Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Front Immunol ; 12: 674048, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054859

RESUMEN

Dendritic cells (DCs) are paramount in initiating and guiding immunity towards a state of activation or tolerance. This bidirectional capacity of DCs sets them at the center stage for treatment of cancer and autoimmune or allergic conditions. Accordingly, many clinical studies use ex vivo DC vaccination as a strategy to boost anti-tumor immunity or to suppress immunity by including vitamin D3, NF-κB inhibitors or retinoic acid to create tolerogenic DCs. As harvesting DCs from patients and differentiating these cells in vitro is a costly and cumbersome process, in vivo targeting of DCs has huge potential as nanoparticulate platforms equipped with activating or tolerogenic adjuvants can modulate DCs in their natural environment. There is a rapid expansion of the choices of nanoparticles and activation- or tolerance-promoting adjuvants for a therapeutic vaccine platform. In this review we highlight the most recent nanomedical approaches aimed at inducing immune activation or tolerance via targeting DCs, together with novel fundamental insights into the mechanisms inherent to fostering anti-tumor or tolerogenic immunity.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Inmunológica/inmunología , Liposomas , Nanopartículas , Vacunas , Sistemas de Liberación de Medicamentos/métodos , Humanos , Liposomas/química , Liposomas/inmunología , Nanopartículas/química , Vacunas/química , Vacunas/inmunología
2.
Cancer Immunol Immunother ; 70(12): 3435-3449, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33877384

RESUMEN

Specific extracts of selected vegetables (SV) have been shown to benefit the survival of stage IIIb/IV non-small cell lung cancer patients in phase I/II studies and is currently in a phase III trial. However, the underlying mechanism of SV-mediated antitumor immune responses has not been elucidated. Our results indicate that SV modulated the NK and adoptive T cell immune responses in antitumor efficacy. Furthermore, antitumor effects of SV were also mediated by innate myeloid cell function, which requires both TLR and ß-glucan signaling in a MyD88/TRIF and Dectin-1-dependent manner, respectively. Additionally, SV treatment reduced granulocytic myeloid-derived suppressor cell (MDSC) infiltration into the tumor and limited monocytic MDSC toward the M2-like functional phenotype. Importantly, SV treatment enhanced antigen-specific immune responses by augmenting the activation of antigen-specific TH1/TH17 cells in secondary lymphoid organs and proliferative response, as well as by reducing the Treg population in the tumor microenvironment, which was driven by SV-primed activated M-MDSC. Our results support the idea that SV can subvert immune-tolerance state in the tumor microenvironment and inhibit tumor growth. The present study suggests that features, such as easy accessibility, favorable clinical efficacy, no detectable side effects and satisfactory safety make SV a feasible, appealing and convincing adjuvant therapy for the treatment of cancer patients and prevent tumor recurrence and/or metastases.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Nutrientes/inmunología , Extractos Vegetales/inmunología , Microambiente Tumoral/inmunología , Animales , Suplementos Dietéticos , Modelos Animales de Enfermedad , Tolerancia Inmunológica/inmunología , Inmunidad/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Monocitos/inmunología , Células Mieloides/inmunología , Células Supresoras de Origen Mieloide/inmunología , Recurrencia Local de Neoplasia/inmunología , Células TH1/inmunología , Células Th17/inmunología
3.
Int J Mol Sci ; 22(5)2021 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-33806610

RESUMEN

Microglia, the innate immune cells of the CNS, exhibit long-term response changes indicative of innate immune memory (IIM). Our previous studies revealed IIM patterns of microglia with opposing immune phenotypes: trained immunity after a low dose and immune tolerance after a high dose challenge with pathogen-associated molecular patterns (PAMP). Compelling evidence shows that innate immune cells adopt features of IIM via immunometabolic control. However, immunometabolic reprogramming involved in the regulation of IIM in microglia has not been fully addressed. Here, we evaluated the impact of dose-dependent microglial priming with ultra-low (ULP, 1 fg/mL) and high (HP, 100 ng/mL) lipopolysaccharide (LPS) doses on immunometabolic rewiring. Furthermore, we addressed the role of PI3Kγ on immunometabolic control using naïve primary microglia derived from newborn wild-type mice, PI3Kγ-deficient mice and mice carrying a targeted mutation causing loss of lipid kinase activity. We found that ULP-induced IIM triggered an enhancement of oxygen consumption and ATP production. In contrast, HP was followed by suppressed oxygen consumption and glycolytic activity indicative of immune tolerance. PI3Kγ inhibited glycolysis due to modulation of cAMP-dependent pathways. However, no impact of specific PI3Kγ signaling on immunometabolic rewiring due to dose-dependent LPS priming was detected. In conclusion, immunometabolic reprogramming of microglia is involved in IIM in a dose-dependent manner via the glycolytic pathway, oxygen consumption and ATP production: ULP (ultra-low-dose priming) increases it, while HP reduces it.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ib/inmunología , Inmunidad Innata/inmunología , Memoria Inmunológica/inmunología , Adenosina Trifosfato/inmunología , Animales , Glucólisis/inmunología , Tolerancia Inmunológica/inmunología , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/inmunología , Consumo de Oxígeno/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Transducción de Señal/inmunología
4.
Eur J Drug Metab Pharmacokinet ; 46(1): 93-104, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32894450

RESUMEN

BACKGROUND AND OBJECTIVE: Ginseng is usually consumed as a dietary supplement for health care in the normal state or prescribed as a herbal medicine in pathologic conditions. Although metabolic studies of ginseng are commonly performed on healthy organisms, the metabolic characteristics in pathologic organisms remain unexplored. This study aimed to uncover the difference in intestinal metabolism of ginseng between normal and cyclophosphamide-induced immunosuppressed rats and further discuss the potential mechanisms involved. METHODS: Twelve Sprague-Dawley rats (6-8 weeks old) were randomly divided into two groups: the normal group (NG) and immunosuppressed group (ISG). Rats in the NG and ISG groups were intraperitoneally administered normal saline and cyclophosphamide injections (40 mg/kg) on the 1st, 2nd, 3rd and 10th days; on the 12th day, all rats were intragastrically administered ginseng water extract (900 mg/kg). The difference in intestinal metabolism of ginseng was compared using an ultra-high-performance liquid chromatography coupled with quadruple time-of-flight mass spectrometry-based metabolomics approach, and the diversities of gut microbiota were analyzed by 16S rRNA gene sequencing between the two groups. RESULTS: The intestinal metabolomic characteristics of ginseng were significantly different between the normal and immunosuppressed rats, with the ginsenoside F2 (F2), 20S-ginsenoside Rg3 (20(S)-Rg3), pseudo-ginsenoside Rt5 (Pseudo-Rt5), ginsenoside Rd (Rd), ginsenoside Rh1 (Rh1), 20S-ginsenoside Rg1 (20(S)-Rg1), ginsenoside compound K (CK), ginsenoside Rg2 (Rg2) and 20S-panaxatriol (S-PPT) more abundant in immunosuppressed ones (P < 0.05). Additionally, the composition of gut microbiota was remarkably altered in the two groups, with some specific bacterial communities such as Bacteroides spp., Eubacterium spp. and Lachnospiraceae_UCG-010 spp. increased and Bifidobacterium spp. decreased in immunosuppressed rats compared with normal ones. CONCLUSION: The intestinal metabolism of ginseng in immunosuppressed rats was significantly different from that in normal ones, which might be partly attributed to the changes in the intensity of specific gut bacteria. The outcomes of this study could provide scientific data for rationalization of ginseng use as both a dietary supplement and herbal medicine.


Asunto(s)
Tolerancia Inmunológica/efectos de los fármacos , Inmunosupresores/toxicidad , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Panax , Extractos Vegetales/farmacología , Animales , Ciclofosfamida/toxicidad , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Ginsenósidos/aislamiento & purificación , Ginsenósidos/farmacología , Tolerancia Inmunológica/inmunología , Mucosa Intestinal/inmunología , Masculino , Extractos Vegetales/aislamiento & purificación , Ratas , Ratas Sprague-Dawley
5.
Oncol Rep ; 44(5): 1799-1809, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33000284

RESUMEN

Galectin­3 is expressed in various tissues and plays an important role in the tumor microenvironment (TME). Galectin­3 has been found to be overexpressed in a variety of cancers and is associated with tumor progression and metastasis. Over the past decades, emerging evidence has suggested that the TME may induce galectin­3 expression to maintain cellular homeostasis and promote cell survival. Furthermore, galectin­3 regulates immune cell function to promote tumor­driven immunosuppression through several mechanisms. In the TME, intracellular and extracellular galectin­3 has different functions. In addition, it has been reported that galectin­3 is associated with glycolysis and mitochondrial metabolism in tumors, and it is involved in the regulation of relevant signaling pathways, thus promoting cancer cell survival via adapting to the TME. The aim of the present review was to summarize the current knowledge on galectin­3 production and its function in the TME, its effect on TME immunosuppression, its association with tumor metabolism and relevant signaling pathways, and to report common types of cancer in which galectin­3 is highly expressed, in order to ensure a comprehensive understanding of the critical effects of galectin­3 on tumor progression and metastasis.


Asunto(s)
Galectina 3/metabolismo , Tolerancia Inmunológica/inmunología , Neoplasias/inmunología , Microambiente Tumoral/inmunología , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Galectina 3/antagonistas & inhibidores , Glucólisis/efectos de los fármacos , Glucólisis/inmunología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Ratones , Mitocondrias/metabolismo , Metástasis de la Neoplasia/inmunología , Metástasis de la Neoplasia/prevención & control , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Pectinas/farmacología , Pectinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Microambiente Tumoral/efectos de los fármacos
6.
Minerva Pediatr ; 72(5): 408-415, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32686923

RESUMEN

Scientific research, diagnostic tools and clinical experience have shown that children suffering from IgE-mediated fish allergy do not need to follow a strict exclusion diet. In fact, they could tolerate some species of fish, which could be reintroduced in the diet by verifying their tolerance with an oral food challenge in a clinical setting. Consequently, it is possible to look a new insight on diagnosis and management of IgE-mediated fish allergy in children, considering the use of canned tuna in clinical settings. Authors performed a literature search through the Cochrane Library and Medline/PubMed databases. All quantitative and qualitative pediatric studies involving diagnosis and management of IgE-mediated fish allergy and the use of canned tuna in clinical settings were considered. Articles related to allergological and nutritional features of fish, and especially canned tuna, were selected. This research was conducted on May 2020. Canned tuna shows peculiar allergological and nutritional characteristics. Relating to allergy, canning process, characterized by cooking the fish under pressure for a time equal to about 7 hours, can lead a conformational change in parvalbumin, making it less allergenic. In terms of nutrition, canned tuna contains B, D and A vitamins and, above all, omega-3 fatty acids and shows a favourable and significantly sustainable nutritional profile. Lower allergenicity, adequate nutritional value and its rich availability in markets at reasonable costs, could make the use of canned tuna as a solution with an excellent risk/benefit ratio in the field of IgE-mediated fish allergy.


Asunto(s)
Hipersensibilidad a los Alimentos/inmunología , Alimentos en Conserva/efectos adversos , Inmunoglobulina E/inmunología , Atún/inmunología , Animales , Niño , Culinaria/métodos , Alimentos , Humanos , Tolerancia Inmunológica/inmunología , Valor Nutritivo
8.
Nanomedicine (Lond) ; 15(1): 77-92, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31868112

RESUMEN

Aim: We aim to demonstrate that a local nanoparticle-mediated hyperthermia can effectively eliminate tumor-associated Tregs and thereby boost checkpoint blockade-based immunotherapy. Materials & methods: Photothermal therapy (PTT), mediated with systemically administered stealthy iron-oxide nanoparticles, was applied to treat BALB/c mice bearing 4T1 murine breast tumors. Flow cytometry was applied to evaluate both Treg and CD8+ T-cell population. Tumor growth following combination therapy of both PTT and anti-CTLA-4 was further evaluated. Results: Our data reveal that tumor-associated Tregs can be preferentially depleted via iron-oxide nanoparticles-mediated PTT. When combining PTT with anti-CTLA-4 immunotherapy, we demonstrate a significant inhibition of syngeneic 4T1 tumor growth. Conclusion: This study offers a novel strategy to overcome Treg-mediated immunosuppression and thereby to boost cancer immunotherapy.


Asunto(s)
Neoplasias de la Mama/terapia , Antígeno CTLA-4/inmunología , Inmunoterapia , Linfocitos T Reguladores/inmunología , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Linfocitos T CD8-positivos/efectos de los fármacos , Antígeno CTLA-4/antagonistas & inhibidores , Terapia Combinada , Modelos Animales de Enfermedad , Femenino , Compuestos Férricos/química , Compuestos Férricos/farmacología , Humanos , Hipertermia Inducida/métodos , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/inmunología , Ratones , Nanopartículas/química , Fototerapia , Microambiente Tumoral/efectos de los fármacos
9.
Nat Commun ; 10(1): 3031, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31292453

RESUMEN

Maternal immune dysregulation seems to affect fetal or postnatal immune development. Preeclampsia is a pregnancy-associated disorder with an immune basis and is linked to atopic disorders in offspring. Here we show reduction of fetal thymic size, altered thymic architecture and reduced fetal thymic regulatory T (Treg) cell output in preeclamptic pregnancies, which persists up to 4 years of age in human offspring. In germ-free mice, fetal thymic CD4+ T cell and Treg cell development are compromised, but rescued by maternal supplementation with the intestinal bacterial metabolite short chain fatty acid (SCFA) acetate, which induces upregulation of the autoimmune regulator (AIRE), known to contribute to Treg cell generation. In our human cohorts, low maternal serum acetate is associated with subsequent preeclampsia, and correlates with serum acetate in the fetus. These findings suggest a potential role of acetate in the pathogenesis of preeclampsia and immune development in offspring.


Asunto(s)
Acetatos/sangre , Feto/inmunología , Preeclampsia/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Linfocitos T Reguladores/inmunología , Acetatos/administración & dosificación , Acetatos/inmunología , Acetatos/metabolismo , Adulto , Animales , Animales Recién Nacidos , Estudios de Casos y Controles , Desarrollo Infantil , Preescolar , Suplementos Dietéticos , Femenino , Feto/citología , Feto/diagnóstico por imagen , Microbioma Gastrointestinal/inmunología , Vida Libre de Gérmenes/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Lactante , Recién Nacido , Estudios Longitudinales , Intercambio Materno-Fetal/inmunología , Ratones , Tamaño de los Órganos/inmunología , Preeclampsia/sangre , Preeclampsia/diagnóstico , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Efectos Tardíos de la Exposición Prenatal/prevención & control , Estudios Prospectivos , Timo/citología , Timo/diagnóstico por imagen , Timo/crecimiento & desarrollo , Timo/inmunología , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo , Ultrasonografía Prenatal , Adulto Joven , Proteína AIRE
10.
Biochem Biophys Res Commun ; 516(3): 626-631, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31235250

RESUMEN

The disruption of intestinal mucosal immune tolerance can lead to the development of intestinal immune diseases such as food allergy (FA). Regulatory T cells (Tregs) in the mucosa play a critical role in maintaining peripheral immune tolerance in the intestine, and retinoic acid (RA) is absolutely required for the induction of Tregs. We have previously reported that kakkonto, a traditional Japanese herbal medicine, suppresses FA in a murine FA model due to the induction of Tregs in the colonic mucosa. However, the precise molecular mechanisms underlying the induction of Tregs remain unclear. Puerarin, an isoflavone derivative, is a major constituent of kakkonto. Thus, we investigated the effect of puerarin on the induction of Tregs. BALB/c mice were systemically sensitized and then orally challenged with ovalbumin (OVA) as an FA model. Puerarin treatment suppressed the development of allergic diarrhea in FA mice. The gene expression levels of IL-4 and mast cell protease I (mMCP-1) were significantly upregulated in the proximal colon of FA mice but were reduced by puerarin. The proportions of Foxp3+CD4+ cells and CD103+CD11c+ dendritic cells (DCs) were significantly higher among the colonic lamina propria (cLP) cells of puerarin-treated FA mice than among those of untreated FA mice. The gene expression of Aldh1a1, an RA synthesis enzyme, in colonic epithelial cells (CECs) was significantly higher in the puerarin-treated FA mouse colon than in the untreated FA mouse colon. In addition, the preventive effect of puerarin was suppressed in the FA model by pretreatment with LE540, an RA receptor (RAR) antagonist. The induction of Foxp3+CD4+ cells and CD103+CD11c+ DCs by puerarin was reduced by pretreatment with LE540. The present findings indicate that the augmentation of RA production in CECs induced by puerarin enhances the induction of Tregs and suppresses the development of FA in a mouse model. Thus, a natural enhancer of RA production, such as puerarin, has the potential to treat immune diseases attributed to Treg deficiency.


Asunto(s)
Hipersensibilidad a los Alimentos/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Mucosa Intestinal/inmunología , Isoflavonas/farmacología , Linfocitos T Reguladores/inmunología , Tretinoina/inmunología , Animales , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Tolerancia Inmunológica/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Linfocitos T Reguladores/metabolismo , Tretinoina/metabolismo , Vasodilatadores/farmacología
11.
Immunol Invest ; 48(8): 844-859, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30917711

RESUMEN

Tubiechong (Eupolyphaga sinensis) is an important material used in traditional Chinese medicine (TCM). However, the immunoregulation effects of E. sinensis Lyophilized Powder (ESL) are unclear. The in vivo study thus designed to elucidate the immuno-enhancement effects of ESL in immunosuppressed mice induced by cyclophosphamide (CTX). Mice were treated with three doses of ESL (0.5, 1.0 and 2.0 g/kg). Compared with model group, ESL notably increased the immune organ index, mononuclear macrophages function and the level of nature killer cell (NK) (p < 0.05 or p < 0.01), delayed type hypersensitivity (DTH) was also improved (p < 0.05). The level of superoxide dismutase (SOD) and catalase (CAT) were enhanced (p < 0.05), while malonyldialdehyde (MDA) and nitrogen monoxide (NO) were reduced (p < 0.05 or p < 0.01). Meanwhile, cluster determinant (CD)3+ T cell, CD4+ T cell and CD4+/CD8+ ratio were increased (p < 0.01). The cytokines secretion such as interleukin (IL)-2 and tumor necrosis factor alpha (TNF-α) were notably increased (p < 0.05 or p < 0.01), and IL-6 and IL-16 were also enhanced (p < 0.05). Furthermore, ESL significantly inhibited the phosphorylation of c-Jun N-terminal kinase (JNK), down-regulated the expression of Bcl-2 associated X protein (Bax), up-regulated the B cell lymphoma-2 protein (Bcl-2) expression and decreased the Bax/Bcl-2 ratio in spleen tissues (p < 0.05). In brief, all these findings suggest that ESL could effectively improve immune functions via modulating oxidative systems and innate immune cells. Abbreviations: TCM: Traditional Chinese Medicine; ESL: Eupolyphaga sinensis Lyophilized Powder; CCl4: Carbon tetrachloride; ERK: Extracellular regulated protein kinases; CTX: Cyclophosphamide; DTH: Delayed type hypersensitivity; SOD: Superoxide dismutase; CAT: Catalase; MDA: Malonyldialdehyde; NO: Nitrogen monoxide; NK: Nature killer cell; CD: Cluster determinant interleukin; TNF-α: Tumor Necrosis Factor alpha; JNK: c-Jun N-terminal kinase; Bax: Bcl-2 associated X protein; Bcl-2: B cell lymphoma-2 protein; Th1: Type-1 helper; Th2: Type-2 helper; FAMEs: Fatty acid methyl esters; DNFB: 2,4 - Dinitrofluorobenzene; ELISA: Enzyme-linked immuno sorbent assay; MAPK: Mitogen activated protein kinase; Cyt-c: Cytochrome c; SCFAs: Short-chain fatty acids; SDS-PAGE: Sodium dodecyl sulfate polyacrylamide gel electrophoresis.


Asunto(s)
Ciclofosfamida/toxicidad , Sistema Inmunológico/efectos de los fármacos , Tolerancia Inmunológica/efectos de los fármacos , Medicina Tradicional China , Neoptera/química , Polvos/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/metabolismo , Liofilización , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Tolerancia Inmunológica/inmunología , Inmunosupresores/toxicidad , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones Endogámicos ICR , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo
12.
Inflamm Bowel Dis ; 25(7): 1196-1207, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-30794280

RESUMEN

BACKGROUND: Inflammatory bowel disease (IBD) has a remitting and relapsing disease course; however, relatively little is understood regarding how inflammatory damage in acute colitis influences the microbiota, epithelial barrier, and immune function in subsequent colitis. METHODS: Mice were administered trinitrobenzene sulphonic acid (TNBS) via enema, and inflammation was assessed 2 days (d2) or 28 days (d28) later. Colitis was reactivated in some mice by re-treating at 28 days with TNBS and assessing 2 days later (d30). Epithelial responsiveness to secretagogues, microbiota composition, colonic infiltration, and immune activation was compared between all groups. RESULTS: At day 28, the distal colon had healed, mucosa was restored, and innate immune response had subsided, but colonic transepithelial transport (P = 0.048), regulatory T-cell (TREG) infiltration (P = 0.014), adherent microbiota composition (P = 0.0081), and responsiveness of stimulated innate immune bone marrow cells (P < 0.0001 for IL-1ß) differed relative to health. Two days after subsequent instillation of TNBS (d30 mice), the effects on inflammatory damage (P < 0.0001), paracellular permeability (P < 0.0001), and innate immune infiltration (P < 0.0001 for Ly6C+ Ly6G- macrophages) were reduced relative to d2 colitis. However, TREG infiltration was increased (P < 0.0001), and the responsiveness of stimulated T cells in the mesenteric lymph nodes shifted from pro-inflammatory at d2 to immune-suppressive at d30 (P < 0.0001 for IL-10). These effects were observed despite similar colonic microbiota composition and degradation of the mucosal layer between d2 and d30. CONCLUSIONS: Collectively, these results indicate that acute colitis chronically alters epithelial barrier function and both innate and adaptive immune responses. These effects reduce the consequences of a subsequent colitis event, warranting longitudinal studies in human IBD subjects.


Asunto(s)
Inmunidad Adaptativa/inmunología , Permeabilidad de la Membrana Celular/inmunología , Colitis/complicaciones , Epitelio/patología , Tolerancia Inmunológica/inmunología , Inmunidad Innata/inmunología , Inflamación/etiología , Enfermedad Aguda , Animales , Colitis/inducido químicamente , Colitis/patología , Modelos Animales de Enfermedad , Epitelio/inmunología , Epitelio/lesiones , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ácido Trinitrobencenosulfónico/toxicidad
13.
Int J Oncol ; 53(3): 949-960, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30015838

RESUMEN

Melanoma is one of the most fatal and therapy-resistant types of cancer; therefore, identifying novel therapeutic candidates to improve patient survival is an ongoing effort. Previous studies have revealed that pimozide is not sufficient to treat melanoma; therefore, enhancing the treatment is necessary. Indoleamine 2, 3­dioxygenase (IDO) is an immunosuppressive, intracellular rate-limiting enzyme, which contributes to immune tolerance in various tumours, including melanoma, and inhibition of IDO may be considered a novel therapeutic strategy when combined with pimozide. The present study aimed to assess the antitumour activities of pimozide in vitro, and to investigate the effects of pimozide combined with L­methyl-tryptophan (L­MT) in vivo. For in vitro analyses, the B16 melanoma cell line was used. Cell cytotoxicity assay, cell viability assay, wound­healing assay and western blotting were conducted to analyse the effects of pimozide on B16 cells. Furthermore, B16 cell-bearing mice were established as the animal model. Haematoxylin and eosin staining, immunohistochemistry, terminal deoxynucleotidyl transferase dUTP nick end-labelling staining, western blotting and flow cytometry were performed to determine the effects of monotherapy and pimozide and L­MT cotreatment on melanoma. The results demonstrated that pimozide exhibited potent antitumour activity via the regulation of proliferation, apoptosis and migration. Furthermore, the antitumour effects of pimozide were enhanced when combined with L­MT, not only via regulation of proliferation, apoptosis and migration, but also via immune modulation. Notably, pimozide may regulate tumour immunity through inhibiting the activities of signal transducer and activator of transcription (Stat)3 and Stat5. In conclusion, the present study proposed the use of pimozide in combination with the IDO inhibitor, L­MT, as a potential novel therapeutic strategy for the treatment of melanoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Tolerancia Inmunológica/efectos de los fármacos , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Melanoma Experimental/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Humanos , Tolerancia Inmunológica/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Pimozida/farmacología , Pimozida/uso terapéutico , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Triptófano/análogos & derivados , Triptófano/farmacología , Triptófano/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Scand J Immunol ; 87(3)2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29412476

RESUMEN

Taking into consideration the multiparametric nature of systemic lupus erythematosus (SLE), the severity and variability of symptoms and the lack of effective therapeutic approaches, this study took advantage of the recently described role of soluble major histocompatibility complex class II (sMHCII) molecules in maintaining tolerance to the organism and attempted to apply sMHCII proteins as a treatment to murine SLE experimental models in vitro as well as in vivo. After breaking tolerance to DNA in vitro, which was accompanied by development of specific anti-dsDNA antibodies, syngeneic or allogeneic sMHCII molecules, purified from healthy mouse serum, could significantly reduce the specific antibody levels and drive the system towards immunosuppression, as assessed by specific marker analysis on T cells and cytokine production by flow cytometry and ELISA, respectively. The in vivo experimental model consisted of pristane-induced SLE symptoms to BALB/c mice, which developed maximal levels of anti-dsDNA 2 months after pristane inoculation. Syngeneic or allogeneic sMHCII administration could alleviate pristane-induced symptoms, significantly decrease specific anti-dsDNA antibody production and develop immunosuppression to the host, as manifested by increase of CD4 + CTLA-4 +  and CD4 + CD25 +  cell populations in the spleen. Thus, the results presented in this study introduced the ability of sMHCII proteins to suppress specific autoantigen response, opening new areas of research and offering novel therapeutic approaches to SLE with expanding features to other autoimmune diseases.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Autoantígenos/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Tolerancia Inmunológica/inmunología , Inmunoterapia/métodos , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/terapia , Linfocitos T/inmunología , Animales , Antígenos CD4/metabolismo , Antígeno CTLA-4/metabolismo , Células Cultivadas , ADN/inmunología , Modelos Animales de Enfermedad , Terapia de Inmunosupresión , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Lupus Eritematoso Sistémico/inducido químicamente , Ratones , Ratones Endogámicos BALB C , Bazo/citología , Bazo/inmunología , Terpenos/efectos adversos
16.
J Theor Biol ; 442: 1-10, 2018 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-29337259

RESUMEN

Myeloid-derived suppressor cells (MDSCs) belong to immature myeloid cells that are generated and accumulated during the tumor development. MDSCs strongly suppress the anti-tumor immunity and provide conditions for tumor progression and metastasis. In this study, we present a mathematical model based on ordinary differential equations (ODE) to describe tumor-induced immunosuppression caused by MDSCs. The model consists of four equations and incorporates tumor cells, cytotoxic T cells (CTLs), natural killer (NK) cells and MDSCs. We also provide simulation models that evaluate or predict the effects of anti-MDSC drugs (e.g., l-arginine and 5-Fluorouracil (5-FU)) on the tumor growth and the restoration of anti-tumor immunity. The simulated results obtained using our model were in good agreement with the corresponding experimental findings on the expansion of splenic MDSCs, immunosuppressive effects of these cells at the tumor site and effectiveness of l-arginine and 5-FU on the re-establishment of antitumor immunity. Regarding this latter issue, our predictive simulation results demonstrated that intermittent therapy with low-dose 5-FU alone could eradicate the tumors irrespective of their origins and types. Furthermore, at the time of tumor eradication, the number of CTLs prevailed over that of cancer cells and the number of splenic MDSCs returned to the normal levels. Finally, our predictive simulation results also showed that the addition of l-arginine supplementation to the intermittent 5-FU therapy reduced the time of the tumor eradication and the number of iterations for 5-FU treatment. Thus, the present mathematical model provides important implications for designing new therapeutic strategies that aim to restore antitumor immunity by targeting MDSCs.


Asunto(s)
Tolerancia Inmunológica/inmunología , Modelos Inmunológicos , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Algoritmos , Animales , Antineoplásicos/farmacología , Arginina/farmacología , Fluorouracilo/farmacología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Inmunidad/efectos de los fármacos , Inmunidad/inmunología , Células Asesinas Naturales/inmunología , Linfoma/tratamiento farmacológico , Linfoma/inmunología , Linfoma/patología , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Linfocitos T Citotóxicos/inmunología
17.
J Allergy Clin Immunol ; 141(5): 1750-1760.e1, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29128670

RESUMEN

BACKGROUND: Three years of treatment with either sublingual or subcutaneous allergen immunotherapy has been shown to be effective and to induce long-term tolerance. The Gauging Response in Allergic Rhinitis to Sublingual and Subcutaneous Immunotherapy (GRASS) trial demonstrated that 2 years of treatment through either route was effective in suppressing the response to nasal allergen challenge, although it was insufficient for inhibition 1 year after discontinuation. OBJECTIVE: We sought to examine in the GRASS trial the time course of immunologic changes during 2 years of sublingual and subcutaneous immunotherapy and for 1 year after treatment discontinuation. METHODS: We performed multimodal immunomonitoring to assess allergen-specific CD4 T-cell properties in parallel with analysis of local mucosal cytokine responses induced by nasal allergen exposure and humoral immune responses that included IgE-dependent basophil activation and measurement of serum inhibitory activity for allergen-IgE binding to B cells (IgE-facilitated allergen binding). RESULTS: All 3 of these distinct arms of the immune response displayed significant and coordinate alterations during 2 years of allergen desensitization, followed by reversal at 3 years, reflecting a lack of a durable immunologic effect. Although frequencies of antigen-specific TH2 cells in peripheral blood determined by using HLA class II tetramer analysis most closely paralleled clinical outcomes, IgE antibody-dependent functional assays remained inhibited in part 1 year after discontinuation. CONCLUSION: Two years of allergen immunotherapy were effective but insufficient for long-term tolerance. Allergen-specific TH2 cells most closely paralleled the transient clinical outcome, and it is likely that recurrence of the T-cell drivers of allergic immunity abrogated the potential for durable tolerance. On the other hand, the persistence of IgE blocking antibody 1 year after discontinuation might be an early indicator of a protolerogenic mechanism.


