Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Physiol Rev ; 103(2): 1423-1485, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36422994

RESUMEN

The cephalic phase insulin response (CPIR) is classically defined as a head receptor-induced early release of insulin during eating that precedes a postabsorptive rise in blood glucose. Here we discuss, first, the various stimuli that elicit the CPIR and the sensory signaling pathways (sensory limb) involved; second, the efferent pathways that control the various endocrine events associated with eating (motor limb); and third, what is known about the central integrative processes linking the sensory and motor limbs. Fourth, in doing so, we identify open questions and problems with respect to the CPIR in general. Specifically, we consider test conditions that allow, or may not allow, the stimulus to reach the potentially relevant taste receptors and to trigger a CPIR. The possible significance of sweetness and palatability as crucial stimulus features and whether conditioning plays a role in the CPIR are also discussed. Moreover, we ponder the utility of the strict classical CPIR definition based on what is known about the effects of vagal motor neuron activation and thereby acetylcholine on the ß-cells, together with the difficulties of the accurate assessment of insulin release. Finally, we weigh the evidence of the physiological and clinical relevance of the cephalic contribution to the release of insulin that occurs during and after a meal. These points are critical for the interpretation of the existing data, and they support a sharper focus on the role of head receptors in the overall insulin response to eating rather than relying solely on the classical CPIR definition.


Asunto(s)
Insulina , Papilas Gustativas , Humanos , Insulina/metabolismo , Gusto/fisiología , Glucemia/metabolismo , Transducción de Señal
2.
Physiol Rev ; 102(2): 689-813, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486393

RESUMEN

During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.


Asunto(s)
Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Neuronas/fisiología , Animales , Homeostasis/fisiología , Humanos , Transducción de Señal/fisiología
3.
Proc Natl Acad Sci U S A ; 117(13): 7021-7029, 2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32179677

RESUMEN

Described here is the development of gadolinium(III) texaphyrin-platinum(IV) conjugates capable of overcoming platinum resistance by 1) localizing to solid tumors, 2) promoting enhanced cancer cell uptake, and 3) reactivating p53 in platinum-resistant models. Side by side comparative studies of these Pt(IV) conjugates to clinically approved platinum(II) agents and previously reported platinum(II)-texaphyrin conjugates demonstrate that the present Pt(IV) conjugates are more stable against hydrolysis and nucleophilic attack. Moreover, they display high potent antiproliferative activity in vitro against human and mouse cell cancer lines. Relative to the current platinum clinical standard of care (SOC), a lead Gd(III) texaphyrin-Pt(IV) prodrug conjugate emerging from this development effort was found to be more efficacious in subcutaneous (s.c.) mouse models involving both cell-derived xenografts and platinum-resistant patient-derived xenografts. Comparative pathology studies in mice treated with equimolar doses of the lead Gd texaphyrin-Pt(IV) conjugate or the US Food and Drug Administration (FDA)-approved agent oxaliplatin revealed that the conjugate was better tolerated. Specifically, the lead could be dosed at more than three times (i.e., 70 mg/kg per dose) the tolerable dose of oxaliplatin (i.e., 4 to 6 mg/kg per dose depending on the animal model) with little to no observable adverse effects. A combination of tumor localization, redox cycling, and reversible protein binding is invoked to explain the relatively increased tolerability and enhanced anticancer activity seen in vivo. On the basis of the present studies, we conclude that metallotexaphyrin-Pt conjugates may have substantial clinical potential as antitumor agents.


Asunto(s)
Antineoplásicos/administración & dosificación , Metaloporfirinas/administración & dosificación , Oxaliplatino/administración & dosificación , Células A549 , Animales , Antineoplásicos/farmacocinética , Resistencia a Antineoplásicos , Femenino , Células HCT116 , Humanos , Metaloporfirinas/farmacocinética , Ratones Desnudos , Oxaliplatino/farmacocinética , Profármacos/administración & dosificación , Profármacos/farmacocinética , Distribución Tisular , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Proc Natl Acad Sci U S A ; 116(16): 8018-8027, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30923123

RESUMEN

Control of multiple life-critical physiological and behavioral functions requires the hypothalamus. Here, we provide a comprehensive description and rigorous analysis of mammalian intrahypothalamic network architecture. To achieve this at the gray matter region (macroscale) level, macroscale connection (macroconnection) data for the rat hypothalamus were extracted from the primary literature. The dataset indicated the existence of 7,982 (of 16,770 possible) intrahypothalamic macroconnections. Network analysis revealed that the intrahypothalamic macroconnection network (its macroscale subconnectome) is divided into two identical top-level subsystems (or subnetworks), each composed of two nested second-level subsystems. At the top-level, this suggests a deeply integrated network; however, regional grouping of the two second-level subsystems suggested a partial separation between control of physiological functions and behavioral functions. Furthermore, inclusion of four candidate hubs (dominant network nodes) in the second-level subsystem that is associated prominently with physiological control suggests network primacy with respect to this function. In addition, comparison of network analysis with expression of gene markers associated with inhibitory (GAD65) and excitatory (VGLUT2) neurotransmission revealed a significant positive correlation between measures of network centrality (dominance) and the inhibitory marker. We discuss these results in relation to previous understandings of hypothalamic organization and provide, and selectively interrogate, an updated hypothalamus structure-function network model to encourage future hypothesis-driven investigations of identified hypothalamic subsystems.


Asunto(s)
Conectoma , Hipotálamo , Vías Nerviosas , Animales , Biología Computacional , Hipotálamo/anatomía & histología , Hipotálamo/fisiología , Masculino , Modelos Neurológicos , Vías Nerviosas/anatomía & histología , Vías Nerviosas/fisiología , Ratas , Ratas Sprague-Dawley
5.
Mol Pharm ; 16(5): 1799-1812, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-30925839

RESUMEN

Invasive pulmonary aspergillosis is a deadly fungal infection with a high mortality rate, particularly in patients having undergone transplant surgery. Voriconazole, a triazole antifungal pharmaceutical product, is considered as a first-line therapy for invasive pulmonary aspergillosis, and exhibits efficacy even for patients who have failed other antifungal drug therapies. The objective of this study is to develop high potency nanoaggregates of crystalline voriconazole composition for dry powder inhalation using the particle engineering process, thin film freezing. In this study, mannitol at low concentrations acted as a surface texture-modifying agent, and we evaluated the physicochemical and aerodynamic properties of the voriconazole formulations containing different amounts of mannitol. In vitro aerosol performance data demonstrated that powder formulations consisting of 90 to 97% (w/w) voriconazole were the optimum for inhalation with a fine particle fraction (% of delivered dose) as high as 73.6 ± 3.2% and mass median aerodynamic diameter of 3.03 ± 0.17 µm when delivered by a commercially available device. The thin film freezing process enabled phase-separated submicron crystalline mannitol to be oriented such as to modify the surface texture of the crystalline voriconazole nanoaggregates, thus enhancing their aerosolization. Addition of as low as 3% (w/w) mannitol significantly increased the fine particle fraction (% of metered dose) of voriconazole nanoaggregates when compared to compositions without mannitol (40.8% vs 24.6%, respectively). The aerosol performance of the voriconazole nanoaggregates with 5% (w/w) mannitol was maintained for 13 months at 25 °C/60% RH. Therefore, voriconazole nanoaggregates having low amounts of surface texture-modifying mannitol made by thin film freezing are a feasible local treatment option for invasive pulmonary aspergillosis with high aerosolization efficiency and drug loading for dry powder inhalation.


Asunto(s)
Aerosoles/química , Antifúngicos/química , Composición de Medicamentos/métodos , Inhaladores de Polvo Seco , Polvos/química , Voriconazol/química , Administración por Inhalación , Antifúngicos/uso terapéutico , Cristalización , Diseño de Fármacos , Liberación de Fármacos , Estabilidad de Medicamentos , Excipientes/química , Estudios de Factibilidad , Humanos , Aspergilosis Pulmonar Invasiva/tratamiento farmacológico , Manitol/química , Tamaño de la Partícula , Voriconazol/uso terapéutico
6.
AAPS PharmSciTech ; 20(3): 117, 2019 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-30783904

RESUMEN

Orally inhaled products have well-known benefits. They allow for effective local administration of many drugs for the treatment of pulmonary disease, and they allow for rapid absorption and avoidance of first-pass metabolism of several systemically acting drugs. Several challenges remain, however, such as dosing limitations, low and variable deposition of the drug in the lungs, and high drug deposition in the oropharynx region. These challenges have stimulated the development of new delivery technologies. Both formulation improvements and new device technologies have been developed through an improved understanding of the mechanisms of aerosolization and lung deposition. These new advancements in formulations have enabled improved aerosolization by controlling particle properties such as density, size, shape, and surface energy. New device technologies emerging in the marketplace focus on minimizing patient errors, expanding the range of inhaled drugs, improving delivery efficiency, increasing dose consistency and dosage levels, and simplifying device operation. Many of these new technologies have the potential to improve patient compliance. This article reviews how new delivery technologies in the form of new formulations and new devices enhance orally inhaled products.


Asunto(s)
Pulmón/metabolismo , Preparaciones Farmacéuticas/administración & dosificación , Administración por Inhalación , Aerosoles/administración & dosificación , Composición de Medicamentos , Sistemas de Liberación de Medicamentos , Inhaladores de Polvo Seco , Humanos
7.
Am J Physiol Regul Integr Comp Physiol ; 314(6): R811-R823, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29384699

RESUMEN

Hindbrain catecholamine neurons convey gut-derived metabolic signals to an interconnected neuronal network in the hypothalamus and adjacent forebrain. These neurons are critical for short-term glycemic control, glucocorticoid and glucoprivic feeding responses, and glucagon-like peptide 1 (GLP-1) signaling. Here we investigate whether these pathways also contribute to long-term energy homeostasis by controlling obesogenic sensitivity to a high-fat/high-sucrose choice (HFSC) diet. We ablated hindbrain-originating catecholaminergic projections by injecting anti-dopamine-ß-hydroxylase-conjugated saporin (DSAP) into the paraventricular nucleus of the hypothalamus (PVH) of male rats fed a chow diet for up to 12 wk or a HFSC diet for 8 wk. We measured the effects of DSAP lesions on food choices; visceral adiposity; plasma glucose, insulin, and leptin; and indicators of long-term ACTH and corticosterone secretion. We also determined lesion effects on the number of carbohydrate or fat calories required to increase visceral fat. Finally, we examined corticotropin-releasing hormone levels in the PVH and arcuate nucleus expression of neuropeptide Y ( Npy), agouti-related peptide ( Agrp), and proopiomelanocortin ( Pomc). DSAP-injected chow-fed rats slowly increase visceral adiposity but quickly develop mild insulin resistance and elevated blood glucose. DSAP-injected HFSC-fed rats, however, dramatically increase food intake, body weight, and visceral adiposity beyond the level in control HFSC-fed rats. These changes are concomitant with 1) a reduction in the number of carbohydrate calories required to generate visceral fat, 2) abnormal Npy, Agrp, and Pomc expression, and 3) aberrant control of insulin secretion and glucocorticoid negative feedback. Long-term metabolic adaptations to high-carbohydrate diets, therefore, require intact forebrain catecholamine projections. Without them, animals cannot alter forebrain mechanisms to restrain increased visceral adiposity.


Asunto(s)
Catecolaminas/metabolismo , Red Nerviosa/fisiopatología , Obesidad/fisiopatología , Prosencéfalo/fisiopatología , Animales , Glucemia/metabolismo , Dieta , Dopamina beta-Hidroxilasa/antagonistas & inhibidores , Ingestión de Energía , Insulina/sangre , Grasa Intraabdominal/efectos de los fármacos , Leptina/sangre , Masculino , Vías Nerviosas/fisiopatología , Núcleo Hipotalámico Paraventricular , Ratas , Ratas Sprague-Dawley , Saporinas/farmacología
8.
Am J Physiol Regul Integr Comp Physiol ; 315(4): R708-R720, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29847161

RESUMEN

Endogenous intestinal glucagon-like peptide-1 (GLP-1) controls satiation and glucose metabolism via vagal afferent neurons (VANs). Recently, VANs have received increasing attention for their role in brown adipose tissue (BAT) thermogenesis. It is, however, unclear whether VAN GLP-1 receptor (GLP-1R) signaling affects BAT thermogenesis and energy expenditure (EE) and whether this VAN mechanism contributes to energy balance. First, we tested the effect of the GLP-1R agonist exendin-4 (Ex4, 0.3 µg/kg ip) on EE and BAT thermogenesis and whether these effects require VAN GLP-1R signaling using a rat model with a selective Glp1r knockdown (kd) in VANs. Second, we examined the role of VAN GLP-1R in energy balance during chronic high-fat diet (HFD) feeding in VAN Glp1r kd rats. Finally, we used viral transsynaptic tracers to identify the possible neuronal substrates of such a gut-BAT interaction. VAN Glp1r kd attenuated the acute suppressive effects of Ex4 on EE and BAT thermogenesis. Consistent with this finding, the VAN Glp1r kd increased EE and BAT activity, diminished body weight gain, and improved insulin sensitivity compared with HFD-fed controls. Anterograde transsynaptic viral tracing of VANs infected major hypothalamic and hindbrain areas involved in BAT sympathetic regulation. Moreover, retrograde tracing from BAT combined with laser capture microdissection revealed that a population of VANs expressing Glp1r is synaptically connected to the BAT. Our findings reveal a novel role of VAN GLP-1R signaling in the regulation of EE and BAT thermogenesis and imply that through this gut-brain-BAT connection, intestinal GLP-1 plays a role in HFD-induced metabolic syndrome.


Asunto(s)
Tejido Adiposo Pardo/inervación , Sistema Nervioso Autónomo/metabolismo , Encéfalo/metabolismo , Metabolismo Energético , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Intestinos/inervación , Termogénesis , Animales , Sistema Nervioso Autónomo/efectos de los fármacos , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Exenatida/farmacología , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/genética , Incretinas/farmacología , Masculino , Vías Nerviosas/metabolismo , Neuronas Aferentes/metabolismo , Ratas Sprague-Dawley , Transducción de Señal , Termogénesis/efectos de los fármacos
9.
Am J Physiol Regul Integr Comp Physiol ; 312(3): R324-R337, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28077392

RESUMEN

The brain networks connected to the sympathetic motor and sensory innervations of brown (BAT) and white (WAT) adipose tissues were originally described using two transneuronally transported viruses: the retrogradely transported pseudorabies virus (PRV), and the anterogradely transported H129 strain of herpes simplex virus-1 (HSV-1 H129). Further complexity was added to this network organization when combined injections of PRV and HSV-1 H129 into either BAT or WAT of the same animal generated sets of coinfected neurons in the brain, spinal cord, and sympathetic and dorsal root ganglia. These neurons are well positioned to act as sensorimotor links in the feedback circuits that control each fat pad. We have now determined the extent of sensorimotor crosstalk between interscapular BAT (IBAT) and inguinal WAT (IWAT). PRV152 and HSV-1 H129 were each injected into IBAT or IWAT of the same animal: H129 into IBAT and PRV152 into IWAT. The reverse configuration was applied in a different set of animals. We found single-labeled neurons together with H129+PRV152 coinfected neurons in multiple brain sites, with lesser numbers in the sympathetic and dorsal root ganglia that innervate IBAT and IWAT. We propose that these coinfected neurons mediate sensory-sympathetic motor crosstalk between IBAT and IWAT. Comparing the relative numbers of coinfected neurons between the two injection configurations showed a bias toward IBAT-sensory and IWAT-sympathetic motor feedback loops. These coinfected neurons provide a neuroanatomical framework for functional interactions between IBAT thermogenesis and IWAT lipolysis that occurs with cold exposure, food restriction/deprivation, exercise, and more generally with alterations in adiposity.


Asunto(s)
Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/inervación , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/inervación , Corteza Sensoriomotora/citología , Sistema Nervioso Simpático/citología , Tejido Adiposo Pardo/fisiología , Tejido Adiposo Blanco/fisiología , Animales , Cricetinae , Retroalimentación Sensorial , Masculino , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Neuronas/citología , Neuronas/fisiología , Phodopus , Receptor Cross-Talk , Corteza Sensoriomotora/fisiología , Sistema Nervioso Simpático/fisiología
10.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1177-85, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27030665

RESUMEN

The consensus view of the ventromedial nucleus of the hypothalamus (VMH) is that it is a key node in the rodent brain network controlling sympathoadrenal counterregulatory responses to hypoglycemia. To identify the location of hypoglycemia-responsive neurons in the VMH, we performed a high spatial resolution Fos analysis in the VMH of rats made hypoglycemic with intraperitoneal injections of insulin. We examined Fos expression in the four constituent parts of VMH throughout its rostrocaudal extent and determined their relationship to blood glucose concentrations. Hypoglycemia significantly decreased Fos expression only in the dorsomedial and central parts of the VMH, but not its anterior or ventrolateral parts. Moreover, the number of Fos-expressing neurons was significantly and positively correlated in the two responsive regions with terminal blood glucose concentrations. We also measured Fos responses in the paraventricular nucleus of the hypothalamus (PVH) and in several levels of the periaqueductal gray (PAG), which receives strong projections from the VMH. We found the expected and highly significant increase in Fos in the neuroendocrine PVH, which was negatively correlated to terminal blood glucose concentrations, but no significant differences were seen in any part of the PAG. Our results show that there are distinct populations of VMH neurons whose Fos expression is suppressed by hypoglycemia, and their numbers correlate with blood glucose. These findings support a clear division of glycemic control functions within the different parts of the VMH.


Asunto(s)
Glucemia/metabolismo , Hipoglucemia/fisiopatología , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Animales , Progresión de la Enfermedad , Regulación hacia Abajo , Masculino , Especificidad de Órganos , Ratas , Ratas Wistar , Distribución Tisular
11.
Physiology (Bethesda) ; 29(5): 314-24, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25180261

RESUMEN

Hypoglycemia poses a serious threat to the integrity of the brain, owing to its reliance on blood glucose as a fuel. Protecting against hypoglycemia is an extended network of glucose sensors located within the brain and in the periphery that serve to mediate responses restoring euglycemia, i.e., counterregulatory responses. This review examines the various glucose sensory loci involved in hypoglycemic detection, with a particular emphasis on peripheral glucose sensory loci and their contribution to hypoglycemic counterregulation.


Asunto(s)
Glucemia/análisis , Glucosa/metabolismo , Hipoglucemia/diagnóstico , Hipoglucemia/metabolismo , Hipoglucemiantes/uso terapéutico , Animales , Encéfalo/metabolismo , Humanos , Neuronas/metabolismo
12.
Curr Diab Rep ; 14(3): 465, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24510608

RESUMEN

It is now widely accepted that the brain makes important contributions to the dysregulated glucose metabolism, altered feeding behaviors, and the obesity often seen in type 2 diabetes (T2D). Although studies focusing on genetic, cellular, and molecular regulatory elements in pancreas, liver, adipose tissue etc provide a good understanding of how these processes relate to T2D, our knowledge of how brain wiring patterns are organized is much less developed. This article discusses animal studies that illustrate the importance of understanding the network organization of those brain regions most closely implicated in T2D. It will describe the brain networks, as well as the methodologies used to explore them. To illustrate some of the gaps in our knowledge, we will discuss the connectional network of the ventromedial nucleus and its adjacent cell groups in the hypothalamus; structures that are widely recognized as key elements in the brain's ability to control glycemia, feeding, and body weight.


Asunto(s)
Encéfalo/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Red Nerviosa/fisiopatología , Animales , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Red Nerviosa/metabolismo , Red Nerviosa/patología , Núcleo Hipotalámico Ventromedial/metabolismo , Núcleo Hipotalámico Ventromedial/patología , Núcleo Hipotalámico Ventromedial/fisiopatología
13.
AAPS PharmSciTech ; 15(4): 981-93, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24824172

RESUMEN

Recently, inhaled immunosuppressive agents have attracted increasing attention for maintenance therapy following lung transplantation. The rationale for this delivery approach includes a more targeted and localized delivery to the diseased site with reduced systemic exposure, potentially leading to decreased adverse side effects. In this study, the in vitro and in vivo performance of an amorphous formulation prepared by thin film freezing (TFF) and a crystalline micronized formulation produced by milling was compared for tacrolimus (TAC). Despite the relatively large geometric size, the TFF-processed formulation was capable of achieving deep lung delivery due to its low-density, highly porous, and brittle characteristics. When emitted from a Miat® monodose inhaler, TFF-processed TAC formulations exhibited a fine particle fraction (FPF) of 83.3% and a mass median aerodynamic diameter (MMAD) of 2.26 µm. Single-dose 24-h pharmacokinetic studies in rats demonstrated that the TAC formulation prepared by TFF exhibited higher pulmonary bioavailability with a prolonged retention time in the lung, possibly due to decreased clearance (e.g., macrophage phagocytosis), compared to the micronized TAC formulation. Additionally, TFF formulation generated a lower systemic TAC concentration with smaller variability than the micronized formulation following inhalation, potentially leading to reduced side effects related to the drug in systemic circulation.


Asunto(s)
Polvos/administración & dosificación , Polvos/química , Administración por Inhalación , Animales , Disponibilidad Biológica , Química Farmacéutica/métodos , Sistemas de Liberación de Medicamentos/métodos , Inhaladores de Polvo Seco/métodos , Excipientes/administración & dosificación , Excipientes/química , Femenino , Congelación , Inmunosupresores/administración & dosificación , Inmunosupresores/química , Inmunosupresores/farmacocinética , Pulmón/metabolismo , Masculino , Tamaño de la Partícula , Porosidad , Polvos/farmacocinética , Ratas , Ratas Sprague-Dawley , Tacrolimus/farmacocinética , Tecnología Farmacéutica/métodos
14.
PLoS One ; 19(6): e0300624, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38870244

RESUMEN

This observational study evaluated the impact of a sponsor company-provided Patient Support Program (PSP) on discontinuation of adalimumab in adult Australian patients eligible for Pharmaceutical Benefit Scheme (PBS)-reimbursed adalimumab for Rheumatoid Arthritis (RA), Ankylosing Spondylitis (AS), Psoriatic Arthritis (PsA), Crohn's Disease (CD), Ulcerative Colitis (UC), or Hidradenitis Suppurativa (HS). Patients initiating adalimumab between May 2018 and September 2019 were enrolled into two prospective cohorts based on their decision to opt for or decline the PSP (PSP or non-PSP cohorts). In addition, a historical, retrospective Non-PSP cohort was established from the Services Australia 10% PBS dataset by extracting data of patients initiating adalimumab prior to the introduction of adalimumab PSPs and based on adalimumab PBS listing dates (AS: April 2007 to March 2009; PsA/RA: January 2007 to December 2008; CD: January 2009 to December 2010; HS and UC indications not included). Follow-up for all cohorts was 12 months. The primary endpoint was the time to discontinuation, compared between the prospective PSP cohort and the prospective or retrospective Non-PSP cohort. Inverse probability of treatment weighting was used to balance the cohorts. A Cox proportional hazards model indicated no difference in time to discontinuation between the prospective PSP (n = 162) and non-PSP (n = 65) cohorts (HR [95% CI] = 1.256 [0.616-2.563], p = 0.5304). The 12-month adalimumab persistence rates (95% CI) were 78% (69%, 84%) and 82% (67%, 90%), respectively. In contrast, discontinuation was less likely in the prospective PSP (n = 151) compared with the retrospective non-PSP (n = 297) cohort (HR [95% CI] = 0.44 [0.28-0.68], p<0.001). The 12-month persistence rates (95% CI) were 81% (76%, 90%) and 61% (56%, 67%), respectively. Overall, this study suggests that optimal adalimumab persistence can be achieved with either a structured PSP or healthcare support from other sources, but this was not the case more than a decade ago.


Asunto(s)
Adalimumab , Humanos , Adalimumab/uso terapéutico , Adalimumab/administración & dosificación , Femenino , Masculino , Adulto , Persona de Mediana Edad , Australia , Estudios Retrospectivos , Estudios Prospectivos , Antirreumáticos/uso terapéutico , Antirreumáticos/administración & dosificación , Espondilitis Anquilosante/tratamiento farmacológico , Anciano , Privación de Tratamiento
15.
Am J Physiol Endocrinol Metab ; 305(1): E78-88, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23651844

RESUMEN

Blood glucose concentration is tightly regulated by the rate of insulin secretion and clearance, a process partially controlled by sensory neurons serving as metabolic sensors in relevant tissues. The activity of these neurons is regulated by the products of metabolism which regulate transmitter release, and recent evidence suggests that neuronally expressed ion channels of the transient receptor potential (TRP) family function in this critical process. Here, we report the novel finding that the cold and menthol-gated channel TRPM8 is necessary for proper insulin homeostasis. Mice lacking TRPM8 respond normally to a glucose challenge while exhibiting prolonged hypoglycemia in response to insulin. Additionally, Trpm8-/- mice have increased rates of insulin clearance compared with wild-type animals and increased expression of insulin-degrading enzyme in the liver. TRPM8 channels are not expressed in the liver, but TRPM8-expressing sensory afferents innervate the hepatic portal vein, suggesting a TRPM8-mediated neuronal control of liver insulin clearance. These results demonstrate that TRPM8 is a novel regulator of serum insulin and support the role of sensory innervation in metabolic homeostasis.


Asunto(s)
Glucemia/metabolismo , Hipoglucemia/genética , Insulina/metabolismo , Células Receptoras Sensoriales/metabolismo , Canales Catiónicos TRPM/genética , Animales , Plásmidos de Bacteriocinas , Diabetes Mellitus Experimental/metabolismo , Homeostasis/fisiología , Hipoglucemia/metabolismo , Células Secretoras de Insulina/metabolismo , Hígado/irrigación sanguínea , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vena Porta/inervación , Ratas , Canales Catiónicos TRPM/metabolismo
16.
Pharm Res ; 30(3): 813-25, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23229856

RESUMEN

PURPOSE: Inhalation of low-density porous particles enables deep lung delivery with less dependence on device design and patient inspiration. The purpose of this study was to implement Thin Film Freezing (TFF) to investigate a novel approach to dry powder inhalation. METHODS: Powders produced by TFF were evaluated for aerodynamic and geometric particle size by cascade impaction and laser light scattering, respectively. Density measurements were conducted according to USP methods and calculated using data from particle size measurements. Excipient inclusion and its effect on moisture sorption was measured by Dynamic Vapor Sorption (DVS). RESULTS: TFF-produced brittle matrix powders were sheared apart into respirable microparticles using a passive DPI device, producing fine particle fractions (FPF) up to 69% and mass median aerodynamic diameters (MMAD) as low as 2.6 µm. Particles had a mean geometric diameter ranging from 25 µm to 50 µm and mass densities of approximately 0.01 g/cm(3). Powders were susceptible to moisture-induced matrix collapse, capillary forces and electrostatic charging; although formulations containing mannitol or no sugar excipient proved to be more robust. CONCLUSIONS: Aerosolized brittle matrices produced by TFF may prove to be a useful platform for highly efficient pulmonary delivery of thermally labile, highly potent, and poorly soluble drugs.


Asunto(s)
Aerosoles/química , Polvos/química , Administración por Inhalación , Carbohidratos/química , Inhaladores de Polvo Seco , Excipientes/química , Congelación , Humanos , Manitol/química , Tamaño de la Partícula , Porosidad , Agua/química
17.
J Neuroendocrinol ; 35(11): e13251, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37053148

RESUMEN

Throughout the 20th Century, regulatory peptide discovery advanced from the identification of gut hormones to the extraction and characterization of hypothalamic hypophysiotropic factors, and to the isolation and cloning of multiple brain neuropeptides. These discoveries were followed by the discovery of G-protein-coupled and other membrane receptors for these peptides. Subsequently, the systems physiology associated with some of these multiple regulatory peptides and receptors has been comprehensively elucidated and has led to improved therapeutics and diagnostics and their approval by the US Food and Drug Administration. In light of this wealth of information and further potential, it is truly a time of renaissance for regulatory peptides. In this perspective, we review what we have learned from the pioneers in exemplified fields of gut peptides, such as cholecystokinin, enterochromaffin-like-cell peptides, and glucagon, from the trailblazing studies on the key stress hormone, corticotropin-releasing factor, as well as from more recently characterized relaxin-family peptides and receptors. The historical viewpoints are based on our understanding of these topics in light of the earliest phases of research and on subsequent studies and the evolution of knowledge, aiming to sharpen our vision of the current state-of-the-art and those studies that should be prioritized in the future.


Asunto(s)
Neuropéptidos , Relaxina , Hormona Liberadora de Corticotropina , Colecistoquinina , Glucagón
18.
J Neurosci ; 31(50): 18479-91, 2011 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-22171049

RESUMEN

Physiological responses to hypoglycemia, hyperinsulinemia, and hyperglycemia include a critical adrenocortical component that is initiated by hypothalamic control of the anterior pituitary and adrenal cortex. These adrenocortical responses ensure appropriate long-term glucocorticoid-mediated modifications to metabolism. Despite the importance of these mechanisms to disease processes, how hypothalamic afferent pathways engage the intracellular mechanisms that initiate adrenocortical responses to glycemia-related challenges are unknown. This study explores these mechanisms using network- and cellular-level interventions in in vivo and ex vivo rat preparations. Results show that a hindbrain-originating catecholamine afferent system selectively engages a MAP kinase pathway in rat paraventricular hypothalamic CRH (corticotropin-releasing hormone) neuroendocrine neurons shortly after vascular insulin and 2-deoxyglucose challenges. In turn, this MAP kinase pathway can control both neuroendocrine neuronal firing rate and the state of CREB phosphorylation in a reduced ex vivo paraventricular hypothalamic preparation, making this signaling pathway an ideal candidate for coordinating CRH synthesis and release. These results establish the first clear structural and functional relationships linking neurons in known nutrient-sensing regions with intracellular mechanisms in hypothalamic CRH neuroendocrine neurons that initiate the adrenocortical response to various glycemia-related challenges.


Asunto(s)
Catecolaminas/metabolismo , Hipotálamo/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Sistema Hipófiso-Suprarrenal/metabolismo , Rombencéfalo/metabolismo , Animales , Glucemia/metabolismo , Desoxiglucosa/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/efectos de los fármacos , Insulina/farmacología , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Rombencéfalo/efectos de los fármacos
19.
Front Neuroendocrinol ; 31(1): 32-43, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19836412

RESUMEN

Glucose is the primary fuel for the vast majority of cells, and animals have evolved essential cellular, autonomic, endocrine, and behavioral measures to counteract both hypo- and hyperglycemia. A central component of these counterregulatory mechanisms is the ability of specific sensory elements to detect changes in blood glucose and then use that information to produce appropriate counterregulatory responses. Here we focus on the organization of the neural systems that are engaged by glucosensing mechanisms when blood glucose concentrations fall to levels that pose a physiological threat. We employ a classic sensory-motor integrative schema to describe the peripheral, hindbrain, and hypothalamic components that make up counterregulatory mechanisms in the brain. We propose that models previously developed to describe how the forebrain modulates autonomic reflex loops in the hindbrain offer a reasoned framework for explaining how counterregulatory neural mechanisms in the hypothalamus and hindbrain are structured.


Asunto(s)
Glucemia/metabolismo , Encéfalo , Homeostasis/fisiología , Hipoglucemia , Red Nerviosa , Encéfalo/patología , Encéfalo/fisiopatología , Glucosa , Humanos , Hipoglucemia/fisiopatología , Masculino , Venas Mesentéricas , Red Nerviosa/patología , Red Nerviosa/fisiopatología , Neuronas/fisiología , Vena Porta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA