Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 388
Filtrar
1.
PLoS One ; 19(5): e0300413, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38739593

RESUMEN

Castration-resistant prostate cancer (CRPC) is associated with resistance to androgen deprivation therapy, and an increase in the population of neuroendocrine (NE) differentiated cells. It is hypothesized that NE differentiated cells secrete neuropeptides that support androgen-independent tumor growth and induce aggressiveness of adjacent proliferating tumor cells through a paracrine mechanism. The cytochrome b561 (CYB561) gene, which codes for a secretory vesicle transmembrane protein, is constitutively expressed in NE cells and highly expressed in CRPC. CYB561 is involved in the α-amidation-dependent activation of neuropeptides, and contributes to regulating iron metabolism which is often dysregulated in cancer. These findings led us to hypothesize that CYB561 may be a key player in the NE differentiation process that drives the progression and maintenance of the highly aggressive NE phenotype in CRPC. In our study, we found that CYB561 expression is upregulated in metastatic and NE prostate cancer (NEPC) tumors and cell lines compared to normal prostate epithelia, and that its expression is independent of androgen regulation. Knockdown of CYB561 in androgen-deprived LNCaP cells dampened NE differentiation potential and transdifferentiation-induced increase in iron levels. In NEPC PC-3 cells, depletion of CYB561 reduced the secretion of growth-promoting factors, lowered intracellular ferrous iron concentration, and mitigated the highly aggressive nature of these cells in complementary assays for cancer hallmarks. These findings demonstrate the role of CYB561 in facilitating transdifferentiation and maintenance of NE phenotype in CRPC through its involvement in neuropeptide biosynthesis and iron metabolism pathways.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Neoplasias de la Próstata Resistentes a la Castración/genética , Línea Celular Tumoral , Fenotipo , Células Neuroendocrinas/metabolismo , Células Neuroendocrinas/patología , Hierro/metabolismo , Diferenciación Celular , Regulación Neoplásica de la Expresión Génica
2.
Pathol Int ; 74(5): 239-251, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38607250

RESUMEN

Pulmonary neuroendocrine (NE) cells are rare airway epithelial cells. The balance between Achaete-scute complex homolog 1 (ASCL1) and hairy and enhancer of split 1, one of the target molecules of the Notch signaling pathway, is crucial for NE differentiation. Small cell lung cancer (SCLC) is a highly aggressive lung tumor, characterized by rapid cell proliferation, a high metastatic potential, and the acquisition of resistance to treatment. The subtypes of SCLC are defined by the expression status of NE cell-lineage transcription factors, such as ASCL1, which roles are supported by SRY-box 2, insulinoma-associated protein 1, NK2 homeobox 1, and wingless-related integration site signaling. This network reinforces NE differentiation and may induce the characteristic morphology and chemosensitivity of SCLC. Notch signaling mediates cell-fate decisions, resulting in an NE to non-NE fate switch. The suppression of NE differentiation may change the histological type of SCLC to a non-SCLC morphology. In SCLC with NE differentiation, Notch signaling is typically inactive and genetically or epigenetically regulated. However, Notch signaling may be activated after chemotherapy, and, in concert with Yes-associated protein signaling and RE1-silencing transcription factor, suppresses NE differentiation, producing intratumor heterogeneity and chemoresistance. Accumulated information on the molecular mechanisms of SCLC will contribute to further advances in the control of this recalcitrant cancer.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Neoplasias Pulmonares , Receptores Notch , Transducción de Señal , Carcinoma Pulmonar de Células Pequeñas , Humanos , Carcinoma Pulmonar de Células Pequeñas/patología , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Receptores Notch/metabolismo , Células Neuroendocrinas/patología , Células Neuroendocrinas/metabolismo
3.
Science ; 384(6693): 295-301, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38669574

RESUMEN

Airway neuroendocrine (NE) cells have been proposed to serve as specialized sensory epithelial cells that modulate respiratory behavior by communicating with nearby nerve endings. However, their functional properties and physiological roles in the healthy lung, trachea, and larynx remain largely unknown. In this work, we show that murine NE cells in these compartments have distinct biophysical properties but share sensitivity to two commonly aspirated noxious stimuli, water and acid. Moreover, we found that tracheal and laryngeal NE cells protect the airways by releasing adenosine 5'-triphosphate (ATP) to activate purinoreceptive sensory neurons that initiate swallowing and expiratory reflexes. Our work uncovers the broad molecular and biophysical diversity of NE cells across the airways and reveals mechanisms by which these specialized excitable cells serve as sentinels for activating protective responses.


Asunto(s)
Adenosina Trifosfato , Laringe , Células Neuroendocrinas , Reflejo , Tráquea , Animales , Ratones , Células Neuroendocrinas/metabolismo , Laringe/fisiología , Adenosina Trifosfato/metabolismo , Reflejo/fisiología , Tráquea/inervación , Tráquea/citología , Deglución , Pulmón/fisiología , Espiración/fisiología , Agua/metabolismo , Células Receptoras Sensoriales/fisiología , Ratones Endogámicos C57BL
4.
Int J Mol Sci ; 24(17)2023 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-37685880

RESUMEN

Gonadotropin-releasing hormone (GnRH) neurons are key neuroendocrine cells in the brain as they control reproduction by regulating hypothalamic-pituitary-gonadal axis function. In this context, anti-Müllerian hormone (AMH), growth hormone (GH), and insulin-like growth factor 1 (IGF1) were shown to improve GnRH neuron migration and function in vitro. Whether AMH, GH, and IGF1 signaling pathways participate in the development and function of GnRH neurons in vivo is, however, currently still unknown. To assess the role of AMH, GH, and IGF1 systems in the development of GnRH neuron, we evaluated the expression of AMH receptors (AMHR2), GH (GHR), and IGF1 (IGF1R) on sections of ex vivo mice at different development stages. The expression of AMHR2, GHR, and IGF1R was assessed by immunofluorescence using established protocols and commercial antibodies. The head sections of mice were analyzed at E12.5, E14.5, and E18.5. In particular, at E12.5, we focused on the neurogenic epithelium of the vomeronasal organ (VNO), where GnRH neurons, migratory mass cells, and the pioneering vomeronasal axon give rise. At E14.5, we focused on the VNO and nasal forebrain junction (NFJ), the two regions where GnRH neurons originate and migrate to the hypothalamus, respectively. At E18.5, the median eminence, which is the hypothalamic area where GnRH is released, was analyzed. At E12.5, double staining for the neuronal marker ß-tubulin III and AMHR2, GHR, or IGF1R revealed a signal in the neurogenic niches of the olfactory and VNO during early embryo development. Furthermore, IGF1R and GHR were expressed by VNO-emerging GnRH neurons. At E14.5, a similar expression pattern was found for the neuronal marker ß-tubulin III, while the expression of IGF1R and GHR began to decline, as also observed at E18.5. Of note, hypothalamic GnRH neurons labeled for PLXND1 tested positive for AMHR2 expression. Ex vivo experiments on mouse sections revealed differential protein expression patterns for AMHR2, GHR, and IGF1R at any time point in development between neurogenic areas and hypothalamic compartments. These findings suggest a differential functional role of related systems in the development of GnRH neurons.


Asunto(s)
Células Neuroendocrinas , Hormonas Peptídicas , Animales , Ratones , Hormona Antimülleriana , Hormona Liberadora de Gonadotropina , Hormona del Crecimiento , Factor I del Crecimiento Similar a la Insulina , Neuronas , Hormonas Liberadoras de Hormona Hipofisaria , Tubulina (Proteína) , Células Neuroendocrinas/metabolismo
5.
Cell Mol Life Sci ; 80(3): 79, 2023 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-36867267

RESUMEN

Pulmonary neuroendocrine (NE) cells represent a small population in the airway epithelium, but despite this, hyperplasia of NE cells is associated with several lung diseases, such as congenital diaphragmatic hernia and bronchopulmonary dysplasia. The molecular mechanisms causing the development of NE cell hyperplasia remains poorly understood. Previously, we showed that the SOX21 modulates the SOX2-initiated differentiation of epithelial cells in the airways. Here, we show that precursor NE cells start to develop in the SOX2 + SOX21 + airway region and that SOX21 suppresses the differentiation of airway progenitors to precursor NE cells. During development, clusters of NE cells start to form and NE cells mature by expressing neuropeptide proteins, such as CGRP. Deficiency in SOX2 resulted in decreased clustering, while deficiency in SOX21 increased both the numbers of NE ASCL1 + precursor cells early in development, and the number of mature cell clusters at E18.5. In addition, at the end of gestation (E18.5), a number of NE cells in Sox2 heterozygous mice, did not yet express CGRP suggesting a delay in maturation. In conclusion, SOX2 and SOX21 function in the initiation, migration and maturation of NE cells.