Asunto(s)
Alérgenos/administración & dosificación , Alérgenos/inmunología , Rinitis Alérgica Estacional/terapia , Administración Cutánea , Administración Sublingual , Anticuerpos/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Desensibilización Inmunológica/métodos , Humanos , Tolerancia Inmunológica/inmunología , Inmunidad Humoral/inmunología , Inmunoglobulina E/inmunología , Phleum/inmunología , Poaceae/inmunología , Polen/inmunología , Rinitis Alérgica Estacional/inmunología , Células Th2/inmunología
19.
World J Gastroenterol ; 23(38): 6962-6972, 2017 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-29097869

RESUMEN

AIM: To test whether a delayed and short course of rapamycin would induce immunosuppressive effects following allogeneic orthotopic liver transplantation (OLT) in rats. METHODS: Allogeneic OLTs were performed using Dark Agouti livers transplanted into Lewis recipients, and syngeneic OLTs were performed using the Lewis rat strain. Rapamycin (1 mg/kg per day) was administered by gavage from day 4 to day 11 post-transplantation. Lymphocyte cellular compartments were analyzed by flow cytometry in draining lymph nodes, non-draining lymph nodes and the spleen at days 11 and 42 in rapamycin-treated rats, untreated control rats and syngeneic grafted rats. Skin grafts from Dark agouti or from F344 RT were performed at day 30 on liver grafted rats treated with rapamycin. RESULTS: An 8-d course of rapamycin treatment initiated 4 d following transplantation resulted in the survival of grafted rats for more than 100 d. In contrast, untreated rats died of liver failure within 13 to 21 d. The analysis of the cellular compartment revealed an increase in two cellular subpopulations, specifically myeloid-derived suppressor cells (MDSCs) and CD8+CD45RClow T cells, without major modifications in the regulatory T cell (Treg) compartment in treated rats in the early stages after grafting. We evaluated the ability of treated rats to reject third-party allogeneic skin grafts to confirm their immune competence. In contrast, when skin was collected from rats syngeneic to the grafted liver, it was not rejected. CONCLUSION: Our results demonstrate that short and delayed rapamycin treatment allows for tolerance in allogeneic OLT. The results also allowed for the identification of the mechanisms of tolerance induced by rapamycin by identifying MDSCs and CD8+CD45RClow T cells as associated with the state of tolerance.


Asunto(s)
Rechazo de Injerto/prevención & control , Tolerancia Inmunológica/efectos de los fármacos , Inmunosupresores/administración & dosificación , Trasplante de Hígado , Sirolimus/administración & dosificación , Animales , Evaluación Preclínica de Medicamentos , Tolerancia Inmunológica/inmunología , Masculino , Ratas Endogámicas Lew , Trasplante Homólogo
20.
Cell Microbiol ; 19(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28382773

RESUMEN

CD4+ CD25+ FoxP3+ regulatory T cells (Tregs) are key players for maintaining immune tolerance and for reducing the inflammation-mediated tissue damage following infection. However, Tregs also suppress protective immune responses to pathogens (including virus, bacteria, parasites, and fungi) and vaccines and enhance pathogen persistence by inhibiting the activation and functions of both innate and adaptive immune cells such as dendritic cells, macrophages, and T and B lymphocytes and by promoting immunosuppressive environment. Therefore, equilibrium in the Treg number and function is important to ensure pathogen clearance and protection from infection-associated immunopathologies. Recent advances in understanding of Treg influence on the outcome of infection opened new avenues to target them. Various small molecules, pharmacological inhibitors, monoclonal antibodies that target Tregs provided proof of concept in experimental models. The field also benefits from advances in other subjects, particularly oncology and autoimmunity, where Treg-targeted therapies are exploited in the clinic to a greater extent. The future research should aim at translating this preclinical success to human application.


Asunto(s)
Infecciones Bacterianas/inmunología , Tolerancia Inmunológica/inmunología , Micosis/inmunología , Enfermedades Parasitarias/inmunología , Linfocitos T Reguladores/inmunología , Virosis/inmunología , Linfocitos B/inmunología , Células Dendríticas/inmunología , Humanos , Macrófagos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA