Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Inflammopharmacology ; 32(2): 1295-1315, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38512652

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory neurodegenerative disease of the central nervous system that injures the myelin sheath, provoking progressive axonal degeneration and functional impairments. No efficient therapy is available at present to combat such insults, and hence, novel safe and effective alternatives for MS therapy are extremely required. Rutin (RUT) is a flavonoid that exhibits antioxidant, anti-inflammatory, and neuroprotective effects in several brain injuries. The present study evaluated the potential beneficial effects of two doses of RUT in a model of pattern-III lesion of MS, in comparison to the conventional standard drug; dimethyl fumarate (DMF). Demyelination was induced in in male adult C57BL/6 mice by dietary 0.2% (w/w) cuprizone (CPZ) feeding for 6 consecutive weeks. Treated groups received either oral RUT (50 or 100 mg/kg) or DMF (15 mg/kg), along with CPZ feeding, for 6 consecutive weeks. Mice were then tested for behavioral changes, followed by biochemical analyses and histological examinations of the corpus callosum (CC). Results revealed that CPZ caused motor dysfunction, demyelination, and glial activation in demyelinated lesions, as well as significant oxidative stress, and proinflammatory cytokine elevation. Six weeks of RUT treatment significantly improved locomotor activity and motor coordination. Moreover, RUT considerably improved remyelination in the CC of CPZ + RUT-treated mice, as revealed by luxol fast blue staining and transmission electron microscopy. Rutin also significantly attenuated CPZ-induced oxidative stress and inflammation in the CC of tested animals. The effect of RUT100 was obviously more marked than either that of DMF, regarding most of the tested parameters, or even its smaller tested dose. In silico docking revealed that RUT binds tightly within NF-κB at the binding site of the protein-DNA complex, with a good negative score of -6.79 kcal/mol. Also, RUT-Kelch-like ECH-associated protein 1 (Keap1) model clarifies the possible inhibition of Keap1-Nrf2 protein-protein interaction. Findings of the current study provide evidence for the protective effect of RUT in CPZ-induced demyelination and behavioral dysfunction in mice, possibly by modulating NF-κB and Nrf2 signaling pathways. The present study may be one of the first to indicate a pro-remyelinating effect for RUT, which might represent a potential additive benefit in treating MS.


Asunto(s)
Enfermedades Desmielinizantes , Esclerosis Múltiple , Enfermedades Neurodegenerativas , Fármacos Neuroprotectores , Masculino , Animales , Ratones , Esclerosis Múltiple/inducido químicamente , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Cuprizona/efectos adversos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , FN-kappa B/metabolismo , Rutina/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
2.
J Pharmacol Sci ; 153(3): 94-103, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37770161

RESUMEN

The cuprizone (CPZ)-induced demyelination model, an animal model of Multiple sclerosis (MS), is characterized by demyelination and motor dysfunction due to microglial-mediated neuroinflammation. To determine the contribution of microglia to motor function during CPZ-induced demyelination, the microglia of mice in the CPZ-model were depleted using PLX3397 (PLX), an orally bioavailable selective colony stimulating factor 1 receptor inhibitor. PLX treatment aggravated motor dysfunction as shown by the pole, beam walk, ladder walk, and rotarod tests. PLX treatment removed microglia from the superior cerebellar peduncle (SCP), but not from the corpus callosum (CC). Although PLX treatment did not affect the degree of demyelination in both of CC and SCP, the expression of axonal damage marker APP (amyloid precursor protein) was increased. Increased TNF-α, IL-1ß, and iNOS expressions were observed in PLX-treated mice. These results suggest that microglial depletion exacerbates axonal damage and motor dysfunction in CPZ model mice. In this study, we found that microglia contribute to motor function and axon-protective effects in CPZ-induced demyelination.


Asunto(s)
Enfermedades Desmielinizantes , Esclerosis Múltiple , Ratones , Animales , Microglía/metabolismo , Cuprizona/efectos adversos , Enfermedades Desmielinizantes/inducido químicamente , Axones , Esclerosis Múltiple/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
3.
Int J Mol Sci ; 24(23)2023 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-38069420

RESUMEN

Microglia are found pathologically at all stages of multiple sclerosis (MS) lesion development and are hypothesized to contribute to both inflammatory injury and neuroprotection in the MS brain. Transient receptor potential vanilloid 4 (TRPV4) channels are widely expressed, play an important role as environmental sensors, and are involved in calcium homeostasis for a variety of cells. TRPV4 modulates myeloid cell phagocytosis in the periphery and microglial motility in the central nervous system. We hypothesized that TRPV4 deletion would alter microglia phagocytosis in vitro and lessen disease activity and demyelination in experimental autoimmune encephalitis (EAE) and cuprizone-induced demyelination. We found that genetic deletion of TRPV4 led to increased microglial phagocytosis in vitro but did not alter the degree of demyelination or remyelination in the cuprizone mouse model of MS. We also found no difference in disease in EAE following global or microglia-specific deletion of Trpv4. Additionally, lesioned and normal appearing white matter from MS brains exhibited similar TRPV4 expression compared to healthy brain tissue. Taken together, these findings indicate that TRPV4 modulates microglial activity but does not impact disease activity in mouse models of MS, suggesting a muted and/or redundant role in MS pathogenesis.


Asunto(s)
Enfermedades Desmielinizantes , Microglía , Canales Catiónicos TRPV , Animales , Ratones , Cuprizona/efectos adversos , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Microglía/metabolismo , Esclerosis Múltiple/patología , Vaina de Mielina/metabolismo , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
4.
Int J Mol Sci ; 23(24)2022 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-36555733

RESUMEN

Remyelination therapies, which are currently under development, have a great potential to delay, prevent or even reverse disability in multiple sclerosis patients. Several models are available to study the effectiveness of novel compounds in vivo, among which is the cuprizone model. This model is characterized by toxin-induced demyelination, followed by endogenous remyelination after cessation of the intoxication. Due to its high reproducibility and ease of use, this model enjoys high popularity among various research and industrial groups. In this review article, we will summarize recent findings using this model and discuss the potential of some of the identified compounds to promote remyelination in multiple sclerosis patients.


Asunto(s)
Enfermedades Desmielinizantes , Esclerosis Múltiple , Remielinización , Humanos , Animales , Ratones , Cuprizona/efectos adversos , Vaina de Mielina , Enfermedades Desmielinizantes/inducido químicamente , Reproducibilidad de los Resultados , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
5.
Int J Mol Sci ; 23(18)2022 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-36142668

RESUMEN

Maintaining the normal function of oligodendrocyte precursor cells (OPCs) and protecting OPCs from damage is the basis of myelin regeneration in multiple sclerosis (MS). In this paper, we investigated the effect of stemazole, a novel small molecule, on the promotion of oligodendrocyte precursor cell survival and remyelination. The results show that stemazole enhanced the survival rate and the number of clone formation in a dose-dependent manner and decreased the percentage of cell apoptosis. In particular, the number of cell clones was increased up to 6-fold (p < 0.001) in the stemazole group compared with the control group. In vivo, we assessed the effect of stemazole on recovering the motor dysfunction and demyelination induced by cuprizone (CPZ). The results show that stemazole promoted the recovery of motor dysfunction and the repair of myelin sheaths. Compared with the CPZ group, the stemazole group showed a 30.46% increase in the myelin area (p < 0.001), a 37.08% increase in MBP expression (p < 0.01), and a 1.66-fold increase in Olig2 expression (p < 0.001). Histologically, stemazole had a better effect than the positive control drugs. In conclusion, stemazole promoted OPC survival in vitro and remyelination in vivo, suggesting that this compound may be used as a therapeutic agent against demyelinating disease.


Asunto(s)
Enfermedades Desmielinizantes , Células Precursoras de Oligodendrocitos , Remielinización , Animales , Diferenciación Celular , Cuprizona/efectos adversos , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/metabolismo , Modelos Animales de Enfermedad , Hidrazinas , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Oxadiazoles
6.
Inflammopharmacology ; 30(6): 2197-2209, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35948811

RESUMEN

BACKGROUND AND AIM: Multiple sclerosis (MS) is a demyelinating neurodegenerative inflammatory disease affecting mainly young adults. Microgliosis-derived neuroinflammation represents a key hallmark in MS pathology and progression. Nebivolol (Neb) demonstrated antioxidant, anti-inflammatory and neuroprotective properties in several brain pathologies. This study was conducted to investigate the potential neuroprotective effect of Neb in the cuprizone (Cup) model of MS. METHODS: C57Bl/6 mice were fed 0.2% Cup mixed into rodent chow for 5 weeks. Neb (5 and 10 mg/kg/day) was administered by oral gavage during the last 2 weeks. RESULTS: Neb prevented Cup-induced weight loss and motor deficits as evidenced by increased latency to fall in the rotarod test and enhanced locomotor activity as compared to Cup-intoxicated mice. Neb reversed Cup-induced demyelination as confirmed by Luxol fast blue staining and myelin basic protein western blotting. Administration of Neb modulated microglial activation status by suppressing M1 markers (Iba-1, CD86, iNOS, NO and TNF-α) and increasing M2 markers (Arg-1 and IL-10) as compared to Cup-fed mice. Furthermore, Neb hindered NLRP3/caspase-1/IL-18 inflammatory cascade and alleviated oxidative stress by reducing lipid peroxidation, as well as increasing catalase and superoxide dismutase activities. CONCLUSION: These findings suggest the potential neuroprotective effect of Neb in the Cup-induced model of MS in mice, at least partially by virtue of shifting microglia towards M2 phenotype, mitigation of NLRP3 inflammasome activation and alleviation of oxidative stress.


Asunto(s)
Enfermedades Desmielinizantes , Esclerosis Múltiple , Nebivolol , Fármacos Neuroprotectores , Animales , Ratones , Cuprizona/efectos adversos , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/metabolismo , Modelos Animales de Enfermedad , Inflamasomas/metabolismo , Ratones Endogámicos C57BL , Microglía , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Nebivolol/farmacología , Fármacos Neuroprotectores/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Macrófagos , Polaridad Celular
7.
J Biol Chem ; 295(34): 12233-12246, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32647008

RESUMEN

Disorders that disrupt myelin formation during development or in adulthood, such as multiple sclerosis and peripheral neuropathies, lead to severe pathologies, illustrating myelin's crucial role in normal neural functioning. However, although our understanding of glial biology is increasing, the signals that emanate from axons and regulate myelination remain largely unknown. To identify the core components of the myelination process, here we adopted a microarray analysis approach combined with laser-capture microdissection of spinal motoneurons during the myelinogenic phase of development. We identified neuronal genes whose expression was enriched during myelination and further investigated hepatoma-derived growth factor-related protein 3 (HRP3 or HDGFRP3). HRP3 was strongly expressed in the white matter fiber tracts of the peripheral (PNS) and central (CNS) nervous systems during myelination and remyelination in a cuprizone-induced demyelination model. The dynamic localization of HPR3 between axons and nuclei during myelination was consistent with its axonal localization during neuritogenesis. To study this phenomenon, we identified two splice variants encoded by the HRP3 gene: the canonical isoform HRP3-I and a newly recognized isoform, HRP3-II. HRP3-I remained solely in the nucleus, whereas HRP3-II displayed distinct axonal localization both before and during myelination. Interestingly, HRP3-II remained in the nuclei of unmyelinated neurons and glial cells, suggesting the existence of a molecular machinery that transfers it to and retains it in the axons of neurons fated for myelination. Overexpression of HRP3-II, but not of HRP3-I, increased Schwann cell numbers and myelination in PNS neuron-glia co-cultures. However, HRP3-II overexpression in CNS co-cultures did not alter myelination.


Asunto(s)
Axones/metabolismo , Núcleo Celular/metabolismo , Enfermedades Desmielinizantes/metabolismo , Perfilación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/sangre , Neuronas Motoras/metabolismo , Animales , Axones/patología , Núcleo Celular/patología , Técnicas de Cocultivo , Cuprizona/efectos adversos , Cuprizona/farmacología , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/patología , Masculino , Ratones , Neuronas Motoras/patología , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Neuroglía/metabolismo , Neuroglía/patología , Isoformas de Proteínas , Ratas
8.
J Pharmacol Sci ; 144(3): 119-122, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32921392

RESUMEN

Multiple sclerosis (MS) is a demyelinating disease of the central nervous system, characterized by apoptotic death of mature oligodendrocytes, neuroinflammation, and motor dysfunction. A pentacyclic triterpenoid compound, ursolic acid (UA), has various pharmacological activities, such as anti-inflammatory, anti-oxidative, and anti-apoptotic effects. In the present study, we investigated the effects of UA on cuprizone-induced demyelination, which is a model of MS. Oral administration of UA effectively suppressed cuprizone-induced demyelination and motor dysfunction via the enhancement of IGF-1 levels in the demyelinating lesions. Our results suggest that UA might be therapeutically useful for demyelination in MS.


Asunto(s)
Cuprizona/efectos adversos , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/genética , Expresión Génica/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/genética , Triterpenos/administración & dosificación , Triterpenos/farmacología , Regulación hacia Arriba/efectos de los fármacos , Administración Oral , Animales , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/fisiopatología , Modelos Animales de Enfermedad , Ratones , Esclerosis Múltiple/inducido químicamente , Esclerosis Múltiple/fisiopatología , Ácido Ursólico
9.
Proc Natl Acad Sci U S A ; 114(33): E6982-E6991, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28760957

RESUMEN

Proinflammatory mononuclear phagocytes (MPs) play a crucial role in the progression of multiple sclerosis (MS) and other neurodegenerative diseases. Despite advances in neuroimaging, there are currently limited available methods enabling noninvasive detection of MPs in vivo. Interestingly, upon activation and subsequent differentiation toward a proinflammatory phenotype MPs undergo metabolic reprogramming that results in increased glycolysis and production of lactate. Hyperpolarized (HP) 13C magnetic resonance spectroscopic imaging (MRSI) is a clinically translatable imaging method that allows noninvasive monitoring of metabolic pathways in real time. This method has proven highly useful to monitor the Warburg effect in cancer, through MR detection of increased HP [1-13C]pyruvate-to-lactate conversion. However, to date, this method has never been applied to the study of neuroinflammation. Here, we questioned the potential of 13C MRSI of HP [1-13C]pyruvate to monitor the presence of neuroinflammatory lesions in vivo in the cuprizone mouse model of MS. First, we demonstrated that 13C MRSI could detect a significant increase in HP [1-13C]pyruvate-to-lactate conversion, which was associated with a high density of proinflammatory MPs. We further demonstrated that the increase in HP [1-13C]lactate was likely mediated by pyruvate dehydrogenase kinase 1 up-regulation in activated MPs, resulting in regional pyruvate dehydrogenase inhibition. Altogether, our results demonstrate a potential for 13C MRSI of HP [1-13C]pyruvate as a neuroimaging method for assessment of inflammatory lesions. This approach could prove useful not only in MS but also in other neurological diseases presenting inflammatory components.


Asunto(s)
Isótopos de Carbono , Ácido Láctico , Imagen por Resonancia Magnética , Esclerosis Múltiple/diagnóstico por imagen , Esclerosis Múltiple/metabolismo , Animales , Isótopos de Carbono/farmacocinética , Isótopos de Carbono/farmacología , Cuprizona/efectos adversos , Cuprizona/farmacología , Modelos Animales de Enfermedad , Femenino , Ácido Láctico/farmacocinética , Ácido Láctico/farmacología , Ratones , Ratones Transgénicos , Esclerosis Múltiple/inducido químicamente , Esclerosis Múltiple/genética
10.
Int J Mol Sci ; 21(2)2020 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-31963761

RESUMEN

The demyelinating diseases of the central nervous system involve myelin abnormalities, oligodendrocyte damage, and consequent glia activation. Neurotoxicant cuprizone (CPZ) was used to establish a mouse model of demyelination. However, the effects of CPZ on microRNA (miRNA) expression and behavior have not been clearly reported. We analyzed the behavior of mice administered a diet containing 0.2% CPZ for 6 weeks, followed by 6 weeks of recovery. Rotarod analysis demonstrated that the treated group had poorer motor coordination than control animals. This effect was reversed after 6 weeks of CPZ withdrawal. Open-field tests showed that CPZ-treated mice exhibited significantly increased anxiety and decreased exploratory behavior. CPZ-induced demyelination was observed to be alleviated after 4 weeks of CPZ treatment, according to luxol fast blue (LFB) staining and myelin basic protein (MBP) expression. miRNA expression profiling showed that the expression of 240 miRNAs was significantly changed in CPZ-fed mice compared with controls. Furthermore, miR-155-5p and miR-20a-5p upregulations enhanced NgR induction through Smad 2 and Smad 4 suppression in demyelination. Taken together, our results demonstrate that CPZ-mediated demyelination induces behavioral deficits with apparent alterations in miRNA expression, suggesting that differences in miRNA expression in vivo may be new potential therapeutic targets for remyelination.


Asunto(s)
Cuprizona/efectos adversos , Enfermedades Desmielinizantes/psicología , Conducta Exploratoria/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Animales , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/genética , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/efectos de los fármacos , MicroARNs/genética , Prueba de Desempeño de Rotación con Aceleración Constante
11.
Cytokine ; 113: 417-426, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30539784

RESUMEN

Multiple sclerosis (MS) is a disease of the central nervous system (CNS) in which both neuroinflammation and neurodegeneration play critical roles in the pathogenesis of the disease. A growing body of evidence indicates that some antibiotics have anti-inflammatory and neuroprotective properties. Rifampicin, commonly used for the treatment of mycobacteria, has been shown to exert neuroprotective activities in neurodegenerative diseases. In this study, we examined the efficacy of rifampicin on demyelination, gliosis, apoptosis, inflammation, behavioral dysfunction, and biochemical alterations in the cuprizone model of demyelination. For this aim, male C57BL/6J mice were fed a chow containing 0.2% cuprizone (w/w) for 6 weeks to induce reversible demyelination in the corpus callosum. Mice intraperitoneally received serial doses of rifampicin (10, 20, or 40 mg/kg body weight) in the last 7 days of a 6-week period of cuprizone treatment. The results showed that the administration of rifampicin led to the improvement in motor behavioral deficits. In line with this, rifampicin decreased the number of apoptotic cells in the corpus callosum thereby diminishing the expression of cleaved caspase-3 and Bax, as well as increasing Bcl-2. Moreover, rifampicin significantly lowered the levels of interleukin-6, interleukin-1ß, caspase-12 activity, heme oxygenase-1(HO-1), nitric oxide (NO), and malondialdehyde (MDA) in mice treated with cuprizone. Conversely, the activity of glutathione peroxidase (GPx) and the level of ferric reducing ability of plasma (FRAP) were increased in response to the treatment with rifampicin. Histopathological findings demonstrated that rifampicin statistically promoted remyelination and mitigated microgliosis and astrogliosis. It seems that rifampicin is able to be added to the armamentarium of therapies for multiple sclerosis.


Asunto(s)
Conducta Animal/efectos de los fármacos , Cuerpo Calloso , Cuprizona/efectos adversos , Enfermedades Desmielinizantes , Rifampin/farmacología , Animales , Cuerpo Calloso/metabolismo , Cuerpo Calloso/patología , Cuprizona/farmacología , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Masculino , Ratones
12.
J Pharmacol Sci ; 136(2): 93-96, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29409686

RESUMEN

Lysophosphatidic acid (LPA) and LPA1 receptor signaling play a crucial role in the initiation of peripheral nerve injury-induced neuropathic pain through the alternation of pain-related genes/proteins expression and demyelination. However, LPA and its signaling in the brain are still poorly understood. In the present study, we revealed that the LPA5 receptor expression in corpus callosum elevated after the initiation of demyelination, and the hyperalgesia through Aδ-fibers following cuprizone-induced demyelination was mediated by LPA5 signaling. These data suggest that LPA5 signaling may play a key role in the mechanisms underlying neuropathic pain following demyelination in the brain.


Asunto(s)
Cuprizona/efectos adversos , Modelos Animales de Enfermedad , Esclerosis Múltiple/etiología , Esclerosis Múltiple/genética , Neuralgia/etiología , Neuralgia/genética , Receptores del Ácido Lisofosfatídico/fisiología , Transducción de Señal/fisiología , Animales , Cuerpo Calloso/metabolismo , Femenino , Expresión Génica , Lisofosfolípidos/fisiología , Masculino , Ratones Endogámicos , Esclerosis Múltiple/metabolismo , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/metabolismo
13.
Metallomics ; 16(1)2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38178638

RESUMEN

Demyelination within the central nervous system (CNS) is a significant feature of debilitating neurological diseases such as multiple sclerosis and administering the copper-selective chelatorcuprizone to mice is widely used to model demyelination in vivo. Conspicuous demyelination within the corpus callosum is generally attributed to cuprizone's ability to restrict copper availability in this vulnerable brain region. However, the small number of studies that have assessed copper in brain tissue from cuprizone-treated mice have produced seemingly conflicting outcomes, leaving the role of CNS copper availability in demyelination unresolved. Herein we describe our assessment of copper concentrations in brain samples from mice treated with cuprizone for 40 d. Importantly, we applied an inductively coupled plasma mass spectrometry methodology that enabled assessment of copper partitioned into soluble and insoluble fractions within distinct brain regions, including the corpus callosum. Our results show that cuprizone-induced demyelination in the corpus callosum was associated with decreased soluble copper in this brain region. Insoluble copper in the corpus callosum was unaffected, as were pools of soluble and insoluble copper in other brain regions. Treatment with the blood-brain barrier permeant copper compound CuII(atsm) increased brain copper levels and this was most pronounced in the soluble fraction of the corpus callosum. This effect was associated with significant mitigation of cuprizone-induced demyelination. These results provide support for the involvement of decreased CNS copper availability in demyelination in the cuprizone model. Relevance to human demyelinating disease is discussed.


Asunto(s)
Cuprizona , Enfermedades Desmielinizantes , Humanos , Animales , Ratones , Cuprizona/efectos adversos , Cuerpo Calloso , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Cobre/farmacología , Oligodendroglía , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Vaina de Mielina
14.
Prog Neurobiol ; 231: 102532, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37774767

RESUMEN

Multiple sclerosis (MS) pathology features autoimmune-driven neuroinflammation, demyelination, and failed remyelination. Carnosine is a histidine-containing dipeptide (HCD) with pluripotent homeostatic properties that is able to improve outcomes in an animal MS model (EAE) when supplied exogenously. To uncover if endogenous carnosine is involved in, and protects against, MS-related neuroinflammation, demyelination or remyelination failure, we here studied the HCD-synthesizing enzyme carnosine synthase (CARNS1) in human MS lesions and two preclinical mouse MS models (EAE, cuprizone). We demonstrate that due to its presence in oligodendrocytes, CARNS1 expression is diminished in demyelinated MS lesions and mouse models mimicking demyelination/inflammation, but returns upon remyelination. Carns1-KO mice that are devoid of endogenous HCDs display exaggerated neuroinflammation and clinical symptoms during EAE, which could be partially rescued by exogenous carnosine treatment. Worsening of the disease appears to be driven by a central, not peripheral immune-modulatory, mechanism possibly linked to impaired clearance of the reactive carbonyl acrolein in Carns1-KO mice. In contrast, CARNS1 is not required for normal oligodendrocyte precursor cell differentiation and (re)myelin to occur, and neither endogenous nor exogenous HCDs protect against cuprizone-induced demyelination. In conclusion, the loss of CARNS1 from demyelinated MS lesions can aggravate disease progression through weakening the endogenous protection against neuroinflammation.


Asunto(s)
Carnosina , Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Humanos , Ratones , Animales , Esclerosis Múltiple/tratamiento farmacológico , Cuprizona/efectos adversos , Cuprizona/metabolismo , Carnosina/efectos adversos , Carnosina/metabolismo , Enfermedades Neuroinflamatorias , Vaina de Mielina/patología , Oligodendroglía/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología
15.
J Neurosci ; 31(7): 2382-90, 2011 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-21325505

RESUMEN

Myelination is one of the fundamental biological processes in the development of vertebrate nervous system. Disturbance of myelination is found to be associated with progression in many neurological diseases such as multiple sclerosis. Tremendous efforts have been made to develop novel therapeutic agents that prevent demyelination and/or promote remyelination. These efforts need to be accompanied by the development of imaging tools that permit direct quantification of myelination in vivo. In this work, we describe a novel near-infrared fluorescence imaging technique that is capable of direct quantification of myelination in vivo. This technique is developed based on a near-infrared fluorescent probe, 3,3'-diethylthiatricarbocyanine iodide (DBT) that readily enters the brain and specifically binds to myelinated fibers. In vivo imaging studies were first conducted in two animal models of hypermyelination and hypomyelination followed by longitudinal studies in the cuprizone-induced demyelination/remyelination mouse model. Quantitative analysis suggests that DBT is a sensitive and specific imaging probe of myelination, which complements other current myelin-imaging modalities and is of low cost.


Asunto(s)
Vaina de Mielina/fisiología , Espectroscopía Infrarroja Corta , Análisis de Varianza , Animales , Benzotiazoles/metabolismo , Carbocianinas/metabolismo , Cuerpo Calloso/citología , Cuerpo Calloso/metabolismo , Cuprizona/efectos adversos , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/metabolismo , Modelos Animales de Enfermedad , Estudios Longitudinales , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Ratones Transgénicos , Proteína Básica de Mielina/genética , Proteína Proteolipídica de la Mielina , Vaina de Mielina/efectos de los fármacos , Permeabilidad , Unión Proteica/fisiología , Proteínas Proto-Oncogénicas c-akt/genética , Factores de Tiempo
16.
Cells ; 11(7)2022 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-35406658

RESUMEN

Oligodendrocytes are glial cells located in the central nervous system (CNS) that play essential roles in the transmission of nerve signals and in the neuroprotection of myelinated neurons. The dysfunction or loss of oligodendrocytes leads to demyelinating diseases such as multiple sclerosis (MS). To treat demyelinating diseases, the development of a therapy that promotes remyelination is required. In the present study, we established an in vitro method to convert human fibroblasts into induced oligodendrocyte-like cells (iOLCs) in 3 days. The induced cells displayed morphologies and molecular signatures similar to oligodendrocytes after treatment with valproic acid and exposure to the small molecules Y27632, SU9516, and forskolin (FSK). To pursue the development of a cell-free remyelination therapy in vivo, we used a cuprizone-induced demyelinated mouse model. The small molecules (Y27632, SU9516, and FSK) were directly injected into the demyelinated corpus callosum of the mouse brain. This combination of small molecules rescued the demyelination phenotype within two weeks as observed by light and electron microscopy. These results provide a foundation for exploring the development of a treatment for demyelinating diseases via regenerative medicine.


Asunto(s)
Cuprizona , Enfermedades Desmielinizantes , Animales , Cuerpo Calloso , Cuprizona/efectos adversos , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/genética , Ratones , Ratones Endogámicos C57BL , Oligodendroglía/fisiología
17.
Dalton Trans ; 51(27): 10361-10376, 2022 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-35766122

RESUMEN

Copper(II) coordination by bis(cyclohexanone)oxalyldihydrazone (also known as cuprizone), resulting in the formation of an intensely coloured blue complex, was first reported over 70 years ago. The cuprizone reaction has been employed in colourimetric tests for the presence of trace levels of copper. Cuprizone administration in C57BL/6 mice also leads to demyelination over time - a consequence that appears to be due to copper dyshomeostasis - and this has led to use of cuprizone as the leading method for toxicant-induced generation of an animal model of demyelination since its first use in the 1960s. Despite broad interest in cuprizone and its ability to bind copper there have been relatively few studies to structurally characterize the copper coordination properties of this ligand. In the absence of an aqueous medium, such as neat alcohol, copper and cuprizone exclusively form an amorphous green precipitate. Under aqueous conditions, where a large excess of cuprizone (relative to copper) is present, the blue complex that is synonymous with copper-cuprizone coordination is predominantly formed. The blue and green copper-cuprizone products demonstrate poor solubility and present challenges for conventional structure characterization methods, such as X-ray crystallography or nuclear magnetic resonance spectroscopy. By combining mass spectrometry, X-ray absorption spectroscopy, computational chemistry, and other techniques, a self-consistent picture of the copper coordination structures of the blue and green complexes is revealed - confirming that the blue complex is in the Cu(III) state, containing two hydrolyzed cuprizone ligands per metal centre, while the green complex represents an extended oligomeric complex, comprised of repeating Cu(II) centres that lie 4.8 Å apart and are bridged by unhydrolyzed cuprizone donors.


Asunto(s)
Complejos de Coordinación , Enfermedades Desmielinizantes , Animales , Complejos de Coordinación/química , Cobre/química , Cristalografía por Rayos X , Cuprizona/efectos adversos , Enfermedades Desmielinizantes/inducido químicamente , Ligandos , Ratones , Ratones Endogámicos C57BL , Espectroscopía de Absorción de Rayos X
18.
J Mol Histol ; 53(5): 817-831, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35947228

RESUMEN

Multiple sclerosis (MS) has no absolute treatment, and researchers are still exploring to introduce promising therapy for MS. Transcranial direct current stimulation (tDCS), is a safe, non-invasive procedure for brain stimulating which can enhance working memory, cognitive neurohabitation and motor recovery. Here, we evaluated the effects of tDCS treatment and Mesenchymal stem cells (MSCs) transplantation on remyelination ability of a Cuprizone (CPZ)-induced demyelination mouse model. tDCS significantly increased the motor coordination and balance abilities in CPZ + tDCS and CPZ + tDCS + MSCs mice in comparison to the CPZ mice. Luxol fast blue (LFB) staining showed that tDCS and MSCs transplantation could increase remyelination capacity in CPZ + tDCS and CPZ + MSCs mice compared to the CPZ mice. But, the effect of tDCS with MSCs transplantation on remyelination process was larger than each of treatment alone. Immunofluorescence technique indicated that the numbers of Olig2+ cells were increased by tDCS and MSCs transplantation in CPZ + tDCS and CPZ + MSCs mice compared to the CPZ mice. Interestingly, the combination effect of tDCS and MSCs was larger than each of treatment alone on Oligodendrocytes population. MSCs transplantation significantly decreased the TUNEL+ cells in CPZ + MSCs and CPZ + tDCS + MSCs mice in comparison to the CPZ mice. Also, the combination effects of tDCS and MSCs transplantation was much larger than each of treatment alone on increasing the mRNA expression of BDNF and Sox2, while decreasing P53 as compared to CPZ mice. It can be concluded that the combination usage of tDCS and MSCs transplantation enhance remyelination process in CPZ-treated mice by increasing transplanted stem cell homing, oligodendrocyte generation and decreasing apoptosis.


Asunto(s)
Enfermedades Desmielinizantes , Células Madre Mesenquimatosas , Esclerosis Múltiple , Estimulación Transcraneal de Corriente Directa , Animales , Cuprizona/efectos adversos , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/terapia , Modelos Animales de Enfermedad , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inducido químicamente , Esclerosis Múltiple/terapia
19.
Cells ; 11(11)2022 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-35681468

RESUMEN

Oligodendrocytes are the myelinating cells of the central nervous system. The physiological importance of oligodendrocytes is highlighted by diseases such as multiple sclerosis, in which the myelin sheaths are degraded and the axonal signal transmission is compromised. In a healthy brain, spontaneous remyelination is rare, and newly formed myelin sheaths are thinner and shorter than the former ones. The myelination process requires the migration, proliferation, and differentiation of oligodendrocyte precursor cells (OPCs) and is influenced by proteins of the extracellular matrix (ECM), which consists of a network of glycoproteins and proteoglycans. In particular, the glycoprotein tenascin-C (Tnc) has an inhibitory effect on the differentiation of OPCs and the remyelination efficiency of oligodendrocytes. The structurally similar tenascin-R (Tnr) exerts an inhibitory influence on the formation of myelin membranes in vitro. When Tnc knockout oligodendrocytes were applied to an in vitro myelination assay using artificial fibers, a higher number of sheaths per single cell were obtained compared to the wild-type control. This effect was enhanced by adding brain-derived neurotrophic factor (BDNF) to the culture system. Tnr-/- oligodendrocytes behaved differently in that the number of formed sheaths per single cell was decreased, indicating that Tnr supports the differentiation of OPCs. In order to study the functions of tenascin proteins in vivo Tnc-/- and Tnr-/- mice were exposed to Cuprizone-induced demyelination for a period of 10 weeks. Both Tnc-/- and Tnr-/- mouse knockout lines displayed a significant increase in the regenerating myelin sheath thickness after Cuprizone treatment. Furthermore, in the absence of either tenascin, the number of OPCs was increased. These results suggest that the fine-tuning of myelin regeneration is regulated by the major tenascin proteins of the CNS.


Asunto(s)
Enfermedades Desmielinizantes , Células Precursoras de Oligodendrocitos , Tenascina , Animales , Cuprizona/efectos adversos , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Tenascina/metabolismo
20.
Adv Clin Exp Med ; 31(9): 999-1009, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35587221

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS). Most exercise studies concentrate on the impact of exercise on cardiovascular system; this study aims to present the effects of exercise of varying intensity on the nervous system. Most recently in MS, positive outcomes were obtained with resistance and high-intensity exercises. This study also analyzes the effects of a prior conditioning program before the induction of demyelination and subsequent neuroprotective effects of such program. OBJECTIVES: To study and determine the neuroprotective and remyelinating effects of different intensity of aquatic exercise and a preconditioning exercise program on demyelination induced by oral administration of cuprizone (Cup). MATERIAL AND METHODS: Six groups of animals, each containing 6 rats, were used in the study. The groups were as follows: group I - control group; group II - Cup group; group III - treated with methylprednisolone (MP); group IV - treated with low-intensity exercise (LIE), free swimming for 40 min and high-intensity exercise (HIE); group V - treated with a resistance of 9% body weight and free swimming for 40 min; group VI - treated with preconditioning exercise (free swimming for 40 min for 3 weeks) before Cup administration followed by the same exercise protocol as for group V. All data were analyzed using one-way analysis of variance (ANOVA) with Tukey's test, by means of SigmaPlot v. 14.5 software. RESULTS: Similarly to the MP group, group VI showed a positive outcome. A value of p < 0.001 was considered statistically significant. Also, group VI showed improved areas of remyelination in histopathology, an increased expression of myelin basic protein (MBP), reduced expression of glial fibrillary acidic protein (GFAP) in corpus callosum, and improved gene expression of brain-derived neurotrophic factor (BDNF) in the hippocampus region. CONCLUSIONS: General fitness achieved through a preconditioning program combined with HIE showed neuroprotective effects, as evidenced by increased areas of remyelination and improved neuronal plasticity, observed mostly in group VI (conditioning+HIE).


Asunto(s)
Enfermedades Desmielinizantes , Esclerosis Múltiple , Fármacos Neuroprotectores , Remielinización , Animales , Factor Neurotrófico Derivado del Encéfalo , Cuprizona/efectos adversos , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/prevención & control , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/efectos adversos , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Metilprednisolona , Ratones , Ratones Endogámicos C57BL , Proteína Básica de Mielina/efectos adversos , Proteína Básica de Mielina/metabolismo , Plasticidad Neuronal , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA