHB. The results revealed that tLNP/siHBV significantly reduced the expression of viral antigens and DNA (up to 3log10 reduction; vs PBS) in dose- and time-dependent manners at single-dose or multi-dose frequencies, with satisfactory safety profiles. Further studies showed that tLNP/siHBVIL2 enables additive antigenic and immune control of the virus, via introducing potent HBsAg clearance through RNAi and triggering strong HBV-specific CD4+ and CD8+ T cell responses by expressed mIL-2 protein. By adopting tLNP as nucleic acid nanocarriers, the co-delivery of siHBV and mIL-2 mRNA enables synergistic antigenic and immune control of HBV, thus offering a promising translational therapeutic strategy for treating CHB.


Sujet(s)
Virus de l'hépatite B , Interleukine-2 , Nanoparticules , Petit ARN interférent , Animaux , Souris , Virus de l'hépatite B/génétique , Interleukine-2/génétique , Interleukine-2/immunologie , Interleukine-2/pharmacologie , Humains , Petit ARN interférent/génétique , Petit ARN interférent/administration et posologie , Nanoparticules/composition chimique , ARN messager/génétique , Hépatite B chronique/thérapie , Hépatite B chronique/génétique , Hépatite B chronique/virologie , Interférence par ARN , Hépatite B/thérapie , Hépatite B/génétique , Hépatite B/virologie , Thérapie par l'interférence par ARN , Liposomes
4.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38731828

RÉSUMÉ

HLA-DR-positive NK cells, found in both healthy individuals and patients with different inflammatory diseases, are characterized as activated cells. However, data on their capacity for IFNγ production or cytotoxic response vary between studies. Thus, more precise investigation is needed of the mechanisms related to the induction of HLA-DR expression in NK cells, their associations with NK cell differentiation stage, and functional or metabolic state. In this work, HLA-DR-expressing NK cell subsets were investigated using transcriptomic analysis, metabolic activity assays, and analysis of intercellular signaling cascades. We demonstrated that HLA-DR+CD56bright NK cells were characterized by a proliferative phenotype, while HLA-DR+CD56dim NK cells exhibited features of adaptive cells and loss of inhibitory receptors with increased expression of MHC class II trans-activator CIITA. The activated state of HLA-DR-expressing NK cells was confirmed by higher levels of ATP and mitochondrial mass observed in this subset compared to HLA-DR- cells, both ex vivo and after stimulation in culture. We showed that HLA-DR expression in NK cells in vitro can be induced both through stimulation by exogenous IL-2 and IL-21, as well as through auto-stimulation by NK-cell-produced IFNγ. At the intracellular level, HLA-DR expression depended on the activation of STAT3- and ERK1/2-mediated pathways, with subsequent activation of isoform 3 of the transcription factor CIITA. The obtained results broaden the knowledge about HLA-DR-positive NK cell appearance, diversity, and functions, which might be useful in terms of understanding the role of this subset in innate immunity and assessing their possible implications in NK cell-based therapy.


Sujet(s)
Différenciation cellulaire , Antigènes HLA-DR , Interféron gamma , Cellules tueuses naturelles , Cellules tueuses naturelles/métabolisme , Cellules tueuses naturelles/immunologie , Humains , Antigènes HLA-DR/métabolisme , Antigènes HLA-DR/génétique , Interféron gamma/métabolisme , Antigènes CD56/métabolisme , Activation des lymphocytes/immunologie , Interleukine-2/métabolisme , Interleukine-2/pharmacologie , Cellules cultivées , Protéines nucléaires , Transactivateurs
5.
CPT Pharmacometrics Syst Pharmacol ; 13(6): 1067-1078, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38676306

RÉSUMÉ

Regulatory T cells (Tregs) are essential for maintaining immune homeostasis by serving as negative regulators of adaptive immune system effector cell responses. Reduced production or function of Tregs has been implicated in several human autoimmune diseases. The cytokine interleukin 2 plays a central role in promoting Treg differentiation, survival, and function in vivo and may therefore have therapeutic benefits for autoimmune diseases. mRNA-6231 is an investigational, lipid nanoparticle-encapsulated, mRNA-based therapy that encodes a modified human interleukin 2 mutein fused to human serum albumin (HSA-IL2m). Herein, we report the development of a semi-mechanistic kinetic-pharmacodynamic model to quantify the relationship between subcutaneous dose(s) of mRNA-6231, HSA-IL2m protein expression, and Treg expansion in nonhuman primates. The nonclinical kinetic-pharmacodynamic model was extrapolated to humans using allometric scaling principles and the physiological basis of pharmacological mechanisms to predict the clinical response to therapy a priori. Model-based simulations were used to inform the dose selection and design of the first-in-human clinical study (NCT04916431). The modeling approach used to predict human responses was validated when data became available from the phase I clinical study. This validation indicates that the approach is valuable in informing clinical decision-making.


Sujet(s)
Interleukine-2 , ARN messager , Humains , Interleukine-2/pharmacocinétique , Interleukine-2/génétique , Interleukine-2/pharmacologie , Interleukine-2/administration et posologie , Animaux , ARN messager/génétique , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Nanoparticules , Modèles biologiques , Mâle , Liposomes
6.
Cancer Discov ; 14(7): 1206-1225, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38563906

RÉSUMÉ

IL2 signals pleiotropically on diverse cell types, some of which contribute to therapeutic activity against tumors, whereas others drive undesired activity, such as immunosuppression or toxicity. We explored the theory that targeting of IL2 to CD8+ T cells, which are key antitumor effectors, could enhance its therapeutic index. To this aim, we developed AB248, a CD8 cis-targeted IL2 that demonstrates over 500-fold preference for CD8+ T cells over natural killer and regulatory T cells (Tregs), which may contribute to toxicity and immunosuppression, respectively. AB248 recapitulated IL2's effects on CD8+ T cells in vitro and induced selective expansion of CD8+T cells in primates. In mice, an AB248 surrogate demonstrated superior antitumor activity and enhanced tolerability as compared with an untargeted IL2Rßγ agonist. Efficacy was associated with the expansion and phenotypic enhancement of tumor-infiltrating CD8+ T cells, including the emergence of a "better effector" population. These data support the potential utility of AB248 in clinical settings. Significance: The full potential of IL2 therapy remains to be unlocked. We demonstrate that toxicity can be decoupled from antitumor activity in preclinical models by limiting IL2 signaling to CD8+ T cells, supporting the development of CD8+ T cell-selective IL2 for the treatment of cancer. See related article by Kaptein et al. p. 1226.


Sujet(s)
Lymphocytes T CD8+ , Interleukine-2 , Animaux , Lymphocytes T CD8+/immunologie , Interleukine-2/pharmacologie , Souris , Humains , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Femelle , Tumeurs/immunologie , Tumeurs/traitement médicamenteux
7.
Zhongguo Zhong Yao Za Zhi ; 49(4): 1028-1043, 2024 Feb.
Article de Chinois | MEDLINE | ID: mdl-38621910

RÉSUMÉ

This study aims to decipher the mechanism of Buzhong Yiqi Decoction(BZYQD) in the treatment of spleen deficiency syndrome via gut microbiota. The mouse models of spleen deficiency syndrome were established by fecal microbiota transplantation(FMT, from patients with spleen deficiency syndrome) and administration of Sennae Folium(SF, 10 g·kg~(-1)), respectively, and treated with BZYQD for 5 d. The pseudosterile mice(administrated with large doses of antibiotics) and the mice transplanted with fecal bacteria from healthy human were taken as the controls. The levels of IgA, interleukin(IL)-2, IL-1ß, interferon(IFN)-γ, tumor necrosis factor-alpha(TNF-α), and 5-hydroxytryptamine(5-HT) in the intestinal tissue of two models were measured by enzyme-linked immunosorbent assay, and the CD8~+/CD3~+ ratio was determined by flow cytometry. The composition and changes of the gut microbiota were determined by 16S rRNA high-throughput sequencing and qPCR. Furthermore, the correlation analysis was performed to study the mediating role of gut microbiota in the treatment. The results showed that BZYQD elevated the IgA level, lowered the IL-1ß, TNF-α, and 5-HT levels, and decreased the CD8~+/CD3~+ ratio in the intestinal tissue of the two models. Moreover, BZYQD had two-way regulatory effects on the levels of IL-2 and IFN-γ. BZYQD inhibited the overgrowth and reduced the richness of gut microbiota in the SF model, and improved the gut microbiota structure in the two models. Algoriphagus, Mycobacterium, and CL500_29_marine_group were the common differential genera in the two models compared with the control. Acinetobacter, Parabacteroides, and Ruminococcus were the differential genera unique to the FMT model, and Sphingorhabdus, Lactobacillus, and Anaeroplasma were the unique differential genera in the SF model. BZYQD was capable of regulating all these genera. The qPCR results showed that BZYQD increased the relative abundance of Akkermansia muciniphila and decreased that of Bacteroides uniformis in the two models. The correlation analysis revealed that the levels of above intestinal cytokines were significantly correlated with characteristic gut microorganisms in different mo-dels. The IL-1ß level had a significantly positive correlation with Acinetobacter and CL500_29_marine_group in the two models, while the different levels of IL-2 and IFN-γ in the two models may be related to its different gut microbiota structures. In conclusion, BZYQD could regulate the disordered gut microbiota structure in different animal models of spleen deficiency syndrome to improve the intestinal immune status, which might be one of the mechanisms of BZYQD in treating spleen deficiency syndrome.


Sujet(s)
Microbiome gastro-intestinal , Rate , Humains , Souris , Animaux , Facteur de nécrose tumorale alpha/pharmacologie , ARN ribosomique 16S/génétique , Interleukine-2/pharmacologie , Sérotonine , Immunoglobuline A/pharmacologie
8.
Int J Hematol ; 119(6): 626-630, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38581458

RÉSUMÉ

As multiple myeloma (MM) progresses, immune effector cells decrease in number and function and become exhausted. This remains an insurmountable clinical issue that must be addressed by development of novel modalities to revitalize anti-MM immunity. Human Vγ9Vδ2 T (Vδ2+ γδ T) cells serve as the first line of defense against pathogens as well as tumors and can be expanded ex vivo from peripheral blood mononuclear cells (PBMCs) upon treatment with amino-bisphosphonates in combination with IL-2. Here, we demonstrated that next-generation immunomodulators called cereblon E3 ligase modulators (CELMoDs), as well as lenalidomide and pomalidomide, expanded Th1-like Vδ2+ γδ T cells from PBMCs in the presence of zoledronic acid (ZA). However, the expansion of Th1-like Vδ2+ γδ T cells by these immunomodulatory drugs was abolished under IL-2 blockade, although IL-2 production was induced in PBMCs. BTN3A1 triggers phosphoantigen presentation to γδ T-cell receptors and is required for γδ T-cell expansion and activation. ZA but not these immunomodulatory drugs upregulated BTN3A1 in monocytes. These results suggest that immunomodulatory drugs and ZA have cooperative roles in expansion of Th1-like Vδ2+ γδ T cells, and provide the important knowledge for clinical application of human Vδ2+ γδ T cells as effector cells.


Sujet(s)
Diphosphonates , Imidazoles , Activation des lymphocytes , Myélome multiple , Récepteur lymphocytaire T antigène, gamma-delta , Thalidomide , Acide zolédronique , Acide zolédronique/pharmacologie , Humains , Récepteur lymphocytaire T antigène, gamma-delta/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Myélome multiple/traitement médicamenteux , Myélome multiple/immunologie , Diphosphonates/pharmacologie , Imidazoles/pharmacologie , Thalidomide/analogues et dérivés , Thalidomide/pharmacologie , Butyrophilines , Interleukine-2/pharmacologie , Lénalidomide/pharmacologie , Ubiquitin-protein ligases , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines adaptatrices de la transduction du signal , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/immunologie , Agranulocytes/métabolisme , Antigènes CD
9.
J Clin Invest ; 134(5)2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38426502

RÉSUMÉ

Immune tolerance to allogenic transplanted tissues remains elusive, and therapeutics promoting CD4+FOXP3+ Tregs are required to achieve this ultimate goal. In this issue of the JCI, Efe and colleagues engineered an Fc domain fused to a human mutein IL-2 (mIL-2-Fc) bearing mutations that confer preferential binding to the high-affinity IL-2 receptor expressed on Tregs. In vivo mIL-2-Fc therapy effectively heightened mouse, monkey, and human Treg numbers, promoted tolerance to minor antigen mismatched skin grafts in mice, and synergized with immunosuppressive drugs used in the clinic. These findings warrant clinical trials that assess the efficacy of mIL-2-Fc in transplantation.


Sujet(s)
Interleukine-2 , Tolérance à la transplantation , Souris , Humains , Animaux , Tolérance à la transplantation/génétique , Interleukine-2/génétique , Interleukine-2/pharmacologie , Lymphocytes T régulateurs , Immunosuppresseurs , Tolérance immunitaire
10.
J Leukoc Biol ; 115(6): 1108-1117, 2024 05 29.
Article de Anglais | MEDLINE | ID: mdl-38374693

RÉSUMÉ

A well-documented Achilles heel of current cancer immunotherapy approaches is T cell exhaustion within solid tumor tissues. The proinflammatory cytokine interleukin (IL)-23 has been utilized to augment chimeric antigen receptor (CAR) T cell survival and tumor immunity. However, in-depth interrogation of molecular events downstream of IL-23/IL-23 receptor signaling is hampered by a paucity of suitable cell models. The current study investigates the differential contribution of IL-2 and IL-23 to the maintenance and differentiation of the IL-23 responsive Kit225 T-cell line. We observed that IL-23 enhanced cellular fitness and survival but was insufficient to drive proliferation. IL-23 rapidly induced phosphorylation of STAT1, STAT3, and STAT4, and messenger RNA expression of IL17A, the archetypal effector cytokine of T helper 17 (Th17) cells, but not their lineage markers RORC and NCR1. These observations suggest that IL-23 endowed Th17/ILC3-like effector function but did not promote their differentiation. In contrast, spontaneous differentiation of Kit225 cells toward a Th17/ILC3-like phenotype was induced by prolonged IL-2 withdrawal. This was marked by strongly elevated basal IL17A and IL17F expression and the secretion of IL-17. Together, our data present Kit225 cells as a valuable model for studying the interplay between cytokines and their contribution to T cell survival, proliferation, and differentiation.


Sujet(s)
Différenciation cellulaire , Interleukine-23 , Interleukine-2 , Cellules Th17 , Humains , Lignée cellulaire , Prolifération cellulaire , Survie cellulaire , Interleukine-17/métabolisme , Interleukine-17/immunologie , Interleukine-2/pharmacologie , Interleukine-23/métabolisme , Interleukine-23/immunologie , Transduction du signal , Cellules Th17/immunologie
11.
Front Immunol ; 15: 1325024, 2024.
Article de Anglais | MEDLINE | ID: mdl-38420118

RÉSUMÉ

In chickens, γδ T cells represent a large fraction of peripheral T cells; however, their function remains largely unknown. Here, we describe the selective in vitro expansion of γδ T cells from total splenocytes by stimulation with the cytokines IL-2 and IL-12. Under these conditions, γδ T cells proliferated preferentially and reached frequencies of >95% within three weeks. Although IL-2 alone also triggered proliferation, an increased proliferation rate was observed in combination with IL-12. Most of the expanded cells were γδ TCR and CD8 double-positive. Splenocytes sorted into TCR1+CD8+, TCR1highCD8-, and TCR1lowCD8- subsets proliferated well upon dual stimulation with IL-2/IL-12, indicating that none of the three γδ T cell subsets require bystander activation for proliferation. TCR1+CD8+ cells maintained CD8 surface expression during stimulation, whereas CD8- subpopulations showed varied levels of CD8 upregulation, with the highest upregulation observed in the TCR1high subset. Changes in the γδ T-cell receptor repertoire during cell culture from day 0 to day 21 were analyzed by next-generation sequencing of the γδ variable regions. Overall, long-term culture led to a restricted γ and δ chain repertoire, characterized by a reduced number of unique variable region clonotypes, and specific V genes were enriched at day 21. On day 0, the δ chain repertoire was highly diverse, and the predominant clonotypes differed between animals, while the most frequent γ-chain clonotypes were shared between animals. However, on day 21, the most frequent clonotypes in both the γ and δ chain repertoires were different between animals, indicating that selective expansion of dominant clonotypes during stimulation seems to be an individual outcome. In conclusion, IL-2 and IL-12 were sufficient to stimulate the in vitro outgrowth of γδ T cells. Analyses of the TCR repertoire indicate that the culture leads to an expansion of individual T cell clones, which may reflect previous in vivo activation. This system will be instrumental in studying γδ T cell function.


Sujet(s)
Poulets , Interleukine-2 , Animaux , Interleukine-2/pharmacologie , Interleukine-12 , Récepteur lymphocytaire T antigène, gamma-delta/génétique , Techniques de culture cellulaire
12.
Cancer Med ; 13(3): e6909, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38317590

RÉSUMÉ

BACKGROUND: The potent immune effects of interleukin-2 (IL-2) for cancer therapy can be increased by genetic fusion of IL-2 to the Fc domain of an antibody (IL-2-Fc) or tumor targeted by genetic fusion to a whole antibody known as an immunocytokine (ICK). METHODS: An anti-CEA ICK (M5A-IL-2) was compared to an IL-2-Fc fusion protein using tumor therapy and PET imaging in CEA transgenic immunocompetent mice bearing CEA positive colon or breast tumors. Combination with stereotactic radiation therapy (SRT) was performed with either ICK or IL-2-Fc. RESULTS: ICK and IL-2-Fc had comparable antitumor effects in both tumor models, although ICK had higher tumor uptake and slower blood clearance than an IL-2-Fc. Analysis of IFNγ+ /CD8+ and FoxP3+ /CD4+ T cells revealed higher levels of IFNγ-producing CD8+ T cells in ICK treated mice versus more efficient Treg elimination in IL-2-Fc treated mice. No significant or lasting toxicity was detected for either agent. Combination therapies with SRT revealed comparable efficacy and induction of immune memory for both ICK and IL-2-Fc when mice were rechallenged post-therapy. CONCLUSIONS: IL-2-Fc had comparable antitumor efficacy to CEA-targeted M5A-IL-2 ICK, while both fusion proteins induced immune memory when combined with SRT. Differences in the therapeutic mechanisms of both agents were observed.


Sujet(s)
Tumeurs , Radiochirurgie , Souris , Animaux , Interleukine-2/pharmacologie , Lymphocytes T CD8+ , Tumeurs/thérapie , Anticorps , Souris transgéniques
13.
Biomaterials ; 306: 122475, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38306733

RÉSUMÉ

Although tumor-infiltrating T lymphocytes (TIL-Ts) play a crucial role in solid tumor immunotherapy, their clinical application has been limited because of the immunosuppressive microenvironment. Herein, we developed an injectable hydrogel microsphere-integrated training court (MS-ITC) to inspire the function of TIL-Ts and amplify TIL-Ts, through grafting with anti-CD3 and anti-CD28 antibodies and bovine serum albumin nanoparticles encapsulated with IL-7 and IL-15. MS-ITC provided the T-cell receptor and co-stimulatory signals required for TIL-Ts activation and IL-7/IL-15 signals for TIL-Ts expansion. Afterward, the MS-ITC was injected locally into the osteosarcoma tumor tissue in mice. MS-ITC suppressed the growth of primary osteosarcoma by more than 95 %, accompanied with primed and expanded TIL-Ts in the tumor tissues, compromising significantly increased CD8+ T and memory T cells, thereby enhancing the anti-tumor effect. Together, this work provides an injectable hydrogel microsphere-integrated training platform capable of inspiring TIL-Ts potential for a range of solid tumor immunotherapy.


Sujet(s)
Interleukine-15 , Tumeurs , Animaux , Souris , Hydrogels , Interleukine-7 , Microsphères , Cytotoxicité immunologique , Lymphocytes TIL , Lymphocytes T , Interleukine-2/pharmacologie , Activation des lymphocytes , Microenvironnement tumoral
14.
Drug Discov Ther ; 18(1): 24-33, 2024 Mar 20.
Article de Anglais | MEDLINE | ID: mdl-38382992

RÉSUMÉ

Maintenance of pregnancy is highly dependent on the maternal immune system. High levels of regulatory T cells (Tregs) accumulate in the maternal placenta to suppress immunoreactivity against fetal antigens. We assessed whether Astragalus root (AsR) and AsR-containing Kampo medicines modulate immunoreactivity and thereby increase mouse litter size. AsR-exposed murine splenocytes exhibited significantly increased IL-2 secretion. In AsR-exposed mice, total Tregs were significantly increased, whereas cytotoxic T lymphocyte antigen 4 (CTLA-4)-positive Tregs were decreased in AsR-exposed mice. Tregs express IL-2 receptor subunit alpha and are activated by IL-2. CTLA-4 interacts with B7 expressed in antigen-presenting cells (APCs) with high affinity, and CTLA-4/B7 signaling plays a critical role in inhibiting APC activity, thereby suppressing CD4+ T cell proliferation and activation. The decrease in CTLA-4+ Tregs in AsR-exposed mice is thought to induce an increase in CD4+ T cells, leading to increased IL-2 secretion from CD4+ T cells followed by Treg activation. Th17 cells prevent trophoblast apoptosis, resulting in trophoblast invasion into the decidua. AsR increases Th17 cells, thereby inducing dose-dependent increases in litter size. Although Keishikaogito (KO)- and Ogikenchuto (OK)-exposed mice exhibited increased IL-2 secretion and splenic Tregs, KO also increased CTLA-4+ Tregs. Therefore, KO promoted immunosuppression by increasing CTLA-4+ Tregs, which induced a decrease in Th17 and exerted little effect on litter size. Therefore, an increase in both Tregs and Th17 cells can be considered necessary for embryo implantation and pregnancy maintenance.


Sujet(s)
Interleukine-2 , Lymphocytes T régulateurs , Grossesse , Femelle , Souris , Animaux , Antigène CTLA-4 , Interleukine-2/pharmacologie , Cellules Th17 , Implantation embryonnaire , Maintien de la grossesse
15.
Front Immunol ; 15: 1309509, 2024.
Article de Anglais | MEDLINE | ID: mdl-38352877

RÉSUMÉ

Immunotherapy of tumors plays a pivotal role in the current treatment of cancer. While interleukin 2 (IL-2) demonstrated its efficacy as an immunotherapeutic drug in the early days, its short blood circulation time poses challenges in maintaining effective therapeutic concentrations. Additionally, IL-2's activation of regulatory T cells can counteract its anti-cancer effects. Therefore, the primary goal of this study was to formulate IL-2-carrying nanoparticles via boron-nitrogen coordination between methoxy poly (ethylene glycol) block poly-[(N-2-hydroxyethyl)-aspartamide]phenylboronic acid (mPEG-b-PHEA-PBA, P-PBA) and poly (L-lysine) (PLL). These nanoparticles are intended to be used in combination with CDK4/6 inhibitors to address the short blood circulation time of IL-2, reduce its immunosuppressive effects, and enhance the overall immune response. The envisaged outcome is a sustained and potent therapeutic effect, offering a novel and promising combination therapy strategy for tumor immunotherapy.


Sujet(s)
Tumeurs du côlon , Nanoparticules , Pipérazines , Pyridines , Humains , Interleukine-2/pharmacologie , Interleukine-2/usage thérapeutique , Tumeurs du côlon/traitement médicamenteux , Tumeurs du côlon/anatomopathologie , Immunité
16.
Discov Med ; 36(180): 140-149, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38273754

RÉSUMÉ

BACKGROUND: Endometritis is a condition usually resulted from the bacterial infection of uterus, causing pelvic disease, sepsis, shock, uterine necrosis and even death if it is inappropriately treated. The aim of this study is to explore the pathogenesis of endometritis, and investigate whether the combination of doxycycline and metronidazole offers stronger protection against lipopolysaccharide (LPS)-induced endometritis, and decipher more about the mechanisms underlying endometritis-related pyroptosis. METHODS: Sprague-Dawley (SD) rats were divided into five groups (n = 8 per group): control, model, metronidazole, doxycycline, and combination groups. In control group, the rats were injected with saline, while in other groups, lipopolysaccharide was injected into uterus of the rats to establish endometritis. Hematoxylin-eosin (H&E) staining was performed as part of the histopathological examination of endometrium. The integrity of chromatin and pyroptosis were evaluated by terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling (TUNEL) assay. Western blot and quantitative real-time reverse-transcription polymerase chain reaction (qRT-PCR) were performed to ascertain the activation of toll-like receptors (TLR4)/nuclear factor-kappa B (NF-κB) pathway by detecting protein levels of phosphorylated p50 (p-p50)/p50, phosphorylated nuclear factor-kappa B (p-NF-κB)/NF-κB, phosphorylated IkappaB (p-IκB), and TLR4 protein and mRNA. Development of pyroptosis was also detected by determining the levels of caspase-1 and caspase-5 through Western blot and qRT-PCR. Enzyme-linked immunosorbent assay (ELISA) was used to detect levels of interleukin (IL)-1ß, IL-18, IL-2, IL-4, IL-6 and tumor necrosis factor alpha (TNF-α), and flow cytometry was adopted to determine T-helper (Th)1 and Th2 cell percentage to assess the extent of pyroptosis and Th1/Th2 imbalance. RESULTS: The uterine of the model group exhibited pathological alterations and higher degree of cell apoptosis. Compared with the control rats, model group showed lower protein levels of p-p50/p50 (p < 0.001), p-NF-κB/NF-κB (p < 0.001), p-IκB (p < 0.001), and TLR4 protein (p < 0.001) and mRNA (p < 0.001). Elevated levels of caspase-1 (p < 0.001), caspase-5 (p < 0.001), IL-1ß (p < 0.001), IL-18 (p < 0.001), IL-2 (p < 0.01), TNF-α (p < 0.05) and Th1/Th2 (p < 0.001) as well as reduced levels of IL-4 (p < 0.05) and IL-6 (p < 0.01) were observed in the model group, which could however be reversed by metronidazole (p < 0.01) or doxycycline (p < 0.01), with a more significant effect detected if a combination of the two drugs was administered (p < 0.01). CONCLUSIONS: The combination of doxycycline and metronidazole protects against rat endometritis by inhibiting TLR4/NF-κB pathway-mediated inflammation and suppressing pyroptosis.


Sujet(s)
Endométrite , Facteur de transcription NF-kappa B , Humains , Femelle , Rats , Animaux , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/pharmacologie , Endométrite/traitement médicamenteux , Interleukine-18/pharmacologie , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme , Métronidazole/usage thérapeutique , Métronidazole/pharmacologie , Doxycycline/pharmacologie , Transduction du signal , Facteur de nécrose tumorale alpha/pharmacologie , Lipopolysaccharides/pharmacologie , Interleukine-6/métabolisme , Pyroptose , Interleukine-2/pharmacologie , Interleukine-4/pharmacologie , Rat Sprague-Dawley , Caspases/métabolisme , Caspases/pharmacologie , ARN messager/génétique
17.
BioDrugs ; 38(2): 227-248, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-37999893

RÉSUMÉ

The interleukin-2 (IL-2) cytokine plays a crucial role in regulating immune responses and maintaining immune homeostasis. Its immunosuppressive effects have been harnessed therapeutically via administration of low cytokine doses. Low-dose IL-2 has shown promise in the treatment of various autoimmune and inflammatory diseases; however, the clinical use of IL-2 is complicated by its toxicity, its pleiotropic effects on both immunostimulatory and immunosuppressive cell subsets, and its short serum half-life, which collectively limit the therapeutic window. As a result, there remains a considerable need for IL-2-based autoimmune disease therapies that can selectively target regulatory T cells with minimal off-target binding to immune effector cells in order to prevent cytokine-mediated toxicities and optimize therapeutic efficacy. In this review, we discuss exciting advances in IL-2 engineering that are empowering the development of novel therapies to treat autoimmune conditions. We describe the structural mechanisms of IL-2 signaling, explore current applications of IL-2-based compounds as immunoregulatory interventions, and detail the progress and challenges associated with clinical adoption of IL-2 therapies. In particular, we focus on protein engineering approaches that have been employed to optimize the regulatory T-cell bias of IL-2, including structure-guided or computational design of cytokine mutants, conjugation to polyethylene glycol, and the development of IL-2 fusion proteins. We also consider future research directions for enhancing the translational potential of engineered IL-2-based therapies. Overall, this review highlights the immense potential to leverage the immunoregulatory properties of IL-2 for targeted treatment of autoimmune and inflammatory diseases.


Sujet(s)
Maladies auto-immunes , Interleukine-2 , Humains , Interleukine-2/génétique , Interleukine-2/pharmacologie , Interleukine-2/usage thérapeutique , Maladies auto-immunes/traitement médicamenteux , Lymphocytes T régulateurs , Cytokines , Immunothérapie
18.
Trends Cancer ; 10(4): 332-346, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38129234

RÉSUMÉ

There is considerable interest in developing more effective programmed cell death (PD)-1 combination therapies against cancer. One major obstacle to these efforts is a dysfunctional/exhausted state of CD8 T cells, which PD-1 monotherapy is not able to overcome. Recent studies have highlighted that PD-1+ T cell factor (TCF)-1+ stem-like CD8 T cells are not fate locked into the exhaustion program and their differentiation trajectory can be changed by interleukin (IL)-2 signals. Modifying the CD8 T cell exhaustion program and generating better effectors from stem-like CD8 T cells by IL-2 form the fundamental immunological basis for combining IL-2 with PD-1 therapy. Many versions of IL-2-based products are being tested and each product should be carefully evaluated for its ability to modulate dysfunctional states of anti-tumor CD8 T cells.


Sujet(s)
Tumeurs , Récepteur-1 de mort cellulaire programmée , Humains , Récepteur-1 de mort cellulaire programmée/métabolisme , Interleukine-2/pharmacologie , Interleukine-2/usage thérapeutique , Interleukine-2/métabolisme , Lymphocytes T CD8+ , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Différenciation cellulaire
19.
Oncoimmunology ; 13(1): 2293511, 2024.
Article de Anglais | MEDLINE | ID: mdl-38125721

RÉSUMÉ

Anti-PD-1 antibody therapy has achieved success in tumor treatment; however, the duration of its clinical benefits are typically short. The functional state of intratumoral CD8+ T cells substantially affects the efficacy of anti-PD-1 antibody therapy. Understanding how intratumoral CD8+ T cells change will contribute to the improvement in anti-PD-1 antibody therapy. In this study, we found that tumor growth was not arrested after the late administration of anti-PD-1 antibody and that the antitumor function of CD8+ T cells decreased with tumor progression. The results of the RNA sequencing of CD8+ T cells infiltrating the tumor site on days 7 and 14 showed that the cell adhesion molecule Lymphocyte Function-associated Antigen-1 (LFA-1) participates in regulating the antitumor function of CD8+ T cells and that decreased LFA-1 expression in intratumoral CD8+ T cells is associated with tumor progression. By analyzing the Gene Expression Omnibus (GEO) database and our results, we found that the antitumor function of intratumoral CD8+ T cells with high LFA-1 expression was stronger. The formation of immune synapses is impaired in Itgal-si CD8+ T cells, resulting in decreased anti-tumor function. LFA-1 expression in intratumoral CD8+ T cells is regulated by the IL-2/STAT5 pathway. The combination of IL-2 and anti-PD-1 antibody effectively enhanced LFA-1 expression and the antitumor function of intratumoral CD8+ T cells. The adoptive transfer of OT-1 T cells overexpressing LFA-1, STAT5A, or STAT5B resulted in higher antitumor function, deferred tumor growth, and prolonged survival. These findings indicate that LFA-1-mediated immune synapse acts as a regulator of the antitumor function of intratumoral CD8+ T cells, which can be applied to improve anti-PD-1 antibody therapy.


Sujet(s)
Lymphocytes T CD8+ , Tumeurs , Humains , Antigène-1 associé à la fonction du lymphocyte/métabolisme , Interleukine-2/pharmacologie , Facteur de transcription STAT-5/métabolisme , Molécules d'adhérence cellulaire
20.
ACS Nano ; 18(1): 770-782, 2024 Jan 09.
Article de Anglais | MEDLINE | ID: mdl-38113242

RÉSUMÉ

Interleukin-2 (IL-2) used in multiple sclerosis (MS) therapy modulates the balance between regulatory T (Treg) cells and effector T (Teff) cells. However, the off-target activation of Teff cells by IL-2 limits its clinical application. Therefore, a rapidly prepared immunoswitch nanomodulator termed aT-IL2C NPs was developed, which specifically recognized Treg cells with high TIGIT expression thanks to the presence of an anti-TIGIT and an IL-2/JES6-1 complex (IL2C) being delivered to Treg cells but not to Teff cells with low TIGIT expression. Then, IL2C released IL-2 due to the specific expression of the high-affinity IL-2 receptor on Treg cells, thus enabling the active targeting and selective proliferation of Treg cells. Moreover, the anti-TIGIT of aT-IL2C NPs selectively inhibited the proliferation of Teff cells while leaving the proliferation of Treg cells unaffected. In addition, since the IL-2 receptor on Teff cells had medium-affinity, the IL2C hardly released IL-2 to Teff cells, thus enabling the inhibition of Teff cell proliferation. The treatment of experimental autoimmune encephalomyelitis (EAE) mice with aT-IL2C NPs ameliorated the severity of the EAE and restored white matter integrity. Collectively, this work described a potential promising agent for effective MS therapy.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Sclérose en plaques , Souris , Animaux , Lymphocytes T régulateurs , Sclérose en plaques/traitement médicamenteux , Sclérose en plaques/métabolisme , Interleukine-2/pharmacologie , Interleukine-2/usage thérapeutique , Interleukine-2/métabolisme , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Prolifération cellulaire , Souris de lignée C57BL
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE