Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 825
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Front Cell Infect Microbiol ; 14: 1437207, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39386167

RESUMO

Introduction: Pulmonary tuberculosis (PTB) remains one of the deadliest infectious diseases. Understanding PTB immunity is of potential value for exploring immunotherapy for treating chemotherapy-resistant PTB. CD4+CD25+Foxp3+ regulatory T cells (Tregs) are key players that impair immune responses to Mycobacteria tuberculosis (MTB). Currently, the intrinsic factors governing Treg expansion and influencing the immunosuppressive attributes of Tregs in PTB patients are far from clear. Methods: Here, we employed flow cytometry to determine the frequency of Tregs and the expression of B and T lymphocyte attenuator (BTLA) and its ligand, herpesvirus entry mediator (HVEM), on Tregs in patients with active PTB. Furthermore, the expression of conventional T cells and of programmed death-ligand 1 (PD-L1) and programmed death-1 (PD-1) on Tregs in patients with active PTB was determined. We then examined the characteristics of BTLA/HVEM expression and its correlation with Treg frequency and PD-L1 and PD-1 expression on Tregs in PTB patients. Results: The frequency of Tregs was increased in PTB patients and it had a relevance to PTB progression. Intriguingly, the axis of cosignal molecules, BTLA and HVEM, were both downregulated on the Tregs of PTB patients compared with healthy controls (HCs), which was the opposite of their upregulation on conventional T cells. Unexpectedly, their expression levels were positively correlated with the frequency of Tregs, respectively. These seemingly contradictory results may be interpreted as follows: the downregulation of BTLA and HVEM may alleviate BTLA/HVEM cis-interaction-mediated coinhibitory signals pressing on naïve Tregs, helping their activation, while the BTLA/HVEM axis on effector Tregs induces a costimulatory signal, promoting their expansion. Certainly, the mechanism underlying such complex effects remains to be explored. Additionally, PD-L1 and PD-1, regarded as two of the markers characterizing the immunosuppressive attributes and differentiation potential of Tregs, were upregulated on the Tregs of PTB patients. Further analysis revealed that the expression levels of BTLA and HVEM were positively correlated with the frequency of PD-1+Tregs and PD-L1+Tregs, respectively. Conclusion: Our study illuminated distinct characteristics of BTLA/HVEM axis expression on Tregs and uncovered its impact on the expansion and attributes of Tregs in patients with active PTB. Therefore, blockade of the BTLA/HVEM axis may be a promising potential pathway to reduce Treg expansion for the improvement of anti-MTB immune responses.


Assuntos
Antígeno B7-H1 , Receptor de Morte Celular Programada 1 , Receptores Imunológicos , Membro 14 de Receptores do Fator de Necrose Tumoral , Linfócitos T Reguladores , Tuberculose Pulmonar , Humanos , Linfócitos T Reguladores/imunologia , Receptores Imunológicos/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Tuberculose Pulmonar/imunologia , Masculino , Feminino , Adulto , Receptor de Morte Celular Programada 1/metabolismo , Pessoa de Meia-Idade , Antígeno B7-H1/metabolismo , Mycobacterium tuberculosis/imunologia , Adulto Jovem , Citometria de Fluxo
2.
Front Immunol ; 15: 1411392, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39351218

RESUMO

Haploidentical hematopoietic cell transplantation (haplo-HCT) is associated with an increased risk of allograft rejection. Here, we employed a major histocompatibility complex (MHC)-mismatched allogeneic HCT (allo-HCT) murine model to better understand the role of Gal-1 in immune tolerance. Transplanted mice were classified into either rejected or engrafted based on donor chimerism levels. We noted significantly higher frequencies of CD4+ T cells, CD8+ T cells, natural killer cells, IFN-γ and TNF-α producing CD4+ T cells, and IFN-γ producing dendritic cells and macrophages in rejected mice. Conversely, we found significantly increased frequencies of regulatory T cells (Tregs), predominantly Helios+, IL-10-producing CD4+ T cells, type 1 regulatory (Tr1) cells, and the proportion of Tr1+Gal-1+ cells in engrafted mice. Further, Gal-1 specific blockade in Tregs reduced suppression of effector T cells in engrafted mice. Lastly, effector T cells from engrafted mice were more prone to undergo apoptosis. Collectively, we have shown that Gal-1 may favor HSC engraftment in an MHC-mismatched murine model. Our results demonstrate that Gal-1-expressing Tregs, especially at earlier time points post-transplant, are associated with inducing immune tolerance and stable mixed chimerism after HCT.


Assuntos
Galectina 1 , Transplante de Células-Tronco Hematopoéticas , Linfócitos T Reguladores , Animais , Camundongos , Galectina 1/imunologia , Galectina 1/metabolismo , Linfócitos T Reguladores/imunologia , Camundongos Endogâmicos C57BL , Rejeição de Enxerto/imunologia , Transplante Homólogo , Complexo Principal de Histocompatibilidade/imunologia , Sobrevivência de Enxerto/imunologia , Camundongos Endogâmicos BALB C , Tolerância Imunológica
3.
Cell Rep Med ; : 101751, 2024 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-39357524

RESUMO

Although oncolytic adenoviruses are widely studied for their direct oncolytic activity and immunomodulatory role in cancer immunotherapy, the immunosuppressive feedback loop induced by oncolytic adenoviruses remains to be studied. Here, we demonstrate that type V adenovirus (ADV) induces the polarization of tumor-associated macrophages (TAMs) to the M2 phenotype and increases the infiltration of regulatory T cells (Tregs) in the tumor microenvironment (TME). By selectively compensating for these deficiencies, thymosin alpha 1 (Tα1) reprograms "M2-like" TAMs toward an antitumoral phenotype, thereby reprogramming the TME into a state more beneficial for antitumor immunity. Moreover, ADVTα1 is constructed by harnessing the merits of all the components for the aforementioned combinatorial therapy. Both exogenously supplied and adenovirus-produced Tα1 orchestrate TAM reprogramming and enhance the antitumor efficacy of ADV via CD8+ T cells, showing promising prospects for clinical translation. Our findings provide inspiration for improving oncolytic adenovirus combination therapy and designing oncolytic engineered adenoviruses.

4.
Int Immunopharmacol ; 142(Pt A): 113105, 2024 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-39260310

RESUMO

Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal cell carcinoma and has a poor prognosis. Despite the impressive advancements in treating ccRCC using immune checkpoint (IC) blockade, such as PD-1/PD-L1 inhibitors, a considerable number of ccRCC patients experience adaptive resistance. Therefore, exploring new targetable ICs will provide additional treatment options for ccRCC patients. We comprehensively analyzed multi-omics data and performed functional experiments, such as pathologic review, bulk transcriptome data, single-cell sequencing data, Western blotting, immunohistochemistry and in vitro/in vivo experiments, to explore novel immunotherapeutic targets in ccRCC. It was found that immune-related genes VSIG4, SAA1, CD7, FOXP3, IL21, TNFSF13B, BATF, CD72, MZB1, LTB, CCL25 and KLRK1 were significantly upregulated in ccRCC (Student's t test and p-value < 0.05; 36 normal and 267 ccRCC tissues in raining cohort; 36 normal and 266 ccRCC tissues in validation cohort) and correlated with the poor prognosis of ccRCC patients (Wald test and p-value < 0.05 in univariate cox analysis; log-rank test and p-value < 0.05 in Kaplan-Meier method; 267 patients in training cohort and 266 in validation cohort). In particular, we found the novel IC target VSIG4 was specifically expressed in inhibitory immune cells M2-biased tumor-associated macrophages (TAMs), conventional dendritic cell 2 (cDC2) cells, and cycling myeloid cells in ccRCC microenvironment. Moreover, VSIG4 showed a closely relation with resistance of Ipilimumab/Nivolumab immunotherapy in ccRCC. Furthermore, VSIG4 promoted the infiltration of M2 macrophages, Tregs, and cDC2 in ccRCC tissues. VSIG4+ TAMs and VSIG4+ cDC2s may be a kind of immune cell subtypes related to immunosuppression. VSIG4 may play similar roles with other IC ligands, as it is highly expressed on the surface of antigen-presenting cells and ccRCC cells to inhibit T cells activity and facilitate immune escape. Targeting IC gene VSIG4 may provide a novel immunotherapeutic strategy to ccRCC patients with resistance to existing targeted therapy options.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Macrófagos , Linfócitos T Reguladores , Microambiente Tumoral , Carcinoma de Células Renais/imunologia , Humanos , Neoplasias Renais/imunologia , Neoplasias Renais/patologia , Neoplasias Renais/tratamento farmacológico , Microambiente Tumoral/imunologia , Linfócitos T Reguladores/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Animais , Linhagem Celular Tumoral , Masculino , Camundongos , Feminino , Regulação Neoplásica da Expressão Gênica , Prognóstico
5.
bioRxiv ; 2024 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-39345626

RESUMO

Background: This work seeks to understand whether IL15-incorporating treatments improve response to radiotherapy and uncover mechanistic rationale for overcoming resistance to IL15 agonism using novel therapeutic combinations. Experimental Design: Orthotopic tumor models of PDAC were used to determine response to treatment. IL15-/- and Rag1-/- mouse models were employed to determine dependence on IL15 and CTLs, respectively. Flow cytometry was used to assess immune cell frequency and activation state. Phospho-proteomic analyses were used to characterize intracellular signaling pathways. Results: We show that the combination of radiation therapy (RT) and an IL15/IL15Ra fusion complex (denoted IL15c) fails to confer anti-tumor efficacy; however, a CD8-driven anti-tumor immune response is elicited with the concurrent administration of an aCD25 Treg-depleting antibody. Using IL15-/- and Rag1-/- mice, we demonstrate that response to RT + IL15c + aCD25 is dependent on both IL15 and CTLs. Furthermore, despite an equivalent survival benefit following treatment with RT + IL15c + aCD25 and combination RT + PD1-IL2v, a novel immunocytokine with PD-1 and IL2Rßγ binding domains, CTL immunophenotyping and phospho-proteomic analysis of intracellular metabolites showed significant upregulation of activation and functionality in CD8 T cells treated with RT + PD1-IL2v. Finally, we show the immunostimulatory response to RT + PD1-IL2v is significantly diminished with a concurrent lack of TCF+ CD8 T cell generation in the absence of functional IL15 signaling. Conclusions: Our results are illustrative of a mechanism wherein unimpeded effector T cell activation through IL2Rß signaling and Treg inhibition are necessary in mediating an anti-tumor immune response.

6.
Bioorg Med Chem Lett ; 112: 129945, 2024 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-39222889

RESUMO

Regulatory T (Treg) cells play a central role in immune homeostasis. Forkhead box P3 (Foxp3), a hallmark molecule in Treg cells, is a vital transcription factor for their development and function. Studies have shown that degradation of the Foxp3 could provide therapeutic benefits in achieving effective anti-tumor immunity. In this study, we designed three PROTAC molecules, P60-L1-VHL, P60-L2-VHL, and P60-L3-VHL, based on a 15-mer peptide inhibitor of Foxp3 (P60), and explored their potential in regulating Foxp3 expression and function. Our data show that, among these molecules, P60-L3-VHL can inhibit the expression and nuclear localization of Foxp3 in HEK 293 T and HeLa cells, respectively. Meanwhile, use of proteasome inhibitor in P60-L3-VHL treated cells revealed an increased Foxp3 expression, indicating that P60-L3-VHL mediates the inhibition of Foxp3 through its degradation in the proteasome pathway. We further substantiate that P60-L3-VHL reduces the differentiation and Foxp3 expression in the in-vitro activated Treg cells. Overall, our findings suggest that P60-L3-VHL inhibits the differentiation of Treg cells by degrading the Foxp3, which may have potential implications in cancer immunotherapy.


Assuntos
Fatores de Transcrição Forkhead , Proteólise , Humanos , Fatores de Transcrição Forkhead/metabolismo , Proteólise/efeitos dos fármacos , Células HEK293 , Células HeLa , Linfócitos T Reguladores/efeitos dos fármacos , Relação Estrutura-Atividade , Estrutura Molecular , Descoberta de Drogas , Relação Dose-Resposta a Droga , Complexo de Endopeptidases do Proteassoma/metabolismo , Quimera de Direcionamento de Proteólise
7.
Mol Ther Oncol ; 32(3): 200861, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39328291

RESUMO

High-grade gliomas (HGGs) and glioblastomas (GBMs) are the most aggressive and lethal brain tumors. The current standard of care (SOC) includes gross safe surgical resection followed by chemoradiotherapy. The main chemotherapeutic agents are the DNA-alkylating agent temozolomide (TMZ) and adjuvants. Due to the outdated therapeutic protocols and lack of specific treatments, there is an urgent and rising need to improve our understanding of tumor biology and design more effective therapeutic strategies. In vitro models are essential for investigating glioma biology and testing novel therapeutic approaches. While using commercially available and patient-derived glioma cell lines for in vitro studies is common practice, they exhibit several limitations, including failing to maintain the genetic and phenotypic diversity of primary tumors, undergo genetic drift over time, and often lacking the invasive and stem-like characteristics of patient tumors. These limitations can lead to inconsistent and non-reproducible results, hampering translational research progress. In this study, we established a novel primary murine HGG cell line, isolated from an immunocompetent HGG-bearing RCAS/T-va mouse. We characterized the transcriptome and phenotype to ensure that this cell line resembles the nature of HGGs and retains the ability to reprogram primary murine T lymphocytes.

8.
J Transl Med ; 22(1): 861, 2024 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-39334238

RESUMO

BACKGROUND: Gene methylation and the immune-related tumor microenvironment (TME) are highly correlated in tumor progression and therapeutic efficacy. Although both of them can be used to predict the clinical outcomes of colorectal cancer (CRC) patients, their predictive value is still unsatisfactory. Whether a combination risk model comprising these two prediction parameters performs better predictive effectiveness than independent factor is still unclear. Methylated Septin9 (mSEPT9) is an early diagnosis biomarker of CRC, in this study, we aimed to investigate mSEPT9-related biomarkers of immunosuppressive TME and identify the value of the combination risk model in predicting the clinical outcomes of CRC. METHODS: Immunofluorescence staining was performed to clarify the correlation between intratumoral IL-10+ Treg infiltration and mSEPT9 in peripheral blood. Survival time, response to 5-fluorouracil (5-FU)-based chemotherapy and PD-1 blockade, and the probability of recurrence or metastasis were analyzed in study (197 CRC samples) and validation (195 CRC samples) sets to evaluate the efficacy of combination risk model. Potential mechanisms were explored by mRNA sequencing. RESULTS: Hypermethylated SEPT9 in the peripheral blood of patients with CRC (stage I-III, and stage IV with resectable M1) before radical resection was positively correlated with high intratumoral IL-10+ Treg infiltration. The high-risk model revealed poor overall survival and progression-free survival, inferior therapeutic response to 5-FU-based chemotherapy and PD-1 blockade, and high probability of recurrence or metastasis. The underlying mechanisms may be associated with the increase in mSEPT9 and mediation of IL-10 via methionine metabolic reprogramming-induced infiltration of IL-10+ Tregs in the TME, which promotes tumor progression and resistance to 5-FU-based chemotherapy and PD-1 blockade. CONCLUSIONS: The combination risk model of peripheral mSETP9 and intratumoral IL-10+ Treg infiltration in CRC can effectively predict prognosis, responsiveness to 5-FU-based chemotherapy and PD-1 blockade, and the probability of recurrence or metastasis. Therefore, this model can be used for precision treatment of CRC.


Assuntos
Neoplasias Colorretais , Metilação de DNA , Interleucina-10 , Nomogramas , Septinas , Linfócitos T Reguladores , Humanos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/imunologia , Septinas/genética , Septinas/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Linfócitos T Reguladores/imunologia , Masculino , Feminino , Pessoa de Meia-Idade , Resultado do Tratamento , Microambiente Tumoral/imunologia , Prognóstico , Idoso , Fluoruracila/uso terapêutico
9.
Artigo em Inglês | MEDLINE | ID: mdl-39297473

RESUMO

BACKGROUND: Dendrobine is a bioactive alkaloid isolated from Dendrobium nobile. Studies have evaluated the anti-tumor effect of dendrobine in cancers, including lung cancer. However, the mechanism of dendrobine inhibiting tumors requires further study. METHODS: Bioinformatics was performed to screen the potential targets of dendrobine. The in-tersection of dendrobine and lung cancer targets was performed for KEGG analysis. CCK-8 was used to detect cell viability after dendrobine treatment. A xenograft mouse model was es-tablished to explore the effect of dendrobine on lung cancer. The percentages of PD-L1+, CD4+, CD8+, CD11b+, CD25+FOXP3+ cells, the expression of Ki-67 and caspase-3, the ex-pression of pathway-related proteins, the levels of IL-2, IFN-γ, and TGF-ß, and the changes of indicators of liver and renal function were measured. RESULTS: Dendrobine regulated the PD1/PD-L1 checkpoint signaling pathway and affected the occurrence and development of lung cancer. Dendrobine decreased the cell viability of lung cancer. Dendrobine and anti-PD-L1 decreased tumor growth, increased caspase-3 expression, and reduced Ki-67 expression in tumor tissues. Dendrobine and anti-PD-L1 suppressed pro-tein expression of PD-L1, p-JAK1/JAK1, and p-JAK2/JAK2 in tumor tissues. Greatly, den-drobine and anti-PD-L1 decreased the percentages of PD-L1+, CD11b+, and CD25+FOXP3+ cells, increased the percentages of CD4+ and CD8+cells, and enhanced the levels of IL-2, IFN-γ, and TGF-ß in tumor tissues. Dendrobine demonstrated no hepatorenal toxicity to the tumor mice. The combination of dendrobine and anti-PD-L1 greatly strengthened the effects of dendrobine on tumors. CONCLUSION: Dendrobine inhibited tumor immune escape by suppressing the PD-1/PD-L1 checkpoint pathway, thus restricting tumor growth of lung cancer.

10.
Mol Immunol ; 175: 31-39, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39298996

RESUMO

Peptide-based anticancer vaccines have shown some efficacy in generating cancer-specific immune responses in various cancer studies, but clinical success is limited, one of the reasons is due to its prone degradation and weak immunogenicity. So some tumor epitope peptide vaccines often require coupling or forming fusion proteins with corresponding protein carriers to enhance their stability and immunogenicity. Given the scarcity of validated carriers for clinical trials, there is an urgent requirement for the development of novel protein carrier. Our previous work has demonstrated that VEGF165b mutant could be used as an effective immunization adjunct to enhance anti-tumor immune response. By analyzing and evaluating the gene structure of VEGF, we speculated that mVEGF165b has the potential to be utilized as a tumor peptide vaccine carrier. An mVEGF165b-MUC1 chimeric tumor vaccine was produced by fusing the MUC1 peptide ((MUC1, a T-cell epitope dominant peptide from Mucin1) to the C-terminus of mVEGF165b, expressing the fusing protein in pichia yeast, followed by purification with a HiTrap heparin affinity chromatography column. We found that immunizing mice with mVEGF165b-MUC1 fusion protein induced high-titer antibodies against VEGF in a preventive context, which in turn reduced the proportion of Tregs and further stimulated mice to produce T-cell responses specific to mucin1. The high-titer VEGF antibody stimulated by mVEGF165b also promoted tumor blood vessel maturation and facilitated T-cell infiltration. In conclusion,immunized with mVEGF165b-MUC1 protein are beneficial for eliciting immune responses targeting Mucin1, mVEGF165b have the potential to be utilized as a peptide tumor vaccine carrier.

11.
Biomark Res ; 12(1): 97, 2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-39227959

RESUMO

Immunotherapy has shown promising anti-tumor effects across various tumors, yet it encounters challenges from the inhibitory tumor immune microenvironment (TIME). Infiltrating regulatory T cells (Tregs) are important contributors to immunosuppressive TIME, limiting tumor immunosurveillance and blocking effective anti-tumor immune responses. Although depletion or inhibition of systemic Tregs enhances the anti-tumor immunity, autoimmune sequelae have diminished expectations for the approach. Herein, we summarize emerging strategies, specifically targeting tumor-infiltrating (TI)-Tregs, that elevate the capacity of organisms to resist tumors by reprogramming their phenotype. The regulatory mechanisms of Treg reprogramming are also discussed as well as how this knowledge could be utilized to develop novel and effective cancer immunotherapies.

12.
Front Immunol ; 15: 1426657, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39234236

RESUMO

Our goal is to improve the outcomes of cancer immunotherapy by targeting FOXP3+ T-regulatory (Treg) cells with a next generation of antisense oligonucleotides (ASO), termed FOXP3 AUMsilence ASO. We performed in vitro experiments with human healthy donor PBMC and clinical samples from patients with lung cancer, mesothelioma and melanoma, and tested our approach in vivo using ASO FOXP3 in syngeneic murine cancer models and in humanized mice. ASO FOXP3 had no effects on cell viability or cell division, did not affect expression of other FOXP members, but decreased expression of FOXP3 mRNA in PBMC by 54.9% and in cancer samples by 64.7%, with corresponding 41.0% (PBMC) and 60.0% (cancer) decreases of Treg numbers (all p<0.0001). Hence, intratumoral Treg were more sensitive to the effects of ASO FOXP3 than peripheral blood Tregs. Isolated human Treg, incubated with ASO FOXP3 for 3.5 hours, had significantly impaired suppressive function (66.4%) versus Scramble control. In murine studies, we observed a significant inhibition of tumor growth, while 13.6% (MC38) to 22% (TC1) of tumors were completely resorbed, in conjunction with ~50% decrease of Foxp3 mRNA by qPCR and decreased numbers of intratumoral Tregs. In addition, there were no changes in FOXP3 mRNA expression or in the numbers of Tregs in draining lymph nodes and in spleens of tumor bearing mice, confirming that intratumoral Treg had enhanced sensitivity to ASO FOXP3 in vivo compared to other Treg populations. ASO FOXP3 Treg targeting in vivo and in vitro was accompanied by significant downregulation of multiple exhaustion markers, and by increased expression of perforin and granzyme-B by intratumoral T cells. To conclude, we report that targeting the key Treg transcription factor FOXP3, with ASO FOXP3, has a powerful anti-tumoral effect and enhances T cell response in vitro and in vivo.


Assuntos
Fatores de Transcrição Forkhead , Oligonucleotídeos Antissenso , Linfócitos T Reguladores , Animais , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/genética , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Humanos , Camundongos , Feminino , Neoplasias/imunologia , Neoplasias/terapia , Linhagem Celular Tumoral , Camundongos Endogâmicos C57BL , Imunoterapia/métodos
13.
Artigo em Inglês | MEDLINE | ID: mdl-39284778

RESUMO

BACKGROUND: Idiopathic inflammatory myopathy (IIM) is a systemic autoimmune disease characterized by skeletal muscle involvement. This study aimed to investigate the role of adenosine receptor signalling pathways in the development of experimental autoimmune myositis (EAM). METHODS: An ecto-5'-nucleotidase (CD73) inhibitor, adenosine receptor agonists, a hypoxia-inducible factor-1α (HIF-1α) inhibitor or a vehicle were administered to control and EAM mice. Murine splenic CD4+ or regulatory T cells (Tregs) were isolated using magnetic beads and subsequently stimulated with an adenosine A2B receptor agonist, a HIF-1α inhibitor, or vehicle in vitro. In cross-sectional studies, we collected 64 serum samples (69% female, 49 ± 9 years), 63 peripheral blood samples (70% female, 50 ± 11 years), and 34 skeletal muscle samples (71% female, 63 ± 6 years) from patients with IIM. Additionally, 35 serum samples and 30 peripheral blood samples were obtained from age- and sex-matched healthy controls, and six quadriceps muscle samples were collected from patients with osteoarthritis to serve as the normal group. RESULTS: Patients with IIM exhibited increased CD73 [dermatomyositis (DM), polymyositis (PM): P < 0.01; immune-mediated necrotizing myopathy (IMNM): P < 0.0001] and adenosine deaminase (ADA) expression (DM: P < 0.001; PM, IMNM: P < 0.0001) in the skeletal muscles, and serum ADA levels [56.7 (95% CI: 53.7, 58.7) vs. 198.8 (95% CI: 186.2, 237.3) ng/µL, P < 0.0001]. Intervention with a CD73 inhibitor exacerbated (P = 0.0461), whereas adenosine receptor agonists (A1: P = 0.0009; A2B: P < 0.0001; A3: P = 0.0001) and the HIF-1α inhibitor (P = 0.0044) alleviated skeletal muscle injury in EAM mice. Elevated expression of programmed cell death protein-1 (PD1: P = 0.0023) and T-cell immunoglobulin and mucin-domain containing-3 (TIM3: P < 0.0001) in skeletal muscles of patients with IIM were correlated with creatine kinase levels (PD1, r = 0.7072, P < 0.0001; TIM3, r = 0.4808, P = 0.0046). PD1+CD4+ (r = 0.3243, P = 0.0115) and PD1+CD8+ (r = 0.3959, P = 0.0017) T cells were correlated with Myositis Disease Activity Assessment Visual Analogue Scale scores (muscle) in IIM. The exhausted Tregs were identified in the skeletal muscles of patients with IIM. Activation of the A2B adenosine receptor downregulated HIF-1α (protein or mRNA level, P < 0.01), resulting in decreased T helper cell 17 (Th17) (13.58% vs. 5.43%, P = 0.0201) and phosphorylated-signal transducer and activator of transcription 3 (p-STAT3)+ Th17 (16.32% vs. 6.73%, P = 0.0029), decreased exhausted Tregs (PD1+ Tregs: 53.55% vs. 40.28%, P = 0.0005; TIM3+ Tregs: 3.93% vs. 3.11%, P = 0.0029), and increased Tregs (0.45% vs. 2.89%, P = 0.0006) in EAM mice. CONCLUSIONS: The exhausted T cells may be pathogenic in IIM, and the activation of adenosine A2B receptor signalling pathway can regulate Th17/Treg balance and inhibit Tregs exhaustion, thereby slowing EAM disease progression.

14.
Exp Mol Pathol ; 139: 104924, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39208564

RESUMO

AIMS: Phytocannabinoids and terpenes from Cannabis sativa have demonstrated limited anti-inflammatory and analgesic effects in several inflammatory conditions. In the current study, we test the hypothesis that phytocannabinoids exert immunomodulatory effects in vitro by decreasing inflammatory cytokine expression and activation. KEY METHODS: CD3/CD28 and lipopolysaccharide activated peripheral blood mononuclear cells (PBMCs) from healthy donors (n = 6) were treated with phytocannabinoid compounds and terpenes in vitro. Flow cytometry was used to determine regulatory T cell (Treg) and T helper 17 (Th17) cell responses to treatments. Cell pellets were harvested for qRT-PCR gene expression analysis of cytokines, cell activation markers, and inflammation-related receptors. Cell culture supernatants were analysed by ELISA to quantify IL-6, TNF-α and IL-10 secretion. MAIN FINDINGS: In an initial screen of 20 µM cannabinoids and terpenes which were coded to blind investigators, cannabigerol (GL4a), caryophyllene oxide (GL5a) and gamma-terpinene (GL6a) significantly reduced cytotoxicity and gene expression levels of IL6, IL10, TNF, TRPV1, CNR1, HTR1A, FOXP3, RORC and NFKΒ1. Tetrahydrocannabinol (GL7a) suppression of T cell activation was associated with downregulation of RORC and NFKΒ1 gene expression and reduced IL-6 (p < 0.0001) and IL10 (p < 0.01) secretion. Cannabidiol (GL1b) significantly suppressed activation of Tregs (p < 0.05) and Th17 cells (p < 0.05) in a follow-on in vitro dose-response study. IL-6 (p < 0.01) and IL-10 (p < 0.01) secretion was significantly reduced with 50 µM cannabidiol. SIGNIFICANCE: The study provides the first evidence that cannabidiol and tetrahydrocannabinol suppress extracellular expression of both anti- and pro-inflammatory cytokines in an in vitro PBMC model of inflammation.


Assuntos
Anti-Inflamatórios , Canabinoides , Linfócitos T Reguladores , Terpenos , Células Th17 , Humanos , Células Th17/efeitos dos fármacos , Células Th17/imunologia , Células Th17/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Anti-Inflamatórios/farmacologia , Terpenos/farmacologia , Canabinoides/farmacologia , Citocinas/metabolismo , Citocinas/genética , Inflamação/tratamento farmacológico , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/imunologia , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Cannabis/química , Células Cultivadas , Ativação Linfocitária/efeitos dos fármacos
15.
Front Oncol ; 14: 1373820, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39104717

RESUMO

Objective: To explore the mechanism by which Tregs promote the progression of colorectal cancer by inducing tumor-associated macrophages to polarize into M2 type via ICOS. Methods: Postoperative pathological tissues and clinical pathological data of 268 colorectal cancer patients who underwent initial surgery were collected. Immunohistochemistry (IHC) was used to detect the expression levels of ICOS, CD163 (a marker for M2 macrophages), and Foxp3 (a marker for Tregs) in cancerous, adjacent non-tumorous, and normal tissues. The relationship of ICOS, M2 macrophages, and Tregs in CRC with clinical pathological characteristics and pre-surgical tumor markers (such as CEA and CA199) was explored. Results: The expression levels of M2 macrophages and Tregs increased with tumor progression, while ICOS expression showed a decreasing trend. Compared to adjacent and normal tissues, the expression levels of ICOS, M2 macrophages, and Tregs were higher in CRC tissues. The expression levels of M2 macrophages and Tregs were significantly positively correlated with tumor markers, while ICOS expression was significantly negatively correlated. Conclusion: Tumor-associated m2 macrophages induced by Tregs and ICOS participate in the dynamic balance of the colorectal cancer tumor microenvironment, and their interaction affects colorectal carcinogenesis and progression. High levels of ICOS are associated with better long-term survival rates.

16.
Sci Rep ; 14(1): 19906, 2024 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-39191849

RESUMO

Ibrutinib (IB) is a tyrosine kinase inhibitor (TKI) that has immunomodulatory action and can be used as second-line therapy for steroid-refractory or steroid-resistant chronic Graft versus Host Disease (cGVHD). Mesenchymal stromal cells (MSCs) are distributed throughout the body and their infusion has also been explored as a second-line therapeutic alternative for the treatment of cGVHD. Considering the currently unknown effects of IB on endogenous MSCs, as well as the possible combined use of IB and MSCs for cGVHD, we investigated whether adipose tissue-derived MSCs present IB-targets, as well as the consequences of treating MSCs with this drug, regarding cell viability, proliferation, phenotype, and anti-inflammatory potential. Interestingly, we show for the first time that MSCs express several IB target genes. Also of note, the treatment of such cells with this TKI elevated the levels of CD90 and CD105 surface proteins, as well as VCAM-1. Furthermore, IB-treated MSCs presented increased mRNA expression of the anti-inflammatory genes PD-L1, TSG-6, and IL-10. However, continued exposure to IB, even at low doses, compromised the viability of MSCs. These data indicate that the use of IB can stimulate an anti-inflammatory profile in MSCs, but also that a continued exposure to IB can compromise MSC viability over time.


Assuntos
Adenina , Tecido Adiposo , Proliferação de Células , Sobrevivência Celular , Células-Tronco Mesenquimais , Piperidinas , Pirazóis , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Proliferação de Células/efeitos dos fármacos , Humanos , Piperidinas/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Tecido Adiposo/citologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Pirazóis/farmacologia , Fenótipo , Pirimidinas/farmacologia , Anti-Inflamatórios/farmacologia , Células Cultivadas
17.
Expert Rev Vaccines ; 23(1): 812-829, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39186325

RESUMO

INTRODUCTION: Cancer vaccines stimulate the activation of specific humoral and cellular adaptive responses against cancer cells.Antibodies generated post vaccination can be isolated and further selected to develop highly specific and potent monoclonal antibodies (mAbs) against tumor-associated antigens. AREAS COVERED: This review describes different types of cancer vaccines, the process of the generation of the mAb NEO-201 from the Hollinshead cancer vaccine platform, the characterization of the antigen recognized by NEO-201, the ability of NEO-201 to bind and mediate the killing of cancer cells and immunosuppressive cells (gMDSCs and Tregs) through ADCC and CDC, NEO-201 preclinical and clinical toxicity and efficacy. EXPERT OPINION: To overcome the problem of poor clinical efficacy of cancer vaccines, due to the activity of immunosuppressive cells, cancer vaccines could be combined with other immunotherapeutics able to deplete immunosuppressive cells. Results from clinical trials, employing NEO-201 alone or in combination with pembrolizumab, showed that durable stabilization of disease after treatment was due to the ability of NEO-201 to target and reduce the percentage of circulating Tregs and gMDSCs.These findings provide compelling support to combine NEO-201 with cancer vaccines to reintegrate their ability to elicit a robust and durable immune adaptive response against cancer.


Assuntos
Anticorpos Monoclonais , Antígenos de Neoplasias , Vacinas Anticâncer , Neoplasias , Humanos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Neoplasias/imunologia , Neoplasias/terapia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/farmacologia , Animais , Antígenos de Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Imunoterapia/métodos
18.
Int J Mol Sci ; 25(13)2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-39000453

RESUMO

Regulatory T cells (Tregs) possess unique immunosuppressive activity among CD4-positive T cells. Tregs are ubiquitously present in mammals and function to calm excessive immune responses, thereby suppressing allergies or autoimmune diseases. On the other hand, due to their immunosuppressive function, Tregs are thought to promote cancer progression. The tumor microenvironment (TME) is a multicellular system composed of many cell types, including tumor cells, infiltrating immune cells, and cancer-associated fibroblasts (CAFs). Within this environment, Tregs are recruited by chemokines and metabolic factors and impede effective anti-tumor responses. However, in some cases, their presence can also improve patient's survival rates. Their functional consequences may vary across tumor types, locations, and stages. An in-depth understanding of the precise roles and mechanisms of actions of Treg is crucial for developing effective treatments, emphasizing the need for further investigation and validation. This review aims to provide a comprehensive overview of the complex and multifaceted roles of Tregs within the TME, elucidating cellular communications, signaling pathways, and their impacts on tumor progression and highlighting their potential anti-tumor mechanisms through interactions with functional molecules.


Assuntos
Progressão da Doença , Neoplasias , Linfócitos T Reguladores , Microambiente Tumoral , Humanos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Microambiente Tumoral/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/metabolismo , Animais , Transdução de Sinais , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/imunologia , Fibroblastos Associados a Câncer/patologia
19.
Cancers (Basel) ; 16(13)2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-39001455

RESUMO

T-cell-mediated anti-tumoral responses may have significant clinical relevance as a biomarker for response to immunotherapy. The value of peripheral blood pre-existing tumor antigen-specific T cells (PreI+) as a predictive immunotherapy biomarker in NSCLC patients was investigated, along with the frequency of various circulating immune cells. Fifty-two treatment-naïve, stage III/IV NSCLC patients, treated with front-line immune checkpoint inhibitors (ICI)-containing regimens were enrolled. PreI was calculated as the percentages of CD3+IFNγ+ cells after in vitro co-cultures of PBMCs with peptides against four different Tumor-Associated Antigens (TAA). Immunophenotyping of peripheral blood immune cells was performed using multicolor flow cytometry. PreI+ T cells were detected in 44% of patients. Median overall survival (OS) was significantly higher in PreI+ patients compared to PreI- patients (not reached vs. 321 days, respectively; p = 0.014). PreI+ patients had significantly higher numbers of possible exhausted CD3+CD8+PD-1+ cells and lower percentages of immunosuppressive Tregs compared to PreI- patients. Additionally, patients with PreI+ and low numbers of peripheral blood M-MDSCs had a significant survival advantage compared to the rest of the patients. Thus, combining pre-existing tumor antigen-specific immunity before initiation of ICI in NSCLC patients with selected immune-suppressive cells could identify patients who have a favorable clinical outcome when treated with ICI-containing regimens.

20.
Adv Sci (Weinh) ; 11(36): e2310304, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39072947

RESUMO

Despite the success of immunotherapy in treating hepatocellular carcinoma (HCC), HCC remains a severe threat to health. Here, a crucial transcription factor, SOX12, is revealed that induces the immunosuppression of liver tumor microenvironment. Overexpressing SOX12 in HCC syngeneic models increases intratumoral regulatory T-cell (Treg) infiltration, decreases CD8+T-cell infiltration, and hastens HCC metastasis. Hepatocyte-specific SOX12 knockout attenuates DEN/CCl4-induced HCC progression and metastasis, whereas hepatocyte-specific SOX12 knock-in accelerates these effects. Mechanistically, SOX12 transcriptionally activates C-C motif chemokine ligand 22 (CCL22) expression to promote the recruitment and suppressive activity of Tregs. Moreover, SOX12 transcriptionally upregulates CD274 expression to suppress CD8+T-cell infiltration. Either knockdown of CCL22 or PD-L1 dampens SOX12-mediated HCC metastasis. Blocking of CC chemokine receptor 4 (CCR4), a receptor for CCL22, by inhibitor C-021 or Treg-specific knockout of CCR4 inhibits SOX12-mediated HCC metastasis. Transforming growth factor-ß1 (TGF-ß1)/TGFßR1-Smad2/3/4 is identified as a key upstream signaling for SOX12 overexpression in HCC cells. Combining C-021 or TGFßR1 inhibitor galunisertib with anti-PD-L1 exhibits an enhanced antitumor effect in two HCC models. Collectively, the findings demonstrate that SOX12 contributes to HCC immunosuppression through the CCL22/CCR4-Treg and PD-L1-CD8+T axes. Blocking of CCR4 or TGFßR1 improves the efficacy of anti-PD-L1 in SOX12-mediated HCC.


Assuntos
Carcinoma Hepatocelular , Progressão da Doença , Neoplasias Hepáticas , Fatores de Transcrição SOXC , Linfócitos T Reguladores , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Animais , Camundongos , Linfócitos T Reguladores/imunologia , Fatores de Transcrição SOXC/genética , Fatores de Transcrição SOXC/metabolismo , Modelos Animais de Doenças , Microambiente Tumoral/imunologia , Microambiente Tumoral/genética , Humanos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Masculino , Tolerância Imunológica/genética , Tolerância Imunológica/imunologia , Linhagem Celular Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA