Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.003
Filtrar
1.
J Endocrinol ; 261(3)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38593829

RESUMEN

Pancreatic alpha cell activity and glucagon secretion lower as glucose levels increase. While part of the decrease is regulated by glucose itself, paracrine signaling by their neighboring beta and delta cells also plays an important role. Somatostatin from delta cells is an important local inhibitor of alpha cells at high glucose. Additionally, urocortin 3 (UCN3) is a hormone that is co-released from beta cells with insulin and acts locally to potentiate somatostatin secretion from delta cells. UCN3 thus inhibits insulin secretion via a negative feedback loop with delta cells, but its role with respect to alpha cells and glucagon secretion is not understood. We hypothesize that the somatostatin-driven glucagon inhibition at high glucose is regulated in part by UCN3 from beta cells. Here, we use a combination of live functional Ca2+ and cAMP imaging as well as direct glucagon secretion measurement, all from alpha cells in intact mouse islets, to determine the contributions of UCN3 to alpha cell behavior. Exogenous UCN3 treatment decreased alpha cell Ca2+ and cAMP levels and inhibited glucagon release. Blocking endogenous UCN3 signaling increased alpha cell Ca2+ by 26.8 ± 7.6%, but this did not result in increased glucagon release at high glucose. Furthermore, constitutive deletion of Ucn3 did not increase Ca2+ activity or glucagon secretion relative to controls. UCN3 is thus capable of inhibiting mouse alpha cells, but, given the subtle effects of endogenous UCN3 signaling on alpha cells, we propose that UCN3-driven somatostatin may serve to regulate local paracrine glucagon levels in the islet instead of inhibiting gross systemic glucagon release.


Asunto(s)
Células Secretoras de Glucagón , Glucagón , Comunicación Paracrina , Urocortinas , Animales , Urocortinas/metabolismo , Urocortinas/genética , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/efectos de los fármacos , Ratones , Glucagón/metabolismo , Glucosa/metabolismo , Calcio/metabolismo , Masculino , Ratones Endogámicos C57BL , AMP Cíclico/metabolismo , Somatostatina/farmacología , Somatostatina/metabolismo
2.
Am J Physiol Endocrinol Metab ; 326(4): E537-E544, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38477876

RESUMEN

There is increasing evidence linking bitter taste receptor (BTR) signaling to gut hormone secretion and glucose homeostasis. However, its effect on islet hormone secretion has been poorly characterized. This study investigated the effect of the bitter substance, denatonium benzoate (DB), on hormone secretion from mouse pancreatic islets and INS-1 832/13 cells. DB (0.5-1 mM) augmented insulin secretion at both 2.8 mM and 16.7 mM glucose. This effect was no longer present at 5 mM DB likely due to the greater levels of cellular apoptosis. DB-stimulated insulin secretion involved closure of the KATP channel, activation of T2R signaling in beta-cells, and intraislet glucagon-like peptide-1 (GLP-1) release. DB also enhanced glucagon and somatostatin secretion, but the underlying mechanism was less clear. Together, this study demonstrates that the bitter substance, DB, is a strong potentiator of islet hormone secretion independent of glucose. This observation highlights the potential for widespread off-target effects associated with the clinical use of bitter-tasting substances.NEW & NOTEWORTHY We show that the bitter substance, denatonium benzoate (DB), stimulates insulin, glucagon, somatostatin, and GLP-1 secretion from pancreatic islets, independent of glucose, and that DB augments insulin release via the KATP channel, bitter taste receptor signaling, and intraislet GLP-1 secretion. Exposure to a high dose of DB (5 mM) induces cellular apoptosis in pancreatic islets. Therefore, clinical use of bitter substances to improve glucose homeostasis may have unintended negative impacts beyond the gut.


Asunto(s)
Islotes Pancreáticos , Compuestos de Amonio Cuaternario , Gusto , Ratones , Animales , Glucagón/farmacología , Insulina/farmacología , Glucosa/farmacología , Péptido 1 Similar al Glucagón/farmacología , Somatostatina/farmacología , Adenosina Trifosfato/farmacología
3.
J Neurosci ; 44(17)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38438258

RESUMEN

Acetylcholine (ACh) is released from basal forebrain cholinergic neurons in response to salient stimuli and engages brain states supporting attention and memory. These high ACh states are associated with theta oscillations, which synchronize neuronal ensembles. Theta oscillations in the basolateral amygdala (BLA) in both humans and rodents have been shown to underlie emotional memory, yet their mechanism remains unclear. Here, using brain slice electrophysiology in male and female mice, we show large ACh stimuli evoke prolonged theta oscillations in BLA local field potentials that depend upon M3 muscarinic receptor activation of cholecystokinin (CCK) interneurons (INs) without the need for external glutamate signaling. Somatostatin (SOM) INs inhibit CCK INs and are themselves inhibited by ACh, providing a functional SOM→CCK IN circuit connection gating BLA theta. Parvalbumin (PV) INs, which can drive BLA oscillations in baseline states, are not involved in the generation of ACh-induced theta, highlighting that ACh induces a cellular switch in the control of BLA oscillatory activity and establishes an internally BLA-driven theta oscillation through CCK INs. Theta activity is more readily evoked in BLA over the cortex or hippocampus, suggesting preferential activation of the BLA during high ACh states. These data reveal a SOM→CCK IN circuit in the BLA that gates internal theta oscillations and suggest a mechanism by which salient stimuli acting through ACh switch the BLA into a network state enabling emotional memory.


Asunto(s)
Acetilcolina , Colecistoquinina , Ratones Endogámicos C57BL , Ritmo Teta , Ritmo Teta/efectos de los fármacos , Ritmo Teta/fisiología , Animales , Masculino , Ratones , Femenino , Acetilcolina/farmacología , Acetilcolina/metabolismo , Colecistoquinina/farmacología , Colecistoquinina/metabolismo , Interneuronas/fisiología , Interneuronas/efectos de los fármacos , Somatostatina/metabolismo , Somatostatina/farmacología , Amígdala del Cerebelo/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Complejo Nuclear Basolateral/fisiología , Complejo Nuclear Basolateral/efectos de los fármacos , Red Nerviosa/fisiología , Red Nerviosa/efectos de los fármacos , Receptor Muscarínico M3/fisiología , Receptor Muscarínico M3/metabolismo , Parvalbúminas/metabolismo
4.
Int J Biol Sci ; 20(2): 801-817, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38169563

RESUMEN

Somatostatin analogues (SSTA) are first-line pharmacological treatment choice for acromegaly, which received satisfying tumor shrinkage and normalization of growth hormone. However, there are still patients unresponsive to SSTA, and the underline mechanism remains unknown. Besides, there is no evidence regarding the role of endoplasmic reticulum stress (ERS) and its transmission in SSTA resistance, which also require investigation. Primary growth hormone adenoma cells and cell lines were treated with SSTA; autophagy double-labeled LC3 (mRFP-GFP) adenovirus transfection, flow cytometry sorting, western blotting, calcium imaging as well as immunofluorescence staining were used to determine ERS and autophagy signal transmission; xenograft and syngeneic tumor in vivo model were exploited to confirm the ERS signal transmission mediated effect. Our results revealed that SSTA induces ERS in pituitary growth hormone (GH) adenoma cells. The ERS signals can be intercellularly transmitted, leading to less responsible to SSTA treatment. Moreover, SSTA stimulates inositol triphosphate (IP3) elevation, mediating ERS intercellular transfer. In addition, connexin 36 tunnels ERS transmission, and its blocker, Quinine, exhibits a synergistic effect with SSTA treating GH adenoma. Our study provided insight into ERS intercellular transmission mediated SSTA resistance, which could be translated into clinical usage to improve SSTA efficiency in GH adenoma treatment.


Asunto(s)
Adenoma , Neoplasias Hipofisarias , Humanos , Somatostatina/farmacología , Somatostatina/uso terapéutico , Hormona del Crecimiento/metabolismo , Hormona del Crecimiento/uso terapéutico , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Proteína delta-6 de Union Comunicante , Adenoma/tratamiento farmacológico , Estrés del Retículo Endoplásmico
5.
Oncologist ; 29(5): e643-e654, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38206830

RESUMEN

BACKGROUND: In advanced neuroendocrine tumors (NET), antiproliferative treatment options beyond somatostatin analogs remain limited. Temozolomide (TMZ) has shown efficacy in NET alone or combined with other drugs. MATERIALS AND METHODS: SONNET (NCT02231762) was an open, multicenter, prospective, phase II study to evaluate lanreotide autogel 120 mg (LAN) plus TMZ in patients with progressive advanced/metastatic grade 1/2 gastroenteropancreatic (GEP) NET or of unknown primary. Patients could be enrolled at first-line or higher therapy line. The primary endpoint was disease control rate ([DCR], rate of stable disease [SD], partial [PR], and complete response [CR]) at 6 months of LAN and TMZ. Patients with nonfunctioning (NF) NET without progression at 6 months were randomized to 6-month LAN maintenance or watch and wait, patients with functioning (F)-NET with clinical benefit (PR, SD) continued on LAN. RESULTS: Fifty-seven patients were recruited. The majority of patients received the study drug at second or higher treatment line and had an NET G2. DCR at 6 months LAN and TMZ was 73.5%. After 6 months of further LAN maintenance, 54.5% of patients with F-NET and 71.4% with NF-NET had SD or PR vs 41.7% with NF-NET on observation only. LAN and TMZ were effective in all subgroups analyzed. At 12 months of follow-up, median progression-free survival was 11.1 months. Median serum chromogranin A decreased except in NF-NET on observation. O6-methylguanine DNA methyltransferase promoter methylation appeared to better reflect TMZ response than loss of gene expression. During combination therapy, the most frequent treatment-emergent adverse events grade 3/4 reported were nausea (14%), thrombocytopenia (12.3%), and neutropenia (8.8%). Four deaths were reported resulting from severe adverse events not considered related to study medication. CONCLUSIONS: LAN plus TMZ is a treatment option for patients with progressive GEP-NET with more aggressive biological profile showing a manageable safety profile.


Asunto(s)
Neoplasias Intestinales , Tumores Neuroendocrinos , Neoplasias Pancreáticas , Péptidos Cíclicos , Somatostatina , Somatostatina/análogos & derivados , Temozolomida , Humanos , Temozolomida/farmacología , Temozolomida/uso terapéutico , Temozolomida/administración & dosificación , Somatostatina/uso terapéutico , Somatostatina/farmacología , Somatostatina/administración & dosificación , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/patología , Masculino , Femenino , Persona de Mediana Edad , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Anciano , Péptidos Cíclicos/uso terapéutico , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/administración & dosificación , Neoplasias Intestinales/tratamiento farmacológico , Neoplasias Intestinales/patología , Adulto , Estudios Prospectivos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Anciano de 80 o más Años
6.
Eur J Clin Invest ; 54(4): e14147, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38071418

RESUMEN

BACKGROUND: Polycystic liver disease (PLD) is a common extrarenal manifestation of autosomal dominant polycystic kidney disease (ADPKD). Bile acids may play a role in PLD pathogenesis. We performed a post-hoc exploratory analysis of bile acids in ADPKD patients, who had participated in a trial on the effect of a somatostatin analogue. Our hypothesis was that serum bile acid levels increase in PLD, and that lanreotide, which reduces liver growth, may also reduce bile acid levels. Furthermore, in PLD, urinary excretion of bile acids might contribute to renal disease. METHODS: With liquid chromatography-mass spectrometry, 11 bile acids in serum and 6 in urine were quantified in 105 PLD ADPKD patients and 52 age-, sex-, mutation- and eGFR-matched non-PLD ADPKD patients. Sampling was done at baseline and after 120 weeks of either lanreotide or standard care. RESULTS: Baseline serum levels of taurine- and glycine-conjugated bile acids were higher in patients with larger livers. In PLD patients, multiple bile acids decreased upon treatment with lanreotide but remained stable in untreated subjects. Changes over time did not correlate with changes in liver volume. Urine bile acid levels did not change and did not correlate with renal disease progression. CONCLUSION: In ADPKD patients with PLD, baseline serum bile acids were associated with liver volume. Lanreotide reduced bile acid levels and has previously been shown to reduce liver volume. However, in this study, the decrease in bile acids was not associated with the change in liver volume.


Asunto(s)
Quistes , Hepatopatías , Péptidos Cíclicos , Riñón Poliquístico Autosómico Dominante , Somatostatina/análogos & derivados , Humanos , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/complicaciones , Riñón Poliquístico Autosómico Dominante/patología , Hígado/patología , Hepatopatías/tratamiento farmacológico , Hepatopatías/complicaciones , Somatostatina/uso terapéutico , Somatostatina/farmacología , Ácidos y Sales Biliares
7.
Dev Neurobiol ; 84(1): 3-17, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38072668

RESUMEN

Transient expression of somatostatin (SST) has been observed in the olfactory epithelium (OE) and nerves of chick embryos. Intense expression of SST in these regions on embryonic days (E) 5-8 coincides with the migration of neurons producing gonadotropin-releasing hormone (GnRH) from the OE to the forebrain (FB), suggesting that SST plays a role in the development of GnRH neurons. Using in ovo electroporation of small interfering RNA, we found that the suppression of SST mRNA in the olfactory placode (OP) of E3.5 chick embryos significantly reduced the number of GnRH and Islet-1-immunoreactive neurons in the nasal region without affecting the entry of GnRH neurons into the FB at E5.5-6. SST knockdown did not lead to changes in the number of apoptotic, proliferating, or HuC/D-positive neuronal cells in the OE; therefore, it is possible that SST is involved in the neurogenesis/differentiation of GnRH neurons and OP-derived GnRH-negative migratory neurons. In whole OP explant cultures, we also found that SST or its analog octreotide treatment significantly increased the number of migratory GnRH neurons and the migratory distance from the explants. The co-application of an SST antagonist blocked the octreotide-induced increase in the number of GnRH neurons. Furthermore, the fasciculation of polysialylated neural cell adhesion molecule-immunoreactive fibers emerging from the explants was dependent on octreotide. Taken together, our results provide evidence that SST exerts facilitatory effects on the development of neurons expressing GnRH or Islet-1 and on GnRH neuronal migration, in addition to olfactory-related fiber fasciculation.


Asunto(s)
Hormona Liberadora de Gonadotropina , Octreótido , Animales , Embrión de Pollo , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Octreótido/metabolismo , Octreótido/farmacología , Fasciculación/metabolismo , Neuronas/fisiología , Somatostatina/farmacología , Somatostatina/metabolismo , Movimiento Celular/fisiología
8.
Neuropeptides ; 103: 102399, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38118293

RESUMEN

OBJECTIVE: To determine whether somatostatin (SST) could be a cortico-brainstem neurotransmitter involved in producing the headache of migraine. BACKGROUND: There is evidence to support the idea that a cortico-brainstem-trigeminal nucleus neuraxis might be responsible for producing migraine headache; we have suggested that SST may be one of the neurotransmitters involved. METHODS: Rats were anesthetised and prepared for recording neurons in either the periaqueductal gray matter (PAG) or nucleus raphe magnus (NRM), as well as the trigeminal nucleus caudalis (TNC). The dura mater and facial skin were stimulated electrically or mechanically. SST, the SST agonist L054264 and the SST antagonist CYN54806 were injected intravenously, by microinjection, or by iontophoresis into the PAG or NRM. Cortical neuronal activity was provoked by cortical spreading depression (CSD) or light flash (LF) and was monitored by recording cortical blood flow (CBF). RESULTS: Intravenous injection of SST: (a) selectively decreased the responses of TNC neurons to stimulation of the dura, but not skin, for up to 5 h; (b) decreased the ongoing discharge rate of TNC neurons while simultaneously increasing the discharge rate of neurons in either brainstem nucleus and; (c) prevented, or reversed, the effect of CSD and LF on brainstem and trigeminal neuron discharge rates. CSD and LF decreased the discharge rate of neurons in both brainstem nuclei and increased the discharge rate of TNC neurons. These effects were reversed by L054264 and mimicked by CYN54806. Injections of L054264 into the PAG or NRM reduced the response of TNC neurons to dural stimulation and skin stimulation differentially, depending on the nucleus injected. Injections of CYN54806 into either brainstem nucleus potentiated the responses of TNC neurons to dural and skin stimulation, but without a marked differential effect. CONCLUSIONS: These results imply that SST could be a neurotransmitter in a pathway responsible for migraine pain.


Asunto(s)
Trastornos Migrañosos , Ratas , Animales , Dolor , Cefalea , Somatostatina/farmacología , Neurotransmisores
9.
Discov Med ; 35(179): 1086-1092, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38058074

RESUMEN

BACKGROUND: Pancreatic cancer (PC), a commonly recognized malignancy, arises within the digestive tract. Somatostatin (SOM) is a regulatory peptide that acts on secretion in vivo. Several studies have shown that SOM has inhibitory effects on various cancers. This work aims to probe the inhibitory effect, and mechanism of SOM action, on the epithelial-mesenchymal transition (EMT) of PC cells. METHODS: First, the effects of SOM and transforming growth factor-ß (TGF-ß) on the proliferation of PC cells was determined by Cell Counting Kit-8 (CCK-8) assay. Next, we assessed the impact of SOM and TGF-ß on the metastasis and apoptosis of PC cells using transwell assays and flow cytometry. Finally, we evaluated the effects of SOM and TGF-ß on the expression of EMT-related proteins, apoptosis-related proteins, and proteins related to the TGF-ß/Smad signaling pathway in PC cells using western blot analysis. RESULTS: SOM suppressed the growth and metastasis of PC cells, and facilitated their apoptosis (p < 0.05). Moreover, SOM reversed pro-apoptotic effects of TGF-ß (p < 0.05). Specifically, SOM increased the expression of Cysteine-aspartic acid protease 3 (Caspase-3) and Bcl-2-associated X protein (Bax) proteins while reducing the expression of B-cell lymphoma 2 (Bcl-2) protein (p < 0.05). SOM also reversed the TGF-ß-induced EMT process. The TGF-ß1, Smad2, and Smad3 proteins in PC cells treated with SOM were significantly down-regulated (p < 0.05). CONCLUSIONS: SOM suppressed the EMT progression in PC cells through its regulation of the TGF-ß/Smad signaling pathway.


Asunto(s)
Neoplasias Pancreáticas , Transducción de Señal , Humanos , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Factor de Crecimiento Transformador beta , Transición Epitelial-Mesenquimal , Línea Celular Tumoral , Neoplasias Pancreáticas/tratamiento farmacológico , Movimiento Celular , Somatostatina/farmacología
10.
Neuropharmacology ; 238: 109638, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37482180

RESUMEN

The prefrontal cortex (PFC) regulates drinking behaviors and affective changes following chronic alcohol use. PFC activity is dynamically modulated by local inhibitory interneurons (INs), which can be divided into non-overlapping groups with distinct functional roles. Within deeper layers of neocortex, INs that express either parvalbumin or somatostatin directly inhibit pyramidal cells. By contrast, the plurality of all remaining INs express vasoactive intestinal peptide (VIP), reside within superficial layers, and preferentially target other types of INs. While recent studies have described adaptations to PFC parvalbumin-INs and somatostatin-INs in alcohol use models, whether ethanol or drinking affect the physiology of PFC VIP-INs has not been reported. To address this gap, we used genetically engineered female and male mice to target VIP-INs in layers 1-3 of prelimbic PFC for whole-cell patch-clamp electrophysiology. We found that ethanol (20 mM, ∼0.09 BEC/90 mg/dL) application to PFC brain slices enhances VIP-IN excitability. We next examined effects following chronic drinking by providing mice with 4 weeks of intermittent access (IA) ethanol two-bottle choice in the home cage. In these studies, VIP-INs from female and male IA ethanol mice displayed reduced excitability relative to cells from water-only controls. Finally, we assessed whether these effects continue into abstinence. After 7-13 days without ethanol, the hypo-excitability of VIP-INs from male IA ethanol mice persisted, whereas cells from female IA ethanol mice were not different from their controls. Together, these findings illustrate that acute ethanol enhances VIP-IN excitability and suggest these cells undergo pronounced homeostatic changes following long-term drinking.


Asunto(s)
Neocórtex , Péptido Intestinal Vasoactivo , Ratones , Masculino , Femenino , Animales , Péptido Intestinal Vasoactivo/farmacología , Péptido Intestinal Vasoactivo/metabolismo , Parvalbúminas , Potenciales de Acción , Interneuronas/fisiología , Etanol/farmacología , Corteza Prefrontal , Neocórtex/metabolismo , Somatostatina/farmacología , Somatostatina/metabolismo
11.
No Shinkei Geka ; 51(4): 706-715, 2023 Jul.
Artículo en Japonés | MEDLINE | ID: mdl-37491068

RESUMEN

Transsphenoidal surgery is the first-line treatment for most functioning pituitary neuroendocrine tumors(PitNETs). Medical therapies are usually chosen for patients with residual or refractory tumors after surgery or contraindications to surgery. Dopamine agonists(DA)are the first-line treatment for prolactinomas. Somatostatin analogs are the first line of therapy for GH- and TSH-producing PitNETs. In severe hypercortisolemia due to ACTH-producing PitNETs, adrenal enzyme inhibitors such as 11ß-hydroxylase inhibitors should be started immediately, as marked hypercortisolemia leads to serious opportunistic infections. Pasireotide and DA are usually administered to treat mild hypercortisolemia. Based on the histological pattern of secretory granules, somatotroph, lactotroph, and corticotroph tumors can be divided into two subtypes: densely granulated and sparsely granulated. Densely granulated lactotroph tumors tend to be resistant to DA. In contrast, densely granulated somatotroph and corticotroph tumors express high levels of somatostatin receptors and are more responsive to somatostatin analogs. Since ACTH-producing PitNETs express SSTR5 without SSTR2, the second-generation somatostatin analog, pasireotide, is effective against ACTH-producing PitNETs.


Asunto(s)
Tumores Neuroendocrinos , Neoplasias Hipofisarias , Humanos , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/cirugía , Tumores Neuroendocrinos/patología , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/cirugía , Neoplasias Hipofisarias/patología , Somatostatina/uso terapéutico , Somatostatina/farmacología , Hormona Adrenocorticotrópica
12.
Neuropharmacology ; 237: 109622, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37307896

RESUMEN

Somatostatin (SST) is a neuropeptide widely expressed in the central nervous system with dense expression in limbic regions such as the extended amygdala. It has recently gained attention for playing a role in modulating alcohol use disorders and co-morbid neuropsychiatric disorders. However, the role of SST in the central nucleus of the amygdala (CeA), a key region for neuropeptide regulation of alcohol and anxiety related behaviors, in alcohol consumption has not been assessed. In this work we perform an initial examination of the interaction between the CeA SST system and binge ethanol intake. Binge intake is a dangerous pattern of excessive ethanol consumption associated with health complications and the transition into alcohol dependence. We use the Drinking in the Dark (DID) model of binge intake in C57BL/6J male and female mice to examine: 1) the impact of 3 DID cycles on CeA SST expression; 2) the effect of intra-CeA SST injection on binge-like ethanol consumption; and 3) if the SST receptor 2 or 4 (SST2R or SST4R) mediate any effect on consumption. Our results show binge-like ethanol intake decreases SST expression in the CeA, but not neighboring basolateral amygdala. We further found intra-SST CeA administration reduces binge ethanol intake. This decrease was replicated by the administration of an SST4R agonist. These effects were not sex-dependent. Overall, this work lends further support for SST playing a role in alcohol related behaviors and as a potential therapeutic target.


Asunto(s)
Alcoholismo , Consumo Excesivo de Bebidas Alcohólicas , Núcleo Amigdalino Central , Femenino , Ratones , Masculino , Animales , Núcleo Amigdalino Central/metabolismo , Alcoholismo/metabolismo , Ratones Endogámicos C57BL , Etanol , Consumo de Bebidas Alcohólicas/metabolismo , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Somatostatina/farmacología , Somatostatina/metabolismo
13.
Molecules ; 28(4)2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36838810

RESUMEN

New strategies facilitate the design of cyclic peptides which can penetrate the brain. We have designed a bicyclic peptide, OL-CTOP, composed of the sequences of a selective µ-opioid receptor antagonist, CTOP (f-cyclo(CYwOTX)T) (X = penicillamine, Pen; O = ornithine) and odorranalectin, OL (YASPK-cyclo(CFRYPNGVLAC)T), optimized its solid-phase synthesis and demonstrated its ability for nose-to-brain delivery and in vivo activity. The differences in reactivity of Cys and Pen thiol groups protected with trityl and/or acetamidomethyl protecting groups toward I2 in different solvents were exploited for selective disulfide bond formation on the solid phase. Both the single step and the sequential strategy applied to macrocyclization reactions generated the desired OL-CTOP, with the sequential strategy yielding a large quantity and better purity of crude OL-CTOP. Importantly, intranasally (i.n.s.) administered OL-CTOP dose-dependently antagonized the analgesic effect of morphine administered to mice through the intracerebroventricular route and prevented morphine-induced respiratory depression. In summary, the results demonstrate the feasibility of our solid-phase synthetic strategy for the preparation of the OL-CTOP bicyclic peptide containing two disulfide bonds and reveal the potential of odorranalectin for further modifications and the targeted delivery to the brain.


Asunto(s)
Técnicas de Síntesis en Fase Sólida , Somatostatina , Ratones , Animales , Administración Intranasal , Somatostatina/farmacología , Receptores Opioides mu , Péptidos/farmacología , Morfina/farmacología
14.
J Neurosci ; 43(3): 419-432, 2023 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-36639888

RESUMEN

We tested the hypothesis that dorsal cervical epidural electrical stimulation (CEES) increases respiratory activity in male and female anesthetized rats. Respiratory frequency and minute ventilation were significantly increased when CEES was applied dorsally to the C2-C6 region of the cervical spinal cord. By injecting pseudorabies virus into the diaphragm and using c-Fos activity to identify neurons activated during CEES, we found neurons in the dorsal horn of the cervical spinal cord in which c-Fos and pseudorabies were co-localized, and these neurons expressed somatostatin (SST). Using dual viral infection to express the inhibitory Designer Receptors Exclusively Activated by Designer Drugs (DREADD), hM4D(Gi), selectively in SST-positive cells, we inhibited SST-expressing neurons by administering Clozapine N-oxide (CNO). During CNO-mediated inhibition of SST-expressing cervical spinal neurons, the respiratory excitation elicited by CEES was diminished. Thus, dorsal cervical epidural stimulation activated SST-expressing neurons in the cervical spinal cord, likely interneurons, that communicated with the respiratory pattern generating network to effect changes in ventilation.SIGNIFICANCE STATEMENT A network of pontomedullary neurons within the brainstem generates respiratory behaviors that are susceptible to modulation by a variety of inputs; spinal sensory and motor circuits modulate and adapt this output to meet the demands placed on the respiratory system. We explored dorsal cervical epidural electrical stimulation (CEES) excitation of spinal circuits to increase ventilation in rats. We identified dorsal somatostatin (SST)-expressing neurons in the cervical spinal cord that were activated (c-Fos-positive) by CEES. CEES no longer stimulated ventilation during inhibition of SST-expressing spinal neuronal activity, thereby demonstrating that spinal SST neurons participate in the activation of respiratory circuits affected by CEES. This work establishes a mechanistic foundation to repurpose a clinically accessible neuromodulatory therapy to activate respiratory circuits and stimulate ventilation.


Asunto(s)
Médula Cervical , Neuronas , Frecuencia Respiratoria , Animales , Femenino , Masculino , Ratas , Médula Cervical/fisiología , Estimulación Eléctrica/métodos , Neuronas/fisiología , Proteínas Proto-Oncogénicas c-fos , Somatostatina/metabolismo , Somatostatina/farmacología , Médula Espinal/fisiología , Frecuencia Respiratoria/fisiología
15.
Biochem Pharmacol ; 209: 115419, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36693436

RESUMEN

Since the conventional and adjuvant analgesics have limited effectiveness frequently accompanied by serious side effects, development of novel, potent pain killers for chronic neuropathic and inflammatory pain conditions is a big challenge. Somatostatin (SS) regulates endocrine, vascular, immune and neuronal functions, cell proliferation through 5 Gi protein-coupled receptors (SST1-SST5). SS released from the capsaicin-sensitive peptidergic sensory nerves mediates anti-inflammatory and antinociceptive effects without endocrine actions via SST4. The therapeutic use of the native SS is limited by its diverse biological actions and short plasma elimination half-life. Therefore, SST4 selective SS analogues could be promising analgesic and anti-inflammatory drug candidates with new mode of action. TT-232 is a cyclic heptapeptide showing great affinity to SST4 and SST1. Here, we report the in silico SST4 receptor binding mechanism, in vitro binding (competition assay) and cAMP- decreasing effect of TT-232 in SST4-expressing CHO cells, as well as its analgesic and anti-inflammatory actions in chronic neuropathic pain and arthritis models using wildtype and SST4-deficient mice. TT-232 binds to SST4 with similar interaction energy (-11.03 kcal/mol) to the superagonist J-2156, displaces somatostatin from SST4 binding (10 nM to 30 µM) and inhibits forskolin-stimulated cAMP accumulation (EC50: 371.6 ± 58.03 nmol; Emax: 78.63 ± 2.636 %). Its i.p. injection (100, 200 µg/kg) results in significant, 35.7 % and 50.4 %, analgesic effects upon single administration in chronic neuropathic pain and repeated injection in arthritis models in wildtype, but not in SST4-deficient mice. These results provide evidence that the analgesic effect of TT-232 is mediated by SST4 activation, which might open novel drug developmental potentials. Chemical compounds Chemical compounds studied in this article TT-232 (PubChem CID: 74053735).


Asunto(s)
Artritis , Neuralgia , Cricetinae , Ratones , Animales , Cricetulus , Somatostatina/metabolismo , Somatostatina/farmacología , Receptores de Somatostatina/metabolismo , Analgésicos/uso terapéutico , Antiinflamatorios/farmacología , Neuralgia/tratamiento farmacológico , Artritis/tratamiento farmacológico
16.
Biochem Biophys Res Commun ; 643: 30-38, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36586156

RESUMEN

Breast cancer is the most commonly diagnosed cancer and a leading cause of cancer-related death among women worldwide. Somatostatin (SST) and Cannabinoids have an anti-proliferative and pro-apoptotic effect, but the mechanisms of their actions remain elusive. In the present study, we have evaluated the effects of SST, Cannabidiol (CBD) alone or in combination on receptor expression, cell proliferation and apoptosis and related downstream signalling pathways in MDA-MB-231 and MCF-7 breast cancer cells. The results presented here demonstrate the cell type and agonist-dependent changes in receptor expression at the cell membrane, inhibition of cell proliferation and increased apoptosis following treatment with SST and CBD alone and in combination. In comparison to MDA-MB-231 cells, MCF-7 cells treated with SST alone and in combination with CBD exhibited inhibition of phosphorylated Protein Kinase B (pAKT) and phosphorylated-Phosphoinositide 3-Kinase (pPI3K) expression. Importantly, inhibition of PI3K/AKT activation was accompanied by enhanced PTEN expression in MCF-7 cells. These results highlight the possible interaction between SSTR and CBR subtypes with the implication in the modulation of receptor expression, cell viability and signal transduction pathways in a breast cancer cell type-dependent manner.


Asunto(s)
Neoplasias de la Mama , Cannabidiol , Femenino , Humanos , Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Cannabidiol/farmacología , Línea Celular Tumoral , Proliferación Celular , Células MCF-7 , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Somatostatina/farmacología
17.
Brain ; 146(2): 629-644, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-35867870

RESUMEN

Premature infants with germinal matrix haemorrhage-intraventricular haemorrhage (GMH-IVH) suffer from neurobehavioural deficits as they enter childhood and adolescence. Yet the underlying mechanisms remain unclear. Impaired development and function of interneurons contribute to neuropsychiatric disorders. Therefore, we hypothesized that the occurrence of IVH would reduce interneuron neurogenesis in the medial ganglionic eminence and diminish the population of parvalbumin+ and somatostatin+ cortical interneurons. Because Sonic Hedgehog promotes the production of cortical interneurons, we also postulated that the activation of Sonic Hedgehog signalling might restore neurogenesis, cortical interneuron population, and neurobehavioural function in premature newborns with IVH. These hypotheses were tested in a preterm rabbit model of IVH and autopsy samples from human preterm infants. We compared premature newborns with and without IVH for intraneuronal progenitors, cortical interneurons, transcription factors regulating neurogenesis, single-cell transcriptome of medial ganglionic eminence and neurobehavioural functions. We treated premature rabbit kits with adenovirus expressing Sonic Hedgehog (Ad-Shh) or green fluorescence protein gene to determine the effect of Sonic Hedgehog activation on the interneuron production, cortical interneuron population and neurobehaviour. We discovered that IVH reduced the number of Nkx2.1+ and Dlx2+ progenitors in the medial ganglionic eminence of both humans and rabbits by attenuating their proliferation and inducing apoptosis. Moreover, IVH decreased the population of parvalbumin+ and somatostatin+ neurons in the frontal cortex of both preterm infants and kits relative to controls. Sonic Hedgehog expression and the downstream transcription factors, including Nkx2.1, Mash1, Lhx6 and Sox6, were also reduced in kits with IVH. Consistent with these findings, single-cell transcriptomic analyses of medial ganglionic eminence identified a distinct subpopulation of cells exhibiting perturbation in genes regulating neurogenesis, ciliogenesis, mitochondrial function and MAPK signalling in rabbits with IVH. More importantly, restoration of Sonic Hedgehog level by Ad-Shh treatment ameliorated neurogenesis, cortical interneuron population and neurobehavioural function in kits with IVH. Additionally, Sonic Hedgehog activation alleviated IVH-induced inflammation and several transcriptomic changes in the medial ganglionic eminence. Taken together, IVH reduced intraneuronal production and cortical interneuron population by downregulating Sonic Hedgehog signalling in both preterm rabbits and humans. Notably, activation of Sonic Hedgehog signalling restored interneuron neurogenesis, cortical interneurons and cognitive function in rabbit kits with IVH. These findings highlight disruption in cortical interneurons in IVH and identify a novel therapeutic strategy to restore cortical interneurons and cognitive function in infants with IVH. These studies can accelerate the development of new therapies to enhance the neurodevelopmental outcome of survivors with IVH.


Asunto(s)
Proteínas Hedgehog , Parvalbúminas , Animales , Recién Nacido , Humanos , Conejos , Niño , Proteínas Hedgehog/metabolismo , Parvalbúminas/metabolismo , Parvalbúminas/farmacología , Recien Nacido Prematuro , Factores de Transcripción/genética , Cognición , Hemorragia , Interneuronas/metabolismo , Somatostatina/metabolismo , Somatostatina/farmacología
18.
Comput Math Methods Med ; 2022: 1221190, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36267315

RESUMEN

The present study was designed to observe the effect of quadruple therapy combined with probiotics on Helicobacter pylori-related peptic ulcer. The patients in the control group (n = 90) were given regular quadruple therapy including proton pump inhibitor ilaprazole enteric-coated tablet + two antibiotics amoxicillin dispersible tablet and metronidazole tablet + colloidal bismuth pectin capsule for 2 weeks. Patients in the study group (n = 90) were given abovementioned quadruple therapy combined with probiotics live combined Bifidobacterium, Lactobacillus, and Enterococcus Capsules, oral for 2 weeks. Then Hp clearance rate, recurrence rate, levels of gastrointestinal hormone makers, and advance reactions between two groups were compared. At the 2nd week after the treatment, the Helicobacter pylori clearance rate in the study group (87.79%) was significantly higher than the control group (78.89%), and the total recurrence rate in the study group (6.67%) was significantly lower than the control group (13.33%) (P < 0.05). Serum gastrin and motilin expression were lower, and somatostatin expressions was significantly higher than those in the control group (P < 0.05). There was no significant difference in the total incidence of adverse reactions between the two groups (P > 0.05). In summary, quadruple therapy combined with probiotics in the treatment of Helicobacter pylori-related peptic ulcer can improve the Helicobacter pylori clearance rate, reduce the Helicobacter pylori recurrence rate, and is beneficial to improving the level of gastrointestinal hormones, with certain safety.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Úlcera Péptica , Probióticos , Humanos , Infecciones por Helicobacter/tratamiento farmacológico , Bismuto/farmacología , Bismuto/uso terapéutico , Metronidazol/farmacología , Metronidazol/uso terapéutico , Inhibidores de la Bomba de Protones/farmacología , Gastrinas/farmacología , Gastrinas/uso terapéutico , Motilina/farmacología , Motilina/uso terapéutico , Comprimidos Recubiertos/farmacología , Comprimidos Recubiertos/uso terapéutico , Quimioterapia Combinada , Úlcera Péptica/tratamiento farmacológico , Úlcera Péptica/microbiología , Amoxicilina/uso terapéutico , Amoxicilina/farmacología , Antibacterianos/uso terapéutico , Probióticos/uso terapéutico , Pectinas/farmacología , Pectinas/uso terapéutico , Somatostatina/farmacología , Somatostatina/uso terapéutico
19.
Best Pract Res Clin Endocrinol Metab ; 36(6): 101684, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35931640

RESUMEN

Acromegaly is a chronic disease with an increased mortality in case of persistently active disease. The treatment of acromegaly is mainly based on the surgical resection of the GH secreting pituitary tumor and, in cases with persistent disease, on the medical therapy with first generation somatostatin analogues (first gen-SSAs). Data from national registries, meta-analysis and epidemiology studies showed that 24%-65% of acromegaly patients treated with first gen-SSA did not reach the control of disease, requiring second line therapies, as the second gen-SSAs and the GH receptor antagonist. According to the high efficacy of these treatments and their molecular mechanisms of action, the choice of second line therapies should be personalized. In this review, we summarize the evidence on clinical, molecular and morphological aspects that may predict the response to second line therapies, in order to integrate and translate in the clinical practice for a patient-tailored therapeutic approach.


Asunto(s)
Acromegalia , Adenoma , Hormona de Crecimiento Humana , Neoplasias Hipofisarias , Humanos , Adenoma/cirugía , Acromegalia/tratamiento farmacológico , Somatostatina/uso terapéutico , Somatostatina/farmacología , Hormona de Crecimiento Humana/uso terapéutico , Factor I del Crecimiento Similar a la Insulina , Resultado del Tratamiento
20.
BMC Anesthesiol ; 22(1): 202, 2022 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-35780092

RESUMEN

BACKGROUND: Norepinephrine (NE) is a α1-adrenergic mediated vasopressor and a key player in the treatment of perioperative hypotension. Apart from modulating systemic hemodynamics, NE may also affect regional blood flow, such as the hepatic circulation, which contains a wide variety of adrenergic receptors. It may alter regional vascular tonus and hepatic blood flow (HBF) by reducing portal vein flow (PVF) or hepatic arterial flow (HAF). The aim of this study was to assess the effects of NE on HBF. METHODS: Patients scheduled for pancreaticoduodenectomy were included. All patients received standardized anesthetic care using propofol and remifentanil and were hemodynamically stabilized using a goal-directed hemodynamic strategy guided by Pulsioflex™. On surgical indication, somatostatin (SOMATO) was given to reduce pancreatic secretion. HBF measurements were performed using transit-time ultrasound (Medistim™). Baseline hemodynamic and HBF measurements were made after pancreatectomy, at T1. Afterwards, NE infusion was initiated to increase mean arterial pressure (MAP) by 10 - 20% of baseline MAP (T2) and by 20 - 30% of baseline MAP (T3). HBF and hemodynamic measurements were performed simultaneously at these three time-points. RESULTS: A total of 28 patients were analyzed. Administration of NE significantly increased MAP but had no effect on cardiac index. NE infusion reduced total HBF in all patients (p < 0.01) by a reduction HAF (p < 0.01), while the effect on PVF remained unclear. Post-hoc analysis showed that SOMATO-treated patients had a significant lower PVF at baseline (p < 0.05), which did not change during NE infusion. In these patients, reduction of total HBF was primarily related to a reduction of HAF (p < 0.01). In untreated patients, NE infusion reduced total HBF both by a reduction HAF (p < 0.01) and PVF (p < 0.05). CONCLUSION: Administration of NE reduced total HBF, by decreasing HAF, while the effect on PVF remained unclear. SOMATO-treated patients had a lower PVF at baseline, which remained unaffected during NE infusion. In these patients the decrease in total HBF with NE was entirely related to the decrease in HAF. In SOMATO-untreated patients PVF also significantly decreased with NE. TRIAL REGISTRATION: Study protocol EC: 2019/0395. EudraCT n°: 2018-004,139-66 (25 - 03 - 2019). Clin.trail.gov: NCT03965117 (28 - 05 - 2019).


Asunto(s)
Circulación Hepática , Norepinefrina , Hemodinámica , Humanos , Hígado/irrigación sanguínea , Circulación Hepática/fisiología , Norepinefrina/farmacología , Somatostatina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA