Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Neurobiol Dis ; 171: 105802, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35753626

RESUMEN

ß-thalassemia is associated with multiple hematological and cerebrovascular symptoms linked to a hypercoagulable state that has not been fully replicated in animal models for the development of stroke treatments. Herein we compared the physiological properties and responses to transient cerebral hypoxia-ischemia (tHI) between six-month-old wildtype and heterozygous Th3/+ mice, a model of non-transfusion-dependent ß-thalassemia intermedia (ß-TI). We found that Th3/+ mice developed microcytic anemia, splenomegaly, higher platelet counts, and increased platelet-erythrocyte plus erythrocyte-leukocyte aggregates. Furthermore, Th3/+ mice showed diminished cerebrovascular reactivity (CVR) and cortical oxygen saturation under repetitive hypercapnic challenges. When subjected to a sub-threshold tHI insult, platelets and leukocytes in Th3/+ mice adhered to the cerebrovascular wall or formed aggregates, while their counterparts flew through smoothly in wildtype mice. Subsequently, Th3/+ mice showed increased fibrin deposition around cerebral blood vessels and larger infarction than wildtype mice, especially in female Th3/+ mice. Collectively these results showed that Th3/+ mice mimic key clinical features and a propensity to thromboembolism in ß-TI patients. The hypercoagulable state in Th3/+ mice is likely caused by multiple hematological and CVR anomalies that are similar, but are not identical to those in the mouse model of sickle cell anemia. As such, we suggest that Th3/+ mice are a useful model to study the pathological mechanisms and prophylactic stroke treatments in thalassemia patients.


Asunto(s)
Hipoxia-Isquemia Encefálica , Accidente Cerebrovascular , Talasemia beta , Animales , Modelos Animales de Enfermedad , Femenino , Hipoxia-Isquemia Encefálica/complicaciones , Ratones , Accidente Cerebrovascular/complicaciones , Talasemia beta/complicaciones , Talasemia beta/patología
2.
J Neurosci ; 40(49): 9386-9400, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33127853

RESUMEN

Growing evidence suggests that early-life interactions among genetic, immune, and environment factors may modulate neurodevelopment and cause psycho-cognitive deficits. Maternal immune activation (MIA) induces autism-like behaviors in offspring, but how it interplays with perinatal brain injury (especially birth asphyxia or hypoxia ischemia [HI]) is unclear. Herein we compared the effects of MIA (injection of poly[I:C] to dam at gestational day 12.5), HI at postnatal day 10, and the combined MIA/HI insult in murine offspring of both sexes. We found that MIA induced autistic-like behaviors without microglial activation but amplified post-HI NFκB signaling, pro-inflammatory responses, and brain injury in offspring. Conversely, HI neither provoked autistic-like behaviors nor concealed them in the MIA offspring. Instead, the dual MIA/HI insult added autistic-like behaviors with diminished synaptic density and reduction of autism-related PSD-95 and Homer-1 in the hippocampus, which were missing in the singular MIA or HI insult. Further, the dual MIA/HI insult enhanced the brain influx of Otx2-positive monocytes that are associated with an increase of perineuronal net-enwrapped parvalbumin neurons. Using CCR2-CreER mice to distinguish monocytes from the resident microglia, we found that the monocytic infiltrates gradually adopted a ramified morphology and expressed the microglial signature genes (Tmem119, P2RY12, and Sall1) in post-MIA/HI brains, with some continuing to express the proinflammatory cytokine TNFα. Finally, genetic or pharmacological obstruction of monocytic influx significantly reduced perineuronal net-enwrapped parvalbumin neurons and autistic-like behaviors in MIA/HI offspring. Together, these results suggest a pathologic role of monocytes in the two-hit (immune plus neonatal HI) model of neurodevelopmental defects.SIGNIFICANCE STATEMENT In autism spectrum disorders (ASDs), prenatal infection or maternal immune activation (MIA) may act as a primer for multiple genetic and environmental factors to impair neurodevelopment. This study examined whether MIA cooperates with neonatal cerebral hypoxia ischemia to promote ASD-like aberrations in mice using a novel two-hit model. It was shown that the combination of MIA and neonatal hypoxia ischemia produces autistic-like behaviors in the offspring, and has synergistic effects in inducing neuroinflammation, monocytic infiltrates, synaptic defects, and perineuronal nets. Furthermore, genetic or pharmacological intervention of the MCP1-CCR2 chemoattractant pathway markedly reduced monocytic infiltrates, perineuronal nets, and autistic-like behaviors. These results suggest reciprocal escalation of immune and neonatal brain injury in a subset of ASD that may benefit from monocyte-targeted treatments.


Asunto(s)
Trastorno Autístico/inmunología , Trastorno Autístico/psicología , Conducta Animal , Discapacidades del Desarrollo/inmunología , Discapacidades del Desarrollo/psicología , Monocitos/inmunología , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/psicología , Femenino , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , FN-kappa B , Parvalbúminas/genética , Poli I-C , Densidad Postsináptica , Embarazo , Transducción de Señal , Conducta Social
3.
J Cell Mol Med ; 25(19): 9434-9446, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34448532

RESUMEN

Naphthalimide derivatives have multiple biological activities, including antitumour and anti-inflammatory activities. We previously synthesized several naphthalimide derivatives; of them, compound 5 was found to exert the strongest inhibitory effect on human DNA topoisomerase II activity. However, the effects of naphthalimide derivatives on platelet activation have not yet been investigated. Therefore, the mechanism underlying the antiplatelet activity of compound 5 was determined in this study. The data revealed that compound 5 (5-10 µM) inhibited collagen- and convulxin- but not thrombin- or U46619-mediated platelet aggregation, suggesting that compound 5 is more sensitive to the inhibition of glycoprotein VI (GPVI) signalling. Indeed, compound 5 could inhibit the phosphorylation of signalling molecules downstream of GPVI, followed by the inhibition of calcium mobilization, granule release and GPIIb/IIIa activation. Moreover, compound 5 prevented pulmonary embolism and prolonged the occlusion time, but tended to prolong the bleeding time, indicating that it can prevent thrombus formation but may increase bleeding risk. This study is the first to demonstrate that the naphthalimide derivative compound 5 exerts antiplatelet and antithrombotic effects. Future studies should modify compound 5 to synthesize more potent and efficient antiplatelet agents while minimizing bleeding risk, which may offer a therapeutic potential for cardiovascular diseases.


Asunto(s)
Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Naftalimidas/farmacología , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Trombosis/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Inmunohistoquímica , Masculino , Ratones , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Microvasos/patología , Estructura Molecular , Naftalimidas/química , Agregación Plaquetaria/efectos de los fármacos , Transducción de Señal , Trombosis/tratamiento farmacológico , Trombosis/etiología , Trombosis/patología
4.
Neurobiol Dis ; 148: 105200, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33248237

RESUMEN

Hypoxia-inducible factor-1α (HIF1α) is a major regulator of cellular adaptation to hypoxia and oxidative stress, and recent advances of prolyl-4-hydroxylase (P4H) inhibitors have produced powerful tools to stabilize HIF1α for clinical applications. However, whether HIF1α provokes or resists neonatal hypoxic-ischemic (HI) brain injury has not been established in previous studies. We hypothesize that systemic and brain-targeted HIF1α stabilization may have divergent effects. To test this notion, herein we compared the effects of GSK360A, a potent P4H inhibitor, in in-vitro oxygen-glucose deprivation (OGD) and in in-vivo neonatal HI via intracerebroventricular (ICV), intraperitoneal (IP), and intranasal (IN) drug-application routes. We found that GSK360A increased the erythropoietin (EPO), heme oxygenase-1 (HO1) and glucose transporter 1 (Glut1) transcripts, all HIF1α target-genes, and promoted the survival of neurons and oligodendrocytes after OGD. Neonatal HI insult stabilized HIF1α in the ipsilateral hemisphere for up to 24 h, and either ICV or IN delivery of GSK360A after HI increased the HIF1α target-gene transcripts and decreased brain damage. In contrast, IP-injection of GSK360A failed to reduce HI brain damage, but elevated the risk of mortality at high doses, which may relate to an increase of the kidney and plasma EPO, leukocytosis, and abundant vascular endothelial growth factor (VEGF) mRNAs in the brain. These results suggest that brain-targeted HIF1α-stabilization is a potential treatment of neonatal HI brain injury, while systemic P4H-inhibition may provoke unwanted adverse effects.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Glicina/análogos & derivados , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Hipoxia-Isquemia Encefálica/metabolismo , Neuronas/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Quinolonas/farmacología , Administración Intranasal , Animales , Animales Recién Nacidos , Supervivencia Celular/efectos de los fármacos , Eritropoyetina/genética , Transportador de Glucosa de Tipo 1/efectos de los fármacos , Transportador de Glucosa de Tipo 1/genética , Glicina/farmacología , Hemo Oxigenasa (Desciclizante)/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inyecciones Intraperitoneales , Inyecciones Intraventriculares , Neuronas/metabolismo , Oligodendroglía/metabolismo , Ratas
5.
Genes Dev ; 27(11): 1272-87, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23723414

RESUMEN

Neural stem cells (NSCs) reside in widespread regions along the lateral ventricle and generate diverse olfactory bulb (OB) interneuron subtypes in the adult mouse brain. Molecular mechanisms underlying their regional diversity, however, are not well understood. Here we show that the homeodomain transcription factor Gsx2 plays a crucial role in the region-specific control of adult NSCs in both persistent and injury-induced neurogenesis. In the intact brain, Gsx2 is expressed in a regionally restricted subset of NSCs and promotes the activation and lineage progression of stem cells, thereby controlling the production of selective OB neuron subtypes. Moreover, Gsx2 is ectopically induced in damaged brains outside its normal expression domains and is required for injury-induced neurogenesis in the subventricular zone (SVZ). These results demonstrate that mobilization of adult NSCs is controlled in a region-specific manner and that distinct mechanisms operate in continuous and injury-induced neurogenesis in the adult brain.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Ventrículos Laterales/citología , Ventrículos Laterales/lesiones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Células Madre Adultas/metabolismo , Animales , Linaje de la Célula , Ventrículos Laterales/metabolismo , Ratones , Células-Madre Neurales/clasificación , Bulbo Olfatorio/citología , Especificidad de Órganos , Nicho de Células Madre , Factores de Transcripción/metabolismo
6.
J Neuroinflammation ; 15(1): 45, 2018 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-29452577

RESUMEN

BACKGROUND: Polymerase δ-interacting protein 2 (Poldip2) is a multifunctional protein that regulates vascular extracellular matrix composition and matrix metalloproteinase (MMP) activity. The blood-brain barrier (BBB) is a dynamic system assembled by endothelial cells, basal lamina, and perivascular astrocytes, raising the possibility that Poldip2 may be involved in maintaining its structure. We investigated the role of Poldip2 in the late BBB permeability induced by cerebral ischemia. METHODS: Transient middle cerebral artery occlusion (tMCAO) was induced in Poldip2+/+ and Poldip2+/- mice. The volume of the ischemic lesion was measured in triphenyltetrazolium chloride-stained sections. BBB breakdown was evaluated by Evans blue dye extravasation. Poldip2 protein expression was evaluated by western blotting. RT-PCR, zymography, and ELISAs were used to measure mRNA levels, activity, and protein levels of cytokines and MMPs. Cultured astrocytes were transfected with Poldip2 siRNA, and mRNA levels of cytokines were evaluated as well as IκBα protein degradation. RESULTS: Cerebral ischemia induced the expression of Poldip2. Compared to Poldip2+/+ mice, Poldip2+/- animals exhibited decreased Evans blue dye extravasation and improved survival 24 h following stroke. Poldip2 expression was upregulated in astrocytes exposed to oxygen and glucose deprivation (OGD) and siRNA-mediated downregulation of Poldip2 abrogated OGD-induced IL-6 and TNF-α expression. In addition, siRNA against Poldip2 inhibited TNF-α-induced IκBα degradation. TNF-α, IL-6, MCP-1, VEGF, and MMP expression induced by cerebral ischemia was abrogated in Poldip2+/- mice. The protective effect of Poldip2 depletion on the increased permeability of the BBB was partially reversed by systemic administration of TNF-α. CONCLUSIONS: Poldip2 is upregulated following ischemic stroke and mediates the breakdown of the BBB by increasing cerebral cytokine production and MMP activation. Therefore, Poldip2 appears to be a promising novel target for the development of therapeutic strategies to prevent the development of cerebral edema in the ischemic brain.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevención & control , Permeabilidad Capilar/fisiología , Proteínas Mitocondriales/deficiencia , Neuroprotección/fisiología , Proteínas Nucleares/deficiencia , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Isquemia Encefálica/diagnóstico por imagen , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
7.
J Neurosci ; 36(6): 2027-43, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26865625

RESUMEN

Growth-associated protein 43 (GAP43), a protein kinase C (PKC)-activated phosphoprotein, is often implicated in axonal plasticity and regeneration. In this study, we found that GAP43 can be induced by the endotoxin lipopolysaccharide (LPS) in rat brain astrocytes both in vivo and in vitro. The LPS-induced astrocytic GAP43 expression was mediated by Toll-like receptor 4 and nuclear factor-κB (NF-κB)- and interleukin-6/signal transducer and activator of transcription 3 (STAT3)-dependent transcriptional activation. The overexpression of the PKC phosphorylation-mimicking GAP43(S41D) (constitutive active GAP43) in astrocytes mimicked LPS-induced process arborization and elongation, while application of a NF-κB inhibitory peptide TAT-NBD or GAP43(S41A) (dominant-negative GAP43) or knockdown of GAP43 all inhibited astrogliosis responses. Moreover, GAP43 knockdown aggravated astrogliosis-induced microglial activation and expression of proinflammatory cytokines. We also show that astrogliosis-conditioned medium from GAP43 knock-down astrocytes inhibited GAP43 phosphorylation and axonal growth, and increased neuronal damage in cultured rat cortical neurons. These proneurotoxic effects of astrocytic GAP43 knockdown were accompanied by attenuated glutamate uptake and expression of the glutamate transporter excitatory amino acid transporter 2 (EAAT2) in LPS-treated astrocytes. The regulation of EAAT2 expression involves actin polymerization-dependent activation of the transcriptional coactivator megakaryoblastic leukemia 1 (MKL1), which targets the serum response elements in the promoter of rat Slc1a2 gene encoding EAAT2. In sum, the present study suggests that astrocytic GAP43 mediates glial plasticity during astrogliosis, and provides beneficial effects for neuronal plasticity and survival and attenuation of microglial activation. SIGNIFICANCE STATEMENT: Astrogliosis is a complex state in which injury-stimulated astrocytes exert both protective and harmful effects on neuronal survival and plasticity. In this study, we demonstrated for the first time that growth-associated protein 43 (GAP43), a well known growth cone protein that promotes axonal regeneration, can be induced in rat brain astrocytes by the proinflammatory endotoxin lipopolysaccharide via both nuclear factor-κB and signal transducer and activator of transcription 3-mediated transcriptional activation. Importantly, LPS-induced GAP43 mediates plastic changes of astrocytes while attenuating astrogliosis-induced microglial activation and neurotoxicity. Hence, astrocytic GAP43 upregulation may serve to indicate beneficial astrogliosis after CNS injury.


Asunto(s)
Astrocitos/efectos de los fármacos , Proteína GAP-43/biosíntesis , Proteína GAP-43/genética , Gliosis/genética , Microglía/efectos de los fármacos , FN-kappa B/genética , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/patología , Factor de Transcripción STAT3/genética , Receptor Toll-Like 4/genética , Animales , Citocinas/biosíntesis , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/genética , Activación de Macrófagos/efectos de los fármacos , Neuronas , Fosforilación , Ratas , Ratas Sprague-Dawley , Transactivadores/biosíntesis , Transactivadores/genética , Factores de Transcripción
8.
Stroke ; 48(12): 3347-3355, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29127268

RESUMEN

BACKGROUND AND PURPOSE: The effects of lytic stroke therapy in patients with sickle cell anemia are unknown, although a recent study suggested that coexistent sickle cell anemia does not increase the risk of cerebral hemorrhage. This finding calls for systemic analysis of the effects of thrombolytic stroke therapy, first in humanized sickle mice, and then in patients. There is also a need for additional predictive markers of sickle cell anemia-associated vasculopathy. METHODS: We used Doppler ultrasound to examine the carotid artery of Townes sickle mice tested their responses to repetitive mild hypoxia-ischemia- and transient hypoxia-ischemia-induced stroke at 3 or 6 months of age, respectively. We also examined the effects of tPA (tissue-type plasminogen activator) treatment in transient hypoxia-ischemia-injured sickle mice. RESULTS: Three-month-old sickle cell (SS) mice showed elevated resistive index in the carotid artery and higher sensitivity to repetitive mild hypoxia-ischemia-induced cerebral infarct. Six-month-old SS mice showed greater resistive index and increased flow velocity without obstructive vasculopathy in the carotid artery. Instead, the cerebral vascular wall in SS mice showed ectopic expression of PAI-1 (plasminogen activator inhibitor-1) and P-selectin, suggesting a proadhesive and prothrombotic propensity. Indeed, SS mice showed enhanced leukocyte and platelet adherence to the cerebral vascular wall, broader fibrin deposition, and higher mortality after transient hypoxia-ischemia. Yet, post-transient hypoxia-ischemia treatment with tPA reduced thrombosis and mortality in SS mice. CONCLUSIONS: Sickle mice are sensitive to hypoxia/ischemia-induced cerebral infarct but benefit from thrombolytic treatment. An increased resistive index in carotid arteries may be an early marker of sickle cell vasculopathy.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Fibrinolíticos/uso terapéutico , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/tratamiento farmacológico , Activador de Tejido Plasminógeno/uso terapéutico , Anemia de Células Falciformes/diagnóstico por imagen , Animales , Arterias Carótidas/diagnóstico por imagen , Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Enfermedades de las Arterias Carótidas/fisiopatología , Infarto Cerebral/diagnóstico por imagen , Infarto Cerebral/genética , Circulación Cerebrovascular , Masculino , Ratones , Selectina-P/biosíntesis , Selectina-P/genética , Adhesividad Plaquetaria , Serpina E2/biosíntesis , Serpina E2/genética , Accidente Cerebrovascular/diagnóstico por imagen , Ultrasonografía Doppler
9.
Acta Neuropathol ; 134(4): 585-604, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28725968

RESUMEN

Treatment of acute ischemic stroke with the thrombolytic tissue plasminogen activator (tPA) can significantly improve neurological outcomes; however, thrombolytic therapy is associated with an increased risk of intra-cerebral hemorrhage (ICH). Previously, we demonstrated that during stroke tPA acting on the parenchymal side of the neurovascular unit (NVU) can increase blood-brain barrier (BBB) permeability and ICH through activation of latent platelet-derived growth factor-CC (PDGF-CC) and signaling by the PDGF receptor-α (PDGFRα). However, in vitro, activation of PDGF-CC by tPA is very inefficient and the mechanism of PDGF-CC activation in the NVU is not known. Here, we show that the integrin Mac-1, expressed on brain microglia/macrophages (denoted microglia throughout), acts together with the endocytic receptor LRP1 in the NVU to promote tPA-mediated activation of PDGF-CC. Mac-1-deficient mice (Mac-1-/-) are protected from tPA-induced BBB permeability but not from permeability induced by intracerebroventricular injection of active PDGF-CC. Immunofluorescence analysis demonstrates that Mac-1, LRP1, and the PDGFRα all localize to the NVU of arterioles, and following middle cerebral artery occlusion (MCAO) Mac-1-/- mice show significantly less PDGFRα phosphorylation, BBB permeability, and infarct volume compared to wild-type mice. Bone-marrow transplantation studies indicate that resident CD11b+ cells, but not bone-marrow-derived leukocytes, mediate the early activation of PDGF-CC by tPA after MCAO. Finally, using a model of thrombotic stroke with late thrombolysis, we show that wild-type mice have an increased incidence of spontaneous ICH following thrombolysis with tPA 5 h after MCAO, whereas Mac-1-/- mice are resistant to the development of ICH even with late tPA treatment. Together, these results indicate that Mac-1 and LRP1 act as co-factors for the activation of PDGF-CC by tPA in the NVU, and suggest a novel mechanism for tightly regulating PDGFRα signaling in the NVU and controlling BBB permeability.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Permeabilidad Capilar/fisiología , Linfocinas/metabolismo , Microglía/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Arteriolas/efectos de los fármacos , Arteriolas/metabolismo , Arteriolas/patología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Antígeno CD11b/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Modelos Animales de Enfermedad , Femenino , Fibrinolíticos/efectos adversos , Fibrinolíticos/farmacología , Leucocitos/metabolismo , Leucocitos/patología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Antígeno de Macrófago-1/genética , Antígeno de Macrófago-1/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Receptores de LDL/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/patología , Activador de Tejido Plasminógeno/efectos adversos , Activador de Tejido Plasminógeno/farmacología , Proteínas Supresoras de Tumor/metabolismo
10.
Eur J Neurosci ; 43(2): 245-57, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26547131

RESUMEN

Mitochondria play a critical role in various pathways of regulated cell death. Here we propose a novel method for detection of initial derangement of mitochondria in degenerating and dying neuronal cells. The method is based on our recent finding that antibodies directed against the cannabinoid type 1 receptor (CB1) also bind the mitochondrial stomatin-like protein 2 (SLP2) that belongs to an inner mitochondrial membrane protein complex. It is well established that SLP2 regulates mitochondrial biogenesis and respiratory functions. We now show that anti-CB1 antibodies recognize conformational epitopes but not the linear amino acid sequence of SLP2. In addition we found that anti-CB1 serum mostly labels swollen mitochondria with early or advanced stages of pathology in mouse brain while other proteins of the complex may mask epitopes of SLP2 in the normal mitochondria. Although neurons and endothelial cells in healthy brains contain occasional immunopositive mitochondria detectable with anti-CB1 serum, their numbers increase significantly after hypoxic insults in parallel with signs of cellular damage. Moreover, use of electron microscopy suggests relocation of SLP2 from its normal functional position in the inner mitochondrial membrane into the mitochondrial matrix in pathological cells. Thus, SLP2-like immunolabeling serves as an in situ histochemical target detecting early derangement of mitochondria. Anti-CB1 serum is crucial for this purpose because available anti-SLP2 antibodies do not provide selective labeling of mitochondria in the fixed tissue. This new method of detecting mitochondrial dysfunction can benefit the in vitro research of human diseases and developmental disorders by enabling analysis in live animal models.


Asunto(s)
Encéfalo/inmunología , Encéfalo/ultraestructura , Inmunohistoquímica/métodos , Proteínas de la Membrana/inmunología , Mitocondrias/ultraestructura , Proteínas Mitocondriales/inmunología , Proteínas del Tejido Nervioso/inmunología , Neuronas/inmunología , Neuronas/ultraestructura , Receptor Cannabinoide CB1/inmunología , Animales , Anticuerpos , Muerte Celular , Hipoxia de la Célula , Epítopos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL
11.
J Neurosci ; 34(49): 16467-81, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25471584

RESUMEN

Intrauterine infection (chorioamnionitis) aggravates neonatal hypoxic-ischemic (HI) brain injury, but the mechanisms linking systemic inflammation to the CNS damage remain uncertain. Here we report evidence for brain influx of T-helper 17 (TH17)-like lymphocytes to coordinate neuroinflammatory responses in lipopolysaccharide (LPS)-sensitized HI injury in neonates. We found that both infants with histological chorioamnionitis and rat pups challenged by LPS/HI have elevated expression of the interleukin-23 (IL-23) receptor, a marker of early TH17 lymphocytes, in the peripheral blood mononuclear cells. Post-LPS/HI administration of FTY720 (fingolimod), a sphingosine-1-phosphate receptor agonist that blocks lymphocyte trafficking, mitigated the influx of leukocytes through the choroid plexus and acute induction of nuclear factor-κB signaling in the brain. Subsequently, the FTY720 treatment led to attenuated blood-brain barrier damage, fewer cluster of differentiation 4-positive, IL-17A-positive T-cells in the brain, less proinflammatory cytokine, and better preservation of growth and white matter functions. The FTY720 treatment also provided dose-dependent reduction of brain atrophy, rescuing >90% of LPS/HI-induced brain tissue loss. Interestingly, FTY720 neither opposed pure-HI brain injury nor directly inhibited microglia in both in vivo and in vitro models, highlighting its unique mechanism against inflammation-sensitized HI injury. Together, these results suggest that the dual hit of systemic inflammation and neonatal HI injury triggers early onset of the TH17/IL-17-mediated immunity, which causes severe brain destruction but responds remarkably to the therapeutic blockade of lymphocyte trafficking.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Hipoxia-Isquemia Encefálica/patología , Hipoxia-Isquemia Encefálica/prevención & control , Inflamación/prevención & control , Activación de Linfocitos/efectos de los fármacos , Linfocitos/efectos de los fármacos , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Animales , Animales Recién Nacidos , Atrofia/tratamiento farmacológico , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Corioamnionitis/tratamiento farmacológico , Corioamnionitis/metabolismo , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Clorhidrato de Fingolimod , Humanos , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Recién Nacido , Lipopolisacáridos , Linfocitos/citología , FN-kappa B/metabolismo , Embarazo , Glicoles de Propileno/uso terapéutico , Ratas , Receptores de Interleucina/metabolismo , Esfingosina/farmacología , Esfingosina/uso terapéutico , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Sustancia Blanca/efectos de los fármacos
12.
Glia ; 63(7): 1138-54, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25690886

RESUMEN

The aryl hydrocarbon receptor (AhR) regulates peripheral immunity; but its role in microglia-mediated neuroinflammation in the brain remains unknown. Here, we demonstrate that AhR mediates both anti-inflammatory and proinflammatory effects in lipopolysaccharide (LPS)-activated microglia. Activation of AhR by its ligands, formylindolo[3,2-b]carbazole (FICZ) or 3-methylcholanthrene (3MC), attenuated LPS-induced microglial immune responses. AhR also showed proinflammatory effects, as evidenced by the findings that genetic silence of AhR ameliorated the LPS-induced microglial immune responses and LPS-activated microglia-mediated neurotoxicity. Similarly, LPS-induced expressions of tumor necrosis factor α (TNFα) and inducible nitric oxide synthase (iNOS) were reduced in the cerebral cortex of AhR-deficient mice. Intriguingly, LPS upregulated and activated AhR in the absence of AhR ligands via the MEK1/2 signaling pathway, which effects were associated with a transient inhibition of cytochrome P450 1A1 (CYP1A1). Although AhR ligands synergistically enhance LPS-induced AhR activation, leading to suppression of LPS-induced microglial immune responses, they cannot do so on their own in microglia. Chromatin immunoprecipitation results further revealed that LPS-FICZ co-treatment, but not LPS alone, not only resulted in co-recruitment of both AhR and NFκB onto the κB site of TNFα gene promoter but also reduced LPS-induced AhR binding to the DRE site of iNOS gene promoter. Together, we provide evidence showing that microglial AhR, which can be activated by LPS, exerts bi-directional effects on the regulation of LPS-induced neuroinflammation, depending on the availability of external AhR ligands. These findings confer further insights into the potential link between environmental factors and the inflammatory brain disorders.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Microglía/fisiología , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Muerte Celular/fisiología , Línea Celular , Células Cultivadas , Corteza Cerebral/inmunología , Cromatina/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Técnicas de Silenciamiento del Gen , Lipopolisacáridos , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Ratones Endogámicos BALB C , Ratones Noqueados , Neuronas/fisiología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores de Hidrocarburo de Aril/genética , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
13.
Stroke ; 46(7): 1947-55, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26060244

RESUMEN

BACKGROUND AND PURPOSE: Hypoperfusion-induced thrombosis is an important mechanism for postsurgery stroke and cognitive decline, but there are no perioperative neuroprotectants to date. This study investigated whether prophylactic application of Edaravone, a free radical scavenger already used in treating ischemic stroke in Japan, can prevent infarct and cognitive deficits in a murine model of transient cerebral hypoxia-ischemia. METHODS: Adult male C57BL/6 mice were subjected to transient hypoxic-ischemic (tHI) insult that consists of 30-minute occlusion of the unilateral common carotid artery and exposure to 7.5% oxygen. Edaravone or saline was prophylactically applied to compare their effects on cortical oxygen saturation, blood flow, coagulation, oxidative stress, metabolites, and learning-memory using methods that include photoacoustic imaging, laser speckle contrast imaging, solid-state NMR, and Morris water maze. The effects on infarct size by Edaravone application at different time points after tHI were also compared. RESULTS: Prophylactic administration of Edaravone (4.5 mg/kg×2, IP, 1 hour before and 1 hour after tHI) improved vascular reperfusion, oxygen saturation, and the maintenance of brain metabolites, reducing oxidative stress, thrombosis, white-matter injury, and learning impairment after tHI insult. Delayed Edaravone treatment after 3 h post-tHI became unable to reduce infarct size. CONCLUSIONS: Acute application of Edaravone may be a useful strategy to prevent postsurgery stroke and cognitive impairment, especially in patients with severe carotid stenosis.


Asunto(s)
Antipirina/análogos & derivados , Depuradores de Radicales Libres/administración & dosificación , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Atención Perioperativa , Animales , Antipirina/administración & dosificación , Edaravona , Hipoxia-Isquemia Encefálica/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Atención Perioperativa/métodos , Distribución Aleatoria
14.
Cereb Cortex ; 23(5): 1218-29, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22556277

RESUMEN

Intrauterine infection exacerbates neonatal hypoxic-ischemic (HI) brain injury and impairs the development of cerebral cortex. Here we used low-dose lipopolysaccharide (LPS) pre-exposure followed by unilateral cerebral HI insult in 7-day-old rats to study the pathogenic mechanisms. We found that LPS pre-exposure blocked the HI-induced proteolytic activity of tissue-type plasminogen activator (tPA), but significantly enhanced NF-κB signaling, microglia activation, and the production of pro-inflammatory cytokines in newborn brains. Remarkably, these pathogenic responses were all blocked by intracerebroventricular injection of a stable-mutant form of plasminogen activator protein-1 called CPAI. Similarly, LPS pre-exposure amplified, while CPAI therapy mitigated HI-induced blood-brain-barrier damage and the brain tissue loss with a therapeutic window at 4 h after the LPS/HI insult. The CPAI also blocks microglia activation following a brain injection of LPS, which requires the contribution by tPA, but not the urinary-type plasminogen activator (uPA), as shown by experiments in tPA-null and uPA-null mice. These results implicate the nonproteolytic tPA activity in LPS/HI-induced brain damage and microglia activation. Finally, the CPAI treatment protects near-normal motor and white matter development despite neonatal LPS/HI insult. Together, because CPAI blocks both proteolytic and nonproteolytic tPA neurotoxicity, it is a promising therapeutics of neonatal HI injury either with or without infection.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/prevención & control , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/prevención & control , Lipopolisacáridos , Inhibidor 1 de Activador Plasminogénico/farmacología , Activador de Tejido Plasminógeno/metabolismo , Animales , Animales Recién Nacidos , Encefalitis/inducido químicamente , Encefalitis/metabolismo , Encefalitis/prevención & control , Hipoxia-Isquemia Encefálica/inducido químicamente , Ratones , Ratas
15.
Biomed Pharmacother ; 173: 116404, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38471275

RESUMEN

High-fat diet (HFD)-induced fatty liver disease is a deteriorating risk factor for Alzheimer's disease (AD). Mitigating fatty liver disease has been shown to attenuate AD-like pathology in animal models. However, it remains unclear whether enhancing Aß clearance through immunotherapy would in turn attenuate HFD-induced fatty liver or whether its efficacy would be compromised by long-term exposure to HFD. Here, the therapeutic potentials of an anti-Aß antibody, NP106, was investigated in APP/PS1 mice by HFD feeding for 44 weeks. The data demonstrate that NP106 treatment effectively reduced Aß burden and pro-inflammatory cytokines in HFD-fed APP/PS1 mice and ameliorated HFD-aggravated cognitive impairments during the final 18 weeks of the study. The rejuvenating characteristics of microglia were evident in APP/PS1 mice with NP106 treatment, namely enhanced microglial Aß phagocytosis and attenuated microglial lipid accumulation, which may explain the benefits of NP106. Surprisingly, NP106 also reduced HFD-induced hyperglycemia, fatty liver, liver fibrosis, and hepatic lipids, concomitant with modifications in the expressions of genes involved in hepatic lipogenesis and fatty acid oxidation. The data further reveal that brain Aß burden and behavioral deficits were positively correlated with the severity of fatty liver disease and fasting serum glucose levels. In conclusion, our study shows for the first time that anti-Aß immunotherapy using NP106, which alleviates AD-like disorders in APP/PS1 mice, ameliorates fatty liver disease. Minimizing AD-related pathology and symptoms may reduce the vicious interplay between central AD and peripheral fatty liver disease, thereby highlighting the importance of developing AD therapies from a systemic disease perspective.


Asunto(s)
Enfermedad de Alzheimer , Hígado Graso , Hepatopatías , Ratones , Animales , Precursor de Proteína beta-Amiloide/metabolismo , Ratones Transgénicos , Dieta Alta en Grasa/efectos adversos , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Hepatopatías/metabolismo , Hígado Graso/metabolismo , Modelos Animales de Enfermedad , Péptidos beta-Amiloides/metabolismo
16.
Stroke ; 44(9): 2623-2627, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23881953

RESUMEN

BACKGROUND AND PURPOSE: Plasminogen activator inhibitor-I (PAI-1), a ≈50-kDa serine protease inhibitor, markedly reduces the extravascular toxicity of tissue-type plasminogen activator in experimental hypoxic-ischemic (HI) brain injury of newborns. However, the current treatment with PAI-1 requires intracerebroventricle injection to cross the blood-brain barrier, which is an invasive procedure of limited clinical potential. Thus, we tested whether intranasal administration of PAI-1 can bypass blood-brain barrier and mitigate neonatal HI brain injury. METHODS: Rat pups were subjected to HI, with or without lipopolysaccharide pre-exposure, followed by intranasal delivery of a stable-mutant form of PAI-1 (CPAI). RESULTS: Immunoblotting showed that CPAI sequentially entered the olfactory bulbs and cerebral cortex after intranasal delivery and reduced ≈75% of brain atrophy in HI or lipopolysaccharide-sensitized HI injury. Mechanistically, CPAI attenuated HI-induced plasminogen activators and lipopolysaccharide/HI-induced nuclear factor-κB signaling, neuroinflammation, and blood-brain barrier permeability. CONCLUSIONS: Intranasal delivery of CPAI is an effective treatment of experimental HI brain injury of newborns. Clinical application of this experimental therapy merits further investigation.


Asunto(s)
Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Inhibidor 1 de Activador Plasminogénico/administración & dosificación , Inactivadores Plasminogénicos/administración & dosificación , Inactivadores Plasminogénicos/uso terapéutico , Administración Intranasal , Animales , Animales Recién Nacidos , Atrofia , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Esquema de Medicación , Femenino , Hipoxia-Isquemia Encefálica/patología , Hipoxia-Isquemia Encefálica/fisiopatología , Masculino , Inhibidor 1 de Activador Plasminogénico/uso terapéutico , Ratas , Ratas Wistar , Inhibidores de Serina Proteinasa/administración & dosificación , Inhibidores de Serina Proteinasa/uso terapéutico
17.
Front Neurol ; 14: 1215876, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37822524

RESUMEN

Patients with sickle cell anemia (SCA) have a high incidence of ischemic stroke, but are usually excluded from thrombolytic therapy due to concerns for cerebral hemorrhage. Maladaptation to cerebral ischemia may also contribute to the stroke propensity in SCA. Here we compared post-stroke cortical collateral circulation in transgenic sickle (SS) mice, bone marrow grafting-derived SS-chimera, and wildtype (AA) controls, because collateral circulation is a critical factor for cell survival within the ischemic penumbra. Further, it has been shown that SS mice develop poorer neo-collateral perfusion after limb ischemia. We used the middle cerebral artery (MCA)-targeted photothrombosis model in this study, since it is better tolerated by SS mice and creates a clear infarct core versus peri-infarct area. Compared to AA mice, SS mice showed enlarged infarction and lesser endothelial proliferation after photothrombosis. SS-chimera showed anemia, hypoxia-induced erythrocyte sickling, and attenuated recovery of blood flow in the ipsilateral cortex after photothrombosis. In AA chimera, cerebral blood flow in the border area between MCA and the anterior cerebral artery (ACA) and posterior cerebral artery (PCA) trees improved from 44% of contralateral level after stroke to 78% at 7 d recovery. In contrast, blood flow in the MCA-ACA and MCA-PCA border areas only increased from 35 to 43% at 7 d post-stroke in SS chimera. These findings suggest deficits of post-stroke collateral circulation in SCA. Better understanding of the underpinnings may suggest novel stroke therapies for SCA patients.

18.
Thromb Res ; 225: 63-72, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37030187

RESUMEN

INTRODUCTION: Paclitaxel is a microtubule-stabilizing drug used to treat several types of cancer, including ovarian and breast cancer. Because of its antiproliferative effect on vascular smooth muscle cells, balloons and stents are coated with paclitaxel for use in coronary revascularization and prevention of in-stent restenosis (ISR). However, mechanisms underlying ISR are complicated. Platelet activation is one of the major causes of ISR after percutaneous coronary intervention. Although the antiplatelet activity of paclitaxel was noted in rabbit platelets, the effect of paclitaxel on platelets remains unclear. This study investigated whether paclitaxel exhibits antiplatelet activity in human platelets. METHODS AND RESULTS: Paclitaxel inhibited platelet aggregation induced by collagen but not that induced by thrombin, arachidonic acid, or U46619, suggesting that paclitaxel is more sensitive to the inhibition of collagen-induced platelet activation. Moreover, paclitaxel blocked collagen receptor glycoprotein (GP) VI downstream signaling molecules, including Lyn, Fyn, PLCγ2, PKC, Akt, and MAPKs. However, paclitaxel did not directly bind to GPVI and cause GPVI shedding, as detected by surface plasmon resonance and flow cytometry, respectively, indicating that paclitaxel may interfere with GPVI downstream signaling molecules, such as Lyn and Fyn. Paclitaxel also prevented granule release and GPIIbIIIa activation induced by collagen and low convulxin doses. Moreover, paclitaxel attenuated pulmonary thrombosis and delayed platelet thrombus formation in mesenteric microvessels without significantly affecting hemostasis. CONCLUSION: Paclitaxel exerts antiplatelet and antithrombotic effects. Thus, paclitaxel may provide additional benefits beyond its antiproliferative effect when used in drug-coated balloons and drug-eluting stents for coronary revascularization and prevention of ISR.


Asunto(s)
Reestenosis Coronaria , Stents Liberadores de Fármacos , Intervención Coronaria Percutánea , Animales , Humanos , Conejos , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Stents Liberadores de Fármacos/efectos adversos , Fibrinolíticos , Reestenosis Coronaria/etiología , Reestenosis Coronaria/terapia , Resultado del Tratamiento , Stents/efectos adversos , Intervención Coronaria Percutánea/efectos adversos , Colágeno , Angiografía Coronaria/efectos adversos
19.
Toxics ; 10(11)2022 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-36355934

RESUMEN

Pulmonary fibrosis, a chronic lung disease caused by progressive deterioration of lung tissue, is generated by several factors including genetic and environmental ones. In response to long-term exposure to environmental stimuli, aberrant tissue repair and epithelial cell-to- mesenchymal cell transition (EMT) trigger the subsequent progression of pulmonary fibrotic diseases. The Aryl hydrocarbon receptor (AhR) is a transcription factor that is activated by ligands providing lung dysfunction when activated by environmental toxins, such as polycyclic aromatic hydrocarbons. Our previous study demonstrated that AhR mediates α-SMA expression by directly binding to the α-SMA (fibroblast differentiation marker) promoter, suggesting the role of AhR in mediating fibrogenic progression. Here we follow the hypothesis that macrophage infiltrated microenvironments may trigger inflammation and subsequent fibrosis. We studied the expression of cytokines in RAW 264.7 cells by AhR activation through an ELISA assay. To investigate molecular events, migration, western blotting and zymography assays were carried out. We found that AhR agonists such as TCDD, IP and FICZ, promote the migration and induce inflammatory mediators such as TNF-α and G-CSF, MIP-1α, MIP-1ß and MIP-2. These cytokines arbitrate EMT marker expression such as E-cadherin, fibronectin, and vimentin in pulmonary epithelial cells. Expression of proteins of MMPs in mouse macrophages was determined by zymography, showing the caseinolytic activity of MMP-1 and the gelatinolytic action of MMP-2 and MMP-9. Taken together, the present study showed that AhR activated macrophages create an inflammatory microenvironment which favours the fibrotic progression of pulmonary epithelial cells. Such production of inflammatory factors was accomplished by affecting the Wnt/ß-catenin signalling pathway, thereby creating a microenvironment which enhances the epithelial-mesenchymal transition, leading to fibrosis of the lung.

20.
Biomedicines ; 10(4)2022 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-35453528

RESUMEN

Spinal cord injury (SCI) promotes brain inflammation; conversely, brain injury promotes spinal neuron loss. There is a need to identify molecular biomarkers and therapeutic targets for central nervous system (CNS) injury. CDGSH iron-sulfur structural domain 2 (CISD2), an NF-κB antagonist, is downregulated after injury in vivo and in vitro. We aimed to examine the diagnostic value of CISD2 in patients with CNS insult. Plasma and cerebrospinal fluid (CSF) CISD2 levels were decreased in 13 patients with CNS insult and were negatively correlated with plasma IL6 levels (associated with disease severity; r = −0.7062; p < 0.01). SCI-induced inflammatory mediators delivered through CSF promoted mouse brain inflammation at 1 h post-SCI. Anti-CISD2 antibody treatment exacerbated SCI-induced inflammation in mouse spine and brain. Lipopolysaccharide-stimulated siCISD2-transfected EOC microglial cells exhibited proinflammatory phenotypes (enhanced M1 polarization, decreased M2 polarization, and increased intranuclear NF-κB p65 translocation). Plasma and CSF CISD2 levels were increased in three patients with CNS insult post-therapeutic hypothermia. CISD2 levels were negatively correlated with plasma and CSF levels of inflammatory mediators. CISD2 inhibition and potentiation experiments in cells, animals, and humans revealed CISD2 as a biomarker for CNS insult and upregulation of CISD2 anti-inflammatory properties as a potential therapeutic strategy for CNS insult.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA