Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.814
Filtrar
1.
Mol Pharm ; 21(5): 2456-2472, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38568423

RESUMEN

Variability of the gastrointestinal tract is rarely reflected in in vitro test protocols but often turns out to be crucial for the oral dosage form performance. In this study, we present a generation method of dissolution profiles accounting for the variability of fasted gastric conditions. The workflow featured 20 biopredictive tests within the physiological variability. The experimental array was constructed with the use of the design of experiments, based on three parameters: gastric pH and timings of the intragastric stress event and gastric emptying. Then, the resulting dissolution profiles served as a training data set for the dissolution process modeling with the machine learning algorithms. This allowed us to generate individual dissolution profiles under a customizable gastric pH and motility patterns. For the first time ever, we used the method to successfully elucidate dissolution properties of two dosage forms: pellet-filled capsules and bare pellets of the marketed dabigatran etexilate product Pradaxa. We showed that the dissolution of capsules was triggered by mechanical stresses and thus was characterized by higher variability and a longer dissolution onset than observed for pellets. Hence, we proved the applicability of the method for the in vitro and in silico characterization of immediate-release dosage forms and, potentially, for the improvement of in vitro-in vivo extrapolation.


Asunto(s)
Cápsulas , Dabigatrán , Ayuno , Vaciamiento Gástrico , Dabigatrán/química , Dabigatrán/administración & dosificación , Dabigatrán/farmacología , Cápsulas/química , Vaciamiento Gástrico/fisiología , Vaciamiento Gástrico/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Solubilidad , Liberación de Fármacos , Administración Oral , Simulación por Computador , Estómago/fisiología , Estómago/efectos de los fármacos
2.
Am J Physiol Gastrointest Liver Physiol ; 326(5): G622-G630, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38375576

RESUMEN

Biopsychosocial factors are associated with disorders of gut-brain interaction (DGBI) and exacerbate gastrointestinal symptoms. The mechanisms underlying pathophysiological alterations of stress remain unclear. Corticotropin-releasing hormone (CRH) is a central regulator of the hormonal stress response and has diverse impact on different organ systems. The aim of the present study was to investigate the effects of peripheral CRH infusion on meal-related gastrointestinal symptoms, gastric electrical activity, and gastric sensorimotor function in healthy volunteers (HVs). In a randomized, double-blinded, placebo-controlled, crossover study, we evaluated the effects of CRH on gastric motility and sensitivity. HVs were randomized to receive either peripheral-administered CRH (100 µg bolus + 1 µg/kg/h) or placebo (saline), followed by at least a 7-day washout period and assignment to the opposite treatment. Tests encompassed saliva samples, gastric-emptying (GE) testing, body surface gastric mapping (BSGM, Gastric Alimetry; Alimetry) to assess gastric myoelectrical activity with real-time symptom profiling, and a gastric barostat study to assess gastric sensitivity to distention and accommodation. Twenty HVs [13 women, mean age 29.2 ± 5.3 yr, body mass index (BMI) 23.3 ± 3.8 kg/m2] completed GE tests, of which 18 also underwent BSGM measurements during the GE tests. The GE half-time decreased significantly after CRH exposure (65.2 ± 17.4 vs. 78.8 ± 24.5 min, P = 0.02) with significantly increased gastric amplitude [49.7 (34.7-55.6) vs. 31.7 (25.7-51.0) µV, P < 0.01], saliva cortisol levels, and postprandial symptom severity. Eleven HVs also underwent gastric barostat studies on a separate day. However, the thresholds for discomfort during isobaric distensions, gastric compliance, and accommodation did not differ between CRH and placebo.NEW & NOTEWORTHY In healthy volunteers, peripheral corticotropin-releasing hormone (CRH) infusion accelerates gastric-emptying rate and increases postprandial gastric response, accompanied by a rise in symptoms, but does not alter gastric sensitivity or meal-induced accommodation. These findings underscore a significant link between stress and dyspeptic symptoms, with CRH playing a pivotal role in mediating these effects.


Asunto(s)
Hormona Liberadora de Corticotropina , Estudios Cruzados , Vaciamiento Gástrico , Voluntarios Sanos , Estómago , Humanos , Femenino , Masculino , Hormona Liberadora de Corticotropina/metabolismo , Hormona Liberadora de Corticotropina/administración & dosificación , Hormona Liberadora de Corticotropina/farmacología , Adulto , Método Doble Ciego , Estómago/efectos de los fármacos , Estómago/fisiología , Vaciamiento Gástrico/efectos de los fármacos , Adulto Joven , Saliva/metabolismo
3.
Food Funct ; 15(4): 2221-2233, 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38318756

RESUMEN

Motilin is an important hormonal regulator in the migrating motor complex (MMC). Free fatty acid receptor-1 (FFAR1, also known as GPR40) has been reported to stimulate motilin release in human duodenal organoids. However, how FFAR1 regulates gastric motility in vivo is unclear. This study investigated the role of FFAR1 in the regulation of gastric contractions and its possible mechanism of action using Suncus murinus. Firstly, intragastric administration of oleic acid (C18:1, OA), a natural ligand for FFAR1, stimulated phase II-like contractions, followed by phase III-like contractions in the fasted state, and the gastric emptying rate was accelerated. The administration of GW1100, an FFAR1 antagonist, inhibited the effects of OA-induced gastric contractions. Intravenous infusion of a ghrelin receptor antagonist (DLS) or serotonin 4 (5-HT4) receptor antagonist (GR125487) inhibited phase II-like contractions and prolonged the onset of phase III-like contractions induced by OA. MA-2029, a motilin receptor antagonist, delayed the occurrence of phase III-like contractions. In vagotomized suncus, OA did not induce phase II-like contractions. In addition, OA promoted gastric emptying through a vagal pathway during the postprandial period. However, OA did not directly act on the gastric body to induce contractions in vitro. In summary, this study indicates that ghrelin, motilin, 5-HT, and the vagus nerve are involved in the role of FFAR1 regulating MMC. Our findings provide novel evidence for the involvement of nutritional factors in the regulation of gastric motility.


Asunto(s)
Ácidos Grasos no Esterificados , Motilidad Gastrointestinal , Humanos , Animales , Ácidos Grasos no Esterificados/farmacología , Motilina/metabolismo , Motilina/farmacología , Complejo Mioeléctrico Migratorio/fisiología , Estómago/fisiología , Musarañas/metabolismo
5.
Adv Mater ; 36(19): e2312735, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38290128

RESUMEN

Devices interfacing with biological tissues can provide valuable insights into function, disease, and metabolism through electrical and mechanical signals. However, certain neuromuscular tissues, like those in the gastrointestinal tract, undergo significant strains of up to 40%. Conventional inextensible devices cannot capture the dynamic responses in these tissues. This study introduces electrodes made from poly(3,4-ethylenedioxythiophene):poly(styrenesulfonate) (PEDOT:PSS) and polydimethylsiloxane (PDMS) that enable simultaneous monitoring of electrical and mechanical responses of gut tissue. The soft PDMS layers conform to tissue surfaces during gastrointestinal movement. Dopants, including Capstone FS-30 and polyethylene glycol, are explored to enhance the conductivity, electrical sensitivity to strain, and stability of the PEDOT:PSS. The devices are fabricated using shadow masks and solution-processing techniques, providing a faster and simpler process than traditional clean-room-based lithography. Tested on ex vivo mouse colon and human stomach, the device recorded voltage changes of up to 300 µV during contraction and distension consistent with muscle activity, while simultaneously recording resistance changes of up to 150% due to mechanical strain. These devices detect and respond to chemical stimulants and blockers, and can induce contractions through electrical stimulation. They hold great potential for studying and treating complex disorders like irritable bowel syndrome and gastroparesis.


Asunto(s)
Dimetilpolisiloxanos , Poliestirenos , Animales , Ratones , Poliestirenos/química , Humanos , Dimetilpolisiloxanos/química , Contracción Muscular/fisiología , Electrodos , Tracto Gastrointestinal/fisiología , Estómago/fisiología , Colon/fisiología , Conductividad Eléctrica , Polímeros/química , Fenómenos Electrofisiológicos , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Tiofenos/química , Tiofenos/farmacología
6.
Neurogastroenterol Motil ; 36(2): e14723, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38062544

RESUMEN

BACKGROUND: Despite evidence that slow-wave dysrhythmia in the stomach is associated with clinical conditions such as gastroparesis and functional dyspepsia, there is still no widely available device for long-term monitoring of gastric electrical signals. Actionable biomarkers of gastrointestinal health are critically needed, and an implantable slow-wave monitoring device could aid in the establishment of causal relationships between symptoms and gastric electrophysiology. Recent developments in the area of wireless implantable gastric monitors demonstrate potential, but additional work and validation are required before this potential can be realized. METHODS: We hypothesized that translating an existing implantable cardiac monitoring device, the Reveal LINQ™ (Medtronic), would present a more immediate solution. Following ethical approval and laparotomy in anesthetized pigs (n = 7), a Reveal LINQ was placed on the serosal surface of the stomach, immediately adjacent to a validated flexible-printed-circuit (FPC) electrical mapping array. Data were recorded for periods of 7.5 min, and the resultant signal characteristics from the FPC array and Reveal LINQ were compared. KEY RESULTS: The Reveal LINQ device recorded slow waves in 6/7 subjects with a comparable period (p = 0.69), signal-to-noise ratio (p = 0.58), and downstroke width (p = 0.98) to the FPC, but with reduced amplitude (p = 0.024). Qualitatively, the Reveal LINQ slow-wave signal lacked the prolonged repolarization phase present in the FPC signals. CONCLUSIONS & INFERENCES: These findings suggest that existing cardiac monitors may offer an efficient solution for the long-term monitoring of slow waves. Translation toward implantation now awaits.


Asunto(s)
Motilidad Gastrointestinal , Gastroparesia , Porcinos , Humanos , Animales , Motilidad Gastrointestinal/fisiología , Estómago/fisiología , Fenómenos Electrofisiológicos
7.
Acta Biomater ; 173: 167-183, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37984627

RESUMEN

The complex mechanics of the gastric wall facilitates the main digestive tasks of the stomach. However, the interplay between the mechanical properties of the stomach, its microstructure, and its vital functions is not yet fully understood. Importantly, the pig animal model is widely used in biomedical research for preliminary or ethically prohibited studies of the human digestion system. Therefore, this study aims to thoroughly characterize the mechanical behavior and microstructure of the porcine stomach. For this purpose, multiple quasi-static mechanical tests were carried out with three different loading modes, i.e., planar biaxial extension, radial compression, and simple shear. Stress-relaxation tests complemented the quasi-static experiments to evaluate the deformation and strain-dependent viscoelastic properties. Each experiment was conducted on specimens of the complete stomach wall and two separate layers, mucosa and muscularis, from each of the three gastric regions, i.e., fundus, body, and antrum. The significant preconditioning effects and the considerable regional and layer-specific differences in the tissue response were analyzed. Furthermore, the mechanical experiments were complemented with histology to examine the influence of the microstructural composition on the macrostructural mechanical response and vice versa. Importantly, the shear tests showed lower stresses in the complete wall compared to the single layers which the loose network of submucosal collagen might explain. Also, the stratum arrangement of the muscularis might explain mechanical anisotropy during tensile tests. This study shows that gastric tissue is characterized by a highly heterogeneous microstructure with regional variations in layer composition reflecting not only functional differences but also diverse mechanical behavior. STATEMENT OF SIGNIFICANCE: Unfortunately, only few experimental data on gastric tissue are available for an adequate material parameter and model estimation. The present study therefore combines layer- and region-specific stomach wall mechanics obtained under multiple loading conditions with histological insights into the heterogeneous microstructure. On the one hand, the extensive data sets of this study expand our understanding of the interplay between gastric mechanics, motility and functionality, which could help to identify and treat associated pathologies. On the other hand, such data sets are of high relevance for the constitutive modeling of stomach tissue, and its application in the field of medical engineering, e.g., in the development of surgical staplers and the improvement of bariatric surgical interventions.


Asunto(s)
Colágeno , Estómago , Porcinos , Animales , Humanos , Estómago/fisiología , Modelos Animales , Colágeno/química , Anisotropía , Pruebas Mecánicas , Fenómenos Biomecánicos , Estrés Mecánico
8.
IEEE Trans Biomed Eng ; 71(4): 1289-1297, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37971910

RESUMEN

OBJECTIVE: Bioelectrical 'slow waves' regulate gastrointestinal contractions. We aimed to confirm whether the pyloric sphincter demarcates slow waves in the intact stomach and duodenum. METHODS: We developed and validated novel anatomically-specific electrode cradles and analysis techniques which enable high-resolution slow wave mapping across the in vivo gastroduodenal junction. Cradles housed flexible-printed-circuit and custom cradle-specific electrode arrays during acute porcine experiments (N = 9; 44.92 kg ± 8.49 kg) and maintained electrode contact with the gastroduodenal serosa. Simultaneous gastric and duodenal slow waves were filtered independently after determining suitable organ-specific filters. Validated algorithms calculated slow wave propagation patterns and quantitative descriptions. RESULTS: Butterworth filters, with cut-off frequencies (0.0167 - 2) Hz and (0.167 - 3.33) Hz, were optimal filters for gastric and intestinal slow wave signals, respectively. Antral slow waves had a frequency of (2.76 ± 0.37) cpm, velocity of (4.83 ± 0.21) mm·s-1, and amplitude of (1.13 ± 0.24) mV, before terminating at the quiescent pylorus that was (46.54 ± 5.73) mm wide. Duodenal slow waves had a frequency of (18.13 ± 0.56) cpm, velocity of (11.66 ± 1.36) mm·s-1, amplitude of (0.32 ± 0.03) mV, and originated from a pacemaker region (7.24 ± 4.70) mm distal to the quiescent zone. CONCLUSION: Novel engineering methods enable measurement of in vivo electrical activity across the gastroduodenal junction and provide qualitative and quantitative definitions of slow wave activity. SIGNIFICANCE: The pylorus is a clinical target for a range of gastrointestinal motility disorders and this work may inform diagnostic and treatment practices.


Asunto(s)
Motilidad Gastrointestinal , Marcapaso Artificial , Animales , Porcinos , Motilidad Gastrointestinal/fisiología , Estómago/fisiología , Electrodos , Procesamiento de Señales Asistido por Computador
9.
J Neural Eng ; 20(6)2024 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-38100816

RESUMEN

Objective.Neural regulation of gastric motility occurs partly through the regulation of gastric bioelectrical slow waves (SWs) and phasic contractions. The interaction of the tissues and organs involved in this regulatory process is complex. We sought to infer the relative importance of cellular mechanisms in inhibitory neural regulation of the stomach by enteric neurons and the interaction of inhibitory and excitatory electrical field stimulation.Approach.A novel mathematical model of gastric motility regulation by enteric neurons was developed and scenarios were simulated to determine the mechanisms through which enteric neural influence is exerted. This model was coupled to revised and extended electrophysiological models of gastric SWs and smooth muscle cells (SMCs).Main results.The mathematical model predicted that regulation of contractile apparatus sensitivity to intracellular calcium in the SMC was the major inhibition mechanism of active tension development, and that the effect on SW amplitude depended on the inhibition of non-specific cation currents more than the inhibition of calcium-activated chloride current (kiNSCC= 0.77 vs kiAno1= 0.33). The model predicted that the interaction between inhibitory and excitatory neural regulation, when applied with simultaneous and equal intensity, resulted in an inhibition of contraction amplitude almost equivalent to that of inhibitory stimulation (79% vs 77% decrease), while the effect on frequency was overall excitatory, though less than excitatory stimulation alone (66% vs 47% increase).Significance.The mathematical model predicts the effects of inhibitory and excitatory enteric neural stimulation on gastric motility function, as well as the effects when inhibitory and excitatory enteric neural stimulation interact. Incorporation of the model into organ-level simulations will provide insights regarding pathological mechanisms that underpin gastric functional disorders, and allow forin silicotesting of the effects of clinical neuromodulation protocols for the treatment of these disorders.


Asunto(s)
Calcio , Estómago , Estómago/fisiología , Miocitos del Músculo Liso , Neuronas , Contracción Muscular/fisiología
10.
Artículo en Inglés | MEDLINE | ID: mdl-38082999

RESUMEN

Gastric rhythmic contractions are regulated by bioelectrical events known as slow waves (SW). Abnormal SW activity is associated with gastric motility disorders. Gastric pacing is a potential treatment method to restore rhythmic SW activity. However, to date, the efficacy of gastric pacing is inconsistent and the underlying mechanisms of gastric pacing are poorly understood. Optical mapping is widely used in cardiac electrophysiology studies. Its immunity to pacing artifacts offers a distinct advantage over conventional electrical mapping for studying pacing. In the present study, we first found that optical mapping can image pacing-induced virtual electrode polarization patterns in the stomach (adjacent regions of depolarized and hyperpolarized tissue). Second, we found that elicited SWs usually (15 of 16) originated from the depolarized areas of the stimulated region (virtual cathodes). To our knowledge, this is the first direct observation of virtual electrode polarization patterns in the stomach. Conclusions: Optical mapping can image virtual electrode polarization patterns during gastric pacing with high spatial resolution.Clinical Relevance- Gastric pacing is a potential therapeutic method for gastric motility disorders. This study provides direct observation of virtual electrode polarization pattern during gastric pacing and improves our understanding of the mechanisms underlying gastric pacing..


Asunto(s)
Marcapaso Artificial , Estómago , Estómago/diagnóstico por imagen , Estómago/fisiología , Electrodos
11.
Artículo en Inglés | MEDLINE | ID: mdl-38082764

RESUMEN

Cervical vagus nerve stimulation (cVNS) is a promising neuromodulation therapy for treating symptoms of disease in peripheral organs. The rat is a common animal model for studying and trialing new applications of cVNS therapy, but the stomach and its activity in rats is less well characterized than other animals, such as pigs. We sought to investigate the effects of acute, in vivo cVNS on gastric bioelectrical activity as an intermediate step to computational modeling of the effects of cVNS on gastric smooth muscle electromechanical coupling. Here we show a method of detecting bioelectrical gastric slow wave events using a non-linear energy operator. The marked events are compared to the underlying bioelectrical slow wave activity.The mean propagation velocity before stimulation was 0.79 ± 0.31 mm s-1, and the mean interval was 17.4 ± 1.4 s. During cVNS, there was a significant increase in velocity (1.02 ± 0.69 mm s-1; p < 0.001), and decrease in interval (15.4 ± 2.9 s; p = 0.0196). At stimulation onset, premature slow waves were induced at an ectopic pacemaker location and waves originating at the ectopic and initial pacemaker sites continued to collide following the cessation of cVNS.This work forms the basis for more thorough investigation of the effects of cVNS on gastric bioelectrical slow wave activity and consequential smooth muscle contractions in rats. A better understanding of the effects of cVNS on gastric function will allow the refinement of cVNS therapy to target the stomach, and avoid off-target effects on the stomach.Clinical relevance- This work presents a signal processing and analysis approach for the investigation of cervical vagus nerve stimulation on gastric bioelectrical activity in rats. Vagus nerve stimulation may enable the control and amelioration of hunger, gastric emptying, or functional gastric disorders.


Asunto(s)
Marcapaso Artificial , Estimulación del Nervio Vago , Ratas , Animales , Porcinos , Estómago/fisiología
12.
Nature ; 624(7990): 130-137, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37993711

RESUMEN

The termination of a meal is controlled by dedicated neural circuits in the caudal brainstem. A key challenge is to understand how these circuits transform the sensory signals generated during feeding into dynamic control of behaviour. The caudal nucleus of the solitary tract (cNTS) is the first site in the brain where many meal-related signals are sensed and integrated1-4, but how the cNTS processes ingestive feedback during behaviour is unknown. Here we describe how prolactin-releasing hormone (PRLH) and GCG neurons, two principal cNTS cell types that promote non-aversive satiety, are regulated during ingestion. PRLH neurons showed sustained activation by visceral feedback when nutrients were infused into the stomach, but these sustained responses were substantially reduced during oral consumption. Instead, PRLH neurons shifted to a phasic activity pattern that was time-locked to ingestion and linked to the taste of food. Optogenetic manipulations revealed that PRLH neurons control the duration of seconds-timescale feeding bursts, revealing a mechanism by which orosensory signals feed back to restrain the pace of ingestion. By contrast, GCG neurons were activated by mechanical feedback from the gut, tracked the amount of food consumed and promoted satiety that lasted for tens of minutes. These findings reveal that sequential negative feedback signals from the mouth and gut engage distinct circuits in the caudal brainstem, which in turn control elements of feeding behaviour operating on short and long timescales.


Asunto(s)
Regulación del Apetito , Tronco Encefálico , Ingestión de Alimentos , Retroalimentación Fisiológica , Alimentos , Saciedad , Estómago , Regulación del Apetito/fisiología , Tronco Encefálico/citología , Tronco Encefálico/fisiología , Ingestión de Alimentos/fisiología , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Neuronas/metabolismo , Hormona Liberadora de Prolactina/metabolismo , Saciedad/fisiología , Núcleo Solitario/citología , Núcleo Solitario/fisiología , Estómago/fisiología , Gusto/fisiología , Factores de Tiempo , Animales , Ratones
13.
Physiol Behav ; 272: 114374, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37806511

RESUMEN

Several reports have demonstrated that depressive disorder is related to somatic symptoms including gastrointestinal or genitourinary alterations. The pathophysiological mechanisms underlying the gastrointestinal or genitourinary alterations associated with the depression are still not fully understood. Therefore, this study aimed to evaluate the motor activity of gastrointestinal (fundus of stomach and duodenum) and genitourinary tract (bladder) in a stress-based animal model of depression. Adult male mice were submitted to uncontrollable and unpredictable stress (learned helplessness model), controllable stress and non-stressful situations (control). Then, animals were euthanized and the fundus of stomach, duodenum segments or whole bladder were isolated and mounted in a standard organ bath preparation. We evaluated the contractile effects induced by KCl 80 mM for 5 min or carbachol (acetylcholine receptor agonist). The relaxant effects of isoproterenol (ß-adrenoceptor agonist) were also checked. Animals submitted to the learned helplessness model developed a helpless (depressive-like behavior) or resilient (does not exhibit depressive-like behavior) phenotype. The contractions induced by carbachol were diminished in fundus of stomach isolated from helpless and resilient animals. The isoproterenol-induced fundus of stomach relaxation was reduced in resilient but not helpless mice. The contractions/relaxation of duodenum segments isolated from helpless or resilient animals were not altered. Both helpless and resilient animals showed an increase in the bladder contractions induced by carbachol while the relaxant effects of isoproterenol were reduced when compared to control. Conversely, mice underwent a controllable stress situation did not exhibit alterations in the fundus of stomach or duodenum contraction/relaxation induced by pharmacological agents although a decrease in the bladder contraction induced by carbachol was found. In conclusion, incontrollable and unpredictable stress and not depressive phenotype (helpless animals) or controllable stress could be related to the alterations in motor activity of the fundus of stomach and bladder.


Asunto(s)
Depresión , Vejiga Urinaria , Ratones , Masculino , Animales , Carbacol/farmacología , Isoproterenol/farmacología , Estómago/fisiología , Contracción Muscular/fisiología , Duodeno
14.
Mol Pharm ; 20(11): 5416-5428, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37878746

RESUMEN

The TIM-1 gastrointestinal model is one of the most advanced in vitro systems currently available for biorelevant dissolution testing. This technology, the initial version of which was developed nearly 30 years ago and has been subject to a number of significant updates over this period, simulates the dynamic environment of the human gastrointestinal tract, including pH, transfer times, secretion of bile, enzymes, and electrolytes. In the pharmaceutical industry, the TIM-1 system is used to support drug product design and provide a biopredictive assessment of drug product performance. Typically, the bioaccessibility data sets generated by TIM-1 experiments are used to qualitatively compare formulation performance, and the use of bioaccessibility data as inputs for physiologically based pharmacokinetic (PBPK) modeling for quantitative predictions is limited. To expand the utility of the TIM-1 model beyond standard bioaccessibility measurements (which define the fraction available for absorption), we have developed a computational tool, TIM-1 Data Explorer, to describe the fluid and mass balance within the TIM-1 system. The use of this tool allows a detailed inspection and in-depth interpretation of the experimental data. In addition to mass balance calculation, this model also can be used to describe the critical processes a drug substance would undergo during a TIM-1 experiment, such as dissolution, precipitation on transfer from the stomach to duodenum, and redissolution. The TIM-1 Data Explorer was validated in two case studies. In the first case study with paracetamol, we have shown the ability of the simulator to adequately describe mass transfer events within the TIM-1 system, and in the second study with a weakly basic in-house compound, PF-07059013, the TIM-1 Data Explorer was successfully used to describe dissolution and precipitation processes.


Asunto(s)
Tracto Gastrointestinal , Estómago , Humanos , Simulación por Computador , Duodeno , Absorción Intestinal/fisiología , Modelos Biológicos , Estómago/fisiología
15.
J Physiol ; 601(21): 4751-4766, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37772988

RESUMEN

A monosynaptic pathway connects the substantia nigra pars compacta (SNpc) to neurons of the dorsal motor nucleus of the vagus (DMV). This monosynaptic pathway modulates the vagal control of gastric motility. It is not known, however, whether this nigro-vagal pathway also modulates the tone and motility of the proximal colon. In rats, microinjection of retrograde tracers in the proximal colon and of anterograde tracers in SNpc showed that bilaterally labelled colonic-projecting neurons in the DMV received inputs from SNpc neurons. Microinjections of the ionotropic glutamate receptor agonist, NMDA, in the SNpc increased proximal colonic motility and tone, as measured via a strain gauge aligned with the colonic circular smooth muscle; the motility increase was inhibited by acute subdiaphragmatic vagotomy. Upon transfection of SNpc with pAAV-hSyn-hM3D(Gq)-mCherry, chemogenetic activation of nigro-vagal nerve terminals by brainstem application of clozapine-N-oxide increased the firing rate of DMV neurons and proximal colon motility; both responses were abolished by brainstem pretreatment with the dopaminergic D1-like antagonist SCH23390. Chemogenetic inhibition of nigro-vagal nerve terminals following SNpc transfection with pAAV-hSyn-hM4D(Gi)-mCherry decreased the firing rate of DMV neurons and inhibited proximal colon motility. These data suggest that a nigro-vagal pathway modulates activity of the proximal colon motility tonically via a discrete dopaminergic synapse in a manner dependent on vagal efferent nerve activity. Impairment of this nigro-vagal pathway may contribute to the severely reduced colonic transit and prominent constipation observed in both patients and animal models of parkinsonism. KEY POINTS: Substantia nigra pars compacta (SNpc) neurons are connected to the dorsal motor nucleus of the vagus (DMV) neurons via a presumed direct pathway. Brainstem neurons in the lateral DMV innervate the proximal colon. Colonic-projecting DMV neurons receive inputs from neurons of the SNpc. The nigro-vagal pathway modulates tone and motility of the proximal colon via D1-like receptors in the DMV. The present study provides the mechanistic basis for explaining how SNpc alterations may lead to a high rate of constipation in patients with Parkinson's Disease.


Asunto(s)
Estómago , Sustancia Negra , Humanos , Ratas , Animales , Estómago/fisiología , Ratas Sprague-Dawley , Sustancia Negra/metabolismo , Nervio Vago/fisiología , Motilidad Gastrointestinal/fisiología , Colon , Estreñimiento/metabolismo
16.
Neurogastroenterol Motil ; 35(11): e14674, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37702071

RESUMEN

BACKGROUND: Coping with the ingested food, the gastric regions of fundus, corpus, and antrum display different motility patterns. Intrinsic components of such patterns involving mechanosensitive enteric neurons (MEN) have been described in the guinea pig gastric corpus but are poorly understood in the fundus and antrum. METHODS: To elucidate mechanosensitive properties of myenteric neurons in the gastric fundus and antrum, membrane potential imaging using Di-8-ANEPPS was applied. A small-volume injection led to neuronal compression. We analyzed the number of MEN and their firing frequency in addition to the involvement of selected mechanoreceptors. To characterize the neurochemical phenotype of MEN, we performed immunohistochemistry. KEY RESULTS: In the gastric fundus, 16% of the neurons reproducibly responded to mechanical stimulation and thus were MEN. Of those, 83% were cholinergic and 19% nitrergic. In the antrum, 6% of the neurons responded to the compression stimulus, equally distributed among cholinergic and nitrergic MEN. Defunctionalizing the sensory extrinsic afferents led to a significant drop in the number of MEN in both regions. CONCLUSION: We provided evidence for MEN in the gastric fundus and antrum and further investigated mechanoreceptors. However, the proportions of the chemical phenotypes of the MEN differed significantly between both regions. Further investigations of synaptic connections of MEN are crucial to understand the hardwired neuronal circuits in the stomach.


Asunto(s)
Fundus Gástrico , Neuronas , Cobayas , Humanos , Animales , Neuronas/fisiología , Estómago/fisiología , Intestino Delgado , Colinérgicos , Antro Pilórico
17.
Comput Biol Med ; 165: 107384, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37633085

RESUMEN

Gastric motility is coordinated by bioelectric slow waves (SWs) and dysrhythmic SW activity has been linked with motility disorders. Magnetogastrography (MGG) is the non-invasive measurement of the biomagnetic fields generated by SWs. Dysrhythmia identification using MGG is currently challenging because source models are not well developed and the impact of anatomical variation is not well understood. A novel method for the quantitative spatial co-registration of serosal SW potentials, MGG, and geometric models of anatomical structures was developed and performed on two anesthetized pigs to verify feasibility. Electrode arrays were localized using electromagnetic transmitting coils. Coil localization error for the volume where the stomach is normally located under the sensor array was assessed in a benchtop experiment, and mean error was 4.2±2.3mm and 3.6±3.3° for a coil orientation parallel to the sensor array and 6.2±5.7mm and 4.5±7.0° for a perpendicular coil orientation. Stomach geometries were reconstructed by fitting a generic stomach to up to 19 localization coils, and SW activation maps were mapped onto the reconstructed geometries using the registered positions of 128 electrodes. Normal proximal-to-distal and ectopic SW propagation patterns were recorded from the serosa and compared against the simultaneous MGG measurements. Correlations between the center-of-gravity of normalized MGG and the mean position of SW activity on the serosa were 0.36 and 0.85 for the ectopic and normal propagation patterns along the proximal-distal stomach axis, respectively. This study presents the first feasible method for the spatial co-registration of MGG, serosal SW measurements, and subject-specific anatomy. This is a significant advancement because these data enable the development and validation of novel non-invasive gastric source characterization methods.


Asunto(s)
Motilidad Gastrointestinal , Estómago , Animales , Porcinos , Motilidad Gastrointestinal/fisiología , Estómago/fisiología , Fenómenos Electrofisiológicos/fisiología , Electrodos , Abdomen
18.
AAPS J ; 25(5): 76, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37498389

RESUMEN

The rate and extent of drug dissolution and absorption from a solid oral dosage form depend largely on the fluid volume along the gastrointestinal tract. Hence, a model built upon the gastric fluid volume profiles can help to predict drug dissolution and subsequent absorption. To capture the great inter- and intra-individual variability (IAV) of the gastric fluid volume in fasted human, a stochastic differential equation (SDE)-based mixed effects model was developed and compared with the ordinary differential equation (ODE)-based model. Twelve fasted healthy adult subjects were enrolled and had their gastric fluid volume measured before and after consumption of 240 mL of water at pre-determined intervals for up to 2 hours post ingestion. The SDE- and ODE-based mixed effects models were implemented and compared using extended Kalman filter algorithm via NONMEM. The SDE approach greatly improved the goodness of fit compared with the ODE counterpart. The proportional and additive measurement error of the final SDE model decreased from 14.4 to 4.10% and from 17.6 to 4.74 mL, respectively. The SDE-based mixed effects model successfully characterized the gastric volume profiles in the fasted healthy subjects, and provided a robust approximation of the physiological parameters in the very dynamic system. The remarkable IAV could be further separated into system dynamics terms and measurement error terms in the SDE model instead of only empirically attributing IAV to measurement errors in the traditional ODE method. The system dynamics were best captured by the random fluctuations of gastric emptying coefficient Kge.


Asunto(s)
Tracto Gastrointestinal , Estómago , Humanos , Adulto , Estómago/fisiología , Tracto Gastrointestinal/metabolismo , Ayuno/fisiología , Vaciamiento Gástrico/fisiología , Liberación de Fármacos
19.
J Physiol ; 601(14): 2853-2875, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37154244

RESUMEN

Perinatal high-fat diet (pHFD) exposure alters the development of vagal neurocircuits that control gastrointestinal (GI) motility and reduce stress resiliency in offspring. Descending oxytocin (OXT; prototypical anti-stress peptide) and corticotropin releasing factor (CRF; prototypical stress peptide) inputs from the paraventricular nucleus (PVN) of the hypothalamus to the dorsal motor nucleus of the vagus (DMV) modulate the GI stress response. How these descending inputs, and their associated changes to GI motility and stress responses, are altered following pHFD exposure are, however, unknown. The present study used retrograde neuronal tracing experiments, cerebrospinal fluid extraction, in vivo recordings of gastric tone, motility and gastric emptying rates, and in vitro electrophysiological recordings from brainstem slice preparations to investigate the hypothesis that pHFD alters descending PVN-DMV inputs and dysregulates vagal brain-gut responses to stress. Compared to controls, rats exposed to pHFD had slower gastric emptying rates and did not respond to acute stress with the expected delay in gastric emptying. Neuronal tracing experiments demonstrated that pHFD reduced the number of PVNOXT neurons that project to the DMV, but increased PVNCRF neurons. Both in vitro electrophysiology recordings of DMV neurons and in vivo recordings of gastric motility and tone demonstrated that, following pHFD, PVNCRF -DMV projections were tonically active, and that pharmacological antagonism of brainstem CRF1 receptors restored the appropriate gastric response to brainstem OXT application. These results suggest that pHFD exposure disrupts descending PVN-DMV inputs, leading to a dysregulated vagal brain-gut response to stress. KEY POINTS: Maternal high-fat diet exposure is associated with gastric dysregulation and stress sensitivity in offspring. The present study demonstrates that perinatal high-fat diet exposure downregulates hypothalamic-vagal oxytocin (OXT) inputs but upregulates hypothalamic-vagal corticotropin releasing factor (CRF) inputs. Both in vitro and in vivo studies demonstrated that, following perinatal high-fat diet, CRF receptors were tonically active at NTS-DMV synapses, and that pharmacological antagonism of these receptors restored the appropriate gastric response to OXT. The current study suggests that perinatal high-fat diet exposure disrupts descending PVN-DMV inputs, leading to a dysregulated vagal brain-gut response to stress.


Asunto(s)
Hormona Liberadora de Corticotropina , Oxitocina , Embarazo , Femenino , Ratas , Animales , Ratas Sprague-Dawley , Dieta Alta en Grasa/efectos adversos , Estómago/fisiología , Motilidad Gastrointestinal , Nervio Vago/fisiología
20.
Neuron ; 111(14): 2184-2200.e7, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37192624

RESUMEN

Vagal sensory neurons monitor mechanical and chemical stimuli in the gastrointestinal tract. Major efforts are underway to assign physiological functions to the many distinct subtypes of vagal sensory neurons. Here, we use genetically guided anatomical tracing, optogenetics, and electrophysiology to identify and characterize vagal sensory neuron subtypes expressing Prox2 and Runx3 in mice. We show that three of these neuronal subtypes innervate the esophagus and stomach in regionalized patterns, where they form intraganglionic laminar endings. Electrophysiological analysis revealed that they are low-threshold mechanoreceptors but possess different adaptation properties. Lastly, genetic ablation of Prox2 and Runx3 neurons demonstrated their essential roles for esophageal peristalsis in freely behaving mice. Our work defines the identity and function of the vagal neurons that provide mechanosensory feedback from the esophagus to the brain and could lead to better understanding and treatment of esophageal motility disorders.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal , Esófago , Motilidad Gastrointestinal , Proteínas de Homeodominio , Células Receptoras Sensoriales , Nervio Vago , Animales , Ratones , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Esófago/inervación , Esófago/metabolismo , Esófago/fisiología , Motilidad Gastrointestinal/genética , Motilidad Gastrointestinal/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Mecanorreceptores/fisiología , Neuronas Aferentes/fisiología , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Estómago/inervación , Estómago/metabolismo , Estómago/fisiología , Nervio Vago/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...