Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Blood ; 144(5): 552-564, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38820589

RESUMEN

ABSTRACT: Chronic kidney disease (CKD) is a major contributor to morbidity and mortality in sickle cell disease (SCD). Anemia, induced by chronic persistent hemolysis, is associated with the progressive deterioration of renal health, resulting in CKD. Moreover, patients with SCD experience acute kidney injury (AKI), a risk factor for CKD, often during vaso-occlusive crisis associated with acute intravascular hemolysis. However, the mechanisms of hemolysis-driven pathogenesis of the AKI-to-CKD transition in SCD remain elusive. Here, we investigated the role of increased renovascular rarefaction and the resulting substantial loss of the vascular endothelial protein C receptor (EPCR) in the progressive deterioration of renal function in transgenic SCD mice. Multiple hemolytic events raised circulating levels of soluble EPCR (sEPCR), indicating loss of EPCR from the cell surface. Using bone marrow transplantation and super-resolution ultrasound imaging, we demonstrated that SCD mice overexpressing EPCR were protective against heme-induced CKD development. In a cohort of patients with SCD, plasma sEPCR was significantly higher in individuals with CKD than in those without CKD. This study concludes that multiple hemolytic events may trigger CKD in SCD through the gradual loss of renovascular EPCR. Thus, the restoration of EPCR may be a therapeutic target, and plasma sEPCR can be developed as a prognostic marker for sickle CKD.


Asunto(s)
Anemia de Células Falciformes , Receptor de Proteína C Endotelial , Hemo , Ratones Transgénicos , Insuficiencia Renal Crónica , Animales , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/patología , Anemia de Células Falciformes/metabolismo , Anemia de Células Falciformes/sangre , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/etiología , Receptor de Proteína C Endotelial/metabolismo , Receptor de Proteína C Endotelial/genética , Ratones , Hemo/metabolismo , Humanos , Masculino , Femenino , Hemólisis , Riñón/metabolismo , Riñón/patología
2.
Neurochem Int ; 173: 105660, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38151109

RESUMEN

Traumatic brain injury (TBI) presents a significant global health challenge with no effective therapies developed to date. Regulatory T lymphocytes (Tregs) have recently emerged as a potential therapy due to their critical roles in maintaining immune homeostasis, reducing inflammation, and promoting brain repair. Following TBI, fluctuations in Treg populations and shifts in their functionality have been noted. However, the precise impact of Tregs on the pathophysiology of TBI remains unclear. In this review, we discuss recent advances in understanding the intricate roles of Tregs in TBI and other brain diseases. Increased knowledge about Tregs may facilitate their future application as an immunotherapy target for TBI treatment.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Linfocitos T Reguladores , Humanos , Lesiones Traumáticas del Encéfalo/terapia , Encéfalo , Inflamación
3.
PNAS Nexus ; 2(5): pgad149, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37215630

RESUMEN

White-matter injury in sickle-cell disease (SCD) includes silent cerebral infarction diagnosed by diffusion tensor imaging (DTI), a complication associated with cognitive dysfunction in children with SCD. The link between white-matter injury and cognitive dysfunction has not been fully elucidated. The goal of this study was to define whether cerebrovascular lesions and cognitive function in SCD are linked to neuroaxonal damage and astrocyte activation in humanized Townes' SCD mice homozygous for human sickle hemoglobin S (SS) and control mice homozygous for human normal hemoglobin A (AA). Mice underwent MRI with DTI and cognitive testing, and histology sections from their brains were stained to assess microstructural tissue damage, neuroaxonal damage, and astrocyte activation. Fractional anisotropy, showing microstructural cerebrovascular abnormalities identified by DTI in the white matter, was significantly associated with neuronal demyelination in the SS mouse brain. SS mice had reduced learning and memory function with a significantly lower discrimination index compared with AA control mice in the novel object recognition tests. Neuroaxonal damage in the SS mice was synchronously correlated with impaired neurocognitive function and activation of astrocytes. The interplay between astrocyte function and neurons may modulate cognitive performance in SCD.

4.
Biomedicines ; 11(3)2023 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-36979670

RESUMEN

Occlusion of cerebral blood vessels causes acute cerebral hypoxia-an important trigger of ischemic white matter injury and stroke in sickle cell disease (SCD). While chronic hypoxia triggers compensatory neuroprotection via insulin-like growth factor-1 (IGF-1) and hypoxia inducible factor-1α (HIF-1α), severe bouts of acute hypoxia and subsequent restoration of blood flow (hypoxia/reoxygenation, H/R) overwhelm compensatory mechanisms and cause neuroaxonal damage-identified as white matter lesions-in the brain. The neuroprotective role of IGF-1 in the pathogenesis of white matter injury in SCD has not been investigated; however, it is known that systemic IGF-1 is reduced in individuals with SCD. We hypothesized that IGF-1 supplementation may prevent H/R-induced white matter injury in SCD. Transgenic sickle mice homozygous for human hemoglobin S and exposed to H/R developed white matter injury identified by elevated expression of non-phosphorylated neurofilament H (SMI32) with a concomitant decrease in myelin basic protein (MBP) resulting in an increased SMI32/MBP ratio. H/R-challenge also lowered plasma and brain IGF-1 expression. Human recombinant IGF-1 prophylaxis significantly induced HIF-1α and averted H/R-induced white matter injury in the sickle mice compared to vehicle-treated mice. The expression of the IGF-1 binding proteins IGFBP-1 and IGFBP-3 was elevated in the IGF-1-treated brain tissue indicating their potential role in mediating neuroprotective HIF-1α signaling. This study provides proof-of-concept for IGF-1-mediated neuroprotection in SCD.

5.
CNS Neurosci Ther ; 29(3): 783-788, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36550598

RESUMEN

In the ischemic brain, hypoxia leads to mitochondrial dysfunction, insufficient energy production, and astrocyte activation. Yet, most studies investigating mitochondrial dysfunction in cerebral ischemia have focused exclusively on neurons. This review will highlight the importance of the morphological, molecular, and functional heterogeneity of astrocytes in their role in brain injuries and explore how activated astrocytes exhibit calcium imbalance, reactive oxygen species overproduction, and apoptosis. In addition, special focus will be given to the role of the mitochondrial protein frataxin in activated astrocytes during ischemia and its putative role in the pharmacological management of cerebral ischemia.


Asunto(s)
Isquemia Encefálica , Proteínas Mitocondriales , Humanos , Astrocitos/metabolismo , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Mitocondrias/fisiología , Proteínas Mitocondriales/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Frataxina
6.
Semin Immunopathol ; 45(3): 329-346, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36469056

RESUMEN

Unrestrained excessive inflammatory responses exacerbate ischemic brain injury and impede post-stroke brain recovery. CD4+CD25+Foxp3+ regulatory T (Treg) cells play important immunosuppressive roles to curtail inflammatory responses and regain immune homeostasis after stroke. Accumulating evidence confirms that Treg cells are neuroprotective at the acute stage after stroke and promote brain repair at the chronic phases. The beneficial effects of Treg cells are mediated by diverse mechanisms involving cell-cell interactions and soluble factor release. Multiple types of cells, including both immune cells and non-immune CNS cells, have been identified to be cellular targets of Treg cells. In this review, we summarize recent findings regarding the function of Treg cells in ischemic stroke and the underlying cellular and molecular mechanisms. The protective and reparative properties of Treg cells endorse them as good candidates for immune therapy. Strategies that boost the numbers and functions of Treg cells have been actively developing in the fields of transplantation and autoimmune diseases. We discuss the approaches for Treg cell expansion that have been tested in stroke models. The application of these approaches to stroke patients may bring new hope for stroke treatments.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Linfocitos T Reguladores , Accidente Cerebrovascular/terapia , Inmunosupresores
7.
Blood ; 135(13): 1044-1048, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32043112

RESUMEN

Acute kidney injury (AKI) is a major clinical concern in sickle cell disease (SCD). Clinical evidence suggests that red cell alarmins may cause AKI in SCD, however, the sterile inflammatory process involved has hitherto not been defined. We discovered that hemopexin deficiency in SCD is associated with a compensatory increase in α-1-microglobulin (A1M), resulting in an up to 10-fold higher A1M-to-hemopexin ratio in SCD compared with healthy controls. The A1M-to-hemopexin ratio is associated with markers of hemolysis and AKI in both humans and mice with SCD. Studies in mice showed that excess heme is directed to the kidneys in SCD in a process involving A1M causing AKI, whereas excess heme in controls is transported to the liver as expected. Using genetic and bone marrow chimeric tools, we confirmed that hemopexin deficiency promotes AKI in sickle mice under hemolytic stress. However, AKI was blocked when hemopexin deficiency in sickle mice was corrected with infusions of purified hemopexin prior to the induction of hemolytic stress. This study identifies acquired hemopexin deficiency as a risk factor of AKI in SCD and hemopexin replacement as a potential therapy.


Asunto(s)
Lesión Renal Aguda/etiología , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/genética , Susceptibilidad a Enfermedades , Hemopexina/deficiencia , Lesión Renal Aguda/diagnóstico , Animales , Biopsia , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Eritrocitos/metabolismo , Tasa de Filtración Glomerular , Hemo/metabolismo , Humanos , Pruebas de Función Renal , Ratones , Modelos Biológicos
8.
Br J Haematol ; 182(2): 271-275, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29923176

RESUMEN

Acute chest syndrome (ACS) mortality in sickle cell disease (SCD) rises sharply in young adult patients and mechanism-based prophylaxis is lacking. In SCD, haem oxygenase-1 (HO-1) declines with age and ACS is associated with low HO-1. To test if enhanced HO-1 can reduce ACS mortality, young SCD mice were treated with D3T (3H-1,2-dithiole-3-thione), an activator of nuclear-factor erythroid 2 like 2, which controls HO-1 expression, for 3 months. Following haem-induced ACS, all vehicle-treated mice succumbed to severe lung injury, while D3T-treated mice had significantly improved survival. Blocking HO-1 activity abrogated the D3T effect. Thus HO-1 may be targeted to reduce ACS severity in adult patients.


Asunto(s)
Síndrome Torácico Agudo/prevención & control , Factor 2 Relacionado con NF-E2/fisiología , Síndrome Torácico Agudo/inducido químicamente , Animales , Hematínicos/farmacología , Hemo-Oxigenasa 1/metabolismo , Hemina/toxicidad , Ratones Transgénicos , Oxígeno/sangre , Tionas/farmacología , Tiofenos/farmacología
9.
Brain Struct Funct ; 223(4): 1731-1745, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29204911

RESUMEN

Previous studies have shown that exposure to stressful events can enhance fear memory and anxiety-like behavior as well as increase synaptic plasticity in the rat basolateral amygdala (BLA). We have evidence that repeated unpredictable shock stress (USS) elicits a long-lasting increase in anxiety-like behavior in rats, but the cellular mechanisms mediating this response remain unclear. Evidence from recent morphological studies suggests that alterations in the dendritic arbor or spine density of BLA principal neurons may underlie stress-induced anxiety behavior. Recently, we have shown that the induction of long-term potentiation (LTP) in BLA principal neurons is dependent on activation of postsynaptic D1 dopamine receptors and the subsequent activation of the cyclic adenosine 5'-monophosphate (cAMP)-protein kinase A (PKA) signaling cascade. Here, we have used in vitro whole-cell patch-clamp recording from BLA principal neurons to investigate the long-term consequences of USS on their morphological properties and synaptic plasticity. We provided evidence that the enhanced anxiety-like behavior in response to USS was not associated with any significant change in the morphological properties of BLA principal neurons, but was associated with a changed frequency dependence of synaptic plasticity, lowered LTP induction threshold, and reduced expression of phosphodiesterase type 4 enzymes (PDE4s). Furthermore, pharmacological inhibition of PDE4 activity with rolipram mimics the effects of chronic stress on LTP induction threshold and baseline startle. Our results provide the first evidence that stress both enhances anxiety-like behavior and facilitates synaptic plasticity in the amygdala through a common mechanism of PDE4-mediated disinhibition of cAMP-PKA signaling.


Asunto(s)
Complejo Nuclear Basolateral/patología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Estrés Psicológico/patología , Estimulación Acústica/efectos adversos , Animales , Ansiedad/etiología , Complejo Nuclear Basolateral/fisiopatología , Benzazepinas/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Modelos Animales de Enfermedad , Antagonistas de Dopamina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Estimulación Eléctrica , Técnicas In Vitro , Plasticidad Neuronal/efectos de los fármacos , Técnicas de Placa-Clamp , Inhibidores de Fosfodiesterasa 4/farmacología , Psicoacústica , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reflejo Acústico/efectos de los fármacos , Reflejo Acústico/fisiología , Rolipram/farmacología , Estrés Psicológico/complicaciones , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/etiología
10.
JCI Insight ; 1(4)2016.
Artículo en Inglés | MEDLINE | ID: mdl-27158670

RESUMEN

The prevention of organ damage and early death in young adults is a major clinical concern in sickle cell disease (SCD). However, mechanisms that control adult progression of SCD during the transition from adolescence are poorly defined with no cognate prophylaxis. Here, we demonstrate in a longitudinal cohort of homozygous SCD (SS) mice a link between intravascular hemolysis, vascular inflammation, lung injury, and early death. Prophylactic Nrf2 activation in young SS mice stabilized intravascular hemolysis, reversed vascular inflammation, and attenuated lung edema in adulthood. Enhanced Nrf2 activation in endothelial cells in vitro concurred with the dramatic effect on vascular inflammation in the mice. BM chimeric SS mice lacking Nrf2 expression in nonhematopoietic tissues were created to dissect the role of nonerythroid Nrf2 in SCD progression. The SS chimeras developed severe intravascular hemolysis despite having erythroid Nrf2. In addition, they developed premature vascular inflammation and pulmonary edema and died younger than donor littermates with intact nonhematopoietic Nrf2. Our results reveal a dominant protective role for nonhematopoietic Nrf2 against tissue damage in both erythroid and nonerythroid tissues in SCD. Furthermore, we show that prophylactic augmentation of Nrf2-coordinated cytoprotection effectively impedes onset of the severe adult phenotype of SCD in mice.

11.
Neuropharmacology ; 97: 251-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26032436

RESUMEN

Depression during pregnancy has been linked to in utero stress and is associated with long-lasting symptoms in offspring, including anxiety, helplessness, attentional deficits, and social withdrawal. Depression is diagnosed in 10-20% of expectant mothers, but the impact of antidepressant treatment on offspring development is not well documented, particularly for females. Here, we used a prenatal stress model of maternal depression to test the hypothesis that in utero antidepressant treatment could mitigate the effects of prenatal stress. We also investigated the effects of prenatal stress and antidepressant treatment on gene expression related to GABAergic and serotonergic neurotransmission in the amygdala, which may underlie behavioral effects of prenatal stress. Nulliparous female rats were implanted with osmotic minipumps delivering clinically-relevant concentrations of escitalopram and mated. Pregnant dams were exposed to 12 days of mixed-modality stressors, and offspring were behaviorally assessed in adolescence (postnatal day 28) and adulthood (beyond day 90) to determine the extent of behavioral change. We found that in utero stress exposure, regardless of escitalopram treatment, increased anxiety-like behavior in adolescent females and profoundly influenced amygdala expression of the chloride transporters KCC2 and NKCC1, which regulate GABAergic function. In contrast, prenatal escitalopram exposure alone elevated amygdala expression of 5-HT1A receptors. In adulthood, anxiety-like behavior returned to baseline and gene expression effects in the amygdala abated, whereas deficits emerged in novel object recognition for rats exposed to stress during gestation. These findings suggest prenatal stress causes age-dependent deficits in anxiety-like behavior and amygdala function in female offspring, regardless of antidepressant exposure.


Asunto(s)
Amígdala del Cerebelo/fisiopatología , Ansiedad/fisiopatología , Citalopram/farmacología , Efectos Tardíos de la Exposición Prenatal , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Estrés Psicológico/fisiopatología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/crecimiento & desarrollo , Animales , Ansiedad/etiología , Corticosterona/sangre , Modelos Animales de Enfermedad , Estradiol/sangre , Femenino , Expresión Génica/efectos de los fármacos , Trastornos de la Memoria/etiología , Trastornos de la Memoria/fisiopatología , Embarazo , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1A/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Estrés Psicológico/tratamiento farmacológico , Simportadores/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Cotransportadores de K Cl
12.
J Comp Neurol ; 522(3): 609-25, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24037673

RESUMEN

Regulation of BNSTALG neuronal firing activity is tightly regulated by the opposing actions of the fast outward potassium current, IA , mediated by α subunits of the Kv4 family of ion channels, and the transient inward calcium current, IT . Together, these channels play a critical role in regulating the latency to action potential onset, duration, and frequency, as well as dendritic back-propagation and synaptic plasticity. Previously we have shown that Type I-III BNSTALG neurons express mRNA transcripts for each of the Kv4 α subunits. However, the biophysical properties of native IA channels are critically dependent on the formation of macromolecular complexes of Kv4 channels with a family of chaperone proteins, the potassium channel-interacting proteins (KChIP1-4). Here we used a multidisciplinary approach to investigate the expression and function of Kv4 channels and KChIPs in neurons of the rat BNSTALG . Using immunofluorescence we demonstrated the pattern of localization of Kv4.2, Kv4.3, and KChIP1-4 proteins in the BNSTALG . Moreover, our single-cell reverse-transcription polymerase chain reaction (scRT-PCR) studies revealed that mRNA transcripts for Kv4.2, Kv4.3, and all four KChIPs were differentially expressed in Type I-III BNSTALG neurons. Furthermore, immunoelectron microscopy revealed that Kv4.2 and Kv4.3 channels were primarily localized to the dendrites and spines of BNSTALG neurons, and are thus ideally situated to modulate synaptic transmission. Consistent with this observation, in vitro patch clamp recordings showed that reducing postsynaptic IA in these neurons lowered the threshold for long-term potentiation (LTP) induction. These results are discussed in relation to potential modulation of IA channels by chronic stress.


Asunto(s)
Proteínas de Interacción con los Canales Kv/metabolismo , Neuronas/metabolismo , Núcleos Septales/anatomía & histología , Núcleos Septales/metabolismo , Canales de Potasio Shal/metabolismo , 4-Aminopiridina/farmacología , Análisis de Varianza , Animales , Biofisica , Estimulación Eléctrica , Técnicas In Vitro , Proteínas de Interacción con los Canales Kv/genética , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Microscopía Inmunoelectrónica , Neuronas/clasificación , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , ARN Mensajero , Ratas , Ratas Sprague-Dawley , Canales de Potasio Shal/genética , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/ultraestructura
13.
Front Neurosci ; 7: 156, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24009552

RESUMEN

Corticotrophin-releasing factor (CRF) plays a key role in initiating many of the endocrine, autonomic, and behavioral responses to stress. CRF-containing neurons of the paraventricular nucleus of the hypothalamus (PVN) are classically involved in regulating endocrine function through activation of the stress axis. However, CRF is also thought to play a critical role in mediating anxiety-like responses to environmental stressors, and dysfunction of the CRF system in extra-hypothalamic brain regions, like the bed nucleus of stria terminalis (BNST), has been linked to the etiology of many psychiatric disorders including anxiety and depression. Thus, although CRF neurons of the PVN and BNST share a common neuropeptide phenotype, they may represent two functionally diverse neuronal populations. Here, we employed dual-immunofluorescence, single-cell RT-PCR, and electrophysiological techniques to further examine this question and report that CRF neurons of the PVN and BNST are fundamentally different such that PVN CRF neurons are glutamatergic, whereas BNST CRF neurons are GABAergic. Moreover, these two neuronal populations can be further distinguished based on their electrophysiological properties, their co-expression of peptide neurotransmitters such as oxytocin and arginine-vasopressin, and their cognate receptors. Our results suggest that CRF neurons in the PVN and the BNST would not only differ in their response to local neurotransmitter release, but also in their action on downstream target structures.

14.
Biol Psychiatry ; 74(11): 817-26, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24012328

RESUMEN

BACKGROUND: Striatal-enriched protein tyrosine phosphatase (STEP) is a brain-specific protein tyrosine phosphatase that opposes the development of synaptic strengthening and the consolidation of fear memories. In contrast, stress facilitates fear memory formation, potentially by activating corticotrophin releasing factor (CRF) neurons in the anterolateral cell group of the bed nucleus of the stria terminalis (BNSTALG). METHODS: Here, using dual-immunofluorescence, single-cell reverse transcriptase polymerase chain reaction, quantitative reverse transcriptase polymerase chain reaction, Western blot, and whole-cell patch-clamp electrophysiology, we examined the expression and role of STEP in regulating synaptic plasticity in rat BNSTALG neurons and its modulation by stress. RESULTS: Striatal-enriched protein tyrosine phosphatase was selectively expressed in CRF neurons in the oval nucleus of the BNSTALG. Following repeated restraint stress (RRS), animals displayed a significant increase in anxiety-like behavior, which was associated with a downregulation of STEP messenger RNA and protein expression in the BNSTALG, as well as selectively enhancing the magnitude of long-term potentiation (LTP) induced in Type III, putative CRF neurons. To determine if the changes in STEP expression following RRS were mechanistically related to LTP facilitation, we examined the effects of intracellular application of STEP on the induction of LTP. STEP completely blocked the RRS-induced facilitation of LTP in BNSTALG neurons. CONCLUSIONS: Hence, STEP acts to buffer CRF neurons against excessive activation, while downregulation of STEP after chronic stress may result in pathologic activation of CRF neurons in the BNSTALG and contribute to prolonged states of anxiety. Thus, targeted manipulations of STEP activity might represent a novel treatment strategy for stress-induced anxiety disorders.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Neuronas/enzimología , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Núcleos Septales/enzimología , Estrés Fisiológico , Animales , Masculino , Proteínas Tirosina Fosfatasas no Receptoras/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal
15.
J Neurophysiol ; 110(4): 926-41, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23719209

RESUMEN

Many psychiatric disorders, including anxiety and autism spectrum disorders, have early ages of onset and high incidence in juveniles. To better treat and prevent these disorders, it is important to first understand normal development of brain circuits that process emotion. Healthy and maladaptive emotional processing involve the basolateral amygdala (BLA), dysfunction of which has been implicated in numerous psychiatric disorders. Normal function of the adult BLA relies on a fine balance of glutamatergic excitation and GABAergic inhibition. Elsewhere in the brain GABAergic transmission changes throughout development, but little is known about the maturation of GABAergic transmission in the BLA. Here we used whole cell patch-clamp recording and single-cell RT-PCR to study GABAergic transmission in rat BLA principal neurons at postnatal day (P)7, P14, P21, P28, and P35. GABAA currents exhibited a significant twofold reduction in rise time and nearly 25% reduction in decay time constant between P7 and P28. This corresponded with a shift in expression of GABAA receptor subunit mRNA from the α2- to the α1-subunit. The reversal potential for GABAA receptors transitioned from depolarizing to hyperpolarizing with age, from around -55 mV at P7 to -70 mV by P21. There was a corresponding shift in expression of opposing chloride pumps that influence the reversal, from NKCC1 to KCC2. Finally, we observed short-term depression of GABAA postsynaptic currents in immature neurons that was significantly and gradually abolished by P28. These findings reveal that in the developing BLA GABAergic transmission is highly dynamic, reaching maturity at the end of the first postnatal month.


Asunto(s)
Amígdala del Cerebelo/crecimiento & desarrollo , Potenciales Postsinápticos Inhibidores , Neuronas/fisiología , Receptores de GABA-A/metabolismo , Ácido gamma-Aminobutírico/fisiología , Amígdala del Cerebelo/fisiología , Animales , Femenino , Técnicas In Vitro , Masculino , Plasticidad Neuronal , Ratas , Ratas Sprague-Dawley
16.
Proc Natl Acad Sci U S A ; 109(40): 16330-5, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-22992651

RESUMEN

Corticotropin-releasing factor (CRF) is critical for the endocrine, autonomic, and behavioral responses to stressors, and it has been shown to modulate fear and anxiety. The CRF receptor is widely expressed across a variety of cell types, impeding progress toward understanding the contribution of specific CRF-containing neurons to fear dysregulation. We used a unique CRF-Cre driver transgenic mouse line to remove floxed GABA(A)α1 subunits specifically from CRF neurons [CRF-GABA(A)α1 KO]. This process resulted in mice with decreased GABA(A)α1 expression only in CRF neurons and increased CRF mRNA within the amygdala, bed nucleus of the stria terminalis (BNST) and paraventricular nucleus of the hypothalamus. These mice show normal locomotor and pain responses and no difference in depressive-like behavior or Pavlovian fear conditioning. However, CRF-GABA(A)α1 KO increased anxiety-like behavior and impaired extinction of conditioned fear, coincident with an increase in plasma corticosterone concentration. These behavioral impairments were rescued with systemic or BNST infusion of the CRF antagonist R121919. Infusion of Zolpidem, a GABA(A)α1-preferring benzodiazepine-site agonist, into the BNST of the CRF-GABA(A)α1 KO was ineffective at decreasing anxiety. Electrophysiological findings suggest a disruption in inhibitory current may play a role in these changes. These data indicate that disturbance of CRF containing GABA(A)α1 neurons causes increased anxiety and impaired fear extinction, both of which are symptoms diagnostic for anxiety disorders, such as posttraumatic stress disorder.


Asunto(s)
Ansiedad/fisiopatología , Hormona Liberadora de Corticotropina/metabolismo , Extinción Psicológica/fisiología , Miedo/fisiología , Neuronas/metabolismo , Receptores de GABA-A/deficiencia , Análisis de Varianza , Animales , Ansiedad/metabolismo , Condicionamiento Psicológico/fisiología , Corticosterona/sangre , Cartilla de ADN/genética , Hibridación in Situ , Hibridación Fluorescente in Situ , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Neuronas/fisiología , Técnicas de Placa-Clamp , Piridinas , Pirimidinas , Receptores de GABA-A/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Zolpidem
17.
Neuropharmacology ; 62(4): 1671-83, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22166222

RESUMEN

The anterolateral cell group of the bed nucleus of the stria terminalis (BNST(ALG)) serves as an important relay station in stress circuitry. Limbic inputs to the BNST(ALG) are primarily glutamatergic and activity-dependent changes in this input have been implicated in abnormal behaviors associated with chronic stress and addiction. Significantly, local infusion of acetylcholine (ACh) receptor agonists into the BNST trigger stress-like cardiovascular responses, however, little is known about the effects of these agents on glutamatergic transmission in the BNST(ALG). Here, we show that glutamate- and ACh-containing fibers are found in close association in the BNST(ALG). Moreover, in the presence of the acetylcholinesterase inhibitor, eserine, endogenous ACh release evoked a long-lasting reduction of the amplitude of stimulus-evoked EPSCs. This effect was mimicked by exogenous application of the ACh analog, carbachol, which caused a reversible, dose-dependent, reduction of the evoked EPSC amplitude, and an increase in both the paired-pulse ratio and coefficient of variation, suggesting a presynaptic site of action. Uncoupling of postsynaptic G-proteins with intracellular GDP-ß-S, or application of the nicotinic receptor antagonist, tubocurarine, failed to block the carbachol effect. In contrast, the carbachol effect was blocked by prior application of atropine or M(2) receptor-preferring antagonists, and was absent in M(2)/M(4) receptor knockout mice, suggesting that presynaptic M(2) receptors mediate the effect of ACh. Immunoelectron microscopy studies further revealed the presence of M(2) receptors on axon terminals that formed asymmetric synapses with BNST neurons. Our findings suggest that presynaptic M(2) receptors might be an important modulator of the stress circuit and hence a novel target for drug development.


Asunto(s)
Ácido Glutámico/metabolismo , Neuronas/metabolismo , Receptor Muscarínico M2/metabolismo , Núcleos Septales/metabolismo , Transmisión Sináptica/fisiología , Acetilcolina/metabolismo , Animales , Inhibidores de la Colinesterasa/farmacología , Relación Dosis-Respuesta a Droga , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Ratones , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/metabolismo , Neuronas/efectos de los fármacos , Fisostigmina/farmacología , Ratas , Ratas Sprague-Dawley , Núcleos Septales/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Transmisión Sináptica/efectos de los fármacos
18.
PLoS One ; 6(10): e26065, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22022509

RESUMEN

Fear memory formation is thought to require dopamine, brain-derived neurotrophic factor (BDNF) and zinc release in the basolateral amygdala (BLA), as well as the induction of long term potentiation (LTP) in BLA principal neurons. However, no study to date has shown any relationship between these processes in the BLA. Here, we have used in vitro whole-cell patch clamp recording from BLA principal neurons to investigate how dopamine, BDNF, and zinc release may interact to modulate the LTP induction in the BLA. LTP was induced by either theta burst stimulation (TBS) protocol or spaced 5 times high frequency stimulation (5xHFS). Significantly, both TBS and 5xHFS induced LTP was fully blocked by the dopamine D1 receptor antagonist, SCH23390. LTP induction was also blocked by the BDNF scavenger, TrkB-FC, the zinc chelator, DETC, as well as by an inhibitor of matrix metalloproteinases (MMPs), gallardin. Conversely, prior application of the dopamine reuptake inhibitor, GBR12783, or the D1 receptor agonist, SKF39393, induced robust and stable LTP in response to a sub-threshold HFS protocol (2xHFS), which does not normally induce LTP. Similarly, prior activation of TrkB receptors with either a TrkB receptor agonist, or BDNF, also reduced the threshold for LTP-induction, an effect that was blocked by the MEK inhibitor, but not by zinc chelation. Intriguingly, the TrkB receptor agonist-induced reduction of LTP threshold was fully blocked by prior application of SCH23390, and the reduction of LTP threshold induced by GBR12783 was blocked by prior application of TrkB-FC. Together, our results suggest a cellular mechanism whereby the threshold for LTP induction in BLA principal neurons is critically dependent on the level of dopamine in the extracellular milieu and the synergistic activation of postsynaptic D1 and TrkB receptors. Moreover, activation of TrkB receptors appears to be dependent on concurrent release of zinc and activation of MMPs.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Plasticidad Neuronal , Receptor trkB/metabolismo , Receptores de Dopamina D1/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Animales , Benzazepinas/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Calcio/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dopamina/farmacología , Estimulación Eléctrica , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Metaloproteinasa 17 de la Matriz/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/enzimología , Piperazinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor trkB/genética , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D1/genética , Transducción de Señal/efectos de los fármacos
19.
Psychoneuroendocrinology ; 36(9): 1312-26, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21481539

RESUMEN

Activation of corticotrophin releasing factor (CRF) neurons in the paraventricular nucleus of the hypothalamus (PVN) is necessary for establishing the classic endocrine response to stress, while activation of forebrain CRF neurons mediates affective components of the stress response. Previous studies have reported that mRNA for CRF2 receptor (CRFR2) is expressed in the bed nucleus of the stria terminalis (BNST) as well as hypothalamic nuclei, but little is known about the localization and cellular distribution of CRFR2 in these regions. Using immunofluorescence with confocal microscopy, as well as electron microscopy, we demonstrate that in the BNST CRFR2-immunoreactive fibers represent moderate to strong labeling on axons terminals. Dual-immunofluorescence demonstrated that CRFR2-fibers co-localize oxytocin (OT), but not arginine-vasopressin (AVP), and make perisomatic contacts with CRF neurons. Dual-immunofluorescence and single cell RT-PCR demonstrate that in the hypothalamus, CRFR2 immunoreactivity and mRNA are found in OT, but not in CRF or AVP-neurons. Furthermore, CRF neurons of the PVN and BNST express mRNA for the oxytocin receptor, while the majority of OT/CRFR2 neurons in the hypothalamus do not. Finally, using adenoviral-based anterograde tracing of PVN neurons, we show that OT/CRFR2-immunoreactive fibers observed in the BNST originate in the PVN. Our results strongly suggest that CRFR2 located on oxytocinergic neurons and axon terminals might regulate the release of this neuropeptide and hence might be a crucial part of potential feedback loop between the hypothalamic oxytocin system and the forebrain CRF system that could significantly impact affective and social behaviors, in particular during times of stress.


Asunto(s)
Afecto/fisiología , Hormona Liberadora de Corticotropina/metabolismo , Retroalimentación Fisiológica/fisiología , Hipotálamo/metabolismo , Oxitocina/metabolismo , Núcleos Septales/metabolismo , Estrés Psicológico/metabolismo , Afecto/efectos de los fármacos , Animales , Arginina Vasopresina/metabolismo , Colchicina/administración & dosificación , Colchicina/farmacología , Retroalimentación Fisiológica/efectos de los fármacos , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Inyecciones Intraventriculares , Masculino , Neuroanatomía , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Núcleos Septales/efectos de los fármacos , Núcleos Septales/fisiología , Estrés Psicológico/fisiopatología
20.
Mol Cell Neurosci ; 46(4): 699-709, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21310239

RESUMEN

The activity of neurons in the anterolateral cell group of the bed nucleus of the stria terminalis (BNST(ALG)) plays a critical role in anxiety- and stress-related behaviors. Histochemical studies have suggested that multiple distinct neuronal phenotypes exist in the BNST(ALG). Consistent with this observation, the physiological properties of BNST(ALG) neurons are also heterogeneous, and three distinct cell types can be defined (Types I-III) based primarily on their expression of four key membrane currents, namely I(h), I(A), I(T), and I(K(IR)). Significantly, all four channels are multimeric proteins and can comprise of more than one pore-forming α subunit. Hence, differential expression of α subunits may further diversify the neuronal population. However, nothing is known about the relative expression of these ion channel α subunits in BNST(ALG) neurons. We have addressed this lacuna by combining whole-cell patch-clamp recording together with single-cell reverse transcriptase polymerase chain reaction (scRT-PCR) to assess the mRNA transcript expression for each of the subunits for the four key ion channels in Type I-III neurons of the BNST(ALG.) Here, cytosolic mRNA from single neurons was probed for the expression of transcripts for each of the α subunits of I(h) (HCN1-HCN4), I(T) (Ca(v)3.1-Ca(v)3.3), I(A) (K(v)1.4, K(v)3.4, K(v)4.1-K(v) 4.3) and I(K(IR)) (Kir2.1-Kir2.4). An unbiased hierarchical cluster analysis followed by discriminant function analysis revealed that a positive correlation exists between the physiological and genetic phenotype of BNST(ALG) neurons. Thus, the analysis segregated BNST(ALG) neurons into 3 distinct groups, based on their α subunit mRNA expression profile, which positively correlated with our existing electrophysiological classification (Types I-III). Furthermore, analysis of mRNA transcript expression in Type I-Type III neurons suggested that, whereas Type I and III neurons appear to represent genetically homologous cell populations, Type II neurons may be further subdivided into three genetically distinct subgroups. These data not only validate our original classification scheme, but further refine the classification at the molecular level, and thus identifies novel targets for potential disruption and/or pharmacotherapeutic intervention in stress-related anxiety-like behaviors.


Asunto(s)
Perfilación de la Expresión Génica , Neuronas/clasificación , Neuronas/fisiología , Núcleos Septales/citología , Animales , Análisis por Conglomerados , Canales Iónicos/genética , Canales Iónicos/metabolismo , Masculino , Potenciales de la Membrana/fisiología , Neuronas/citología , Neurotransmisores/metabolismo , Técnicas de Placa-Clamp , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...