Asunto(s)
Células Neuroendocrinas , Factores de Transcripción SOXB1 , Factores de Transcripción SOXB2 , Animales , Ratones , Péptido Relacionado con Gen de Calcitonina , Diferenciación Celular/genética , Epitelio , Hiperplasia , Células Neuroendocrinas/citología , Células Neuroendocrinas/metabolismo
6.
Front Endocrinol (Lausanne) ; 14: 1096365, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36742381

RESUMEN

The fusion of the secretory vesicle with the plasma membrane requires the assembly of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein complexes formed by synaptobrevin, syntaxin-1, and SNAP-25. Within the pathway leading to exocytosis, the transitions between the "open" and "closed" conformations of syntaxin-1 function as a switch for the fusion of vesicles with the plasma membranes; rapid assembly and disassembly of syntaxin-1 clusters on the plasma membrane provide docking and fusion sites for secretory vesicles in neuroendocrine cells; and the fully zippered trans-SNARE complex, which requires the orderly, rapid and accurate binding of syntaxin-1 to other SNARE proteins, play key roles in triggering fusion. All of these reactions that affect exocytosis under physiological conditions are tightly regulated by multiple factors. Here, we review the current evidence for the involvement of syntaxin-1 in the mechanism of neuroendocrine cell exocytosis, discuss the roles of multiple factors such as proteins, lipids, protein kinases, drugs, and toxins in SNARE complex-mediated membrane fusion, and present an overview of syntaxin-1 mutation-associated diseases with a view to developing novel mechanistic therapeutic targets for the treatment of neuroendocrine disorders.


Asunto(s)
Células Neuroendocrinas , Sintaxina 1/genética , Proteínas Qa-SNARE/metabolismo , Células Neuroendocrinas/metabolismo , Proteínas R-SNARE , Exocitosis/fisiología
7.
J Pathol ; 260(1): 43-55, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36752189

RESUMEN

Neuroendocrine (NE) cells comprise ~1% of epithelial cells in benign prostate and prostatic adenocarcinoma (PCa). However, they become enriched in hormonally treated and castration-resistant PCa (CRPC). In addition, close to 20% of hormonally treated tumors recur as small cell NE carcinoma (SCNC), composed entirely of NE cells, which may be the result of clonal expansion or lineage plasticity. Since NE cells do not express androgen receptors (ARs), they are resistant to hormonal therapy and contribute to therapy failure. Here, we describe the identification of glypican-3 (GPC3) as an oncofetal cell surface protein specific to NE cells in prostate cancer. Functional studies revealed that GPC3 is critical to the viability of NE tumor cells and tumors displaying NE differentiation and that it regulates calcium homeostasis and signaling. Since our results demonstrate that GPC3 is specifically expressed by NE cells, patients with confirmed SCNC may qualify for GPC3-targeted therapy which has been developed in the context of liver cancer and displays minimal toxicity due to its tumor-specific expression. © 2023 The Pathological Society of Great Britain and Ireland.


Asunto(s)
Adenocarcinoma , Células Neuroendocrinas , Neoplasias de la Próstata , Masculino , Humanos , Células Neuroendocrinas/metabolismo , Células Neuroendocrinas/patología , Glipicanos/metabolismo , Adenocarcinoma/patología , Recurrencia Local de Neoplasia/patología , Neoplasias de la Próstata/patología , Biomarcadores/metabolismo
8.
Brain Behav Evol ; 98(1): 32-43, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35921812

RESUMEN

In Atlantic salmon (Salmo salar), seasonal photoperiod is shown to regulate the onset of sexual maturation, yet which brain region(s) is involved, and how light information impacts the neuroendocrine system are still not fully understood in teleosts. Detailed knowledge about the photoperiodic regulation of maturation in fish is still missing. In birds, it is shown that gonadotropin-releasing hormone (Gnrh) is located in the same neurons as vertebrate ancient (VA) opsin, suggesting a direct photoreceptive regulation for the onset of sexual maturity. This study presents a comprehensive topographic mapping of gnrh2, gnrh3, kisspeptin 2 (kiss2), gonadotropin-inhibiting hormone (gnih), and VA opsin using in situ hybridization on mature Atlantic salmon brains. Neurons positive for gnrh3 are expressed in the olfactory bulb and ventral telencephalon, while gnrh2-positive neurons are located dorsally in the midbrain tegmentum. Gonadotropin-inhibiting hormone (Gnih)-expressing cell bodies are present in the ventral thalamus and extend caudally to the hypothalamus with kiss2-expressing cells appearing in a lateral position. VA opsin-positive cells are present in the telencephalon, the rostro-dorsal ring of the left habenula, the ventral thalamus, and the midbrain tegmentum. The results show no similar co-location as found in birds, hypothesizing that the photoreceptive modulation of Gnrh in salmon may interact through neuronal networks. The topography analyses of the essential neuroendocrine cells related to sexual maturation in the Atlantic salmon brain show that diencephalic (thalamus, hypothalamus) and midbrain (tegmentum) regions seem central for controlling sexual maturation.


Asunto(s)
Células Neuroendocrinas , Salmo salar , Animales , Opsinas/metabolismo , Salmo salar/metabolismo , Células Neuroendocrinas/metabolismo , Maduración Sexual , Hormona Liberadora de Gonadotropina/metabolismo , Encéfalo/metabolismo , Gonadotropinas/metabolismo
9.
Elife ; 112022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36469459

RESUMEN

Pulmonary neuroendocrine cells (PNECs) are sensory epithelial cells that transmit airway status to the brain via sensory neurons and locally via calcitonin gene-related peptide (CGRP) and γ- aminobutyric acid (GABA). Several other neuropeptides and neurotransmitters have been detected in various species, but the number, targets, functions, and conservation of PNEC signals are largely unknown. We used scRNAseq to profile hundreds of the rare mouse and human PNECs. This revealed over 40 PNEC neuropeptide and peptide hormone genes, most cells expressing unique combinations of 5-18 genes. Peptides are packaged in separate vesicles, their release presumably regulated by the distinct, multimodal combinations of sensors we show are expressed by each PNEC. Expression of the peptide receptors predicts an array of local cell targets, and we show the new PNEC signal angiotensin directly activates one subtype of innervating sensory neuron. Many signals lack lung targets so may have endocrine activity like those of PNEC-derived carcinoid tumors. PNECs are an extraordinarily rich and diverse signaling hub rivaling the enteroendocrine system.


Asunto(s)
Pulmón , Células Neuroendocrinas , Neuropéptidos , Animales , Humanos , Ratones , Células Epiteliales/metabolismo , Pulmón/patología , Células Neuroendocrinas/metabolismo , Neuropéptidos/metabolismo , Análisis de Secuencia de ARN
10.
Int J Mol Sci ; 23(21)2022 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-36362289

RESUMEN

Little is known about the adaptor protein FAM159B. Recently, FAM159B was shown to be particularly expressed in neuroendocrine cells and tissues, such as pancreatic islets and neuroendocrine cells of the bronchopulmonary and gastrointestinal tracts, as well as in different types of neuroendocrine tumours. To gain insights into possible interactions of FAM159B with other proteins and/or receptors, we analysed the co-expression of FAM159B and various neuroendocrine-specific markers in the cancer cell lines BON-1, PC-3, NCI-h82, OH-1, and A431 and also in human pancreatic tissues and pancreatic neuroendocrine tumours. The markers included prominent markers of neuroendocrine differentiation, such as chromogranin A (CgA), neuron-specific enolase (NSE), synaptophysin (SYP), insulinoma-associated protein 1 (INSM1), neural cell adhesion molecule 1 (NCAM1), serotonin (5-HT), somatostatin-14/28 (SST), and several receptors that are typically expressed by neuroendocrine cells, such as dopamine receptor 2 (D2R), somatostatin receptor (SSTR) 1, 2, 3, 4 and 5, and regulator of G-protein signalling 9 (RGS9). FAM159B was expressed evenly throughout the cytosol in all five cancer cell lines. Immunocytochemical and immunohistochemical analyses revealed co-expression of FAM159B with SYP, INSM1, RGS9, D2R, SSTR2, SSTR3, SSTR4, and SSTR5 and strong overlapping co-localisation with NSE. Double-labelling and co-immunoprecipitation Western blot analyses confirmed a direct association between FAM159B and NSE. These results suggest the involvement of FAM159B in several intracellular signalling pathways and a direct or indirect influence on diverse membrane proteins and receptors.


Asunto(s)
Células Neuroendocrinas , Tumores Neuroendocrinos , Neoplasias Pancreáticas , Humanos , Células Neuroendocrinas/metabolismo , Biomarcadores de Tumor/metabolismo , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/metabolismo , Cromogranina A/genética , Cromogranina A/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Represoras/metabolismo
11.
Thorac Cancer ; 13(21): 3076-3079, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36134429

RESUMEN

Diffuse idiopathic pulmonary neuroendocrine cell hyperplasia (DIPNECH), a rare condition, is characterized by pathological proliferation of neuroendocrine cells. Some of them are localized to the airway mucosa, and others locally infiltrate to form tumorlets and nodules. Here, we present a patient with lung adenocarcinoma accompanied by DIPNECH, making the latter difficult to distinguish from multiple pulmonary metastases. The patient, a 72-year-old Japanese woman, was diagnosed as having stage IVA lung adenocarcinoma because she had multiple nodules in both lungs. Mutation of epidermal growth factor receptor gene having been found in the primary tumor, treatment with osimertinib was started. This resulted in shrinkage of the primary tumor, but not the multiple pulmonary nodules. To determine whether these lung nodules were indeed lung metastases, we performed right upper lobectomy with lymphadenectomy and wedge resection of the right lower lobe. On pathological examination, the primary tumor was diagnosed as invasive adenocarcinoma, whereas the multiple pulmonary nodules were diagnosed as DIPNECH manifesting as tumorlets. Therefore, the final diagnosis was stage IA1 lung adenocarcinoma accompanied by DINPECH. The patient had no recurrences 1 year after the operation without any additional treatment. This is a rare case of lung adenocarcinoma accompanied by DIPNECH presenting as multiple pulmonary nodules. DIPNECH should be included in the differential diagnosis of multiple pulmonary nodules.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Nódulos Pulmonares Múltiples , Células Neuroendocrinas , Femenino , Humanos , Anciano , Células Neuroendocrinas/metabolismo , Células Neuroendocrinas/patología , Nódulos Pulmonares Múltiples/patología , Hiperplasia , Adenocarcinoma del Pulmón/patología , Neoplasias Pulmonares/patología
12.
Cell Mol Gastroenterol Hepatol ; 14(5): 1025-1051, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35835391

RESUMEN

BACKGROUND & AIMS: Efforts to characterize the signaling mechanisms that underlie gastroenteropancreatic neoplasms (GEP-NENs) are precluded by a lack of comprehensive models that recapitulate pathogenesis. Investigation into a potential cell-of-origin for gastrin-secreting NENs revealed a non-cell autonomous role for loss of menin in neuroendocrine cell specification, resulting in an induction of gastrin in enteric glia. Here, we investigated the hypothesis that cell autonomous Men1 inactivation in glial fibrillary acidic protein (GFAP)-expressing cells induced neuroendocrine differentiation and tumorigenesis. METHODS: Transgenic GFAPΔMen1 mice were generated by conditional GFAP-directed Men1 deletion in GFAP-expressing cells. Cre specificity was confirmed using a tdTomato reporter. GFAPΔMen1 mice were evaluated for GEP-NEN development and neuroendocrine cell hyperplasia. Small interfering RNA-mediated Men1 silencing in a rat enteric glial cell line was performed in parallel. RESULTS: GFAPΔMen1 mice developed pancreatic NENs, in addition to pituitary prolactinomas that phenocopied the human MEN1 syndrome. GFAPΔMen1 mice exhibited gastric neuroendocrine hyperplasia that coincided with a significant loss of GFAP expression. Men1 deletion induced loss of glial-restricted progenitor lineage markers and an increase in neuroendocrine genes, suggesting a reprogramming of GFAP+ cells. Deleting Kif3a, a mediator of Hedgehog signaling, in GFAP-expressing cells attenuated neuroendocrine hyperplasia by restricting the neuroendocrine cell fate. Similar results in the pancreas were observed when Sox10 was used to delete Men1. CONCLUSIONS: GFAP-directed Men1 inactivation exploits glial cell plasticity in favor of neuroendocrine differentiation.


Asunto(s)
Plasticidad de la Célula , Neuroglía , Animales , Ratones , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Plasticidad de la Célula/genética , Plasticidad de la Célula/fisiología , Gastrinas , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Hedgehog , Hiperplasia/patología , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/patología , Células Neuroendocrinas/metabolismo , Células Neuroendocrinas/patología , Células Neuroendocrinas/fisiología , Neuroglía/metabolismo , Proteínas Proto-Oncogénicas , ARN Interferente Pequeño
13.
Microsc Res Tech ; 85(10): 3309-3315, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35729870

RESUMEN

The vagina is part of the genitalia and constitutes part of the birth canal. Sperm is deposited into vagina, and it acts in transport of sperm. Pregnancy is a complex process involving different physiological changes in the body associated with hormonal and metabolic alterations that control maternal and fetal demands. During pregnancy, neuroendocrine cells in rabbit vagina are considered part of the diffuse neuroendocrine system observed throughout the body. Giant neuroendocrine cells in rabbit vagina during pregnancy have not been observed previously. This study detected the presence of giant neuroendocrine cells in the vagina of the pregnant rabbit. The presence of these cells was demonstrated with the use of different histological techniques, including hematoxylin and eosin, PAS, combined Alcian blue-PAS, Crossmon's trichrome, and the Grimelius silver method. Giant neuroendocrine cells were observed in late-pregnancy intraepithelial sites and on the lamina propria. These cells were characterized by vacuolated basophilic cytoplasm with PAS- and PAS-AB-positive granules. Moreover, neuroendocrine cells exhibited an argyrophilic character. Immunohistochemically, neuroendocrine cells in rabbit vagina during pregnancy demonstrated positive immunoreactivity to neuron-specific enolase (NSE) with different intensities, mild immunoreactivity to the vascular endothelial growth factor (VEGF), and negative immunoreactivity to CD68. HIGHLIGHTS: Neuroendocrine cells on the vagina of the female rabbit during pregnancy are considered part of the diffuse neuroendocrine system. Giant neuroendocrine cells on the vagina of the rabbit during pregnancy have not been observed previously. Giant neuroendocrine cells were demonstrated intraepithelial and on the lamina propria. Immunohistochemically, neuroendocrine cells demonstrated positive immunoreactivity to NSE and VEGF, and negative immunoreactivity to CD68.


Asunto(s)
Células Neuroendocrinas , Factor A de Crecimiento Endotelial Vascular , Azul Alcián , Animales , Eosina Amarillenta-(YS) , Femenino , Hematoxilina , Masculino , Células Neuroendocrinas/metabolismo , Sistemas Neurosecretores , Fosfopiruvato Hidratasa/metabolismo , Embarazo , Conejos , Semen , Plata , Vagina
14.
Aging Cell ; 21(7): e13632, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35653631

RESUMEN

Cellular senescence is characterized by a stable proliferation arrest in response to stresses and the acquisition of a senescence-associated secretory phenotype, called SASP, composed of numerous factors including pro-inflammatory molecules, proteases, and growth factors. The SASP affects the environment of senescent cells, especially during aging, by inducing and modulating various phenotypes such as paracrine senescence, immune cell activity, and extracellular matrix deposition and organization, which critically impact various pathophysiological situations, including fibrosis and cancer. Here, we uncover a novel paracrine effect of the SASP: the neuroendocrine transdifferentiation (NED) of some epithelial cancer cells, evidenced both in the breast and prostate. Mechanistically, this effect is mediated by NF-κB-dependent SASP factors, and leads to an increase in intracellular Ca2+ levels. Consistently, buffering Ca2+ by overexpressing the CALB1 buffering protein partly reverts SASP-induced NED, suggesting that the SASP promotes NED through a SASP-induced Ca2+ signaling. Human breast cancer dataset analyses support that NED occurs mainly in p53 WT tumors and in older patients, in line with a role of senescent cells and its secretome, as they are increasing during aging. In conclusion, our work, uncovering SASP-induced NED in some cancer cells, paves the way for future studies aiming at better understanding the functional link between senescent cell accumulation during aging, NED and clinical patient outcome.


Asunto(s)
Neoplasias de la Mama , Transdiferenciación Celular , FN-kappa B , Anciano , Neoplasias de la Mama/metabolismo , Transdiferenciación Celular/fisiología , Senescencia Celular/genética , Senescencia Celular/fisiología , Humanos , Masculino , FN-kappa B/metabolismo , Células Neuroendocrinas/citología , Células Neuroendocrinas/metabolismo , Secretoma
15.
Nat Commun ; 13(1): 2690, 2022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35577801

RESUMEN

The Notch pathway is a conserved cell-cell communication pathway that controls cell fate decisions. Here we sought to determine how Notch pathway activation inhibits the neuroendocrine cell fate in the lungs, an archetypal process for cell fate decisions orchestrated by Notch signaling that has remained poorly understood at the molecular level. Using intratumoral heterogeneity in small-cell lung cancer as a tractable model system, we uncovered a role for the transcriptional regulators REST and YAP as promoters of the neuroendocrine to non-neuroendocrine transition. We further identified the specific neuroendocrine gene programs repressed by REST downstream of Notch in this process. Importantly, we validated the importance of REST and YAP in neuroendocrine to non-neuroendocrine cell fate switches in both developmental and tissue repair processes in the lungs. Altogether, these experiments identify conserved roles for REST and YAP in Notch-driven inhibition of the neuroendocrine cell fate in embryonic lungs, adult lungs, and lung cancer.


Asunto(s)
Neoplasias Pulmonares , Células Neuroendocrinas , Diferenciación Celular/genética , Humanos , Pulmón/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Células Neuroendocrinas/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo
16.
Am J Pathol ; 192(6): 847-861, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35367201

RESUMEN

Although recent reports have revealed the importance of the inactivation of both RB1 and TP53 in the transformation from lung adenocarcinoma into neuroendocrine carcinoma (NEC), the requirements for complete transformation into NEC have not been elucidated. To investigate alterations in the characteristics associated with the inactivation of RB1/TP53 and define the requirements for transformation into NEC cells, RB1/TP53 double-knockout A549 lung adenocarcinoma cells were established, and additional knockout of REST and transfection of ASCL1 and POU class 3 homeobox transcription factors (TFs) was conducted. More than 60 genes that are abundantly expressed in neural cells and several genes associated with epithelial-to-mesenchymal transition were up-regulated in RB1/TP53 double-knockout A549 cells. Although the expression of chromogranin A and synaptophysin was induced by additional knockout of REST (which mimics the status of most NECs), the expression of another neuroendocrine marker, CD56, and proneural TFs was not induced. However, coexpression of ASCL1 and POU3F4 in RB1/TP53/REST triple-knockout A549 cells induced the expression of not only CD56 but also other proneural TFs (NEUROD1 and insulinoma-associated 1) and induced NEC-like morphology. These findings suggest that the inactivation of RB1 and TP53 induces a state necessary for the transformation of lung adenocarcinoma into NEC and that further inactivation of REST and coexpression of ASCL1 and POU3F4 are the triggers for complete transformation into NEC.


Asunto(s)
Adenocarcinoma del Pulmón , Carcinoma Neuroendocrino , Neoplasias Pulmonares , Células Neuroendocrinas , Carcinoma Pulmonar de Células Pequeñas , Adenocarcinoma del Pulmón/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma Neuroendocrino/genética , Carcinoma Neuroendocrino/metabolismo , Carcinoma Neuroendocrino/patología , Humanos , Recién Nacido , Neoplasias Pulmonares/patología , Células Neuroendocrinas/metabolismo , Factores del Dominio POU/metabolismo , Proteínas de Unión a Retinoblastoma , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética
17.
Zhen Ci Yan Jiu ; 47(4): 305-13, 2022 Apr 25.
Artículo en Chino | MEDLINE | ID: mdl-35486009

RESUMEN

OBJECTIVE: To observe the effect of electroacupuncture (EA) at "Zusanli" (ST36) and "Feishu" (BL13) on the activation and secretion of calcitonin gene-related peptide (CGRP) and 5-hydroxytryptamine (5-HT) of pulmonary neuroendocrine cells (PNECs) and inflammatory response in rats with chronic obstructive pulmonary disease (COPD), so as to explore its underlying mechanisms in treating COPD. METHODS: Male SD rats were randomly divided into normal control, COPD model and EA groups, with 7 rats in each group. The COPD model was established by forced inhale of cigarette smoke for 1 h in a self-made box (1 m×1 m×1 m in volume), twice daily for 12 weeks. EA (4 Hz/20 Hz, 1-3 mA) was applied at bilateral ST36 and BL13 acupoints for 30 min, once a day for 14 consecutive days. The pulmonary function including the forced vital capacity (FVC), forced expiratory volume at 0.1 second (FEV0.1), FEV0.3, FEV0.1/FVC and FEV0.3/FVC was detected using a lung function analyzer for small animals. The lung tissue was sampled for observing histopathological changes by using H.E. staining, for observing expression and distribution of PNECs by Grimelius silver staining, and for detecting the immunoactivity (integrated optical density) of CGRP and 5-HT by using immunohistochemistry. The contents of CGRP, 5-HT, tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and transforming growth factor-ß1 (TGF-ß1) in the bronchoalveolar lavage fluid (BALF) and lung tissue were detected by ELISA, and the correlations between TNF-α and CGRP, IL-1ß and CGRP, TNF-α and 5-HT, and IL-1ß and 5-HT levels were analyzed. The mRNA and protein expression levels of nerve fiber markers of CGRP and purinergic receptor P2X ligand gated ion channel 3 (P2X3) which dominate PNECs in the lung tissue were detected by real-time fluorescence quantitative PCR and Western blot, respectively. RESULTS: Compared with the normal control group, the levels of FVC, FEV0.1, FEV0.3, and the ratios of FEV0.1/FVC and FEV0.3/FVC were significantly decreased (P<0.05, P<0.01), while the immunoactivity of PNECs, CGRP and 5-HT, the contents of CGRP, 5-HT, TNF-α, IL-1ß and TGF-ß1 in the BALF and lung tissue, and the expression levels of CGRP and P2X3 mRNAs and proteins in the lung tissue significantly increased in the COPD model group (P<0.01, P<0.05). Following EA intervention, both the increased and decreased levels of all the indexes mentioned above were reversed (P<0.05, P<0.01) except FEV0.3. H.E. staining showed severe deformed bronchial lumen with thickened wall and alveolar septum, and obvious inflammatory cell infiltration and reduced number of alveolar lumen fusion in the COPD model group, which was mild in the EA group. A positive correlation was found between TNF-α and CGRP, IL-1ß and CGRP, TNF-α and 5-HT,IL-1ß and 5-HT levels in both BALF and lung tissues (P<0.01). CONCLUSION: EA at ST36 and BL13 can improve lung function and reduce inflammatory response in COPD rats, which may be related to its function in inhibiting the activation of PNECs and release of neuroactive substances.


Asunto(s)
Electroacupuntura , Células Neuroendocrinas , Enfermedad Pulmonar Obstructiva Crónica , Animales , Péptido Relacionado con Gen de Calcitonina/genética , Pulmón/metabolismo , Masculino , Células Neuroendocrinas/química , Células Neuroendocrinas/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/terapia , Ratas , Ratas Sprague-Dawley , Serotonina , Factor de Crecimiento Transformador beta1 , Factor de Necrosis Tumoral alfa/metabolismo
18.
Elife ; 112022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35416769

RESUMEN

The mechanism through which the brain senses the metabolic state, enabling an animal to regulate food consumption, and discriminate between nutritional and non-nutritional foods is a fundamental question. Flies choose the sweeter non-nutritive sugar, L-glucose, over the nutritive D-glucose if they are not starved. However, under starvation conditions, they switch their preference to D-glucose, and this occurs independent of peripheral taste neurons. Here, we found that eliminating the TRPγ channel impairs the ability of starved flies to choose D-glucose. This food selection depends on trpγ expression in neurosecretory cells in the brain that express diuretic hormone 44 (DH44). Loss of trpγ increases feeding, alters the physiology of the crop, which is the fly stomach equivalent, and decreases intracellular sugars and glycogen levels. Moreover, survival of starved trpγ flies is reduced. Expression of trpγ in DH44 neurons reverses these deficits. These results highlight roles for TRPγ in coordinating feeding with the metabolic state through expression in DH44 neuroendocrine cells.


Asunto(s)
Proteínas de Drosophila/metabolismo , Células Neuroendocrinas , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Drosophila/fisiología , Drosophila melanogaster/fisiología , Conducta Alimentaria/fisiología , Preferencias Alimentarias , Glucosa/metabolismo , Células Neuroendocrinas/metabolismo , Azúcares/metabolismo
19.
Int J Mol Sci ; 23(3)2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-35163009

RESUMEN

The fusion of membranes is a central part of the physiological processes involving the intracellular transport and maturation of vesicles and the final release of their contents, such as neurotransmitters and hormones, by exocytosis. Traditionally, in this process, proteins, such SNAREs have been considered the essential components of the fusion molecular machinery, while lipids have been seen as merely structural elements. Nevertheless, sphingosine, an intracellular signalling lipid, greatly increases the release of neurotransmitters in neuronal and neuroendocrine cells, affecting the exocytotic fusion mode through the direct interaction with SNAREs. Moreover, recent studies suggest that FTY-720 (Fingolimod), a sphingosine structural analogue used in the treatment of multiple sclerosis, simulates sphingosine in the promotion of exocytosis. Furthermore, this drug also induces the intracellular fusion of organelles such as dense vesicles and mitochondria causing cell death in neuroendocrine cells. Therefore, the effect of sphingosine and synthetic derivatives on the heterologous and homologous fusion of organelles can be considered as a new mechanism of action of sphingolipids influencing important physiological processes, which could underlie therapeutic uses of sphingosine derived lipids in the treatment of neurodegenerative disorders and cancers of neuronal origin such neuroblastoma.


Asunto(s)
Exocitosis/efectos de los fármacos , Células Neuroendocrinas/metabolismo , Esfingosina/metabolismo , Animales , Transporte Biológico , Humanos , Fusión de Membrana , Proteínas SNARE/metabolismo , Esfingosina/farmacología
20.
Molecules ; 27(3)2022 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-35163861

RESUMEN

The aim of this study is to reveal the potential roles of apoptosis markers (Bcl2 and p53), proliferation markers (Ki-67 and CyclD1), and the neuroendocrine marker Chromogranin A as markers for the radioresistance of rectal cancer. Statistically significant differences were found in the expression of p53, Ki-67, and Chromogranin A in groups of patients with and without a favorable prognosis after radiotherapy. The survival analysis revealed that the marker of neuroendocrine differentiation, Chromogranin A, also demonstrated a high prognostic significance, indicating a poor prognosis. Markers of proliferation and apoptosis had no prognostic value for patients who received preoperative radiotherapy. Higher Chromogranin A values were predictors of poor prognosis. The results obtained from studying the Chromogranin A expression suggest that the secretion of biologically active substances by neuroendocrine cells causes an increase in tumor aggressiveness.


Asunto(s)
Adenocarcinoma/patología , Biomarcadores de Tumor/metabolismo , Inmunohistoquímica/métodos , Células Neuroendocrinas/patología , Neoplasias del Recto/patología , Adenocarcinoma/metabolismo , Adenocarcinoma/radioterapia , Adulto , Anciano , Cromogranina A/metabolismo , Ciclina D1/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Antígeno Ki-67/metabolismo , Masculino , Persona de Mediana Edad , Células Neuroendocrinas/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Neoplasias del Recto/metabolismo , Neoplasias del Recto/radioterapia , Tasa de Supervivencia , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA