Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Oncologist ; 29(1): e131-e140, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37531083

RESUMEN

BACKGROUND: This study aimed to evaluate the safety, pharmacokinetics (PKs), and preliminary activity of LY3405105, a covalent inhibitor of cyclin-dependent kinase 7 (CDK7), in patients with advanced solid tumors. MATERIALS AND METHODS: LY3405105 monotherapy was given once daily (QD; part A1) or thrice weekly (TIW; part A2) starting at 1 and 2 mg orally, respectively, and escalated per a Bayesian design in adult patients. The primary endpoint was safety, and secondary endpoints included PKs and antitumor activity. RESULTS: Fifty-four patients were enrolled: 43 in part A1 and 11 in part A2. Seven patients had dose-limiting toxicities, all in part A1 (45 mg: n = 3; 35 mg: n = 3; 25 mg: n = 1). Thirty-five patients (64.8%) reported at least one treatment-related adverse event (TRAE). TRAEs (≥10%) were diarrhea, nausea, fatigue, vomiting, abdominal pain, anemia, asthenia, and decreased platelet count. QD dosing showed sustained exposure with less peak-trough fluctuation compared to TIW dosing. Median time to maximum concentration was 1-2 hours and half-life was 15-19 hours. CDK7-target occupancy in skin and peripheral blood on day 15 was dose-dependent and reached near maximal occupancy of 75% at ≥15 mg QD. The maximum tolerated dose (MTD) was 20 mg QD. Twelve patients in part A1 (27.9%) and 5 patients in part A2 (45.5%) had a best overall response of stable disease. No complete response or partial response was observed. CONCLUSION: The MTD of LY3405105 monotherapy was 20 mg QD. The most common toxicities were gastrointestinal adverse events, myelosuppression, fatigue, and asthenia. Limited clinical activity was observed in this phase I trial, and there are no plans for further development. CLINICALTRIALS.GOV IDENTIFIER: NCT03770494.


Asunto(s)
Antineoplásicos , Neoplasias , Adulto , Humanos , Astenia , Teorema de Bayes , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Inhibidores de Proteínas Quinasas/efectos adversos , Fatiga/inducido químicamente , Quinasas Ciclina-Dependientes , Dosis Máxima Tolerada , Relación Dosis-Respuesta a Droga , Antineoplásicos/efectos adversos
2.
Cancer ; 127(3): 372-380, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33107983

RESUMEN

BACKGROUND: Deregulated Notch signaling is implicated in T-cell acute lymphoblastic leukemia (T-ALL)/T-cell lymphoblastic lymphoma (T-LBL). Crenigacestat (LY3039478) prevents cleavage of Notch proteins and may benefit patients with relapsed/refractory T-ALL/T-LBL. METHODS: JJCB was a multicenter, nonrandomized, open-label, dose-escalation, phase 1 study in adult patients with relapsed/refractory T-ALL/T-LBL. Eligible patients received Crenigacestat orally 3 times per week plus dexamethasone at 24 mg twice daily on days 1 to 5 every other week in a 28-day cycle. The starting level of Crenigacestat was 50 mg, and dose escalation was performed with a modified 3+3 scheme for the estimation of dose-limiting toxicity (DLT) at the recommended dose level. RESULTS: In total, 36 patients with T-ALL (n = 31 [86.1%]) or T-LBL (n = 5 [13.9%]) were treated with Crenigacestat and dexamethasone. Six patients (16.7%) experienced DLTs: 2 of 12 (16.7%) in the 75-mg cohort (grade 4 gastrointestinal hemorrhage and grade 3 nausea, vomiting, and diarrhea), 1 of 15 (6.7%) in the 100-mg cohort (grade 3 diarrhea), and 3 of 3 (100%) in the 125-mg cohort (grade 3 diarrhea, nausea, and vomiting). The maximum tolerated dosewas 75 mg plus 24 mg of dexamethasone daily on days 1 to 5. Twenty-eight patients (77.8%) experienced 1 or more treatment-emergent adverse events related to the study treatment. The best overall response was a confirmed response, with 1 patient (2.8%) having a duration of response of 10.51 months. Six patients (16.7%) achieved stable disease, and 12 patients (33.3%) experienced progressive disease. The remaining 17 patients (47.2%) were not evaluable. The median event-free survival was 1.18 months (95% confidence interval, 0.76-2.14 months) among all groups. A pharmacodynamic analysis showed decreased plasma amyloid ß levels. CONCLUSIONS: Crenigacestat demonstrated limited clinical activity at the recommended dose in adult patients with relapsed/refractory T-ALL/T-LBL.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Benzazepinas/administración & dosificación , Dexametasona/administración & dosificación , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Adulto , Anciano , Benzazepinas/efectos adversos , Benzazepinas/farmacocinética , Dexametasona/efectos adversos , Dexametasona/farmacocinética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
3.
Invest New Drugs ; 39(1): 193-201, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32915419

RESUMEN

Background Crenigacestat is a potent Notch inhibitor that decreases Notch signaling and its downstream biological effects. Here, we report the results from Part F of study 16F-MC-JJCA designed to evaluate the safety, pharmacokinetics (PK), and antitumor activity of crenigacestat with prednisone in advanced or metastatic cancer. The combination was planned to mitigate gastrointestinal toxicities. Methods Eligible patients (Study Part F) received crenigacestat loading dose (75 mg, escalating to 150 mg) administered thrice weekly (TIW) (F1) or twice weekly (BIW) (F2) for 2 weeks during Cycle 1, followed by 50 mg TIW from week 3 onwards. Prednisone was co-administered for 2 weeks in Cycle 1. Results Twenty-eight patients were enrolled; 11 in F1 (median age, 63 years), 17 in F2 (median age, 50 years). Dose-limiting toxicities were Grade 3 increased serum amylase and Grade 2 fatigue in F1, and Grade 4 hypophosphatemia and Grade 3 rash maculo-papular in F2. The maximum tolerated dose was 75 mg in F1 and 100 mg in F2. Best overall response was stable disease (F1, 6 [54.5%] patients; F2, 11 [64.7%] patients). Pharmacokinetic was dose proportional. Prednisone did not modify PK of crenigacestat, and both F1 and F2 achieved pharmacodynamics effects on evaluable tumor tissue samples. Conclusions This study demonstrated the potential use of prednisone to reduce gastrointestinal (GI) toxicities of a Notch inhibitor without affecting its PK. The safety profile observed was consistent with Notch pathway inhibitors, and the maximum tolerated dose was 75 mg TIW and 100 mg BIW in F1 and F2, respectively. ClinicalTrials.gov: NCT01695005.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzazepinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Prednisona/uso terapéutico , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Área Bajo la Curva , Benzazepinas/administración & dosificación , Benzazepinas/efectos adversos , Benzazepinas/farmacocinética , Biomarcadores de Tumor , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Neoplasias/patología , Prednisona/administración & dosificación , Prednisona/efectos adversos
4.
Invest New Drugs ; 39(4): 1001-1010, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33479856

RESUMEN

Background Aurora A kinase (AurA) overexpression likely contributes to tumorigenesis and therefore represents an attractive target for cancer therapeutics. This phase 1 study aimed to determine the safety, pharmacokinetics, and antitumor activity of LY3295668 erbumine, an AurA inhibitor, in patients with locally advanced or metastatic solid tumors. Methods Patients with locally advanced or metastatic solid tumors, Eastern Cooperative Oncology Group performance status 0-1, and disease progression after one to four prior treatment regimens were enrolled. Primary objective was to determine maximum tolerated dose (MTD); secondary objectives included evaluation of the tolerability and safety profile and pharmacokinetics of LY3295668. All patients received twice-daily (BID) oral LY3295668 in 21-day cycles in an ascending-dose schedule. Results Twelve patients were enrolled in phase 1 (25 mg, n = 8; 50 mg, n = 2; 75 mg, n = 2) and one patient was enrolled after. Overall, four patients experienced dose-limiting toxicities (DLTs) within the first cycle (75 mg: Grade 3 diarrhea [one patient], Grade 4 mucositis and Grade 3 corneal deposits [one patient]; 50 mg: mucositis and diarrhea [both Grade 3, one patient]; 25 mg: Grade 3 mucositis [one patient]). Patients exhibiting DLTs had the highest model-predicted exposures at steady state. Mucositis was the most common adverse event (67%), followed by diarrhea, fatigue, alopecia, anorexia, constipation, and nausea. Nine patients had best response of stable disease; the disease control rate was 69%. Conclusions MTD of LY3295668 was 25 mg BID. LY3295668 had a manageable toxicity profile and demonstrated activity in some patients with locally advanced or metastatic solid tumors.Trial registration ClinicalTrials.gov, NCT03092934. Registered March 22, 2017. https://clinicaltrials.gov/ct2/show/NCT03092934 .


Asunto(s)
Aurora Quinasa A/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Piperidinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Adulto , Anciano , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/patología , Piperidinas/efectos adversos , Inhibidores de Proteínas Quinasas/efectos adversos , Pirazoles/efectos adversos , Resultado del Tratamiento
5.
Invest New Drugs ; 39(4): 1089-1098, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33686452

RESUMEN

Notch signaling plays an important role in development and tissue homeostasis. Deregulation of Notch signaling has been implicated in multiple malignancies. Crenigacestat (LY3039478), a potent Notch inhibitor, decreases Notch signaling and its downstream biologic effects. I6F-MC-JJCD was a multicenter, nonrandomized, open-label, Phase 1b study with 5 separate, parallel dose-escalations in patients with advanced or metastatic cancer from a variety of solid tumors, followed by a dose-confirmation phase in prespecified tumor types. This manuscript reports on 3 of 5 groups. The primary objective was to determine the recommended Phase 2 dose of crenigacestat in combination with other anticancer agents (taladegib, LY3023414 [dual inhibitor of phosphoinositide 3-kinase; mechanistic target of rapamycin], or abemaciclib). Secondary objectives included evaluation of safety, tolerability, efficacy, and pharmacokinetics. Patients (N = 63) received treatment between November 2016 and July 2019. Dose-limiting toxicities occurred in 12 patients, mostly gastrointestinal (diarrhea, nausea, vomiting). The maximum-tolerated dose of crenigacestat was 25 mg in Part B (LY3023414), 50 mg in Part C (abemaciclib), and not established in Part A (taladegib) due to toxicities. Patients had at least 1 adverse event (AE) and 75.0-82.6% were ≥ Grade 3 all-causality AEs. No patient had complete or partial response. Disease control rates were 18.8% (Part B) and 26.1% (Part C). The study was terminated before dose confirmation cohorts were triggered. This study demonstrated that crenigacestat combined with different anticancer agents (taladegib, LY3023414, or abemaciclib) was poorly tolerated, leading to lowered dosing and disappointing clinical activity in patients with advanced or metastatic solid tumors. NCT02784795 and date of registration: May 27, 2016.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias/tratamiento farmacológico , Aminopiridinas/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Benzazepinas/administración & dosificación , Bencimidazoles/administración & dosificación , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/patología , Ftalazinas/administración & dosificación , Piridinas/administración & dosificación , Quinolonas/administración & dosificación
6.
Int J Neuropsychopharmacol ; 20(11): 948-955, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29016816

RESUMEN

Evidence over the past decades has found that stress, particularly through the corticosterone stress hormones, produces complex changes in glutamatergic signaling in prefrontal cortex, which leads to the alteration of cognitive processes medicated by this brain region. Interestingly, the effects of stress on glutamatergic transmission appear to be "U-shaped," depending upon the duration and severity of the stressor. These biphasic effects of acute vs chronic stress represent the adaptive vs maladaptive responses to stressful stimuli. Animal studies suggest that the stress-induced modulation of excitatory synaptic transmission involves changes in presynaptic glutamate release, postsynaptic glutamate receptor membrane trafficking and degradation, spine structure and cytoskeleton network, and epigenetic control of gene expression. This review will discuss current findings on the key molecules involved in the stress-induced regulation of prefrontal cortex synaptic physiology and prefrontal cortex-mediated functions. Understanding the molecular and epigenetic mechanisms that underlie the complex effects of stress will help to develop novel strategies to cope with stress-related mental disorders.


Asunto(s)
Trastornos del Conocimiento/etiología , Epigénesis Genética , Estrés Psicológico/complicaciones , Estrés Psicológico/patología , Transmisión Sináptica/fisiología , Animales , Ácido Glutámico/metabolismo , Humanos , Sinapsis/patología , Sinapsis/ultraestructura
7.
J Biol Chem ; 289(36): 25177-85, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25056951

RESUMEN

The monoamine system in the prefrontal cortex has been implicated in various mental disorders and has been the major target of anxiolytics and antidepressants. Clinical studies show that serotonin and norepinephrine reuptake inhibitors (SNRIs) produce better therapeutic effects than single selective reuptake inhibitors, but the underlying mechanisms are largely unknown. Here, we found that low dose SNRIs, by acting on 5-HT(1A) and α2-adrenergic receptors, synergistically reduced AMPA receptor (AMPAR)-mediated excitatory postsynaptic currents and AMPAR surface expression in prefrontal cortex pyramidal neurons via a mechanism involving Rab5/dynamin-mediated endocytosis of AMPARs. The synergistic effect of SNRIs on AMPARs was blocked by inhibition of activator of G protein signaling 3, a G protein modulator that prevents reassociation of G(i) protein α subunit and prolongs the ßγ-mediated signaling pathway. Moreover, the depression of AMPAR-mediated excitatory postsynaptic currents by SNRIs required p38 kinase activity, which was increased by 5-HT(1A) and α2-adrenergic receptor co-activation in an activator of G protein signaling 3-dependent manner. These results have revealed a potential mechanism for the synergy between the serotonin and norepinephrine systems in the regulation of glutamatergic transmission in cortical neurons.


Asunto(s)
Desipramina/farmacología , Fluoxetina/farmacología , Células Piramidales/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Inhibidores de Captación Adrenérgica/farmacología , Animales , Bicuculina , Western Blotting , Proteínas Portadoras/metabolismo , Células Cultivadas , Sinergismo Farmacológico , Dinaminas/metabolismo , Endocitosis/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Técnicas de Placa-Clamp , Corteza Prefrontal/citología , Células Piramidales/metabolismo , Células Piramidales/fisiología , Ratas , Receptor de Serotonina 5-HT1A/metabolismo , Receptores AMPA/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Transducción de Señal/efectos de los fármacos , Transmisión Sináptica/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP rab5/metabolismo
8.
J Biol Chem ; 288(36): 26112-26120, 2013 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-23884421

RESUMEN

The prefrontal cortex (PFC), a key brain region for cognitive and emotional processes, is highly regulated by dopaminergic inputs. The dopamine D4 receptor, which is enriched in PFC, has been implicated in mental disorders, such as attention deficit-hyperactivity disorder and schizophrenia. Recently we have found homeostatic regulation of AMPA receptor-mediated synaptic transmission in PFC pyramidal neurons by the D4 receptor, providing a potential mechanism for D4 in stabilizing cortical excitability. Because stress is tightly linked to adaptive and maladaptive changes associated with mental health and disorders, we examined the synaptic actions of D4 in stressed rats. We found that neural excitability was elevated by acute stress and dampened by repeated stress. D4 activation produced a potent reduction of excitatory transmission in acutely stressed animals and a marked increase of excitatory transmission in repeatedly stressed animals. These effects of D4 targeted GluA2-lacking AMPA receptors and relied on the bi-directional regulation of calcium/calmodulin kinase II activity. The restoration of PFC glutamatergic transmission in stress conditions may enable D4 receptors to serve as a synaptic stabilizer in normal and pathological conditions.


Asunto(s)
Ácido Glutámico/metabolismo , Corteza Prefrontal/metabolismo , Células Piramidales/metabolismo , Receptores de Dopamina D4/metabolismo , Estrés Psicológico/metabolismo , Transmisión Sináptica , Animales , Masculino , Corteza Prefrontal/patología , Corteza Prefrontal/fisiopatología , Células Piramidales/patología , Células Piramidales/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Estrés Psicológico/patología , Estrés Psicológico/fisiopatología
9.
EMBO J ; 29(2): 482-95, 2010 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-19942860

RESUMEN

Central noradrenergic signalling mediates arousal and facilitates learning through unknown molecular mechanisms. Here, we show that the beta(2)-adrenergic receptor (beta(2)AR), the trimeric G(s) protein, adenylyl cyclase, and PKA form a signalling complex with the AMPA-type glutamate receptor subunit GluR1, which is linked to the beta(2)AR through stargazin and PSD-95 and their homologues. Only GluR1 associated with the beta(2)AR is phosphorylated by PKA on beta(2)AR stimulation. Peptides that interfere with the beta(2)AR-GluR1 association prevent this phosphorylation of GluR1. This phosphorylation increases GluR1 surface expression at postsynaptic sites and amplitudes of EPSCs and mEPSCs in prefrontal cortex slices. Assembly of all proteins involved in the classic beta(2)AR-cAMP cascade into a supramolecular signalling complex and thus allows highly localized and selective regulation of one of its major target proteins.


Asunto(s)
Adenilil Ciclasas/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Receptores AMPA/análisis , Receptores AMPA/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Adenilil Ciclasas/análisis , Animales , Canales de Calcio/metabolismo , Células Cultivadas , Corteza Cerebral/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/análisis , Homólogo 4 de la Proteína Discs Large , Electrofisiología , Subunidades alfa de la Proteína de Unión al GTP Gs/análisis , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Regulación de la Expresión Génica , Hipocampo/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/citología , Ratas , Ratas Sprague-Dawley , Receptores AMPA/genética , Receptores Adrenérgicos beta 2/análisis
10.
Pharm Res ; 31(10): 2829-43, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24792830

RESUMEN

PURPOSE: In pharmacokinetic (PK)/pharmacodynamic (PD) modelling and simulations (M&S), omitting dropouts can cause inaccuracies in parameter estimation and clinical trial simulations (CTS). This study examines the impact of different imputation methods for missing data on the interpretation of model results, as well as develops a selection model (where dropout and efficacy are jointly modelled) for use in CTS. METHODS: Missing data were imputed using single and multiple imputation and pattern mixtures methods for a previously reported duloxetine PK/PD model. The probability of dropout was described in the selection model and CTS was conducted with a hypothetical drug to examine the impact of dropout on trial results. RESULTS: The study completion rate was 75% and dropouts were not random. Model parameters obtained with different imputation methods were mostly within 40% (range 0 to 63%) compared to the model without dropouts. CTS showed 0.3 points lower median pain scores and 3% lower coefficient of variation over the 12-week simulations when dropout was included. CONCLUSIONS: Missing data had little impact on the original population PK/PD analyses. Sensitivity analyses for dropouts should be conducted in M&S exercises. The utility of selection models in CTS was explored via a hypothetical case study.


Asunto(s)
Analgésicos/farmacología , Analgésicos/farmacocinética , Modelos Biológicos , Pacientes Desistentes del Tratamiento , Tiofenos/farmacología , Tiofenos/farmacocinética , Analgésicos/uso terapéutico , Simulación por Computador , Interpretación Estadística de Datos , Relación Dosis-Respuesta a Droga , Clorhidrato de Duloxetina , Humanos , Dinámicas no Lineales , Dolor/tratamiento farmacológico , Dimensión del Dolor , Pacientes Desistentes del Tratamiento/estadística & datos numéricos , Probabilidad , Ensayos Clínicos Controlados Aleatorios como Asunto/estadística & datos numéricos , Tiofenos/uso terapéutico
11.
Clin Pharmacol Ther ; 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38864600

RESUMEN

Carbamazepine (CBZ) is the recommended alternative to rifampicin as a CYP3A4 inducer in drug-drug interaction studies. However, the traditional CBZ dosing paradigm can lead to several adverse events (AEs). This study tested a shorter CBZ dosing regimen using the CYP3A4-sensitive index substrate midazolam (MDZ). This was a fixed-sequence arm of an open-label, phase I study (NCT04840888). Healthy participants (n = 15) aged 18-63 years received oral doses of 1.2 mg MDZ alone (Day 1), CBZ b.i.d. alone (100 mg Days 2-4; 200 mg Days 5-7; 300 mg Days 8-10 and 12-13), and 300 mg CBZ b.i.d. plus 1.2 mg MDZ (Days 11 and 14). One participant (6.7%) experienced constipation due to treatment with CBZ plus MDZ on Day 11. One participant (6.7%) experienced urticaria (Days 12-13), and two participants (13.3%) experienced somnolence (Days 8-10) due to treatment with 300 mg CBZ b.i.d. alone. All AEs were mild. For MDZ, the geometric mean (90% CI) ratio (vs. Day 1) of the area under the curve (AUC 0-∞) was 0.28 (0.24-0.31) on Day 11 and 0.26 (0.23-0.29) on Day 14. The AUC (0-12 hours) of CBZ was 114,000 ng∙h/mL on Day 11 and 105,000 ng∙h/mL on Day 14. Steady-state concentrations of CBZ and induction of CYP3A4 were achieved on Day 11. The data are consistent with predictions of physiologically-based pharmacokinetic models in Simcyp. The 9-day dosing regimen for CBZ induction was well-tolerated by healthy participants, supporting the use of a shorter CBZ regimen for CYP3A4 induction studies.

12.
Child Adolesc Psychiatry Ment Health ; 18(1): 57, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773657

RESUMEN

BACKGROUND: The COVID-19 pandemic impacted children, adolescents, and their families, with significant psychosocial consequences. The prevalence of anxiety, depression, and self-injurious behaviors increased in our youth, as well as the number of suicide attempts and hospitalizations related to suicidal ideation. Additionally, parents' mental health saw increasing rates of depression, irritability, and alcohol use combined with worsening family function, child-parent connectedness, positive family expressiveness, and increases in family conflict. In light of these statistics, we created CHATogether (Compassionate Home, Action Together), a pilot family-centered intervention using multi-faceted psychotherapeutic approaches to improve familial communication and relational health between adolescents and their parents. This paper discusses the implementation of the CHATogether intervention at the Adolescent Intensive Outpatient Program (IOP), providing an example of the intervention through an in-depth pilot case, and evaluation of the program's acceptability and feasibility. METHODS: This paper describes a case in detail and evaluation from a total of 30 families that completed CHATogether in the initial pilot. Each family had 4-6 one-hour CHATogether sessions during their 6-week treatment course at the IOP. Before and after CHATogether, adolescents and their parents separately completed a questionnaire designed to explore their perceived family conflicts. After completion of the program, participants completed a brief quality improvement survey to assess their overall experience with CHATogether. In the reported case, the family completed Patient-Reported Outcomes Measurement Information System (PROMIS) depressive and anxiety symptoms scales, Conflict Behavior Questionnaires (CBQ), 9-item Concise Health Risk Tracking Self-Report (CHRT-SR9), and help-seeking attitude from adults during distress and suicide concerns. RESULTS: The pilot case showed a trend of improvement in reported depressive and anxiety symptoms, child-parent conflicts, subfactors of suicide risk including pessimism, helplessness, and despair, help-seeking acceptability from parents for suicide concerns, and the establishment of individualized family relationship goals. Preliminary feedback from participating families demonstrated positive effects on intra-family communication and improvement in the overall family dynamic. Adolescents (n = 30/30) and their parents (n = 30/30) rated "strongly agree" or "agree" that their families had benefited from CHATogether and welcomed participation in future program development. CONCLUSION: This study presents CHATogether as a novel family-centered intervention to address post-pandemic family mental health stress, especially when a family system was disrupted and negatively affected the mental health of children and adolescents. The intervention facilitated positive child-parent communication on a variety of topics, through tools such as emotional expression and help-seeking behavior. The reported pilot case and evaluation suggested CHATogether's acceptability and feasibility in a clinical context. We also provided quality improvement feedback to guide future studies in establishing the efficacy of CHATogether and other similar models of clinical family interventions.

13.
Proc Natl Acad Sci U S A ; 107(51): 22308-13, 2010 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21135234

RESUMEN

Alterations of synaptic transmission have been considered a core feature of mental disorders; thus, we examined the role of dopamine D(4) receptors, which is highly implicated in attention-deficit hyperactivity disorder and schizophrenia, in regulating synaptic functions of prefrontal cortex, a brain region critical for cognitive and emotional processes. We found that D(4) stimulation caused a profound depression or potentiation of AMPA receptor-mediated excitatory synaptic transmission in prefrontal cortex pyramidal neurons when their activity was elevated or dampened, respectively, which was accompanied by a D(4)-induced decrease or increase of AMPARs at synapses. The dual effects of D(4) on AMPAR trafficking and function was dependent on the D(4)-mediated bidirectional regulation of CaMKII activity via coupling to distinct signaling pathways, which provides a unique mechanism for D(4) receptors to serve as a homeostatic synaptic factor to stabilize cortical excitability.


Asunto(s)
Ácido Glutámico/metabolismo , Homeostasis/fisiología , Células Piramidales/metabolismo , Receptores de Dopamina D4/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Animales , Trastorno por Déficit de Atención con Hiperactividad/metabolismo , Humanos , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley , Esquizofrenia/metabolismo
14.
J Am Acad Child Adolesc Psychiatry ; 62(12): 1297-1300, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37088451

RESUMEN

LGBTQ Asian American youth face unique challenges related to their marginalized identities. It is well documented that Asian Americans who need mental health treatment access care at lower rates than White populations.1 Although Asian cultural values are often cited as reasons for decreased help-seeking behavior, research suggests structural barriers including cost, lack of culturally tailored services, and lack of knowledge of available resources as greater contributors to these disparities.1 Asian Americans have also been subject to the "model minority" myth, the stereotype that the community is universally high achieving, rule following, and well adjusted. This false narrative contributes to negative mental health outcomes driven by racial discrimination and homogenizing the Asian American experience. This masks the diversity in mental health needs among Asian Americans. In addition, LGBTQ Asian Americans experience microaggressions, the perception of being "not queer enough," and racism from LGBTQ spaces that often primarily cater to a White population.2.


Asunto(s)
Racismo , Minorías Sexuales y de Género , Humanos , Adolescente , Asiático , Salud Mental , Grupos Minoritarios
15.
Mol Pharmacol ; 81(2): 113-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22072817

RESUMEN

N-Methyl-D-aspartate (NMDA) receptor (NMDAR) hypofunction has been postulated to contribute to the cognitive deficit of schizophrenia. In this study, we examined the effect of lurasidone (Latuda; Dainippon Sumitomo Pharma Co. Ltd., Tokyo, Japan), a newly approved atypical antipsychotic drug (APD), on NMDAR synaptic function in rat frontal cortical pyramidal neurons. In vivo administration of lurasidone produced a significant and selective enhancement of NMDAR-mediated synaptic responses and surface expression of NR2A and NR2B subunits. Lurasidone has high affinity for serotonin 5-HT(1A), 5-HT(2A), and 5-HT(7) receptors and dopamine D(2) receptors. In vivo administration of the 5-HT(7) receptor antagonist (2R)-1-[(3-hydroxyphenyl)sulfonyl]-2 -(2-(4-methyl-1-piperidinyl)ethyl)pyrrolidine (SB-269970) mimicked the enhancing effect of lurasidone on NMDAR responses, whereas the D(2) receptor antagonist haloperidol failed to do so. Previous studies have found that short-term administration of lurasidone reverses the cognitive impairment induced by subchronic administration of phencyclidine (PCP), an NMDAR noncompetitive antagonist. In this study, we found that lurasidone, as well as the prototypical atypical APD clozapine, restored NMDAR-mediated synaptic responses to normal levels in the PCP model of schizophrenia. These results suggest that NMDAR is the potential key molecular target of lurasidone, possibility via antagonizing 5-HT(7) receptors, which is consistent with evidence that 5-HT(7) receptor antagonism contributes to cognitive enhancement by atypical APDs in patients with schizophrenia.


Asunto(s)
Isoindoles/farmacología , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Tiazoles/farmacología , Animales , Antipsicóticos , Clozapina/farmacología , Clozapina/uso terapéutico , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Isoindoles/uso terapéutico , Clorhidrato de Lurasidona , Ratas , Receptor de Serotonina 5-HT2A/efectos de los fármacos , Esquizofrenia/tratamiento farmacológico , Tiazoles/uso terapéutico
16.
J Biol Chem ; 286(28): 24957-65, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21622557

RESUMEN

Aberrant dopamine D(4) receptor function has been implicated in mental illnesses, including schizophrenia and attention deficit-hyperactivity disorder. Recently we have found that D(4) receptor exerts an activity-dependent bi-directional regulation of AMPA receptor (AMPAR)-mediated synaptic currents in pyramidal neurons of prefrontal cortex (PFC) via the dual control of calcium/calmodulin kinase II (CaMKII) activity. In this study, we examined the signaling mechanisms downstream of CaMKII that govern the complex effects of D(4) on glutamatergic transmission. We found that in PFC neurons at high activity state, D(4) suppresses AMPAR responses by disrupting the kinesin motor-based transport of GluR2 along microtubules, which was accompanied by the D(4) reduction of microtubule stability via a mechanism dependent on CaMKII inhibition. On the other hand, in PFC neurons at the low activity state, D(4) potentiates AMPAR responses by facilitating synaptic targeting of GluR1 through the scaffold protein SAP97 via a mechanism dependent on CaMKII stimulation. Taken together, these results have identified distinct signaling mechanisms underlying the homeostatic regulation of glutamatergic transmission by D(4) receptors, which may be important for cognitive and emotional processes in which dopamine is involved.


Asunto(s)
Homeostasis/fisiología , Receptores AMPA/metabolismo , Receptores de Dopamina D4/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Células Cultivadas , Dopamina/genética , Dopamina/metabolismo , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microtúbulos/genética , Microtúbulos/metabolismo , Células Piramidales , Ratas , Ratas Sprague-Dawley , Receptores AMPA/genética , Receptores de Dopamina D4/genética
17.
J Biol Chem ; 286(36): 31852-63, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21757762

RESUMEN

Emerging evidence indicates that amyloid ß peptide (Aß) initially induces subtle alterations in synaptic function in Alzheimer disease. We have recently shown that Aß binds to ß(2) adrenergic receptor (ß(2)AR) and activates protein kinase A (PKA) signaling for glutamatergic regulation of synaptic activities. Here we show that in the cerebrums of mice expressing human familial mutant presenilin 1 and amyloid precursor protein genes, the levels of ß(2)AR are drastically reduced. Moreover, Aß induces internalization of transfected human ß(2)AR in fibroblasts and endogenous ß(2)AR in primary prefrontal cortical neurons. In fibroblasts, Aß treatment also induces transportation of ß(2)AR into lysosome, and prolonged Aß treatment causes ß(2)AR degradation. The Aß-induced ß(2)AR internalization requires the N terminus of the receptor containing the peptide binding sites and phosphorylation of ß(2)AR by G protein-coupled receptor kinase, not by PKA. However, the G protein-coupled receptor kinase phosphorylation of ß(2)AR and the receptor internalization are much slower than that induced by ßAR agonist isoproterenol. The Aß-induced ß(2)AR internalization is also dependent on adaptor protein arrestin 3 and GTPase dynamin, but not arrestin 2. Functionally, pretreatment of primary prefrontal cortical neurons with Aß induces desensitization of ß(2)AR, which leads to attenuated response to subsequent stimulation with isoproterenol, including decreased cAMP levels, PKA activities, PKA phosphorylation of serine 845 on α-amino-2,3-dihydro-5-methyl-3-oxo-4-isoxazolepropanoic acid (AMPA) receptor subunit 1 (GluR1), and AMPA receptor-mediated miniature excitatory postsynaptic currents. This study indicates that Aß induces ß(2)AR internalization and degradation leading to impairment of adrenergic and glutamatergic activities.


Asunto(s)
Péptidos beta-Amiloides/fisiología , Neuronas/metabolismo , Corteza Prefrontal/citología , Receptores Adrenérgicos beta 2/metabolismo , Péptidos beta-Amiloides/genética , Animales , Endocitosis , Humanos , Ratones , Ratones Transgénicos , Presenilina-1/genética , Receptores AMPA/metabolismo , Receptores Adrenérgicos beta 2/análisis
18.
J Biol Chem ; 286(39): 33719-28, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21832090

RESUMEN

Emerging evidence from studies of Huntington disease (HD) pathophysiology suggests that huntingtin (htt) and its associated protein HAP1 participate in intracellular trafficking and synaptic function. However, it is largely unknown whether AMPA receptor trafficking, which is crucial for controlling the efficacy of synaptic excitation, is affected by the mutant huntingtin with polyglutamine expansion (polyQ-htt). In this study, we found that expressing polyQ-htt in neuronal cultures significantly decreased the amplitude and frequency of AMPAR-mediated miniature excitatory postsynaptic current (mEPSC), while expressing wild-type huntingtin (WT-htt) increased mEPSC. AMPAR-mediated synaptic transmission was also impaired in a transgenic mouse model of HD expressing polyQ-htt. The effect of polyQ-htt on mEPSC was mimicked by knockdown of HAP1 and occluded by the dominant negative HAP1. Moreover, we found that huntingtin affected mESPC via a mechanism depending on the kinesin motor protein, KIF5, which controls the transport of GluR2-containing AMPARs along microtubules in dendrites. The GluR2/KIF5/HAP1 complex was disrupted and dissociated from microtubules in the HD mouse model. Together, these data suggest that AMPAR trafficking and function is impaired by mutant huntingtin, presumably due to the interference of KIF5-mediated microtubule-based transport of AMPA receptors. The diminished strength of glutamatergic transmission could contribute to the deficits in movement control and cognitive processes in HD conditions.


Asunto(s)
Mutación , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Receptores AMPA/metabolismo , Potenciales Sinápticos , Transmisión Sináptica , Animales , Células HEK293 , Humanos , Proteína Huntingtina , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Proteínas Nucleares/genética , Péptidos/genética , Péptidos/metabolismo , Transporte de Proteínas/genética , Ratas , Receptores AMPA/genética
19.
Neurobiol Dis ; 46(2): 497-502, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22402331

RESUMEN

Growing evidence suggests that Huntington's disease (HD), a neurodegenerative movement disorder caused by the mutant huntingtin (htt) with an expanded polyglutamine (polyQ) repeat, is associated with the altered intracellular trafficking and synaptic function. GABA(A) receptors, the key determinant of the strength of synaptic inhibition, have been found to bind to the huntingtin associated protein 1 (HAP1). HAP1 serves as an adaptor linking GABA(A) receptors to the kinesin family motor protein 5 (KIF5), controlling the transport of GABA(A) receptors along microtubules in dendrites. In this study, we found that GABA(A)R-mediated synaptic transmission is significantly impaired in a transgenic mouse model of HD expressing polyQ-htt, which is accompanied by the diminished surface expression of GABA(A) receptors. Moreover, the GABA(A)R/HAP1/KIF5 complex is disrupted and dissociated from microtubules in the HD mouse model. These results suggest that GABA(A)R trafficking and function is impaired in HD, presumably due to the interference of KIF5-mediated microtubule-based transport of GABA(A) receptors. The diminished inhibitory synaptic efficacy could contribute to the loss of the excitatory/inhibitory balance, leading to increased neuronal excitotoxicity in HD.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/fisiopatología , Inhibición Neural/fisiología , Receptores de GABA-A/metabolismo , Transmisión Sináptica/fisiología , Animales , Potenciales Postsinápticos Inhibidores/fisiología , Ratones , Ratones Transgénicos , Microtúbulos/metabolismo , Transporte de Proteínas/fisiología
20.
Proc Natl Acad Sci U S A ; 106(33): 14075-9, 2009 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-19666502

RESUMEN

The prefrontal cortex (PFC), a key brain region controlling cognition and emotion, is strongly influenced by stress. While chronic stress often produces detrimental effects on these measures, acute stress has been shown to enhance learning and memory, predominantly through the action of corticosteroid stress hormones. We used a combination of electrophysiological, biochemical, and behavioral approaches in an effort to identify the cellular targets of acute stress. We found that behavioral stressors in vivo cause a long-lasting potentiation of NMDAR- and AMPAR-mediated synaptic currents via glucocorticoid receptors (GRs) selectively in PFC pyramidal neurons. This effect is accompanied by increased surface expression of NMDAR and AMPAR subunits in acutely stressed animals. Furthermore, behavioral tests indicate that working memory, a key function relying on recurrent excitation within networks of PFC neurons, is enhanced by acute stress via a GR-dependent mechanism. These results have identified a form of long-term potentiation of synaptic transmission induced by natural stimuli in vivo, providing a potential molecular and cellular mechanism for the beneficial effects of acute stress on cognitive processes subserved by PFC.


Asunto(s)
Glutamina/metabolismo , Corteza Prefrontal/metabolismo , Corticoesteroides/metabolismo , Animales , Conducta Animal , Electrofisiología/métodos , Glucocorticoides/metabolismo , Hidrocortisona/metabolismo , Potenciación a Largo Plazo , Masculino , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Receptores de Glucocorticoides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA