Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.827
Filtrar
2.
Cell Mol Life Sci ; 81(1): 372, 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39196331

RESUMO

Chronic hepatitis B virus (HBV) infection is a global health problem that substantially increases the risk of developing liver disease. The development of a novel strategy to induce anti-HB seroconversion and achieve a long-lasting immune response against chronic HBV infection remains challenging. Here, we found that chronic HBV infection affected the signaling pathway involved in STING-mediated induction of host immune responses in dendritic cells (DCs) and then generated a lymph node-targeted nanovaccine that co-delivered hepatitis B surface antigen (HBsAg) and cyclic diguanylate monophosphate (c-di-GMP) (named the PP-SG nanovaccine). The feasibility and efficiency of the PP-SG nanovaccine for CHB treatment were evaluated in HBV-carrier mice. Serum samples were analyzed for HBsAg, anti-HBs, HBV DNA, and alanine aminotransferase levels, and liver samples were evaluated for HBV DNA and RNA and HBcAg, accompanied by an analysis of HBV-specific cellular and humoral immune responses during PP-SG nanovaccine treatment. The PP-SG nanovaccine increased antigen phagocytosis and DC maturation, efficiently and safely eliminated HBV, achieved a long-lasting immune response against HBV reinjection, and disrupted chronic HBV infection-induced immune tolerance, as characterized by the generation and multifunctionality of HBV-specific CD8+ T and CD4+ T cells and the downregulation of immune checkpoint molecules. HBV-carrier mice immunized with the PP-SG nanovaccine achieved partial anti-HBs seroconversion. The PP-SG nanovaccine can induce sufficient and persistent viral suppression and achieve anti-HBs seroconversion, rendering it a promising vaccine candidate for clinical chronic hepatitis B therapy.


Assuntos
Células Dendríticas , Antígenos de Superfície da Hepatite B , Vírus da Hepatite B , Hepatite B Crônica , Linfonodos , Proteínas de Membrana , Camundongos Endogâmicos C57BL , Animais , Hepatite B Crônica/imunologia , Hepatite B Crônica/tratamento farmacológico , Camundongos , Células Dendríticas/imunologia , Vírus da Hepatite B/imunologia , Antígenos de Superfície da Hepatite B/imunologia , Linfonodos/imunologia , Linfonodos/efeitos dos fármacos , Proteínas de Membrana/imunologia , Vacinas contra Hepatite B/imunologia , GMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , Feminino , Humanos , Nanopartículas/química , Nanovacinas
3.
Cytokine ; 182: 156722, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39116536

RESUMO

We have previously shown the ability of transamidated gluten (spf) to modulate both innate and adaptive intestinal immunity elicited by wheat gliadin in HLA-DQ8 transgenic mice (DQ8 mice), a model of gluten sensitivity. Herein, we evaluated the influence of spf when administered intragastrically on the immune response to native gliadin in DQ8 mice. To address the issue, we analysed three regimens of antigen administration: before immunisation (pre-treatment), during immunisation (co-treatment) and through breast milk during the lactating phase (suckling treatment). Mice were immunised mucosally by intranasal delivery of digested wheat gliadin along with cholera toxin in multiple doses. After sacrifice, isolated spleen and mesenteric lymph node (MLN) cells were challenged in vitro and the cytokine profile of culture supernatants assessed by ELISA and multiparametric assay. We found that only pre-treatment with spf was effective in down-regulating the gliadin-specific IFN-γ response and only in spleen cells. Interestingly, spf pre-treatment also induced systemic IL-6, IL-17A and TNF-α. By contrast, we found that spf pre-treatment upregulated INF-γ in MLN but also significantly decreased IL-2. In conclusion, our data provide evidence that the preventive intragastric administration of transamidated gluten is able to interfere with the classical cytokine profile induced by gliadin via mucosal immunisation in a transgenic model expressing one of the HLA molecules associated with coeliac disease.


Assuntos
Gliadina , Antígenos HLA-DQ , Camundongos Transgênicos , Triticum , Animais , Gliadina/imunologia , Antígenos HLA-DQ/imunologia , Camundongos , Triticum/imunologia , Feminino , Citocinas/metabolismo , Baço/imunologia , Doença Celíaca/imunologia , Humanos , Toxina da Cólera/farmacologia , Toxina da Cólera/imunologia , Toxina da Cólera/administração & dosagem , Interferon gama/metabolismo , Intestinos/imunologia , Linfonodos/imunologia , Linfonodos/efeitos dos fármacos , Imunização/métodos , Glutens/imunologia , Glutens/administração & dosagem , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-17/metabolismo
4.
Bull Exp Biol Med ; 177(2): 271-273, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-39090464

RESUMO

In female Wistar rats with breast cancer, quantitative changes of pro-oncogenic miRNAs (miR-21, -27a, and -221) and tumor-suppressive miR-429 in the mesenteric lymph node were assessed after photodynamic therapy for breast cancer and after photodynamic therapy followed surgical treatment. The level of pro-oncogenic miR-221 in the mesenteric lymph node decreased, and the level of pro-oncogenic miR-21 increased after photodynamic therapy for breast cancer followed by surgical treatment in comparison with the corresponding parameters after photodynamic therapy alone. The content of tumor-suppressive miR-429 remained reduced, as in the group of animals receiving photodynamic therapy alone.


Assuntos
Linfonodos , MicroRNAs , Fotoquimioterapia , Ratos Wistar , Animais , Feminino , MicroRNAs/genética , MicroRNAs/metabolismo , Linfonodos/patologia , Linfonodos/efeitos dos fármacos , Linfonodos/metabolismo , Fotoquimioterapia/métodos , Ratos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Mesentério/patologia , Fármacos Fotossensibilizantes/uso terapêutico , Fármacos Fotossensibilizantes/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias da Mama/patologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Metástase Linfática
5.
Int J Mol Sci ; 25(16)2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39201318

RESUMO

The skin is a direct target of the air pollutant benzo[a]pyrene (BaP). While its carcinogenic qualities are well-studied, the immunotoxicity of BaP after dermal exposure is less understood. This study examines the immunomodulatory effects of a 10-day epicutaneous BaP application, in environmentally/occupationally relevant doses, by analyzing ex vivo skin immune response (skin explant, epidermal cells and draining lymph node/DLN cell activity), alongside the skin's reaction to sensitization with experimental hapten dinitrochlorobenzene (DNCB). The results show that BaP application disrupts the structure of the epidermal layer and promotes immune cell infiltration in the dermis. BaP exposure led to oxidative stress in epidermal cells, characterized by decreased reduced glutathione and increased AHR and Cyp1A1 expression. Production and gene expression of proinflammatory cytokines (TNF, IL-1ß) by epidermal cells decreased, while IL-10 response increased. Decreased spontaneous production of IFN-γ and IL-17, along with unchanged IL-10, was observed in DLC cells, whereas ConA-stimulated production of these cytokines was elevated. Local immunosuppression caused by BaP application seems to reduce the skin's response to an additional stimulus, evidenced by decreased effector activity of DLN cells three days after sensitization with DNCB. These findings provide new insight into the immunomodulatory effects and health risks associated with skin exposure to BaP.


Assuntos
Benzo(a)pireno , Citocinas , Linfonodos , Benzo(a)pireno/toxicidade , Animais , Ratos , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/metabolismo , Citocinas/metabolismo , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/imunologia , Células Epidérmicas/efeitos dos fármacos , Células Epidérmicas/metabolismo , Epiderme/efeitos dos fármacos , Epiderme/metabolismo , Epiderme/imunologia , Estresse Oxidativo/efeitos dos fármacos , Dinitroclorobenzeno , Masculino , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A1/genética
6.
J Control Release ; 372: 829-845, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38964471

RESUMO

Cancer metastasis and recurrence are obstacles to successful treatment of aggressive cancer. To address this challenge, chemotherapy is indispensable as an essential part of comprehensive cancer treatment, particularly for subsequent therapy after surgical resection. However, small-molecule drugs for chemotherapy always cause inadequate efficacy and severe side effects against cancer metastasis and recurrence caused by lymph node metastases. Here, we developed doxorubicin-carried albumin nanocages (Dox-AlbCages) with appropriate particle sizes and pH/enzyme-responsive drug release for tumor and lymph node dual-targeted therapy by exploiting the inborn transport properties of serum albumin. Inspired by the protein-templated biomineralization and remote loading of doxorubicin into liposomes, we demonstrated the controlled synthesis of Dox-AlbCages via the aggregation or crystallization of doxorubicin and ammonium sulfate within albumin nanocages using a biomineralization strategy. Dox-AlbCages allowed efficient encapsulation of Dox in the core protected by the albumin corona shell, exhibiting favorable properties for enhanced tumor and lymph node accumulation and preferable cellular uptake for tumor-specific chemotherapy. Intriguingly, Dox-AlbCages effectively inhibited tumor growth and metastasis in orthotopic 4T1 breast tumors and prevented postsurgical tumor recurrence and lung metastasis. At the same time, Dox-AlbCages had fewer side effects than free Dox. This nanoplatform provides a facile strategy for designing tumor- and lymph node-targeted nanomedicines for suppressing cancer metastasis and recurrence.


Assuntos
Antibióticos Antineoplásicos , Doxorrubicina , Metástase Linfática , Camundongos Endogâmicos BALB C , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Animais , Feminino , Linhagem Celular Tumoral , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/uso terapêutico , Liberação Controlada de Fármacos , Nanopartículas/administração & dosagem , Nanopartículas/química , Humanos , Camundongos , Linfonodos/patologia , Linfonodos/efeitos dos fármacos , Linfonodos/metabolismo , Sistemas de Liberação de Medicamentos , Albuminas/administração & dosagem , Albuminas/química , Portadores de Fármacos/química
7.
Respir Res ; 25(1): 233, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38840238

RESUMO

BACKGROUND: There is inconclusive evidence to suggest that the expression of programmed cell death ligand 1 (PD-L1) is a putative predictor of response to EGFR-TKI therapy in advanced EGFR-mutant non-small cell lung cancer (NSCLC). We evaluated the heterogeneity in PD-L1 expression in the primary lung site and metastatic lymph nodes to analyze the association between PD-L1 expression and response for patients treated with EGFR-TKI. METHODS: This study reviewed 184 advanced NSCLC patients with EGFR mutations who received first-generation EGFR-TKI as first-line treatment from 2020 to 2021 at Shanghai Chest Hospital. The patients were divided into the primary lung site group (n = 100) and the metastatic lymph nodes group (n = 84) according to the biopsy site. The patients in each group were divided into TPS < 1%, TPS 1-49%, and TPS ≥ 50% groups according to PD-L1 expression. RESULTS: The median PFS was 7 (95% CI: 5.7-8.3) months, and the median OS was 26 (95% CI: 23.5-28.5) months for all patients. No correlation existed between PFS or OS and PD-L1 expression. The median PFS in the primary lung site group was 11 months (95% CI: 9.6-12.4) in the TPS < 1% group, 8 months (95% CI: 6.6-9.4) in TPS 1-49% group, and 4 months (95% CI: 3.2-4.8) in TPS ≥ 50% group, with statistically significant differences (p = 0.000). The median OS of the TPS < 1% group and TPS ≥ 50% group showed a statistically significant difference (p = 0.008) in the primary lung site group. In contrast, PD-L1 expression in the lymph nodes of EGFR-mutant patients was unrelated to PFS or OS after EGFR-TKI therapy. CONCLUSION: PD-L1 expression from the primary lung site might predict clinical benefit from EGFR-TKI, whereas PD-L1 from metastatic lymph nodes did not. TRIAL REGISTRATION: This retrospective study was approved by the Ethics Committee of Shanghai Chest Hospital (ID: IS23060) and performed following the Helsinki Declaration of 1964 (revised 2008).


Assuntos
Antígeno B7-H1 , Carcinoma Pulmonar de Células não Pequenas , Receptores ErbB , Neoplasias Pulmonares , Metástase Linfática , Inibidores de Proteínas Quinases , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Antígeno B7-H1/biossíntese , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Feminino , Masculino , Pessoa de Meia-Idade , Receptores ErbB/biossíntese , Receptores ErbB/genética , Receptores ErbB/metabolismo , Receptores ErbB/antagonistas & inibidores , Idoso , Inibidores de Proteínas Quinases/uso terapêutico , Estudos Retrospectivos , Linfonodos/patologia , Linfonodos/efeitos dos fármacos , Linfonodos/metabolismo , Adulto , Idoso de 80 Anos ou mais , Resultado do Tratamento , Valor Preditivo dos Testes , Mutação , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/análise
8.
Mar Drugs ; 22(6)2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38921545

RESUMO

Deep seawater (DS), obtained from a depth over 200 m, has health benefits due to its rich nutrients and minerals, and intake of DS has shown diverse immunomodulatory effects in allergies and cancer. Therefore, the immunostimulatory effects of Korean mineral-rich seawaters were examined in a cyclophosphamide (CPA)-induced immunosuppression model. Three samples of Korean seawater, namely DS from the East Sea off the coasts of Pohang (PDS) and Uljin (UDS), and seawater from the West Sea off the coast of Boryeong (BS), were collected. The seawaters were abundant in several minerals (calcium, iron, zinc, selenium, etc.). Mice were orally administered the seawaters for 42 days, followed by CPA-induced immunosuppression. The CPA induction reduced the weight of the spleen and lymph nodes; however, the administration of seawaters increased the weight of the lymphoid organs, accompanied by stimulation of natural killer cells' activity and NF-kB-mediated cytokine production (IFNγ, TNFα, IL1ß, IL6, and IL12). The mouse-derived splenocytes showed lymphoproliferation without cytotoxicity in the seawater groups. Histopathological analysis revealed that the seawaters improved the CPA-induced atrophic changes by promoting lymphoproliferation in the spleen and lymph nodes. These results provide useful information for the use of Korean mineral-rich seawaters, particularly PDS and UDS, as alternative immunostimulants under immunosuppressive conditions.


Assuntos
Ciclofosfamida , Água do Mar , Animais , Ciclofosfamida/farmacologia , Camundongos , Minerais/farmacologia , Citocinas/metabolismo , República da Coreia , Terapia de Imunossupressão , Baço/efeitos dos fármacos , Baço/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Masculino , Adjuvantes Imunológicos/farmacologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Imunossupressores/farmacologia , Camundongos Endogâmicos BALB C
9.
J Mater Chem B ; 12(24): 5848-5860, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38775048

RESUMO

Nanoparticles have been regarded as a promising vaccine adjuvant due to their innate immune potentiation and enhanced antigen transport. However, the inefficient infiltration into the lymph node (LN) paracortex of nanoparticles caused by subcapsular sinus (SCS) obstruction is the main challenge in further improvement of nanovaccine immune efficacy. Herein, we propose to overcome paracortex penetration by using nanovaccine to spontaneously and continuously release antigens after retention in the SCS. In detail, we utilized a spontaneous retro-Diels-Alder (r-D-A) reaction linker to connect poly{(2-methyl-2-oxazoline)80-co-[(2-butyl-2-oxazoline)15-r-(2-thioethyl-2-oxazoline)8]} (PMBOxSH) and peptides for the peptide nanovaccine construction. The r-D-A reaction linker can spontaneously break over time, allowing the nanovaccine to release free antigens and adjuvants upon reaching the LN, thereby facilitating the entry of released antigens and adjuvants into the interior of the LNs. We showed that the efficacy of the peptide nanovaccine constructed using this dynamic linker could be significantly improved, thus greatly enhancing the tumor inhibition efficacy in the B16-OVA model. This dynamic-covalent-chemistry-based vaccine strategy may inspire designing more efficient therapeutic vaccines, especially those that require eliciting high-amount T cell responses.


Assuntos
Imunidade Celular , Linfonodos , Nanopartículas , Peptídeos , Animais , Camundongos , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Nanopartículas/química , Peptídeos/química , Peptídeos/farmacologia , Imunidade Celular/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Reação de Cicloadição , Feminino , Tamanho da Partícula , Nanovacinas
10.
Front Immunol ; 15: 1391949, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38765015

RESUMO

Dimethyl fumarate (DMF, Tecfidera) is an oral drug utilized to treat relapsing-remitting multiple sclerosis (MS). DMF treatment reduces disease activity in MS. Gastrointestinal discomfort is a common adverse effect of the treatment with DMF. This study aimed to investigate the effect of DMF administration in the gut draining lymph nodes cells of C57BL6/J female mice with experimental autoimmune encephalomyelitis (EAE), an animal model of MS. We have demonstrated that the treatment with DMF (7.5 mg/kg) significantly reduces the severity of EAE. This reduction of the severity is accompanied by the increase of both proinflammatory and anti-inflammatory mechanisms at the beginning of the treatment. As the treatment progressed, we observed an increasing number of regulatory Foxp3 negative CD4 T cells (Tr1), and anti-inflammatory cytokines such as IL-27, as well as the reduction of PGE2 level in the mesenteric lymph nodes of mice with EAE. We provide evidence that DMF induces a gradual anti-inflammatory response in the gut draining lymph nodes, which might contribute to the reduction of both intestinal discomfort and the inflammatory response of EAE. These findings indicate that the gut is the first microenvironment of action of DMF, which may contribute to its effects of reducing disease severity in MS patients.


Assuntos
Fumarato de Dimetilo , Encefalomielite Autoimune Experimental , Linfonodos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores , Animais , Fumarato de Dimetilo/farmacologia , Fumarato de Dimetilo/uso terapêutico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Linfonodos/imunologia , Linfonodos/efeitos dos fármacos , Camundongos , Feminino , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Mesentério , Citocinas/metabolismo , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Modelos Animais de Doenças
11.
Acta Biomater ; 180: 423-435, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38641183

RESUMO

Communication between tumors and lymph nodes carries substantial significance for antitumor immunotherapy. Remodeling the immune microenvironment of tumor-draining lymph nodes (TdLN) plays a key role in enhancing the anti-tumor ability of immunotherapy. In this study, we constructed a biomimetic artificial lymph node structure composed of F127 hydrogel loading effector memory T (TEM) cells and PD-1 inhibitors (aPD-1). The biomimetic lymph nodes facilitate the delivery of TEM cells and aPD-1 to the TdLN and the tumor immune microenvironment, thus realizing effective and sustained anti-tumor immunotherapy. Exploiting their unique gel-forming and degradation properties, the cold tumors were speedily transformed into hot tumors via TEM cell supplementation. Meanwhile, the efficacy of aPD-1 was markedly elevated compared with conventional drug delivery methods. Our finding suggested that the development of F127@TEM@aPD-1 holds promising potential as a future novel clinical drug delivery technique. STATEMENT OF SIGNIFICANCE: F127@TEM@aPD-1 show unique advantages in cancer treatment. When injected subcutaneously, F127@TEM@aPD-1 can continuously supplement TEM cells and aPD-1 to tumor draining lymph nodes (TdLN) and the tumor microenvironment, not only improving the efficacy of ICB therapy through slow release, but also exhibiting dual regulatory effects on the tumor and TdLN.


Assuntos
Preparações de Ação Retardada , Hidrogéis , Linfonodos , Células T de Memória , Receptor de Morte Celular Programada 1 , Animais , Hidrogéis/química , Hidrogéis/farmacologia , Linfonodos/efeitos dos fármacos , Linfonodos/patologia , Linfonodos/imunologia , Camundongos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Células T de Memória/efeitos dos fármacos , Células T de Memória/imunologia , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacologia , Preparações de Ação Retardada/farmacocinética , Microambiente Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores de Checkpoint Imunológico/farmacologia , Imunoterapia/métodos , Feminino , Camundongos Endogâmicos C57BL , Humanos
12.
J Occup Health ; 66(1)2024 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-38626325

RESUMO

OBJECTIVES: We aimed to analyze the subchronic toxicity and tissue distribution of indium after the intratracheal administration of indium-tin oxide nanoparticles (ITO NPs) to the lungs of rats. METHODS: Male Wistar rats were administered a single intratracheal dose of 10 or 20 mg In/kg body weight (BW) of ITO NPs. The control rats received only an intratracheal dose of distilled water. A subset of rats was periodically euthanized throughout the study from 1 to 20 weeks after administration. Indium concentrations in the serum, lungs, mediastinal lymph nodes, kidneys, liver, and spleen as well as pathological changes in the lungs and kidneys were determined. Additionally, the distribution of ionic indium and indium NPs in the kidneys was analyzed using laser ablation-inductively coupled plasma mass spectrometry. RESULTS: Indium concentrations in the lungs of the 2 ITO NP groups gradually decreased over the 20-week observation period. Conversely, the indium concentrations in the mediastinal lymph nodes of the 2 ITO groups increased and were several hundred times higher than those in the kidneys, spleen, and liver. Pulmonary and renal toxicities were observed histopathologically in both the ITO groups. Both indium NPs and ionic indium were detected in the kidneys, and their distributions were similar to the strong indium signals detected at the sites of inflammatory cell infiltration and tubular epithelial cells. CONCLUSIONS: Our results demonstrate that intratracheal administration of 10 or 20 mg In/kg BW of ITO NPs in male rats produces pulmonary and renal toxicities.


Assuntos
Índio , Rim , Pulmão , Ratos Wistar , Compostos de Estanho , Animais , Masculino , Compostos de Estanho/toxicidade , Compostos de Estanho/administração & dosagem , Pulmão/efeitos dos fármacos , Pulmão/patologia , Ratos , Rim/efeitos dos fármacos , Rim/patologia , Índio/toxicidade , Índio/administração & dosagem , Índio/farmacocinética , Distribuição Tecidual , Testes de Toxicidade Subcrônica , Nanopartículas Metálicas/toxicidade , Nanopartículas Metálicas/administração & dosagem , Nanopartículas/toxicidade , Linfonodos/efeitos dos fármacos
13.
Cell Rep ; 43(5): 114153, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38687643

RESUMO

Gut-draining mesenteric and celiac lymph nodes (mLNs and celLNs) critically contribute to peripheral tolerance toward food and microbial antigens by supporting the de novo induction of regulatory T cells (Tregs). These tolerogenic properties of mLNs and celLNs are stably imprinted within stromal cells (SCs) by microbial signals and vitamin A (VA), respectively. Here, we report that a single, transient gastrointestinal infection in the neonatal, but not adult, period durably abrogates the efficient Treg-inducing capacity of celLNs by altering the subset composition and gene expression profile of celLNSCs. These cells carry information about the early-life pathogen encounter until adulthood and durably instruct migratory dendritic cells entering the celLN with reduced tolerogenic properties. Mechanistically, transiently reduced VA levels cause long-lasting celLN functional impairment, which can be rescued by early-life treatment with VA. Together, our data highlight the therapeutic potential of VA to prevent sequelae post gastrointestinal infections in infants.


Assuntos
Linfonodos , Linfócitos T Reguladores , Vitamina A , Animais , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/efeitos dos fármacos , Vitamina A/farmacologia , Vitamina A/uso terapêutico , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Camundongos , Animais Recém-Nascidos , Tolerância Imunológica/efeitos dos fármacos , Células Dendríticas/imunologia , Camundongos Endogâmicos C57BL , Feminino
14.
J Neuroinflammation ; 21(1): 112, 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38684986

RESUMO

BACKGROUND: Dimethyl fumarate (DMF) is a fumaric acid ester that exhibits immunoregulatory and anti-inflammatory properties. However, the function of DMF in autoimmune uveitis (AU) is incompletely understood, and studies comprehensively exploring the impact of DMF on immune cells are still lacking. METHODS: To explore the function of DMF in uveitis and its underlying mechanisms, we conducted single-cell RNA sequencing (scRNA-seq) on the cervical draining lymph node (CDLN) cells of normal, experimental autoimmune uveitis (EAU), and DMF-treated EAU mice. Additionally, we integrated scRNA-seq data of the retina and CDLNs to identify the potential impact of DMF on ocular immune cell infiltration. Flow cytometry was conducted to verify the potential target molecules of DMF. RESULTS: Our study showed that DMF treatment effectively ameliorated EAU symptoms. The proportional and transcriptional alterations in each immune cell type during EAU were reversed by DMF treatment. Bioinformatics analysis in our study indicated that the enhanced expression of Pim1 and Cxcr4 in EAU was reversed by DMF treatment. Further experiments demonstrated that DMF restored the balance between effector T (Teff) /regulatory T (Treg) cells through inhibiting the pathway of PIM1-protein kinase B (AKT)-Forkhead box O1 (FOXO1). By incorporating the scRNA-seq data of the retina from EAU mice into analysis, our study identified that T cells highly expressing Pim1 and Cxcr4 were enriched in the retina. DMF repressed the ocular infiltration of Teff cells, and this effect might depend on its inhibition of PIM1 and CXCR4 expression. Additionally, our study indicated that DMF might reduce the proportion of plasma cells by inhibiting PIM1 expression in B cells. CONCLUSIONS: DMF effectively attenuated EAU symptoms. During EAU, DMF reversed the Teff/Treg cell imbalance and suppressed the ocular infiltration of Teff cells by inhibiting PIM1 and CXCR4 expression. Thus, DMF may act as a new drug option for the treatment of AU.


Assuntos
Anti-Inflamatórios não Esteroides , Doenças Autoimunes , Fumarato de Dimetilo , Imunossupressores , Retina , Uveíte , Fumarato de Dimetilo/administração & dosagem , Fumarato de Dimetilo/farmacologia , Uveíte/genética , Uveíte/imunologia , Uveíte/terapia , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/terapia , Análise da Expressão Gênica de Célula Única , Modelos Animais de Doenças , Animais , Camundongos , Feminino , Camundongos Endogâmicos C57BL , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/genética , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Transcrição Gênica , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Atlas como Assunto , Imunossupressores/administração & dosagem , Imunossupressores/farmacologia , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacologia , Retina/efeitos dos fármacos , Retina/imunologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia
15.
Int J Pharm ; 656: 124074, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38565406

RESUMO

Tacrolimus (FK506) is an effective therapeutic for transplant rejection in clinical practice, primarily inhibiting rejection by suppressing the activation and proliferation of allogeneic T cells in the lymph nodes (LNs). However, conventional administration methods face challenges in directly delivering free FK506 to the LNs. In this study, we introduce a novel LN-targeted delivery system based on mesoporous silica nanoparticles (MSNs-FK506-MECA79). These particles were designed to selectively target high endothelial venules in LNs; this was achieved through surface modification with MECA79 antibodies. Their mean size and zeta potential were 201.18 ± 5.98 nm and - 16.12 ± 0.36 mV, respectively. Our findings showed that MSNs-FK506-MECA79 could accumulate in LNs and increase the local concentration of FK506 from 28.02 ± 7.71 ng/g to 123.81 ± 76.76 ng/g compared with the free FK506 treatment group. Subsequently, the therapeutic efficacy of MSNs-FK506-MECA79 was evaluated in a skin transplantation model. The treatment with MSNs-FK506-MECA79 could lead to a decrease in the infiltration of T cells in the grafts, a reduction in the grade of rejection, and a significant prolongation of survival. Consequently, this study presents a promising strategy for the active LN-targeted delivery of FK506 and improving the immunotherapeutic effects on transplant rejection.


Assuntos
Rejeição de Enxerto , Imunossupressores , Linfonodos , Nanopartículas , Dióxido de Silício , Tacrolimo , Tacrolimo/administração & dosagem , Tacrolimo/química , Dióxido de Silício/química , Rejeição de Enxerto/prevenção & controle , Rejeição de Enxerto/imunologia , Animais , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Imunossupressores/administração & dosagem , Imunossupressores/química , Imunossupressores/farmacologia , Porosidade , Camundongos Endogâmicos BALB C , Transplante de Pele/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistemas de Liberação de Medicamentos/métodos , Portadores de Fármacos/química
16.
ACS Nano ; 18(17): 11070-11083, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38639726

RESUMO

Effective antitumor immunotherapy depends on evoking a cascade of cancer-immune cycles with lymph nodes (LNs) as the initial sites for activating antitumor immunity, making drug administration through the lymphatic system highly attractive. Here, we describe a nanomedicine with dual responsiveness to pH and enzyme for a programmed activation of antitumor immune through the lymphatic system. The proposed nanomedicine can release the STING agonist diABZI-C2-NH2 in the LNs' acidic environment to activate dendritic cells (DCs) and T cells. Then, the remaining nanomedicine hitchhikes on the activated T cells (PD-1+ T cells) through binding to PD-1, resulting in an effective delivery into tumor tissues owing to the tumor-homing capacity of PD-1+ T cells. The enzyme matrix metalloproteinase-2 (MMP-2) being enriched in tumor tissue triggers the release of PD-1 antibody (aPD-1) which exerts immune checkpoint blockade (ICB) therapy. Eventually, the nanomedicine delivers a DNA methylation inhibitor GSK-3484862 (GSK) into tumor cells, and then the latter combines with granzyme B (GZMB) to trigger tumor cell pyroptosis. Consequently, the pyroptotic tumor cells induce robust immunogenic cell death (ICD) enhancing the DCs maturation and initiating the cascading antitumor immune response. Study on a 4T1 breast tumor mouse model demonstrates the prominent antitumor therapeutic outcome of this nanomedicine through creating a positive feedback loop of cancer-immunity cycles including immune activation in LNs, T cell-mediated drug delivery, ICB therapy, and tumor cell pyroptosis-featured ICD.


Assuntos
Nanomedicina , Animais , Camundongos , Humanos , Imunoterapia , Feminino , Linfonodos/imunologia , Linfonodos/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Antineoplásicos/farmacologia , Antineoplásicos/química , Linhagem Celular Tumoral
17.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-35163272

RESUMO

Polypod-like structured nucleic acids (polypodnas), which are nanostructured DNAs, are useful for delivering cytosine-phosphate guanine oligodeoxynucleotides (CpG ODNs) to antigen-presenting cells (APCs) expressing Toll-like receptor 9 (TLR9) for immune stimulation. Lipid modification is another approach to deliver ODNs to lymph nodes, where TLR9-positive APCs are abundant, by binding to serum albumin. The combination of these two methods can be useful for delivering CpG ODNs to lymph nodes in vivo. In the present study, CpG1668, a phosphodiester-type CpG ODN, was modified with stearic acid (SA) to obtain SA-CpG1668. Tripodna, a polypodna with three pods, was selected as the nanostructured DNA. Tripodnas loaded with CpG1668 or SA-CpG1668 were obtained in high yields. SA-CpG1668/tripodna bound more efficiently to plasma proteins than CpG1668/tripodna and was more efficiently taken up by macrophage-like RAW264.7 cells than CpG1668/tripodna, whereas the levels of tumor necrosis factor-α released from the cells were comparable between the two. After subcutaneous injection into mice, SA-CpG1668/tripodna induced significantly higher interleukin (IL)-12 p40 production in the draining lymph nodes than SA-CpG1668 or CpG1668/tripodna, with reduced IL-6 levels in plasma. These results indicate that the combination of SA modification and nanostructurization is a useful approach for the targeted delivery of CpG ODNs to lymph nodes.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Nanoestruturas/química , Oligodesoxirribonucleotídeos/farmacologia , Adjuvantes Imunológicos/farmacologia , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , DNA/imunologia , Sistemas de Liberação de Medicamentos/métodos , Feminino , Imunização/métodos , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nanoestruturas/uso terapêutico , Conformação de Ácido Nucleico/efeitos dos fármacos , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/metabolismo , Estudo de Prova de Conceito , Células RAW 264.7 , Ácidos Esteáricos/química
18.
Nat Commun ; 13(1): 110, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013252

RESUMO

Microbe-based cancer immunotherapy has recently emerged as a hot topic for cancer treatment. However, serious limitations remain including infection associated side-effect and unsatisfactory outcomes in clinic trials. Here, we fabricate different sizes of nano-formulations derived from yeast cell wall (YCW NPs) by differential centrifugation. The induction of anticancer immunity of our formulations appears to inversely correlate with their size due to the ability to accumulate in tumor-draining lymph node (TDLN). Moreover, we use a percolation model to explain their distribution behavior toward TDLN. The abundance and functional orientation of each effector component are significantly improved not only in the microenvironment in tumor but also in the TDLN following small size YCW NPs treatment. In combination with programmed death-ligand 1 (PD-L1) blockade, we demonstrate anticancer efficiency in melanoma-challenged mice. We delineate potential strategy to target immunosuppressive microenvironment by microbe-based nanoparticles and highlight the role of size effect in microbe-based immune therapeutics.


Assuntos
Imunoterapia/métodos , Linfonodos/efeitos dos fármacos , Melanoma Experimental/terapia , Nanopartículas/administração & dosagem , Saccharomyces cerevisiae/química , Neoplasias Cutâneas/terapia , Aloenxertos , Animais , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Linhagem Celular Tumoral , Parede Celular/química , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica , Injeções Intralesionais , Linfonodos/imunologia , Linfonodos/patologia , Ativação de Macrófagos/efeitos dos fármacos , Melanoma Experimental/genética , Melanoma Experimental/mortalidade , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/química , Tamanho da Partícula , Células RAW 264.7/efeitos dos fármacos , Células RAW 264.7/imunologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/mortalidade , Neoplasias Cutâneas/patologia , Análise de Sobrevida , Quinase Syk/antagonistas & inibidores , Quinase Syk/genética , Quinase Syk/imunologia , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/imunologia , Carga Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
19.
Pharmacol Res ; 176: 106082, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35032662

RESUMO

Patchouli Essential Oil (PEO) has been used as a scent for various healing purposes since the ancient Egyptian period. The primary source of the oil is Pogostemon cablin (PC), a medicinal plant for treating gastrointestinal symptoms. However, the pharmacological function has not been addressed. Here, we report the cancer prevention and gut microbiota (GM) modulating property of PEO and its derivatives patchouli alcohol (PA) and pogostone (PO) in the ApcMin /+ colorectal cancer mice model. We found that PEO, PA, and PO significantly reduced the tumor burden. At the same time, it strengthened the epithelial barrier, evidenced by substantially increasing the number of the goblet and Paneth cells and upregulation of tight junction and adhesion molecules. In addition, PEO, PA, and PO shifted M1 to M2 macrophage phenotypes and remodeled the inflammatory milieu of ApcMin /+ mice. We also found suppression of CD4+CD25+ and stimulation CD4+ CD8+ cells in the spleen, blood, mesenteric lymph nodes (MLNs), and Peyer's patches (PPs) of the treated mice. The composition of the gut microbiome of the drug-treated mice was distinct from the control mice. The drugs stimulated the short-chain fatty acids (SCFAs)-producers and the key SCFA-sensing receptors (GPR41, GPR43, and GPR109a). The activation of SCFAs/GPSs also triggered the alterations of PPAR-γ, PYY, and HSDCs signaling mediators in the treated mice. Our work showed that PEO and its derivatives exert potent anti-cancer effects by modulating gut microbiota and improving the intestinal microenvironment of the ApcMmin /+ mice.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Óleos Voláteis/uso terapêutico , Pogostemon , Animais , Antineoplásicos Fitogênicos/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/microbiologia , Modelos Animais de Doenças , Microbioma Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Linfonodos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Óleos Voláteis/farmacologia , Nódulos Linfáticos Agregados/efeitos dos fármacos , Baço/efeitos dos fármacos
20.
Indian J Pathol Microbiol ; 65(1): 49-54, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35074965

RESUMO

INTRODUCTION: Colorectal cancer is one of the most common malignant tumors and has a relatively poor prognosis. Lymph node involvement is considered the most important prognostic factor. MATERIALS AND METHODS: During a retrospective cohort study, 132 patients with locally advanced rectal cancer who underwent neoadjuvant chemoradiotherapy followed by surgery for resectable rectal cancer from 2010 to 2015 in Sina hospital were reviewed. RESULTS: Multivariable analysis was performed and shown the clinical stage was not a representative factor for disease-free survival (P = 0.187), but Dworak Tumor Regression Grading were significantly associated with higher disease-free survival (P = 0.000) in stage II and stage III. The total number of retrieved lymph nodes and involved lymph nodes in the same clinical stage were statistically associated with higher mean disease-free survival in patients (P = 0.000 in both conditions). CONCLUSION: In the same clinical stage, increasing the Dworak Tumor Regression Grading reduced the risk of rectal cancer recurrence. Increasing total number of retrieved lymph nodes and involved lymph nodes, 2.14 times and 3.87 times increased the risk of recurrence, respectively.


Assuntos
Adenocarcinoma/patologia , Linfonodos/patologia , Terapia Neoadjuvante/normas , Recidiva Local de Neoplasia , Neoplasias Retais/patologia , Adenocarcinoma/classificação , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Intervalo Livre de Doença , Tratamento Farmacológico/normas , Feminino , Humanos , Linfonodos/efeitos dos fármacos , Metástase Linfática/patologia , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante/métodos , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/patologia , Radioterapia/normas , Neoplasias Retais/classificação , Neoplasias Retais/tratamento farmacológico , Neoplasias Retais/terapia , Reto/patologia , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...