Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Genet Metab ; 137(1-2): 153-163, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36049366

RESUMEN

BACKGROUND: Arginase 1 Deficiency (ARG1-D) is a rare, progressive, metabolic disorder that is characterized by devastating manifestations driven by elevated plasma arginine levels. It typically presents in early childhood with spasticity (predominately affecting the lower limbs), mobility impairment, seizures, developmental delay, and intellectual disability. This systematic review aims to identify and describe the published evidence outlining the epidemiology, diagnosis methods, measures of disease progression, clinical management, and outcomes for ARG1-D patients. METHODS: A comprehensive literature search across multiple databases such as MEDLINE, Embase, and a review of clinical studies in ClinicalTrials.gov (with results reported) was carried out per PRISMA guidelines on 20 April 2020 with no date restriction. Pre-defined eligibility criteria were used to identify studies with data specific to patients with ARG1-D. Two independent reviewers screened records and extracted data from included studies. Quality was assessed using the modified Newcastle-Ottawa Scale for non-comparative studies. RESULTS: Overall, 55 records reporting 40 completed studies and 3 ongoing studies were included. Ten studies reported the prevalence of ARG1-D in the general population, with a median of 1 in 1,000,000. Frequently reported diagnostic methods included genetic testing, plasma arginine levels, and red blood cell arginase activity. However, routine newborn screening is not universally available, and lack of disease awareness may prevent early diagnosis or lead to misdiagnosis, as the disease has overlapping symptomology with other diseases, such as cerebral palsy. Common manifestations reported at time of diagnosis and assessed for disease progression included spasticity (predominately affecting the lower limbs), mobility impairment, developmental delay, intellectual disability, and seizures. Severe dietary protein restriction, essential amino acid supplementation, and nitrogen scavenger administration were the most commonly reported treatments among patients with ARG1-D. Only a few studies reported meaningful clinical outcomes of these interventions on intellectual disability, motor function and adaptive behavior assessment, hospitalization, or death. The overall quality of included studies was assessed as good according to the Newcastle-Ottawa Scale. CONCLUSIONS: Although ARG1-D is a rare disease, published evidence demonstrates a high burden of disease for patients. The current standard of care is ineffective at preventing disease progression. There remains a clear need for new treatment options as well as improved access to diagnostics and disease awareness to detect and initiate treatment before the onset of clinical manifestations to potentially enable more normal development, improve symptomatology, or prevent disease progression.


Asunto(s)
Hiperargininemia , Discapacidad Intelectual , Recién Nacido , Humanos , Preescolar , Arginasa/genética , Hiperargininemia/diagnóstico , Hiperargininemia/epidemiología , Hiperargininemia/genética , Convulsiones/diagnóstico , Convulsiones/epidemiología , Convulsiones/etiología , Espasticidad Muscular/diagnóstico , Espasticidad Muscular/epidemiología , Espasticidad Muscular/genética , Arginina/uso terapéutico , Aminoácidos Esenciales , Progresión de la Enfermedad , Nitrógeno
2.
EBioMedicine ; 79: 103974, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35430453

RESUMEN

BACKGROUND: Rheumatoid arthritis (RA) is a progressive degenerative disorder that leads to joint destruction. Available treatments only target the inflammatory component with minimal impact on joint repair. We recently uncovered a previously unappreciated family of pro-resolving mediators, the maresin conjugate in tissue regeneration (MCTR), that display both immunoregulatory and tissue-protective activities. Thus, we queried whether the production of these autacoids is disrupted in RA patients and whether they can be useful in treating joint inflammation and promoting joint repair. METHODS: Using a highly phenotyped RA cohort we evaluated plasma MCTR concentrations and correlated these to clinical markers of disease activity. To evaluate the immunoregulatory and tissue reparative activities we employed both in vivo models of arthritis and organ culture models. FINDINGS: Herein, we observed that plasma MCTR3 concentrations were negatively correlated with joint disease activity and severity in RA patients. Evaluation of the mechanisms engaged by this mediator in arthritic mice demonstrated that MCTR3 reprograms monocytes to confer enduring joint protective properties. Single cell transcriptomic profiling and flow cytometric evaluation of macrophages from mice treated with MCTR3-reprogrammed monocytes revealed a role for Arginase-1 (Arg-1) in mediating their joint reparative and pro-resolving activities. Arg-1 inhibition reversed both the anti-arthritic and tissue reparative actions of MCTR3-reprogrammed monocytes. INTERPRETATION: Our findings demonstrate that circulating MCTR3 levels are negatively correlated with disease in RA. When administered to mice in vivo, MCTR3 displayed both anti-inflammatory and joint reparative activities, protecting both cartilage and bone in murine arthritis. These activities were, at least in part, mediated via the reprogramming of mononuclear phagocyte responses. FUNDING: This work was supported by funding from the European Research Council (ERC) under the European Union's Horizon 2020 research and innovation programme (grant no: 677542) and the Barts Charity (grant no: MGU0343) to J.D. J.D. is also supported by a Sir Henry Dale Fellowship jointly funded by the Wellcome Trust and the Royal Society (grant 107613/Z/15/Z).


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Animales , Antiinflamatorios/farmacología , Arginasa/genética , Artritis Reumatoide/tratamiento farmacológico , Humanos , Macrófagos , Ratones , Monocitos
3.
Cancer Res ; 81(19): 5047-5059, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34301764

RESUMEN

Immune cells regulate tumor growth by mirroring their function as tissue repair organizers in normal tissues. To understand the different facets of immune-tumor collaboration through genetics, spatial transcriptomics, and immunologic manipulation with noninvasive, longitudinal imaging, we generated a penetrant double oncogene-driven autochthonous model of neuroblastoma. Spatial transcriptomic analysis showed that CD4+ and myeloid populations colocalized within the tumor parenchyma, while CD8+ T cells and B cells were peripherally dispersed. Depletion of CD4+ T cells or CCR2+ macrophages, but not B cells, CD8+ T cells, or natural killer (NK) cells, prevented tumor formation. Tumor CD4+ T cells displayed unconventional phenotypes and were clonotypically diverse and antigen independent. Within the myeloid fraction, tumor growth required myeloid cells expressing arginase-1. Overall, these results demonstrate how arginine-metabolizing myeloid cells conspire with pathogenic CD4+ T cells to create permissive conditions for tumor formation, suggesting that these protumorigenic pathways could be disabled by targeting myeloid arginine metabolism. SIGNIFICANCE: A new model of human neuroblastoma provides ways to track tumor formation and expansion in living animals, allowing identification of CD4+ T-cell and macrophage functions required for oncogenesis.


Asunto(s)
Arginasa/genética , Linfocitos T CD4-Positivos/metabolismo , Susceptibilidad a Enfermedades , Células Mieloides/metabolismo , Neuroblastoma/etiología , Neuroblastoma/metabolismo , Animales , Arginasa/metabolismo , Biomarcadores , Células de la Médula Ósea/metabolismo , Linfocitos T CD4-Positivos/inmunología , Línea Celular Tumoral , Biología Computacional/métodos , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Neuroblastoma/patología , Oncogenes , Análisis de la Célula Individual , Transcriptoma
4.
Front Immunol ; 12: 661493, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34025661

RESUMEN

Multiple sclerosis (MS) is a chronic immune-mediated disease characterized by demyelination and neuroaxonal damage in the central nervous system. The etiology is complex and is still not fully understood. Accumulating evidence suggests that our gut microbiota and its metabolites influence the MS pathogenesis. Short-chain fatty acids (SCFAs), such as acetate, propionate and butyrate, are metabolites produced by gut microbiota through fermentation of indigestible carbohydrates. SCFAs and kynurenine metabolites have been shown to have important immunomodulatory properties, and propionate supplementation in MS patients has been associated with long-term clinical improvement. However, the underlying mechanisms of action and its importance in MS remain incompletely understood. We analyzed serum levels of SCFAs and performed targeted metabolomics in relation to biomarkers of inflammation, and clinical and MRI measures in newly diagnosed patients with relapsing-remitting MS before their first disease modifying therapy and healthy controls (HCs). We demonstrated that serum acetate levels were nominally reduced in MS patients compared with HCs. The ratios of acetate/butyrate and acetate/(propionate + butyrate) were significantly lower in MS patients in a multivariate analysis (orthogonal partial least squares discriminant analysis; OPLS-DA). The mentioned ratios and acetate levels correlated negatively with the pro-inflammatory biomarker IFNG, indicating an inverse relation between acetate and inflammation. In contrast, the proportion of butyrate was found higher in MS patients in the multivariate analysis, and both butyrate and valerate correlated positively with proinflammatory cytokines (IFNG and TNF), suggesting complex bidirectional regulatory properties of SCFAs. Branched SCFAs were inversely correlated with clinical disability, at a nominal significance level. Otherwise SCFAs did not correlate with clinical variables or MRI measures. There were signs of an alteration of the kynurenine pathway in MS, and butyrate was positively correlated with the immunomodulatory metabolite 3-hydroxyanthranilic acid. Other variables that influenced the separation between MS and HCs were NfL, ARG1 and IL1R1, D-ribose 5-phosphate, pantothenic acid and D-glucuronic acid. In conclusion, we provide novel results in this rapidly evolving field, emphasizing the complexity of the interactions between SCFAs and inflammation; therefore, further studies are required to clarify these issues before supplementation of SCFAs can be widely recommended.


Asunto(s)
Ácidos Grasos Volátiles/sangre , Voluntarios Sanos , Inflamación/sangre , Esclerosis Múltiple Recurrente-Remitente/sangre , Adulto , Arginasa/genética , Encéfalo/diagnóstico por imagen , Estudios Transversales , Citocinas/genética , Femenino , Expresión Génica , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Inflamación/genética , Inflamación/metabolismo , Imagen por Resonancia Magnética , Masculino , Metabolómica/métodos , Esclerosis Múltiple Recurrente-Remitente/genética , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Receptores de Hidrocarburo de Aril/genética , Médula Espinal/diagnóstico por imagen
5.
Oxid Med Cell Longev ; 2021: 6628957, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33824696

RESUMEN

BACKGROUND: Doxorubicin (DOX) is a commonly used chemotherapeutic drug but is limited in clinical applications by its cardiotoxicity. Neiguan acupoint (PC6) is a well-recognized acupoint for the treatment of cardiothoracic disease. However, whether acupuncture at PC6 could be effective in preventing DOX-induced cardiotoxicity is still unknown. METHODS: A set of experiments were performed with myocardial cells, wild type, inducible nitric oxide synthase knockout (iNOS-/-), and myocardial-specific ablation arginase 2 (Myh6-ARG 2-/-) mice. We investigated the protective effect and the underlying mechanisms for electroacupuncture (EA) against DOX-induced cardiotoxicity by echocardiography, immunostaining, biochemical analysis, and molecular biotechnology in vivo and in vitro analysis. RESULTS: We found that DOX-mediated nitric oxide (NO) production was positively correlated with the iNOS level but has a negative correlation with the arginase 2 (ARG 2) level in both myocardial cells and tissues. Meanwhile, EA at PC6 alleviated cardiac dysfunction and cardiac hypertrophy in DOX-treated mice. EA at PC6 blocked the upregulation of NO production in accompanied with the downregulated iNOS and upregulated ARG 2 levels in myocardial tissue induced by DOX. Furthermore, knockout iNOS prevented cardiotoxicity and EA treatment did not cause the further improvement of cardiac function in iNOS-/- mice treated by DOX. In contrast, deficiency of myocardial ARG 2 aggravated DOX-induced cardiotoxicity and reduced EA protective effect. CONCLUSION: These results suggest that EA treatment at PC6 can prevent DOX-induced cardiotoxicity through modulating NO production by modulating the iNOS/ARG 2 balance in myocardial cells.


Asunto(s)
Antineoplásicos/toxicidad , Arginasa/metabolismo , Doxorrubicina/toxicidad , Electroacupuntura/métodos , Cardiopatías/prevención & control , Óxido Nítrico Sintasa de Tipo II/metabolismo , Puntos de Acupuntura , Animales , Arginasa/genética , Cardiotoxicidad/etiología , Cardiotoxicidad/parasitología , Cardiopatías/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Transducción de Señal
6.
J Ethnopharmacol ; 273: 113998, 2021 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-33689799

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Sophora alopecuroides Linn. (Leguminosae) has been largely used in traditional folk medicine in China as an anti-inflammatory agent and to treat various skin wounds, including sore furunculosis and ulcer (a common type of non-healing wound). The present study aimed to evaluate the effects of S. alopecuroides gel on skin wound healing in rats. MATERIALS AND METHODS: When the rats were anesthetized, full-thickness skin wound was performed on dorsal area by using biopsy punch with 8 mm diameter. Then, wounds received treatment with different doses of S. alopecuroides gel (1.25%, 2.5% and 5%, w/w) once a day with the gel base used as vehicle control and rb-FGF as positive control. Every five animals were sacrificed after 7, 12 days after surgery for histopathology and relevant biochemical indexes analysis. Besides, after RAW 264.7 cells exposure to LPS (1 µg/ml) with or without total extract (25 and 50 µg/ml) for 24 h, the culture supernatant was used for detection of IL-1ß and TNF-α levels using ELISA kits and the protein lysate for western blot analysis. RESULTS: A remarkable wound closure was observed after administration with 5% S. alopecuroides gel with the wound area of 30% and 8.5% as compared to 42% and 19% in the control group on day 7 and 12, respectively. Histological and immunostaining analysis for the wound tissues also revealed that S. alopecuroides promoted the growth of granulation tissue, collagen deposition, cell proliferation and angiogenesis. Meanwhile, it was able to ameliorate inflammatory response and promote the production of TGF-ß. In addition, we also demonstrated that S. alopecuroides inhibited the release of inflammatory mediators and expression of iNOS as well as up-regulated the expression of Arg-1 in LPS-triggered RAW 264.7 cells. CONCLUSIONS: The present study confirmed that S. alopecuroides had a great potential for accelerated wound healing by regulating the over expression of inflammatory response for the first time and provided theoretical basis for the traditional use. It can be used as candidate drug for the treantment of chronic non-healing wounds.


Asunto(s)
Sophora/química , Cicatrización de Heridas/efectos de los fármacos , Animales , Arginasa/genética , Arginasa/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fitoterapia , Células RAW 264.7 , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos
8.
J Nutr ; 151(4): 763-771, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33232463

RESUMEN

BACKGROUND: Genetic variation in arginase may underlie variability in whole blood l-arginine concentrations in unsupplemented and l-arginine-supplemented adults. OBJECTIVES: We aimed to study whether single nucleotide polymorphisms (SNPs) in the arginase 1 (ARG1) and arginase 2 (ARG2) genes are associated with blood l-arginine concentrations in unsupplemented and l-arginine-supplemented individuals. METHODS: In 374 adults (mean ± SD age: 59.6 ± 14.6 y; 180 males), we analyzed SNPs in the ARG1 (rs2246012 and rs2781667) and ARG2 genes (rs3742879 and rs2759757) and their associations with blood l-arginine concentrations. We analyzed associations of haplotypes for the ARG1 gene and for the ARG1 and ARG2 genes combined with blood l-arginine concentrations in supplement users and unsupplemented participants. RESULTS: Of study participants, 120 had low (<42 µmol/L), 133 had medium (42-114 µmol/L), and 121 had high blood l-arginine concentrations (>114 µmol/L); 58 individuals were current l-arginine supplement users. We found a significantly higher prevalence of the minor allele of ARG1 rs2246012 in supplement users with higher blood l-arginine concentrations (P = 0.03). Mean ± SEM l-arginine concentration was 263 ± 9.76 µmol/L in supplement users homozygous for the minor allele of ARG1 rs2246012 (P = 0.004); it was 70.4 ± 25.6 µmol/L in unsupplemented participants homozygous for the minor allele of ARG2 rs3759757 (P = 0.03). The ARG1 haplotype was significantly associated with blood l-arginine concentrations in supplement users (P = 0.046), whereas the combined ARG1/ARG2 haplotype was significantly associated with blood l-arginine concentrations in the cohort as a whole (P = 0.012). CONCLUSIONS: Genetic variability in the ARG1 and ARG2 genes is associated with blood l-arginine concentrations in humans: ARG1 is associated with blood l-arginine concentrations in l-arginine supplement users, whereas ARG2 is associated with blood l-arginine concentrations in unsupplemented participants. Our study is the first to describe a possible functional relation between ARG1 and ARG2 SNPs and blood l-arginine concentrations; genetic variability in ARG1 may explain variation in blood l-arginine concentrations during supplement use and discrepant study results.


Asunto(s)
Arginasa/genética , Arginina/administración & dosificación , Arginina/sangre , Polimorfismo de Nucleótido Simple , Adulto , Anciano , Suplementos Dietéticos , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Haplotipos , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
9.
J Cell Physiol ; 235(12): 9974-9991, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32458472

RESUMEN

Chronic venous ulcer (CVU) is a major cause of chronic wounds of lower extremities and presents a significant financial and resource burden to health care systems worldwide. Defects in the vasculature, matrix deposition, and re-epithelialization are the main histopathological changes believed to impede healing. Supplementation of the amino acid arginine that plays a crucial role in the interactions that occur during inflammation and wound healing was proven clinically to improve acute wound healing probably through enhancing activity of inducible arginase (AI) locally in the wounds. However, the possible mechanism of arginine action and the potential beneficial effects of AI/arginine in human chronic wounds remain unclear. In the present study, using biopsies, taken under local anesthesia, from adult patients (n = 12, mean age 55 years old) with CVUs in lower extremities, we investigated the correlation between AI distribution in CVUs and the histopathological changes, mainly proliferative and vascular changes. Our results show a distinct spatial distribution of AI along the ulcer in the epidermis and in the dermis with the highest level of expression being at the ulcer edge and the least expression towards the ulcer base. The AI cellular immunoreactivity, enzymatic activity, and protein levels were significantly increased towards the ulcer edge. Interestingly, a similar pattern of expression was encountered in the proliferative and the vascular changes with strong correlations between AI and the proliferative activity and vascular changes. Furthermore, AI cellular distribution was associated with increased proliferative activity, inflammation, and vascular changes. Our findings of differential expression of AI along the CVU base, edge, and nearby surrounding skin and its associations with increased proliferative activity and vascular changes provide further support to the AI implication in CVU pathogenesis. The presence of high levels of AI in the epidermis of chronic wounds may serve as a molecular marker of impaired healing and may provide future targets for therapeutic intervention.


Asunto(s)
Arginasa/genética , Úlcera de la Pierna/genética , Isoformas de Proteínas/genética , Úlcera Varicosa/genética , Arginina/metabolismo , Enfermedad Crónica/prevención & control , Femenino , Humanos , Úlcera de la Pierna/fisiopatología , Masculino , Persona de Mediana Edad , Óxido Nítrico Sintasa/genética , Piel/metabolismo , Piel/patología , Úlcera Varicosa/fisiopatología , Venas/metabolismo , Venas/patología , Cicatrización de Heridas/genética
10.
Neuromolecular Med ; 22(3): 341-358, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32048142

RESUMEN

Alzheimer's disease (AD) is a common neurodegenerative disease that is always accompanied by synaptic loss in the brain. Safflower yellow (SY) is the extract of safflower, a traditional Chinese medicine, which has shown neuroprotective effects in recent studies. However, the mechanism of SY in protecting synapses remains unclear. In this study, we are going to study the mechanism of how SY treats AD in terms of synaptic plasticity. We found, via behavioral experiments, that SY treatment could improve the abilities of learning and memory in APP/PS1 mice. In addition, using Golgi staining and HE staining, we found that SY treatment could reduce the loss of dendritic spines in the pathological condition and could maintain the normal physiological state of the cells in cortex and in hippocampus. In addition, the results of immunofluorescence staining and western blotting showed that SY treatment could significantly increase the expression of synapse-related proteins. Moreover, after being treated with SY, the expression of iNOS (marker of M1 microglia) declined remarkably, and the level of Arginase-1 (marker of M2 microglia) increased significantly. Finally, we found BDNF/TrkB/ERK signaling cascade was activated. These results indicate that SY enhances synaptic plasticity in APP/PS1 mice by regulating microglia activation phenotypes and BDNF/TrkB/ERK signaling pathway.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Factor Neurotrófico Derivado del Encéfalo/fisiología , Chalcona/análogos & derivados , Medicamentos Herbarios Chinos/uso terapéutico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Glicoproteínas de Membrana/fisiología , Microglía/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Fitoterapia , Proteínas Tirosina Quinasas/fisiología , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Arginasa/biosíntesis , Arginasa/genética , Corteza Cerebral/química , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Chalcona/uso terapéutico , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/ultraestructura , Modelos Animales de Enfermedad , Donepezilo/farmacología , Donepezilo/uso terapéutico , Inducción Enzimática/efectos de los fármacos , Reacción de Fuga/efectos de los fármacos , Femenino , Hipocampo/química , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Memoria a Largo Plazo/efectos de los fármacos , Memoria a Corto Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/fisiología , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/fisiología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Presenilina-1/genética , Distribución Aleatoria
11.
Front Immunol ; 11: 582998, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33519806

RESUMEN

Alzheimer's disease (AD) includes several hallmarks comprised of amyloid-ß (Aß) deposition, tau neuropathology, inflammation, and memory impairment. Brain metabolism becomes uncoupled due to aging and other AD risk factors, which ultimately lead to impaired protein clearance and aggregation. Increasing evidence indicates a role of arginine metabolism in AD, where arginases are key enzymes in neurons and glia capable of depleting arginine and producing ornithine and polyamines. However, currently, it remains unknown if the reduction of arginase 1 (Arg1) in myeloid cell impacts amyloidosis. Herein, we produced haploinsufficiency of Arg1 by the hemizygous deletion in myeloid cells using Arg1fl/fl and LysMcreTg/+ mice crossed with APP Tg2576 mice. Our data indicated that Arg1 haploinsufficiency promoted Aß deposition, exacerbated some behavioral impairment, and decreased components of Ragulator-Rag complex involved in mechanistic target of rapamycin complex 1 (mTORC1) signaling and autophagy. Additionally, Arg1 repression and arginine supplementation both impaired microglial phagocytosis in vitro. These data suggest that proper function of Arg1 and arginine metabolism in myeloid cells remains essential to restrict amyloidosis.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Amiloidosis/metabolismo , Arginasa/metabolismo , Déficit de la Atención y Trastornos de Conducta Disruptiva/metabolismo , Células Mieloides/fisiología , Animales , Arginasa/genética , Autofagia , Conducta Animal , Modelos Animales de Enfermedad , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Transgénicos , Inflamación Neurogénica , Transducción de Señal
12.
J Clin Invest ; 129(7): 2669-2684, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30990796

RESUMEN

The polarization of macrophages is regulated by transcription factors such as nuclear factor kappa B (NF-κB) and activator protein 1 (AP-1). In this manuscript, we delineated the role of the transcription factor Fos-related antigen 1 (Fra-1) during macrophage activation and development of arthritis. Network level interaction analysis of microarray data derived from Fra-1- or Fra-2-deficient macrophages revealed a central role of Fra-1, but not of Fra-2 in orchestrating the expression of genes related to wound response, toll-like receptor activation and interleukin signaling. Chromatin-immunoprecipitation (ChIP)-sequencing and standard ChIP analyses of macrophages identified arginase 1 (Arg1) as a target of Fra-1. Luciferase reporter assays revealed that Fra-1 down-regulated Arg1 expression by direct binding to the promoter region. Using macrophage-specific Fra-1- or Fra-2- deficient mice, we observed an enhanced expression and activity of Arg1 and a reduction of arthritis in the absence of Fra-1, but not of Fra-2. This phenotype was reversed by treatment with the arginase inhibitor Nω-hydroxy-nor-L-arginine, while ʟ-arginine supplementation increased arginase activity and alleviated arthritis, supporting the notion that reduced arthritis in macrophage-specific Fra-1-deficient mice resulted from enhanced Arg1 expression and activity. Moreover, patients with active RA showed increased Fra-1 expression in the peripheral blood and elevated Fra-1 protein in synovial macrophages compared to RA patients in remission. In addition, the Fra-1/ARG1 ratio in synovial macrophages was related to RA disease activity. In conclusion, these data suggest that Fra-1 orchestrates the inflammatory state of macrophages by inhibition of Arg1 expression and thereby impedes the resolution of inflammation.


Asunto(s)
Arginasa/biosíntesis , Artritis Reumatoide , Regulación Enzimológica de la Expresión Génica , Macrófagos/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Membrana Sinovial/metabolismo , Animales , Arginasa/genética , Antígeno 2 Relacionado con Fos/genética , Antígeno 2 Relacionado con Fos/metabolismo , Humanos , Macrófagos/patología , Masculino , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-fos/genética , Membrana Sinovial/patología
13.
Exp Mol Med ; 50(11): 1-10, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30397194

RESUMEN

Bone marrow-derived mesenchymal stem cells (BMMSCs) are used extensively for cardiac repair and interact with immune cells in the damaged heart. Macrophages are known to be modulated by stem cells, and we hypothesized that priming macrophages with BMMSCs would enhance their therapeutic efficacy. Rat bone marrow-derived macrophages (BMDMs) were stimulated by lipopolysaccharide (LPS) with or without coculture with rat BMCs. In the LPS-stimulated BMDMs, induction of the inflammatory marker iNOS was attenuated, and the anti-inflammatory marker Arg1 was markedly upregulated by coculture with BMMSCs. Myocardial infarction (MI) was induced in rats. One group was injected with BMMSCs, and a second group was injected with MIX (a mixture of BMMSCs and BMDMs after coculture). The reduction in cardiac fibrosis was greater in the MIX group than in the BMC group. Cardiac function was improved in the BMMSC group and was substantially improved in the MIX group. Angiogenesis was better in the MIX group, and anti-inflammatory macrophages were more abundant in the MIX group than in the BMMSC group. In the BMMSCs, interferon regulatory factor 5 (IRF5) was exclusively induced by coculture with macrophages. IRF5 knockdown in BMMSCs failed to suppress inflammatory marker induction in the macrophages. In this study, we demonstrated the successful application of BMDMs primed with BMMSCs as an adjuvant to cell therapy for cardiac repair.


Asunto(s)
Macrófagos/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Infarto del Miocardio/terapia , Animales , Arginasa/genética , Arginasa/metabolismo , Línea Celular , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Ratas Sprague-Dawley
14.
Metallomics ; 10(10): 1501-1509, 2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30206632

RESUMEN

Microglia, the resident immune cells of the central nervous system, can display a pro-inflammatory M1 phenotype or an anti-inflammatory M2 phenotype. Arginase (Arg)-1 expressed in interleukin-4 (IL-4)-induced M2 microglia reduces nitric oxide (NO) production by competing with inducible NO synthase for l-arginine, which contributes to the attenuation of brain inflammation. Although previous studies have indicated that brain zinc promotes M1 activation, the effect of zinc on M2 microglial activation remains to be determined. In the present study, murine primary microglia treated with 10 ng mL-1 IL-4 exhibited increased Arg-1 mRNA expression and levels of intracellular free zinc. Chelation of this increased intracellular free zinc by the cell permeable zinc chelator N,N,N',N'-tetrakis-(2-pyridylmethyl)ethylenediamine (TPEN) aggravated the IL-4-induced mRNA expression and enzymatic activity of Arg-1. However, the cell impermeable zinc chelator CaEDTA had no effect on Arg-1 expression or cytosolic levels of free zinc in IL-4-induced M2-polarized microglia. Furthermore, treatment with IL-4 resulted in upregulation of phagocytic activity in microglia, while administration of TPEN abolished IL-4-induced phagocytic activity. Moreover, this effect was reversed vial-arginine supplementation. These findings suggest that IL-4 induces an increase in intracellular free zinc in microglia, which may act as a negative regulator of IL-4-induced Arg-1 expression, and that such negative regulation is essential for microglial phagocytic activity.


Asunto(s)
Arginasa/metabolismo , Regulación de la Expresión Génica , Interleucina-4/metabolismo , Microglía/efectos de los fármacos , Zinc/farmacología , Animales , Arginasa/genética , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo
15.
Mult Scler Relat Disord ; 25: 5-13, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30014878

RESUMEN

Teminalia chebula (TC) has been traditionally used in the Iranian traditional medicine (ITM) and Ayurvedic medicine primarily for neurologic disorders and inflammation. Mainly, its fruits have been applied for CNS disorders. The effects of Terminalia chebula as herbal medicine with anti-inflammatory and anti-oxidant properties were aimed on lipopolysaccharide (LPS)-induced microglial inflammation. Cytotoxicity of TC extract (0-80) µg/ml on microglial cells was evaluated using the MTT assay. Also, the protective effect of TC extract concentrations with specified amount of LPS-induced mice microglial cells was studied. The concentrations of TNF-α (Tumor Necrosis Factor-α), IL-1ß (Interleukin-1ß), IL-6 and PGE-2 (Prostaglandin-E2) were evaluated using ELISA. Gene expression of TNF-α, IL-1ß, IL-6, COX-2 (Cyclooxygenase-2), iNOS and arginase-1 was also evaluated using the Real-Time PCR method. Nitrite oxide and urea were measured using biochemical methods. The studied concentrations of TC extract did not affect the viability of microglial cells but significantly protected the viability after treatment with LPS. The concentrations and expression levels of pro-inflammatory factors (TNF-α, IL-1ß, IL-6, PGE-2, COX-2) were significantly decreased after TC extract treatment in LPS-induced microglial cells with dose dependent manner. The extract also significantly decreased the levels of nitric oxide, increased urea and down regulated the expression of nitric oxide synthesis while arginase-1 expression was enhanced. Our results suggest that TC extract reduces inflammation in microglial cells and can be used as a potential anti-inflammatory agent in central nervous system inflammatory diseases.


Asunto(s)
Antiinflamatorios/farmacología , Polaridad Celular/efectos de los fármacos , Taninos Hidrolizables/farmacología , Microglía/efectos de los fármacos , Extractos Vegetales/farmacología , Terminalia/química , Animales , Arginasa/genética , Arginasa/metabolismo , Encéfalo/citología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Flavonoides/farmacología , Hidroxibenzoatos/farmacología , Lipopolisacáridos/toxicidad , Ratones , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN Mensajero/metabolismo
16.
RNA Biol ; 15(7): 914-922, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29923457

RESUMEN

Arginase I (ARG1) deficiency is an autosomal recessive urea cycle disorder, caused by deficiency of the enzyme Arginase I, resulting in accumulation of arginine in blood. Current Standard of Care (SOC) for ARG1 deficiency in patients or those having detrimental mutations of ARG1 gene is diet control. Despite diet and drug therapy with nitrogen scavengers, ~25% of patients suffer from severe mental deficits and loss of ambulation. 75% of patients whose symptoms can be managed through diet therapy continue to suffer neuro-cognitive deficits. In our research, we demonstrate in vitro and in vivo that administration of ARG1 mRNA increased ARG1 protein expression and specific activity in relevant cell types, including ARG1-deficient patient cell lines, as well as in wild type mice for up to 4 days. These studies demonstrate that ARG1 mRNA treatment led to increased functional protein expression of ARG1 and subsequently an increase in urea. Hence, ARG1 mRNA therapy could be a potential treatment option to develop for patients.


Asunto(s)
Arginasa/metabolismo , Arginina/metabolismo , Terapia Biológica/métodos , Hiperargininemia/terapia , ARN Mensajero/administración & dosificación , Animales , Arginasa/genética , Células HeLa , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Urea/metabolismo
17.
J Am Heart Assoc ; 7(5)2018 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-29478971

RESUMEN

BACKGROUND: Uninephrectomy (UNX) is performed for various reasons, including kidney cancer or donation. Kidneys being the main site of l-arginine production in the body, we tested whether UNX mediated kidney mass reduction impacts l-arginine metabolism and thereby nitric oxide production and blood pressure regulation in mice. METHODS AND RESULTS: In a first series of experiments, we observed a significant increase in arterial blood pressure 8 days post-UNX in female and not in male mice. Further experimental series were performed in female mice, and the blood pressure increase was confirmed by telemetry. l-citrulline, that is used in the kidney to produce l-arginine, was elevated post-UNX as was also asymmetric dimethylarginine, an inhibitor of nitric oxide synthase that competes with l-arginine and is a marker for renal failure. Interestingly, the UNX-induced blood pressure increase was prevented by supplementation of the diet with 5% of the l-arginine precursor, l-citrulline. Because l-arginine is metabolized in the kidney and other peripheral tissues by arginase-2, we tested whether the lack of this metabolic pathway also compensates for decreased l-arginine production in the kidney and/or for local nitric oxide synthase inhibition and consecutive blood pressure increase. Indeed, upon uninephrectomy, arginase-2 knockout mice (Arg-2-/-) neither displayed an increase in asymmetric dimethylarginine and l-citrulline plasma levels nor a significant increase in blood pressure. CONCLUSIONS: UNX leads to a small increase in blood pressure that is prevented by l-citrulline supplementation or arginase deficiency, 2 measures that appear to compensate for the impact of kidney mass reduction on l-arginine metabolism.


Asunto(s)
Arginina/metabolismo , Presión Sanguínea , Riñón/cirugía , Nefrectomía/efectos adversos , Animales , Arginasa/genética , Arginasa/metabolismo , Arginina/análogos & derivados , Arginina/sangre , Presión Sanguínea/efectos de los fármacos , Citrulina/administración & dosificación , Citrulina/sangre , Femenino , Riñón/metabolismo , Riñón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Tamaño de los Órganos
18.
J Immunol ; 200(1): 286-294, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29180488

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are major regulators of T cell responses in several pathological conditions. Whether MDSCs increase and influence T cell responses in temporary inflammation, such as after vaccine administration, is unknown. Using the rhesus macaque model, which is critical for late-stage vaccine testing, we demonstrate that monocytic (M)-MDSCs and polymorphonuclear (PMN)-MDSCs can be detected using several of the markers used in humans. However, whereas rhesus M-MDSCs lacked expression of CD33, PMN-MDSCs were identified as CD33+ low-density neutrophils. Importantly, both M-MDSCs and PMN-MDSCs showed suppression of T cell proliferation in vitro. The frequency of circulating MDSCs rapidly and transiently increased 24 h after vaccine administration. M-MDSCs infiltrated the vaccine injection site, but not vaccine-draining lymph nodes. This was accompanied by upregulation of genes relevant to MDSCs such as arginase-1, IDO1, PDL1, and IL-10 at the injection site. MDSCs may therefore play a role in locally maintaining immune balance during vaccine-induced inflammation.


Asunto(s)
Subtipo H10N8 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Células Supresoras de Origen Mieloide/inmunología , Neutrófilos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Linfocitos T/inmunología , Animales , Arginasa/genética , Antígeno B7-H1/genética , Proliferación Celular , Regulación de la Expresión Génica , Humanos , Tolerancia Inmunológica , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Interleucina-10/genética , Macaca mulatta , Análisis por Micromatrices , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo , Vacunación
19.
PLoS Negl Trop Dis ; 11(10): e0006025, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29073150

RESUMEN

BACKGROUND: Leishmania uses the amino acid L-arginine as a substrate for arginase, enzyme that produces urea and ornithine, last precursor of polyamine pathway. This pathway is used by the parasite to replicate and it is essential to establish the infection in the mammalian host. L-arginine is not synthesized by the parasite, so its uptake occurs through the amino acid permease 3 (AAP3). AAP3 is codified by two copies genes (5.1 and 4.7 copies), organized in tandem in the parasite genome. One copy presents the expression regulated by L-arginine availability. METHODOLOGY/PRINCIPAL FINDINGS: RNA-seq data revealed 14 amino acid transporters differentially expressed in the comparison of La-WT vs. La-arg- promastigotes and axenic amastigotes. The 5.1 and 4.7 aap3 transcripts were down-regulated in La-WT promastigotes vs. axenic amastigotes, and in La-WT vs. La-arg- promastigotes. In contrast, transcripts of other transporters were up-regulated in the same comparisons. The amount of 5.1 and 4.7 aap3 mRNA of intracellular amastigotes was also determined in sample preparations from macrophages, obtained from BALB/c and C57BL/6 mice and the human THP-1 lineage infected with La-WT or La-arg-, revealing that the genetic host background is also important. We also determined the aap3 mRNA and AAP3 protein amounts of promastigotes and axenic amastigotes in different environmental growth conditions, varying pH, temperature and L-arginine availability. Interestingly, the increase of temperature increased the AAP3 level in plasma membrane and consequently the L-arginine uptake, independently of pH and L-arginine availability. In addition, we demonstrated that besides the plasma membrane localization, AAP3 was also localized in the glycosome of L. amazonensis promastigotes and axenic amastigotes. CONCLUSIONS/SIGNIFICANCE: In this report, we described the differential transcriptional profiling of amino acids transporters from La-WT and La-arg- promastigotes and axenic amastigotes. We also showed the increased AAP3 levels under amino acid starvation or its decrease in L-arginine supplementation. The differential AAP3 expression was determined in the differentiation of promastigotes to amastigotes conditions, as well as the detection of AAP3 in the plasma membrane reflecting in the L-arginine uptake. Our data suggest that depending on the amino acid pool and arginase activity, Leishmania senses and could use an alternative route for the amino acid transport in response to stress signaling.


Asunto(s)
Sistemas de Transporte de Aminoácidos/clasificación , Sistemas de Transporte de Aminoácidos/metabolismo , Arginasa/metabolismo , Arginina/metabolismo , Leishmania/enzimología , Macrófagos/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Animales , Arginasa/genética , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Macrófagos/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células THP-1 , Transcriptoma
20.
Biochem Biophys Res Commun ; 486(2): 458-464, 2017 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-28315684

RESUMEN

Cerebral palsy (CP) is the most common childhood disability worldwide, yet biomarkers for predicting CP are lacking. By subjecting peripheral blood samples from 62 CP patients and 30 healthy controls to Affymetrix GeneChip® PrimeView™ HumanGene Expression Microarray analysis, we identified the novel biomarker B-cell lymphoma 6 (BCL6) as the most upregulated gene in the CP samples. Gastrodin is a traditional Chinese medicine and bioactive compound that promotes adductor angle release, as well as gross and fine motor performance by increasing Gross Motor Function Measure-66 and Fine Motor Function Measure-45 scores. Gastrodin upregulates the mRNA expression of Mgl2 and Mrc1, M2 macrophage markers, and arginase activity, an M2 polarization indicator, in murine RAW264.7 macrophages. Moreover, these effects were blocked by BCL6 siRNA, which also abrogated the protective effects of Gastrodin against hydrogen peroxide-induced apoptosis and death in RAW264.7 cells. Our work identified BCL6 as a novel biomarker for early prediction of CP. Moreover, we demonstrated that Gastrodin not only stimulated polarization toward M2-like macrophages, which promote tissue repair, but also rescued macrophages from oxidative stress, apoptosis and death by inducing BCL6 expression. BCL6-targeted therapeutic strategies have promise for improving motor performance in CP patients.


Asunto(s)
Antioxidantes/uso terapéutico , Alcoholes Bencílicos/uso terapéutico , Parálisis Cerebral/diagnóstico , Parálisis Cerebral/tratamiento farmacológico , Glucósidos/uso terapéutico , Macrófagos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-6/genética , Animales , Apoptosis/efectos de los fármacos , Arginasa/genética , Arginasa/metabolismo , Biomarcadores/metabolismo , Estudios de Casos y Controles , Caspasa 3/genética , Caspasa 3/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Parálisis Cerebral/genética , Parálisis Cerebral/patología , Preescolar , Femenino , Regulación de la Expresión Génica , Humanos , Peróxido de Hidrógeno/antagonistas & inhibidores , Peróxido de Hidrógeno/farmacología , Lactante , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-bcl-6/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Desempeño Psicomotor/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Inmunológicos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA