Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Immunology ; 164(3): 602-616, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34310708

RESUMEN

Asymmetric dimethylarginine (ADMA) is an endogenous nitric oxide synthase (NOS) inhibitor/uncoupler inducing vascular pathology. Vascular pathology is an important factor for the development and progression of CNS pathology of MS, yet the role of ADMA in MS remains elusive. Patients with multiple sclerosis (MS) are reported to have elevated blood levels of ADMA, and mice with experimental autoimmune encephalomyelitis (EAE, an animal model of MS) generated by auto-immunization of myelin oligodendrocyte glycoprotein (MOG) and blood-brain barrier (BBB) disruption by pertussis toxin also had increased blood ADMA levels in parallel with induction of clinical disease. To explore the role of ADMA in EAE pathogenesis, EAE mice were treated with a daily dose of ADMA. It is of special interest that ADMA treatment enhanced the BBB disruption in EAE mice and exacerbated the clinical and CNS disease of EAE. ADMA treatment also induced the BBB disruption and EAE disease in MOG-immunized mice even without pertussis toxin treatment, suggesting the role of ADMA in BBB dysfunction in EAE. T-cell polarization studies also documented that ADMA treatment promotes TH 1- and TH 17-mediated immune responses but without affecting Treg-mediated immune response in EAE mice as well as in in vitro T-cell culture. Taken together, these data, for the first time, document the vascular and immunopathogenic roles of ADMA in EAE, thus pointing to the potential of ADMA-mediated mechanism as a new target of potential therapy for MS.


Asunto(s)
Arginina/análogos & derivados , Barrera Hematoencefálica/patología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Animales , Arginina/metabolismo , Barrera Hematoencefálica/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Ratones , Esclerosis Múltiple/patología , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Glicoproteína Mielina-Oligodendrócito/inmunología , Toxina del Pertussis/administración & dosificación , Toxina del Pertussis/inmunología
2.
PLoS One ; 19(3): e0300095, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38427617

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0210248.].

3.
Front Pharmacol ; 14: 1304697, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38143504

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which initially surfaced in late 2019, often triggers severe pulmonary complications, encompassing various disease mechanisms such as intense lung inflammation, vascular dysfunction, and pulmonary embolism. Currently, however, there's no drug addressing all these mechanisms simultaneously. This study explored the multi-targeting potential of S-nitrosoglutathione (GSNO) and N6022, an inhibitor of GSNO reductase (GSNOR) on markers of inflammatory, vascular, and thrombotic diseases related to COVID-19-induced acute lung disease. For this, acute lung disease was induced in C57BL/6 mice through intranasal administration of recombinant SARS-CoV-2 spike protein S1 domain (SP-S1). The mice exhibited fever, body weight loss, and increased blood levels and lung expression of proinflammatory cytokines (e.g., TNF-α and IL-6) as well as increased vascular inflammation mediated by ICAM-1 and VCAM-1 and lung infiltration by immune cells (e.g., neutrophils, monocytes, and activated cytotoxic and helper T cells). Further, the mice exhibited increased lung hyperpermeability (lung Evans blue extravasation) leading to lung edema development as well as elevated blood coagulation factors (e.g., fibrinogen, thrombin, activated platelets, and von Willebrand factor) and lung fibrin deposition. Similar to the patients with COVID-19, male mice showed more severe disease than female mice, along with higher GSNOR expression in the lungs. Optimization of GSNO by treatment with exogenous GSNO or inhibition of GSNOR by N6022 (or GSNO knockout) protects against SP-S1-induced lung diseases in both genders. These findings provide evidence for the potential efficacies of GSNO and GSNOR inhibitors in addressing the multi-mechanistic nature of SARS-CoV-2 SP-associated acute-lung disease.

4.
Neuroscience ; 458: 31-42, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33493617

RESUMEN

Reactive aldehydes are generated as a toxic end-product of lipid peroxidation under inflammatory oxidative stress condition which is a well-established phenomenon in the pathogenesis of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Alda-1, a selective agonist of mitochondrial aldehyde dehydrogenase 2 (ALDH2), is known to detoxify the reactive aldehydes. In this study, we investigated the effect of Alda-1 on CNS myelin pathology associated with reactive aldehydes and mitochondrial/peroxisomal dysfunctions in a mouse model of EAE. Daily treatment of EAE mice with Alda-1, starting at the peak of disease, ameliorated the clinical manifestation of disease along with the improvement of motor functions. Accordingly, Alda-1 treatment improved demyelination and neuroaxonal degeneration in EAE mice. EAE mice had increased levels of reactive aldehyde species, such as 4-hydroxynonenal (4-HNE), malondialdehyde (MDA), and acrolein (ACL) in the spinal cords and these levels were significantly reduced in Alda-1-treated EAE mice. Furthermore, Alda-1 treatment improved the loss of mitochondrial (OXPHOS) and peroxisomal (PMP70 and catalase) proteins as well as mitochondrial/peroxisomal proliferation factors (PGC-1α and PPARs) in the spinal cords of EAE mice. Taken together, this study demonstrates the therapeutic efficacy of ALDH2-agonist Alda-1 in the abatement of EAE disease through the detoxification of reactive aldehydes, thus suggesting Alda-1 as a potential therapeutic intervention for MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Aldehído Deshidrogenasa Mitocondrial , Aldehídos , Animales , Benzamidas , Benzodioxoles , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL
5.
Redox Biol ; 45: 102053, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34175668

RESUMEN

B cells play both protective and pathogenic roles in T cell-mediated autoimmune diseases by releasing regulatory vs. pathogenic cytokines. B cell-depleting therapy has been attempted in various autoimmune diseases but its efficacy varies and can even worsen symptoms due to depletion of B cells releasing regulatory cytokines along with B cells releasing pathogenic cytokines. Here, we report that S-nitrosoglutathione (GSNO) and GSNO-reductase (GSNOR) inhibitor N6022 drive upregulation of regulatory cytokine (IL-10) and downregulation of pathogenic effector cytokine (IL-6) in B cells and protected against the neuroinflammatory disease of experimental autoimmune encephalomyelitis (EAE). In human and mouse B cells, the GSNO/N6022-mediated regulation of IL-10 vs. IL-6 was not limited to regulatory B cells but also to a broad range of B cell subsets and antibody-secreting cells. Adoptive transfer of B cells from N6022 treated EAE mice or EAE mice deficient in the GSNOR gene also regulated T cell balance (Treg > Th17) and reduced clinical disease in the recipient EAE mice. The data presented here provide evidence of the role of GSNO in shifting B cell immune balance (IL-10 > IL-6) and the preclinical relevance of N6022, a first-in-class drug targeting GSNOR with proven human safety, as therapeutics for autoimmune disorders including multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Animales , Linfocitos B , Citocinas , Encefalomielitis Autoinmune Experimental/genética , Ratones , Ratones Endogámicos C57BL , S-Nitrosoglutatión
6.
PLoS One ; 14(1): e0210248, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30645642

RESUMEN

Long-term exposure to elevated levels of manganese (Mn) causes manganism, a neurodegenerative disorder with Parkinson's disease (PD)-like symptoms. Increasing evidence suggests that leucine-rich repeat kinase 2 (LRRK2), which is highly expressed in microglia and macrophages, contributes to the inflammation and neurotoxicity seen in autosomal dominant and sporadic PD. As gene-environment interactions have emerged as important modulators of PD-associated toxicity, LRRK2 may also mediate Mn-induced inflammation and pathogenesis. In this study, we investigated the role of LRRK2 in Mn-induced toxicity using human microglial cells (HMC3), LRRK2-wild-type (WT) and LRRK2-knockout (KO) RAW264.7 macrophage cells. Results showed that Mn activated LRRK2 kinase by phosphorylation of its serine residue at the 1292 position (S1292) as a marker of its kinase activity in macrophage and microglia, while inhibition with GSK2578215A (GSK) and MLi-2 abolished Mn-induced LRRK2 activation. LRRK2 deletion and its pharmacological inhibition attenuated Mn-induced apoptosis in macrophages and microglia, along with concomitant decreases in the pro-apoptotic Bcl-2-associated X (Bax) protein. LRRK2 deletion also attenuated Mn-induced production of reactive oxygen species (ROS) and the pro-inflammatory cytokine TNF-α. Mn-induced phosphorylation of mitogen-activated protein kinase (MAPK) p38 and ERK signaling proteins was significantly attenuated in LRRK2 KO cells and GSK-treated cells. Moreover, inhibition of MAPK p38 and ERK as well as LRRK2 attenuated Mn-induced oxidative stress and cytotoxicity. These findings suggest that LRRK2 kinase activity plays a critical role in Mn-induced toxicity via downstream activation of MAPK signaling in macrophage and microglia. Collectively, these results suggest that LRRK2 could be a potential molecular target for developing therapeutics to treat Mn-related neurodegenerative disorders.


Asunto(s)
Apoptosis , Biomarcadores/metabolismo , Inflamación/patología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Manganeso/efectos adversos , Microglía/patología , Estrés Oxidativo , Células Cultivadas , Citocinas/metabolismo , Humanos , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Especies Reactivas de Oxígeno
7.
Neurotoxicology ; 65: 280-288, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29183790

RESUMEN

Chronic exposure to manganese (Mn) causes neurotoxicity, referred to as manganism, with common clinical features of parkinsonism. 17ß-estradiol (E2) and tamoxifen (TX), a selective estrogen receptor modulator (SERM), afford neuroprotection in several neurological disorders, including Parkinson's disease (PD). In the present study, we tested if E2 and TX attenuate Mn-induced neurotoxicity in mice, assessing motor deficit and dopaminergic neurodegeneration. We implanted E2 and TX pellets in the back of the neck of ovariectomized C57BL/6 mice two weeks prior to a single injection of Mn into the striatum. One week later, we assessed locomotor activity and molecular mechanisms by immunohistochemistry, real-time quantitative PCR, western blot and enzymatic biochemical analyses. The results showed that both E2 and TX attenuated Mn-induced motor deficits and reversed the Mn-induced loss of dopaminergic neurons in the substantia nigra. At the molecular level, E2 and TX reversed the Mn-induced decrease of (1) glutamate aspartate transporter (GLAST) and glutamate transporter 1 (GLT-1) mRNA and protein levels; (2) transforming growth factor-α (TGF-α) and estrogen receptor-α (ER-α) protein levels; and (3) catalase (CAT) activity and glutathione (GSH) levels, and Mn-increased (1) malondialdehyde (MDA) levels and (2) the Bax/Bcl-2 ratio. These results indicate that E2 and TX afford protection against Mn-induced neurotoxicity by reversing Mn-reduced GLT1/GLAST as well as Mn-induced oxidative stress. Our findings may offer estrogenic agents as potential candidates for the development of therapeutics to treat Mn-induced neurotoxicity.


Asunto(s)
Encéfalo/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Estradiol/farmacología , Intoxicación por Manganeso/prevención & control , Tamoxifeno/farmacología , Sistema de Transporte de Aminoácidos X-AG/biosíntesis , Animales , Catalasa/metabolismo , Receptor alfa de Estrógeno/metabolismo , Femenino , Glutatión/metabolismo , Locomoción/efectos de los fármacos , Malondialdehído/metabolismo , Intoxicación por Manganeso/metabolismo , Ratones , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/patología , Ovariectomía , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor de Crecimiento Transformador alfa/metabolismo , Proteína X Asociada a bcl-2/metabolismo
8.
Neurotoxicology ; 67: 112-120, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29778792

RESUMEN

Exposure to elevated levels of manganese (Mn) causes manganism, a neurological disorder with similar characteristics to those of Parkinson's disease (PD). Valproic acid (VPA), an antiepileptic, is known to inhibit histone deacetylases and exert neuroprotective effects in many experimental models of neurological disorders. In the present study, we investigated if VPA attenuated Mn-induced dopaminergic neurotoxicity and the possible mechanisms involved in VPA's neuroprotection, focusing on modulation of astrocytic glutamate transporters (glutamate aspartate transporter, GLAST and glutamate transporter 1, GLT-1) and histone acetylation in H4 astrocyte culture and mouse models. The results showed that VPA increased promoter activity, mRNA/protein levels of GLAST/GLT-1 and glutamate uptake, and reversed Mn-reduced GLAST/GLT-1 in in vitro astrocyte cultures. VPA also attenuated Mn-induced reduction of GLAST and GLT-1 mRNA/protein levels in midbrain and striatal regions of the mouse brain when VPA (200 mg/kg, i.p., daily, 21 d) was administered 30 min prior to Mn exposure (30 mg/kg, intranasal instillation, daily, 21 d). Importantly, VPA attenuated Mn-induced dopaminergic neuronal damage by reversing Mn-induced decrease of tyrosine hydroxylase (TH) mRNA/protein levels in the nigrostriatal regions. VPA also reversed Mn-induced reduction of histone acetylation in astrocytes as well as mouse brain tissue. Taken together, VPA exerts attenuation against Mn-induced decrease of astrocytic glutamate transporters parallel with reversing Mn-induced dopaminergic neurotoxicity and Mn-reduced histone acetylation. Our findings suggest that VPA could serve as a potential neuroprotectant against Mn neurotoxicity as well as other neurodegenerative diseases associated with excitotoxicity and impaired astrocytic glutamate transporters.


Asunto(s)
Encéfalo/metabolismo , Dopamina/metabolismo , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Manganeso/toxicidad , Ácido Valproico/farmacología , Animales , Anticonvulsivantes/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Encéfalo/efectos de los fármacos , Células Cultivadas , Transportador 1 de Aminoácidos Excitadores/antagonistas & inhibidores , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Transportador 2 de Aminoácidos Excitadores/genética , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
9.
Coron Artery Dis ; 26(7): 604-11, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26180995

RESUMEN

BACKGROUND: There have been few reports on the relationship between the expression of the CD4⁺ T cells producing interferon-γ (Th1)/interleukin-17 (Th17) and degree of atherosclerosis. Thus, we analyzed Th1 and Th17 cell frequencies in patients with noncardiac chest pain (control), stable angina (SA), and acute myocardial infarction (AMI), and compared the complexity of the coronary artery with the SYNTAX score. PATIENTS AND METHODS: This study included 124 patients with a complaint of chest pain who underwent coronary angiography (control: 30 patients, SA: 47 patients, AMI: 47 patients). Peripheral blood was sampled during coronary angiography. Mononuclear cells from patients were stimulated for 4 h ex vivo. After staining with specific antibodies and fluorescence, the frequencies of Th1 and Th17 cells were measured by flow cytometry. The SYNTAX score was calculated by coronary angiography and a web-based calculator. RESULTS: There was no significant difference in the baseline characteristics, except the higher frequencies of hypertension in SA patients (76.1%) and smoking in AMI patients (53.3%). Patients with SA showed a significantly higher frequency of Th1 cells (21.56±9.57%) compared with controls (14.84±8.58%) and patients with AMI (9.04±7.02%) (P<0.001). The frequency of Th17 cells also increased in SA patients (control: 1.90±1.05%, SA: 2.96±1.42%, AMI: 1.32±0.92%, P<0.001). The SYNTAX score was significantly higher in SA patients (SA: 21.51±11.67, AMI: 15.36±8.84, P=0.006) and correlated with the frequencies of Th1 and Th17 cells (r=0.359, P=0.001; r=0.248, P=0.031; respectively). CONCLUSION: Th1 and Th17 cells were related to the development of SA, but not AMI. They could be a useful marker for the complexity of atherosclerosis in coronary artery disease.


Asunto(s)
Angina Estable/inmunología , Enfermedad de la Arteria Coronaria/inmunología , Infarto del Miocardio/inmunología , Células TH1/inmunología , Células Th17/inmunología , Anciano , Angina Estable/diagnóstico por imagen , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Femenino , Citometría de Flujo , Humanos , Interferón gamma/inmunología , Interleucina-17/inmunología , Masculino , Persona de Mediana Edad , Infarto del Miocardio/diagnóstico por imagen , Índice de Severidad de la Enfermedad
10.
Imaging Sci Dent ; 41(4): 189-93, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22232730

RESUMEN

The advent and wide use of antibiotics have decreased the incidence of deep neck infection. When a deep neck infection does occur, however, it can be the cause of significant morbidity and death, resulting in airway obstruction, mediastinitis, pericarditis, epidural abscesses, and major vessel erosion. In our clinic, a patient with diffuse chronic osteomyelitis of mandible and fascial space abscess and necrotic fasciitis due to odontogenic infection at the time of first visit came. We successfully treated the patient by early diagnosis using contrast-enhanced CT and follow up dressing through the appropriate use of radiographic images.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda