Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Zhongguo Zhong Yao Za Zhi ; 48(19): 5304-5314, 2023 Oct.
Artículo en Zh | MEDLINE | ID: mdl-38114120

RESUMEN

This study aims to observe the effects of diosgenin on the expression of mammalian target of rapamycin(mTOR), sterol regulatory element-binding protein-1c(SREBP-1c), heat shock protein 60(HSP60), medium-chain acyl-CoA dehydrogenase(MCAD), and short-chain acyl-CoA dehydrogenase(SCAD) in the liver tissue of the rat model of non-alcoholic fatty liver disease(NAFLD) and explore the mechanism of diosgenin in alleviating NAFLD. Forty male SD rats were randomized into five groups: a control group, a model group, low-(150 mg·kg~(-1)·d~(-1)) and high-dose(300 mg·kg~(-1)·d~(-1)) diosgenin groups, and a simvastatin(4 mg·kg~(-1)·d~(-1)) group. The rats in the control group were fed with a normal diet, while those in the other four groups were fed with a high-fat diet. After feeding for 8 weeks, the body weight of rats in the high-fat diet groups increased significantly. After that, the rats were administrated with the corresponding dose of diosgenin or simvastatin by gavage every day for 8 weeks. The levels of triglyceride(TG), total cholesterol(TC), alanine transaminase(ALT), and aspartate transaminase(AST) in the serum were determined by the biochemical method. The levels of TG and TC in the liver were measured by the enzyme method. Oil-red O staining was employed to detect the lipid accumulation, and hematoxylin-eosin(HE) staining to detect the pathological changes in the liver tissue. The mRNA and protein levels of mTOR, SREBP-1c, HSP60, MCAD, and SCAD in the liver tissue of rats were determined by real-time fluorescence quantitative polymerase chain reaction(RT-qPCR) and Western blot, respectively. Compared with the control group, the model group showed increased body weight, food uptake, liver index, TG, TC, ALT, and AST levels in the serum, TG and TC levels in the liver, lipid deposition in the liver, obvious hepatic steatosis, up-regulated mRNA and protein expression levels of mTOR and SREBP-1c, and down-regulated mRNA and protein expression levels of HSP60, MCAD, and SCAD. Compared with the model group, the rats in each treatment group showed obviously decreased body weight, food uptake, liver index, TG, TC, ALT, and AST levels in the serum, TG and TC levels in the liver, lessened lipid deposition in the liver, ameliorated hepatic steatosis, down-regulated mRNA and protein le-vels of mTOR and SREBP-1c, and up-regulated mRNA and protein levels of HSP60, MCAD, and SCAD. The high-dose diosgenin outperformed the low-dose diosgenin and simvastatin. Diosgenin may prevent and treat NAFLD by inhibiting the expression of mTOR and SREBP-1c and promoting the expression of HSP60, MCAD, and SCAD to reduce lipid synthesis, improving mitochondrial function, and promoting fatty acid ß oxidation in the liver.


Asunto(s)
Diosgenina , Enfermedad del Hígado Graso no Alcohólico , Ratas , Masculino , Animales , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Dieta Alta en Grasa/efectos adversos , Diosgenina/metabolismo , Chaperonina 60/metabolismo , Chaperonina 60/farmacología , Chaperonina 60/uso terapéutico , Ratas Sprague-Dawley , Hígado , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Triglicéridos , ARN Mensajero/metabolismo , Simvastatina/metabolismo , Simvastatina/farmacología , Simvastatina/uso terapéutico , Peso Corporal , Metabolismo de los Lípidos , Mamíferos/genética , Mamíferos/metabolismo
2.
Proc Natl Acad Sci U S A ; 115(51): E11924-E11932, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30509980

RESUMEN

The human chaperonin Hsp60 is thought to play a role in the progression of Alzheimer's disease by mitigating against intracellular ß-amyloid stress. Here, we show that the bacterial homolog GroEL (51% sequence identity) reduces the neurotoxic effects of amyloid-ß(1-42) (Aß42) on human neural stem cell-derived neuronal cultures. To understand the mechanism of GroEL-mediated abrogation of neurotoxicity, we studied the interaction of Aß42 with GroEL using a variety of biophysical techniques. Aß42 binds to GroEL as a monomer with a lifetime of ∼1 ms, as determined from global analysis of multiple relaxation-based NMR experiments. Dynamic light scattering demonstrates that GroEL dissolves small amounts of high-molecular-weight polydisperse aggregates present in fresh soluble Aß42 preparations. The residue-specific transverse relaxation rate profile for GroEL-bound Aß42 reveals the presence of three anchor-binding regions (residues 16-21, 31-34, and 40-41) located within the hydrophobic GroEL-consensus binding sequences. Single-molecule FRET analysis of Aß42 binding to GroEL results in no significant change in the FRET efficiency of a doubly labeled Aß42 construct, indicating that Aß42 samples a random coil ensemble when bound to GroEL. Finally, GroEL substantially slows down the disappearance of NMR visible Aß42 species and the appearance of Aß42 protofibrils and fibrils as monitored by electron and atomic force microscopies. The latter observations correlate with the effect of GroEL on the time course of Aß42-induced neurotoxicity. These data provide a physical basis for understanding how Hsp60 may serve to slow down the progression of Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/efectos de los fármacos , Chaperonina 60/antagonistas & inhibidores , Chaperonina 60/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Fragmentos de Péptidos/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Chaperonina 60/uso terapéutico , Escherichia coli/genética , Escherichia coli/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Humanos , Cinética , Espectroscopía de Resonancia Magnética , Microscopía de Fuerza Atómica , Microscopía Electrónica , Modelos Moleculares , Células-Madre Neurales/efectos de los fármacos , Síndromes de Neurotoxicidad/tratamiento farmacológico , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Agregación Patológica de Proteínas/metabolismo , Unión Proteica , Conformación Proteica , Dominios Proteicos , Coloración y Etiquetado
3.
Bioorg Med Chem Lett ; 29(13): 1665-1672, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31047750

RESUMEN

Current treatments for Mycobacterium tuberculosis infections require long and complicated regimens that can lead to patient non-compliance, increasing incidences of antibiotic-resistant strains, and lack of efficacy against latent stages of disease. Thus, new therapeutics are needed to improve tuberculosis standard of care. One strategy is to target protein homeostasis pathways by inhibiting molecular chaperones such as GroEL/ES (HSP60/10) chaperonin systems. M. tuberculosis has two GroEL homologs: GroEL1 is not essential but is important for cytokine-dependent granuloma formation, while GroEL2 is essential for survival and likely functions as the canonical housekeeping chaperonin for folding proteins. Another strategy is to target the protein tyrosine phosphatase B (PtpB) virulence factor that M. tuberculosis secretes into host cells to help evade immune responses. In the present study, we have identified a series of GroEL/ES inhibitors that inhibit M. tuberculosis growth in liquid culture and biochemical function of PtpB in vitro. With further optimization, such dual-targeting GroEL/ES and PtpB inhibitors could be effective against all stages of tuberculosis - actively replicating bacteria, bacteria evading host cell immune responses, and granuloma formation in latent disease - which would be a significant advance to augment current therapeutics that primarily target actively replicating bacteria.


Asunto(s)
Chaperonina 60/uso terapéutico , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/tratamiento farmacológico , Proteínas Bacterianas/metabolismo , Chaperonina 60/farmacología , Humanos , Modelos Moleculares , Polifarmacología
4.
Biochem Biophys Res Commun ; 498(4): 701-706, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29107690

RESUMEN

HSP60 has been proved to be closely related to atherosclerosis due to its antigenicity. To determine this antigenicity effect, the ApoE-/- mice were fed with western-type diet and HSP60 was administrated orally or subcutaneously (SC) for potential vaccine against atherosclerosis. Here, we observed the ApoE-/- mice with oral HSP60 administration group showed a significant reduction in plaque size at the aortic root; accompanied by increased MSDCs (CD11b+Gr1+) in peripheral blood and spleen which was mostly composed of M-MDSCs (CD11b+LY6G-LY6Chigh), and increased plasma IL-10 and splenic Foxp3, Arg1, iNOS mRNA as well as decreased plasma IFN-γ and splenic T-bet mRNA compared to control group. Surprisingly, ApoE-/- mice with subcutaneous HSP60 administration group showed contrary results and their MDSCs were mostly composed of G-MDSCs (CD11b+LY6G+LY6Clow). As expected, both PBS-oral and PBS-SC groups showed no significant effects on both the immune response and atherosclerotic plaque formation. In contrast, subcutaneous administration of HSP60 causes the opposite response. Thus, we propose the proper method for administering HSP60 as a new immunologic agent for prevention and treatment of atherosclerosis.


Asunto(s)
Apolipoproteínas E/genética , Aterosclerosis/tratamiento farmacológico , Chaperonina 60/uso terapéutico , Factores Inmunológicos/uso terapéutico , Células Supresoras de Origen Mieloide/efectos de los fármacos , Administración a través de la Mucosa , Administración Oral , Animales , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/inmunología , Chaperonina 60/administración & dosificación , Chaperonina 60/inmunología , Citocinas/sangre , Citocinas/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Tolerancia Inmunológica/efectos de los fármacos , Inmunidad Mucosa/efectos de los fármacos , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/inmunología , Inflamación/sangre , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/inmunología , Masculino , Ratones , Ratones Noqueados , Células Supresoras de Origen Mieloide/inmunología , Placa Aterosclerótica/sangre , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/genética , Placa Aterosclerótica/inmunología
5.
Clin Immunol ; 149(3): 307-16, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24090708

RESUMEN

Type 1 diabetes is a chronic immune-mediated disease resulting in destruction of insulin-producing ß-cells. Several studies have been performed aiming to halt disease progression after diagnosis; to reduce the increased diabetes risk in islet-autoantibody positive subjects; and to prevent the onset of ß-cell autoimmunity in subjects genetically at risk but without autoantibodies. Whereas secondary prevention trials failed, trials in newly diagnosed patients have shown partial success in preserving C-peptide. These studies target T-cells and inflammation and make use of antigen-specific immune modulation or stem cell approaches. However, thus far no immune-based therapeutic regimen has cured type 1 diabetes after its clinical onset or has stabilized the decline of C-peptide to achieve the status of an approved drug. This review summarizes immune intervention trials and the current knowledge of DiaPep277® peptide as a form of immune intervention in type 1 diabetes.


Asunto(s)
Chaperonina 60/uso terapéutico , Diabetes Mellitus Tipo 1/terapia , Hipoglucemiantes/uso terapéutico , Inmunoterapia , Islotes Pancreáticos/efectos de los fármacos , Fragmentos de Péptidos/uso terapéutico , Linfocitos T/efectos de los fármacos , Autoanticuerpos/inmunología , Autoinmunidad , Péptido C/inmunología , Péptido C/metabolismo , Ensayos Clínicos como Asunto , Diabetes Mellitus Tipo 1/inmunología , Humanos , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/patología , Linfocitos T/inmunología , Linfocitos T/patología , Resultado del Tratamiento
6.
Clin Immunol ; 149(3): 345-55, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23490422

RESUMEN

Type 1 diabetes (T1D) results from an aberrant immunological response against the insulin-producing beta cells in the islets of the pancreas. The ideal therapy would restore immune balance in a safe and lasting fashion, stopping the process of beta cell decay. The efficacy of immune suppressive agents such as cyclosporin underscores the notion that T1D can in principle be prevented, albeit at an unacceptable long-term safety risk. Immune modulatory drugs such as monoclonal anti-CD3 antibody, on the other hand, have recently had rather disappointing results in phase 3 trials, possibly due to inadequate dosing or choice of inappropriate endpoints. Therefore, it is argued that striking the right balance between safety and efficacy, together with careful trial design, will be paramount in preventing T1D. Here we outline the concept of antigen-specific tolerization as a strategy to safely induce long-term protection against T1D, focusing on available clinical trial data, key knowledge gaps and potential future directions.


Asunto(s)
Autoantígenos/inmunología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Células Secretoras de Insulina/efectos de los fármacos , Anticuerpos Monoclonales/uso terapéutico , Autoantígenos/metabolismo , Complejo CD3/inmunología , Complejo CD3/metabolismo , Chaperonina 60/uso terapéutico , Ensayos Clínicos Fase III como Asunto , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/prevención & control , Glutamato Descarboxilasa/uso terapéutico , Humanos , Tolerancia Inmunológica , Insulina/inmunología , Insulina/metabolismo , Insulina/uso terapéutico , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/patología , Fragmentos de Péptidos/uso terapéutico , Precursores de Proteínas/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología
7.
Front Immunol ; 14: 1162739, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37187739

RESUMEN

Cytokines are secretion proteins that mediate and regulate immunity and inflammation. They are crucial in the progress of acute inflammatory diseases and autoimmunity. In fact, the inhibition of proinflammatory cytokines has been widely tested in the treatment of rheumatoid arthritis (RA). Some of these inhibitors have been used in the treatment of COVID-19 patients to improve survival rates. However, controlling the extent of inflammation with cytokine inhibitors is still a challenge because these molecules are redundant and pleiotropic. Here we review a novel therapeutic approach based on the use of the HSP60-derived Altered Peptide Ligand (APL) designed for RA and repositioned for the treatment of COVID-19 patients with hyperinflammation. HSP60 is a molecular chaperone found in all cells. It is involved in a wide diversity of cellular events including protein folding and trafficking. HSP60 concentration increases during cellular stress, for example inflammation. This protein has a dual role in immunity. Some HSP60-derived soluble epitopes induce inflammation, while others are immunoregulatory. Our HSP60-derived APL decreases the concentration of cytokines and induces the increase of FOXP3+ regulatory T cells (Treg) in various experimental systems. Furthermore, it decreases several cytokines and soluble mediators that are raised in RA, as well as decreases the excessive inflammatory response induced by SARS-CoV-2. This approach can be extended to other inflammatory diseases.


Asunto(s)
Artritis Reumatoide , Chaperonina 60 , Humanos , COVID-19 , Citocinas/metabolismo , Inflamación/tratamiento farmacológico , Péptidos/farmacología , Péptidos/uso terapéutico , SARS-CoV-2/metabolismo , Chaperonina 60/farmacología , Chaperonina 60/uso terapéutico
8.
J Drug Target ; 30(1): 31-45, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33939586

RESUMEN

Heat shock proteins (HSPs), most of which are molecular chaperones, are highly conserved proteins produced by cells under physiological stress or pathological conditions. HSP60 (57-69 kDa) can promote or inhibit cell apoptosis through different mechanisms, and its abnormal expression is also related to tumour cell metastasis and drug resistance. In recent years, HSP60 has received increasing attention in the field of cancer research due to its potential as a diagnostic and prognostic biomarker or therapeutic target. However, in different types of cancer, the specific mechanisms of abnormally expressed HSP60 in tumour carcinogenesis and drug resistance are complicated and still require further study. In this article, we comprehensively review the regulative mechanisms of HSP60 on apoptosis, its applications as a cancer diagnostic biomarker and a therapeutic target, evidence of involvement in tumour resistance and the applications of exosomal HSP60 in liquid biopsy. By evaluating the current findings of HSP60 in cancer research, we highlight some core issues that need to be addressed for the use of HSP60 as a diagnostic or prognostic biomarker and therapeutic target in certain types of cancer.


Asunto(s)
Neoplasias , Apoptosis , Biomarcadores de Tumor/metabolismo , Chaperonina 60/metabolismo , Chaperonina 60/uso terapéutico , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/uso terapéutico , Humanos , Proteínas Mitocondriales , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
9.
Ann Rheum Dis ; 70(12): 2199-206, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21914624

RESUMEN

OBJECTIVES: Mucosal immune therapy with disease-inducing antigens is an effective way to prevent experimental arthritis, but in humans these antigens are unknown. In juvenile idiopathic arthritis, however, T cell recognition of a so-called bystander antigen, heat shock protein 60 (HSP60), is associated with a good prognosis. Recently epitopes derived from HSP60, a microbial peptide (p1) and its self-homologue (p2) were reported to induce tolerogenic T cell responses in vitro in patients with arthritis. A study was undertaken to determine whether mucosal administration of these bystander epitopes can be similarly effective in suppressing arthritis. METHODS: Rats were treated nasally with p1, p2 or phosphate-buffered saline before arthritis induction. Arthritis scores were assessed and peptide-specific proliferative responses, phenotypic analysis, cytokine production and in vitro suppressive capacity of cells were measured in lymph nodes and spleens. CD4 spleen T cells from p1- or p2-treated rats were adoptively transferred into naïve rats that were subsequently injected with complete Freund's adjuvant for arthritis induction. RESULTS: Nasal administration of p1 prevented experimental arthritis whereas treatment with the self-homologue p2 did not. Adoptive transfer of CD4 T cells protected against experimental arthritis. Treatment with p1 increased peptide-specific and self-crossreactive interferon γ (IFNγ) production. Tumour necrosis factor α (TNFα) levels were reduced at the site of inflammation. Forkhead box P3 (FoxP3) expression remained stable but the suppressive capacity of T regulatory cells in p1-treated rats was enhanced. CONCLUSION: p1 immune therapy induces a population of CD4 T cells with reduced TNFα and increased peptide-specific IFNγ production at the site of inflammation. This population expresses FoxP3 and has potent suppressive capacity which, upon transfer, protects against arthritis. The bystander epitope p1 may therefore be a suitable candidate for antigen-specific immunotherapy in arthritis.


Asunto(s)
Artritis Experimental/prevención & control , Efecto Espectador/inmunología , Chaperonina 60/uso terapéutico , Administración Intranasal , Animales , Artritis Experimental/inmunología , Linfocitos T CD4-Positivos/trasplante , Chaperonina 60/administración & dosificación , Chaperonina 60/inmunología , Citocinas/biosíntesis , Epítopos de Linfocito T/administración & dosificación , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/uso terapéutico , Adyuvante de Freund , Inmunidad Mucosa , Inmunoterapia Adoptiva/métodos , Mediadores de Inflamación/metabolismo , Activación de Linfocitos/inmunología , Masculino , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/uso terapéutico , Ratas , Ratas Endogámicas Lew , Bazo/inmunología , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología
10.
Transgenic Res ; 20(2): 221-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20526808

RESUMEN

Transgenic plants are able to express molecules with antigenic properties. In recent years, this has led the pharmaceutical industry to use plants as alternative systems for the production of recombinant proteins. Plant-produced recombinant proteins can have important applications in therapeutics, such as in the treatment of rheumatoid arthritis (RA). In this study, the mycobacterial HSP65 protein expressed in tobacco plants was found to be effective as a treatment for adjuvant-induced arthritis (AIA). We cloned the hsp65 gene from Mycobacterium leprae into plasmid pCAMBIA 2301 under the control of the double 35S promoter from cauliflower mosaic virus. Agrobacterium tumefaciens bearing the pChsp65 plasmid was used to transform tobacco plants. Incorporation of the hsp65 gene was confirmed by PCR, reverse transcription-PCR, histochemistry, and western blot analyses in several transgenic lines of tobacco plants. Oral treatment of AIA rats with the HSP65 protein allowed them to recover body weight and joint inflammation was reduced. Our results suggest a synergistic effect between the HSP65 expressed protein and metabolites presents in tobacco plants.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Proteínas Bacterianas/uso terapéutico , Chaperonina 60/uso terapéutico , Nicotiana/metabolismo , Plantas Modificadas Genéticamente/metabolismo , Administración Oral , Agrobacterium tumefaciens/genética , Animales , Proteínas Bacterianas/administración & dosificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Western Blotting , Chaperonina 60/administración & dosificación , Chaperonina 60/genética , Chaperonina 60/metabolismo , Humanos , Mycobacterium leprae/genética , Mycobacterium leprae/metabolismo , Plantas Modificadas Genéticamente/genética , Plásmidos , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Nicotiana/genética , Resultado del Tratamiento
11.
J Gastroenterol ; 56(5): 442-455, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33782752

RESUMEN

BACKGROUND: We previously showed that supernatants of Lactobacillus biofilms induced an anti-inflammatory response by affecting the secretion of macrophage-derived cytokines, which was abrogated upon immunodepletion of the stress protein GroEL. METHODS: We purified GroEL from L. reuteri and analysed its anti-inflammatory properties in vitro in human macrophages isolated from buffy coats, ex vivo in explants from human biopsies and in vivo in a mouse model of DSS induced intestinal inflammation. As a control, we used GroEL purified (LPS-free) from E. coli. RESULTS: We found that L. reuteri GroEL (but not E. coli GroEL) inhibited pro-inflammatory M1-like macrophages markers, and favored M2-like markers. Consequently, L. reuteri GroEL inhibited pro-inflammatory cytokines (TNFα, IL-1ß, IFNγ) while favouring an anti-inflammatory secretome. In colon tissues from human biopsies, L. reuteri GroEL was also able to decrease markers of inflammation and apoptosis (caspase 3) induced by LPS. In mice, we found that rectal administration of L. reuteri GroEL (but not E. coli GroEL) inhibited all signs of haemorrhagic colitis induced by DSS including intestinal mucosa degradation, rectal bleeding and weight loss. It also decreased intestinal production of inflammatory cytokines (such as IFNγ) while increasing anti-inflammatory IL-10 and IL-13. These effects were suppressed when animals were immunodepleted in macrophages. From a mechanistic point of view, the effect of L. reuteri GroEL seemed to involve TLR4, since it was lost in TRL4-/- mice, and the activation of a non-canonical TLR4 pathway. CONCLUSIONS: L. reuteri GroEL, by affecting macrophage inflammatory features, deserves to be explored as an alternative to probiotics.


Asunto(s)
Chaperonina 60/farmacología , Colon/efectos de los fármacos , Inflamación/prevención & control , Lactobacillus/metabolismo , Animales , Chaperonina 60/uso terapéutico , Colon/fisiopatología , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Limosilactobacillus reuteri/efectos de los fármacos , Limosilactobacillus reuteri/metabolismo , Ratones Endogámicos BALB C , Estadísticas no Paramétricas
12.
Cell Stress Chaperones ; 26(3): 515-525, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33629254

RESUMEN

Hyperinflammation distinguishes COVID-19 patients who develop a slight disease or none, from those progressing to severe and critical conditions. CIGB-258 is a therapeutic option for the latter group of patients. This drug is an altered peptide ligand (APL) derived from the cellular stress protein 60 (HSP60). In preclinical models, this peptide developed anti-inflammatory effects and increased regulatory T cell (Treg) activity. Results from a phase I clinical trial with rheumatoid arthritis (RA) patients indicated that CIGB-258 was safe and reduced inflammation. The aim of this study was to examine specific biomarkers associated with hyperinflammation, some cytokines linked to the cytokine storm granzyme B and perforin in a cohort of COVID-19 patients treated with this peptide. All critically ill patients were under invasive mechanical ventilation and received the intravenous administration of 1 or 2 mg of CIGB-258 every 12 h. Seriously ill patients were treated with oxygen therapy receiving 1 mg of CIGB-258 every 12 h and all patients recovered from their severe condition. Biomarker levels associated with hyperinflammation, such as interleukin (IL)-6, IL-10, tumor necrosis factor (TNF-α), granzyme B, and perforin, significantly decreased during treatment. Furthermore, we studied the ability of CIGB-258 to induce Tregs in COVID-19 patients and found that Tregs were induced in all patients studied. Altogether, these results support the therapeutic potential of CIGB-258 for diseases associated with hyperinflammation. Clinical trial registry: RPCEC00000313.


Asunto(s)
Antiinflamatorios/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Chaperonina 60/uso terapéutico , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Antiinflamatorios/química , COVID-19/sangre , COVID-19/complicaciones , Chaperonina 60/química , Síndrome de Liberación de Citoquinas/sangre , Síndrome de Liberación de Citoquinas/complicaciones , Femenino , Humanos , Inflamación/sangre , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Interleucina-10/sangre , Interleucina-6/sangre , Masculino , Persona de Mediana Edad , SARS-CoV-2/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Factor de Necrosis Tumoral alfa/sangre , Adulto Joven
13.
Diabetes Metab Res Rev ; 25(4): 316-20, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19267355

RESUMEN

In type 1 diabetes mellitus, autoimmune T-cells mount an attack on insulin producing beta-cells eventually causing hyperglycemia. It is hypothesized, that to prevent and treat this disease, one must interfere with the autoimmune process. To avoid hazardous side effects, this intervention should not cause a global immune suppression but immune modulation. DiaPep277 (peptide 277) is a 24 amino acid peptide derived from positions 437-460 in HSP60. It has recently been shown to have an immune modulatory effect on diabetogenic T cells in animal models of diabetes. It has also been recently implicated as a possible auto-antigen in type 1 diabetes. Promising results in animal models led to phase 1 to 3 human clinical trials in patients with type 1 diabetes, the results of which are the focus of this review. A combined analysis of all the adult phase II studies revealed that DiaPep277 significantly inhibits the decline in stimulated C-peptide secretion (thus preserving endogenous insulin secretion). This effect was more pronounced in patients with a high beta-cell reserve at the start of the treatment. Furthermore, opposite trends in glycemic control of DiaPep277 treated patients where noted as opposed to placebo treated patients. The phase III study has began more than 2 years ago in 40 medical centers worldwide, and thus far recruited over 350 patients around the world. If DiaPep277 will prove to be efficacious, it will cause a paradigm shift in the treatment of type 1 diabetes, from treating the subsequent insulin deficiency to addressing the initial autoimmune process that is at the heart of the disease.


Asunto(s)
Chaperonina 60/uso terapéutico , Diabetes Mellitus Tipo 1/terapia , Factores Inmunológicos/uso terapéutico , Células Secretoras de Insulina/inmunología , Péptidos/uso terapéutico , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Péptido C/efectos de los fármacos , Péptido C/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Fragmentos de Péptidos/uso terapéutico , Linfocitos T/inmunología
14.
Autoimmunity ; 51(5): 210-220, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30382756

RESUMEN

Therapeutic efficacy of P277 against type 1 diabetes was extensively investigated and clinically evidenced. Clinical trials Phases I and II concluded promising results, while the data of P277 immunogenicity in Phase III trials represented weak responses that led to abolish medical use. But, a therapeutic performance of P277 cannot be forgotten. So, in order to exploit its therapeutic benefits and improve its immunogenicity, we developed a new analogue VP to optimize therapeutic efficacy and enhancing immunosuppressive modulations. However, new analogue was purified, and then used to immunize diabetic NOD mice to investigate antidiabetic effects through modulation of immunological status. So, DCs immune responses, relative TLRs, MyD88, and NF-Kß1 mRNA expression on DCs and splenocytes under VP effect were tested. Circulating and intracellular cytokines were also evaluated at treated and non-treated mice. Splenic T lymphocytes proliferation (Th1 and Treg cells) were also determined. Results revealed that VP significantly down regulates DCs maturation through TLR2, TLR4, and MyD88 pathways. It also shifts DCs to a tolerogenic polarization through NF-Kß1 pathway that mediates Th1 immunosuppression and enhances iTreg expanding in type1diabetes mice. Meanwhile, we noticed that VP significantly enhances iTreg CD25 + FoxP3+ proliferation. In conclusion, VP showed promising immune potential to modulate immune regulatory responses and shifts DCs to suppress autoreactive Th1 cells which ameliorated immunosuppressive potency in the type1 diabetic mice.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Chaperonina 60/farmacología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Tolerancia Inmunológica/efectos de los fármacos , Inmunosupresores/farmacología , Fragmentos de Péptidos/farmacología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Comunicación Celular/inmunología , Chaperonina 60/genética , Chaperonina 60/inmunología , Chaperonina 60/uso terapéutico , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito B/genética , Femenino , Humanos , Inmunosupresores/uso terapéutico , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Mutagénesis , Subunidad p50 de NF-kappa B/inmunología , Subunidad p50 de NF-kappa B/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/uso terapéutico , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células TH1/inmunología , Células TH1/metabolismo
15.
Cancer Biol Ther ; 6(4): 487-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17457039

RESUMEN

The molecular chaperones Hsp60 and Hsp10 are, according to recent reports, involved in cancer development and progression. We, for instance, have found that their expression varies with distinctive patterns in different malignancies: they are overexpressed in colorectal, exocervical and prostate carcinogenesis, and colorectal cancer progression, but they are downregulated during bronchial carcinogenesis. There is also evidence showing that Hsp60 and Hsp10 can be used as therapeutic agents, for example in rheumatoid arthritis. In view of these findings we want now to call attention to the potential of Hsp60 and Hsp10 in cancer therapy.


Asunto(s)
Chaperonina 10/uso terapéutico , Chaperonina 60/uso terapéutico , Neoplasias/tratamiento farmacológico , Humanos
16.
Med Sci (Paris) ; 23(2): 167-71, 2007 Feb.
Artículo en Francés | MEDLINE | ID: mdl-17291426

RESUMEN

In developed countries the incidence of autoimmune insulin-dependent or type 1 diabetes as the one of all autoimmune diseases has steadily increased over the last decades. Conventional therapy of type 1 diabetes is essentially palliative namely, chronic delivery of exogenous insulin that is associated with major constraints (multiple daily parenteral administration, serious risks linked to hypoglycemic episodes) and incomplete effectiveness in preventing severe degenerative complications. This explains the growing attention on modern therapeutic strategies using biological agents such as CD3 monoclonal antibodies that allow 'reprogramming' the immune system to restore self-tolerance to pancreatic beta cell antigens. This strategy which proved successful in the experimental setting has recently been translated to the clinic with very encouraging results. CD3 antibodies may represent a new category of drugs affording a real cure for autoimmunity namely, inhibiting the pathogenic immune response while preserving the host reactivity to unrelated antigens.


Asunto(s)
Autoantígenos/uso terapéutico , Enfermedades Autoinmunes/terapia , Diabetes Mellitus Tipo 1/terapia , Inmunoterapia/métodos , Autotolerancia/inmunología , Adulto , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Complejo CD3/inmunología , Chaperonina 60/inmunología , Chaperonina 60/uso terapéutico , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Supresión Clonal , Diabetes Mellitus Tipo 1/inmunología , Glutamato Descarboxilasa/inmunología , Glutamato Descarboxilasa/uso terapéutico , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/uso terapéutico , Recién Nacido , Insulina/inmunología , Insulina/uso terapéutico , Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD , Muromonab-CD3/inmunología , Muromonab-CD3/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Subgrupos de Linfocitos T/inmunología , Timectomía/efectos adversos
17.
Diabetes ; 45(9): 1168-72, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8772717

RESUMEN

The development of autoimmune diabetes in the NOD strain of mice (H-2g7) is marked by the presence of T-cells reactive to the p277 peptide of the 6O-kDa heat shock protein (hsp60). We have found that the p277 peptide can be used as a therapeutic vaccine to arrest NOD diabetes. Recently, we found that T-cell autoimmunity to p277 also develops spontaneously in C57BL/KsJ mice (H-2d) during the induction of autoimmune diabetes by a very low dose of the beta-cell toxin streptozotocin (STZ). We now report the inhibition of STZ toxin-induced autoimmune diabetes by p277 peptide therapy. Administration of two doses each of 100 micrograms of peptide p277 in mineral oil given 1 week after toxin induction and 85 days later was most effective. The effect of p277 on STZ toxin-induced diabetes was marked by a shift in p277 autoimmunity from a T-cell proliferative response to the production of anti-p277 antibodies. The anti-p277 antibodies were predominantly of the IgG1 and IgG2b isotypes, known to be regulated by Th2 type cytokines; IgG2a antibody, known to be dependent on interferon (IFN)-gamma, was induced to a much lesser degree. Peptide p277 therapy was specific: treatment of the mice with an immunogenic peptide from the sequence of another antigen, GADp34, failed to prevent the development of diabetes. The GADp34 peptide induced lower titers of specific antibodies, and the antibodies were predominantly of the IgG2a class. Thus, p277 peptide therapy, marked by the induction of Th2-type antibodies, can be effective in toxin-induced autoimmune diabetes.


Asunto(s)
Chaperonina 60/uso terapéutico , Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 1/prevención & control , Hipoglucemiantes/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Autoanticuerpos/clasificación , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inmunología , Relación Dosis-Respuesta a Droga , Glutamato Descarboxilasa/inmunología , Inmunoglobulina G/clasificación , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Estreptozocina/toxicidad , Linfocitos T/efectos de los fármacos , Factores de Tiempo
18.
Diabetes Care ; 38(6): 997-1007, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25998292

RESUMEN

Over the past three decades there have been a number of clinical trials directed at interdicting the type 1 diabetes (T1D) disease process in an attempt to prevent the development of the disease in those at increased risk or to stabilize-potentially even reverse-the disease in people with T1D, usually of recent onset. Unfortunately, to date there has been no prevention trial that has resulted in delay or prevention of T1D. And, trials in people with T1D have had mixed results with some showing promise with at least transient improvement in ß-cell function compared with randomized control groups, while others have failed to slow the decline in ß-cell function when compared with placebo. This Perspective will assess the past and present challenges in this effort and provide an outline for potential future opportunities.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Compuestos de Alumbre/uso terapéutico , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos/uso terapéutico , Suero Antilinfocítico/uso terapéutico , Vacuna BCG/uso terapéutico , Chaperonina 60/uso terapéutico , Diabetes Mellitus Tipo 1/fisiopatología , Predicción , Péptido 1 Similar al Glucagón/uso terapéutico , Humanos , Hipoglucemiantes/uso terapéutico , Células Secretoras de Insulina/fisiología , Interleucina-2/uso terapéutico , Ratones , Fragmentos de Péptidos/uso terapéutico , Proyectos Piloto , Ensayos Clínicos Controlados Aleatorios como Asunto , Proyectos de Investigación , Resultado del Tratamiento
19.
J Immunother ; 38(9): 341-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26448578

RESUMEN

Gliomas are the most common type of brain tumor with poor prognosis. Even after combination treatments including surgery, radiation, and chemotherapy, the median survival is around 15 months, calling for novel approaches such as immunotherapy. To develop novel therapeutic approaches, we tried to prepare a candidate vaccine by mixing the recombinant mycobacterial heat-shock protein 65 (HSP65) with GL261 glioma tissue lysate (GTL). Our data showed that HSP65-GTL induced potent cytotoxic T lymphocyte and prolonged the survival of mice bearing GL261 gliomas. Furthermore, HSP65 or HSP65-GTL upregulated mRNA expressions of RORγt and interleukin-17A in spleen cells or draining lymph node cells, respectively, and enhanced the ratios of brain-infiltrating Th17 cells and inflammatory cells, indicating that the antitumor effect of HSP65-GTL was associated with Th17-type immunity.


Asunto(s)
Proteínas Bacterianas , Vacunas contra el Cáncer , Chaperonina 60 , Glioma/inmunología , Células Th17/inmunología , Animales , Antígenos de Neoplasias/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/uso terapéutico , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral , Chaperonina 60/genética , Chaperonina 60/inmunología , Chaperonina 60/uso terapéutico , Citotoxicidad Inmunológica , Femenino , Glioma/metabolismo , Glioma/terapia , Inmunoterapia , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico
20.
Hum Vaccin Immunother ; 10(4): 838-46, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24165565

RESUMEN

Type 1 diabetes (T1D) results from autoimmune destruction of the pancreatic ß-cells. Current T1D therapies are exclusively focused on regulating glycemia rather than the underlying immune response. A handful of trials have sought to alter the clinical course of T1D using various broad immune-suppressors, e.g., cyclosporine A and azathioprine.(1-3) The effect on ß-cell preservation was significant, however, these therapies were associated with unacceptable side-effects. In contrast, more recent immunomodulators, such as anti-CD3 and antigenic therapies such as DiaPep277, provide a more targeted immunomodulation and have been generally well-tolerated and safe; however, as a monotherapy there appear to be limitations in terms of therapeutic benefit. Therefore, we argue that this new generation of immune-modifying agents will likely work best as part of a combination therapy. This review will summarize current immune-modulating therapies under investigation and discuss how to move the field of immunotherapy in T1D forward.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Factores Inmunológicos/uso terapéutico , Complejo CD3/inmunología , Chaperonina 60/uso terapéutico , Quimioterapia Combinada/métodos , Humanos , Insulina/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA