Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
JAMA Netw Open ; 7(6): e2414122, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38857050

RESUMEN

Importance: Neurological manifestations during acute SARS-CoV-2-related multisystem inflammatory syndrome in children (MIS-C) are common in hospitalized patients younger than 18 years and may increase risk of new neurocognitive or functional morbidity. Objective: To assess the association of severe neurological manifestations during a SARS-CoV-2-related hospital admission with new neurocognitive or functional morbidities at discharge. Design, Setting, and Participants: This prospective cohort study from 46 centers in 10 countries included patients younger than 18 years who were hospitalized for acute SARS-CoV-2 or MIS-C between January 2, 2020, and July 31, 2021. Exposure: Severe neurological manifestations, which included acute encephalopathy, seizures or status epilepticus, meningitis or encephalitis, sympathetic storming or dysautonomia, cardiac arrest, coma, delirium, and stroke. Main Outcomes and Measures: The primary outcome was new neurocognitive (based on the Pediatric Cerebral Performance Category scale) and/or functional (based on the Functional Status Scale) morbidity at hospital discharge. Multivariable logistic regression analyses were performed to examine the association of severe neurological manifestations with new morbidity in each SARS-CoV-2-related condition. Results: Overall, 3568 patients younger than 18 years (median age, 8 years [IQR, 1-14 years]; 54.3% male) were included in this study. Most (2980 [83.5%]) had acute SARS-CoV-2; the remainder (588 [16.5%]) had MIS-C. Among the patients with acute SARS-CoV-2, 536 (18.0%) had a severe neurological manifestation during hospitalization, as did 146 patients with MIS-C (24.8%). Among survivors with acute SARS-CoV-2, those with severe neurological manifestations were more likely to have new neurocognitive or functional morbidity at hospital discharge compared with those without severe neurological manifestations (27.7% [n = 142] vs 14.6% [n = 356]; P < .001). For survivors with MIS-C, 28.0% (n = 39) with severe neurological manifestations had new neurocognitive and/or functional morbidity at hospital discharge compared with 15.5% (n = 68) of those without severe neurological manifestations (P = .002). When adjusting for risk factors in those with severe neurological manifestations, both patients with acute SARS-CoV-2 (odds ratio, 1.85 [95% CI, 1.27-2.70]; P = .001) and those with MIS-C (odds ratio, 2.18 [95% CI, 1.22-3.89]; P = .009) had higher odds of having new neurocognitive and/or functional morbidity at hospital discharge. Conclusions and Relevance: The results of this study suggest that children and adolescents with acute SARS-CoV-2 or MIS-C and severe neurological manifestations may be at high risk for long-term impairment and may benefit from screening and early intervention to assist recovery.


Asunto(s)
COVID-19 , Hospitalización , Enfermedades del Sistema Nervioso , SARS-CoV-2 , Síndrome de Respuesta Inflamatoria Sistémica , Humanos , COVID-19/complicaciones , COVID-19/epidemiología , Niño , Femenino , Masculino , Preescolar , Hospitalización/estadística & datos numéricos , Adolescente , Estudios Prospectivos , Síndrome de Respuesta Inflamatoria Sistémica/epidemiología , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/epidemiología , Lactante , Índice de Severidad de la Enfermedad
2.
Neurosci Lett ; 743: 135567, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33352286

RESUMEN

Coronavirus disease 2019 (COVID-19) usually leads to a mild infectious disease course in children, but serious complications may occur in conjunction with both acute infection and associated phenomena such as the multisystem inflammatory syndrome in children (MIS-C). Neurological symptoms, which have been predominantly reported in adults, range from mild headache to seizure, peripheral neuropathy, stroke, demyelinating disorders, and encephalopathy. Similar to respiratory and cardiac manifestations of COVID-19, neurological complications present differently based on age and underlying comorbidities. This review provides a concise overview of the neurological conditions seen in the context of COVID-19, as well as potential mechanisms and long-term implications of COVID-19 in the pediatric population from literature reviews and primary data collected at NewYork-Presbyterian Morgan Stanley Children's Hospital.


Asunto(s)
COVID-19/complicaciones , Enfermedades del Sistema Nervioso/virología , Niño , Preescolar , Femenino , Humanos , Masculino , SARS-CoV-2
4.
Cancer Res ; 77(18): 5095-5106, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28916678

RESUMEN

High doses of ionizing radiation induce acute damage to epithelial cells of the gastrointestinal (GI) tract, mediating toxicities restricting the therapeutic efficacy of radiation in cancer and morbidity and mortality in nuclear disasters. No approved prophylaxis or therapy exists for these toxicities, in part reflecting an incomplete understanding of mechanisms contributing to the acute radiation-induced GI syndrome (RIGS). Guanylate cyclase C (GUCY2C) and its hormones guanylin and uroguanylin have recently emerged as one paracrine axis defending intestinal mucosal integrity against mutational, chemical, and inflammatory injury. Here, we reveal a role for the GUCY2C paracrine axis in compensatory mechanisms opposing RIGS. Eliminating GUCY2C signaling exacerbated RIGS, amplifying radiation-induced mortality, weight loss, mucosal bleeding, debilitation, and intestinal dysfunction. Durable expression of GUCY2C, guanylin, and uroguanylin mRNA and protein by intestinal epithelial cells was preserved following lethal irradiation inducing RIGS. Oral delivery of the heat-stable enterotoxin (ST), an exogenous GUCY2C ligand, opposed RIGS, a process requiring p53 activation mediated by dissociation from MDM2. In turn, p53 activation prevented cell death by selectively limiting mitotic catastrophe, but not apoptosis. These studies reveal a role for the GUCY2C paracrine hormone axis as a novel compensatory mechanism opposing RIGS, and they highlight the potential of oral GUCY2C agonists (Linzess; Trulance) to prevent and treat RIGS in cancer therapy and nuclear disasters. Cancer Res; 77(18); 5095-106. ©2017 AACR.


Asunto(s)
Rayos gamma/efectos adversos , Tracto Gastrointestinal/efectos de la radiación , Síndrome del Colon Irritable/prevención & control , Traumatismos Experimentales por Radiación/prevención & control , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Receptores de Péptidos/metabolismo , Animales , Apoptosis/efectos de la radiación , Proliferación Celular/efectos de la radiación , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Neoplasias del Colon/radioterapia , Femenino , Hormonas Gastrointestinales/metabolismo , Humanos , Síndrome del Colon Irritable/enzimología , Síndrome del Colon Irritable/etiología , Linfoma/enzimología , Linfoma/patología , Linfoma/radioterapia , Masculino , Melanoma Experimental/enzimología , Melanoma Experimental/patología , Melanoma Experimental/radioterapia , Ratones , Ratones Endogámicos C57BL , Péptidos Natriuréticos/metabolismo , Comunicación Paracrina/efectos de la radiación , Traumatismos Experimentales por Radiación/enzimología , Traumatismos Experimentales por Radiación/etiología , Receptores de Enterotoxina , Transducción de Señal/efectos de la radiación , Células Tumorales Cultivadas
5.
Toxins (Basel) ; 9(9)2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28895923

RESUMEN

There is a geographic inequality in the incidence of colorectal cancer, lowest in developing countries, and greatest in developed countries. This disparity suggests an environmental contribution to cancer resistance in endemic populations. Enterotoxigenic bacteria associated with diarrheal disease are prevalent in developing countries, including enterotoxigenic E. coli (ETEC) producing heat-stable enterotoxins (STs). STs are peptides that are structurally homologous to paracrine hormones that regulate the intestinal guanylyl cyclase C (GUCY2C) receptor. Beyond secretion, GUCY2C is a tumor suppressor universally silenced by loss of expression of its paracrine hormone during carcinogenesis. Thus, the geographic imbalance in colorectal cancer, in part, may reflect chronic exposure to ST-producing organisms that restore GUCY2C signaling silenced by hormone loss during transformation. Here, mice colonized for 18 weeks with control E. coli or those engineered to secrete ST exhibited normal growth, with comparable weight gain and normal stool water content, without evidence of secretory diarrhea. Enterotoxin-producing, but not control, E. coli, generated ST that activated colonic GUCY2C signaling, cyclic guanosine monophosphate (cGMP) production, and cGMP-dependent protein phosphorylation in colonized mice. Moreover, mice colonized with ST-producing E. coli exhibited a 50% reduction in carcinogen-induced colorectal tumor burden. Thus, chronic colonization with ETEC producing ST could contribute to endemic cancer resistance in developing countries, reinforcing a novel paradigm of colorectal cancer chemoprevention with oral GUCY2C-targeted agents.


Asunto(s)
Neoplasias Colorrectales , Enterotoxinas , Escherichia coli/metabolismo , Animales , Vacunas contra el Cáncer , Neoplasias Colorrectales/epidemiología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/prevención & control , Países en Desarrollo , Humanos , Ratones , Receptores de Enterotoxina/metabolismo , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo
6.
Cancer Prev Res (Phila) ; 10(6): 345-354, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28396341

RESUMEN

Guanylate cyclase C (GUCY2C) is a tumor-suppressing receptor silenced by loss of expression of its luminocrine hormones guanylin and uroguanylin early in colorectal carcinogenesis. This observation suggests oral replacement with a GUCY2C agonist may be an effective targeted chemoprevention agent. Linaclotide is an FDA-approved oral GUCY2C agonist formulated for gastric release, inducing fluid secretion into the small bowel to treat chronic idiopathic constipation. The ability of oral linaclotide to induce a pharmacodynamic response in epithelial cells of the colorectum in humans remains undefined. Here, we demonstrate that administration of 0.87 mg of oral linaclotide daily for 7 days to healthy volunteers, after oral colon preparation with polyethylene glycol solution (MoviPrep), activates GUCY2C, resulting in accumulation of its product cyclic (c)GMP in epithelial cells of the cecum, transverse colon, and distal rectum. GUCY2C activation by oral linaclotide was associated with homeostatic signaling, including phosphorylation of vasodilator-stimulated phosphoprotein and inhibition of proliferation quantified by reduced Ki67-positive epithelial cells. In the absence of the complete oral colonoscopy preparation, linaclotide did not alter cGMP production in epithelial cells of the colorectum, demonstrating that there was an effect related to the laxative preparation. These data show that the current FDA-approved formulation of oral linaclotide developed for small-bowel delivery to treat chronic idiopathic constipation is inadequate for reliably regulating GUCY2C in the colorectum to prevent tumorigenesis. The study results highlight the importance of developing a novel GUCY2C agonist formulated for release and activity targeted to the large intestine for colorectal cancer prevention. Cancer Prev Res; 10(6); 345-54. ©2017 AACR.


Asunto(s)
Colon/efectos de los fármacos , Neoplasias Colorrectales/prevención & control , Agonistas de la Guanilato Ciclasa C/farmacología , Péptidos/farmacología , Receptores de Enterotoxina/metabolismo , Recto/efectos de los fármacos , Administración Oral , Animales , Moléculas de Adhesión Celular/metabolismo , Colon/diagnóstico por imagen , Colonoscopía , GMP Cíclico/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Hormonas Gastrointestinales/metabolismo , Agonistas de la Guanilato Ciclasa C/uso terapéutico , Voluntarios Sanos , Humanos , Antígeno Ki-67/metabolismo , Proteínas de Microfilamentos/metabolismo , Péptidos Natriuréticos/metabolismo , Péptidos/uso terapéutico , Fosfoproteínas/metabolismo , Fosforilación , Polietilenglicoles/farmacología , Recto/diagnóstico por imagen
7.
Cancer Res ; 76(2): 339-46, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26773096

RESUMEN

Obesity is a well-known risk factor for colorectal cancer but precisely how it influences risks of malignancy remains unclear. During colon cancer development in humans or animals, attenuation of the colonic cell surface receptor guanylyl cyclase C (GUCY2C) that occurs due to loss of its paracrine hormone ligand guanylin contributes universally to malignant progression. In this study, we explored a link between obesity and GUCY2C silencing in colorectal cancer. Using genetically engineered mice on different diets, we found that diet-induced obesity caused a loss of guanylin expression in the colon with subsequent GUCY2C silencing, epithelial dysfunction, and tumorigenesis. Mechanistic investigations revealed that obesity reversibly silenced guanylin expression through calorie-dependent induction of endoplasmic reticulum stress and the unfolded protein response in intestinal epithelial cells. In transgenic mice, enforcing specific expression of guanylin in intestinal epithelial cells restored GUCY2C signaling, eliminating intestinal tumors associated with a high calorie diet. Our findings show how caloric suppression of the guanylin-GUCY2C signaling axis links obesity to negation of a universal tumor suppressor pathway in colorectal cancer, suggesting an opportunity to prevent colorectal cancer in obese patients through hormone replacement with the FDA-approved oral GUCY2C ligand linaclotide.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Hormonas Gastrointestinales/metabolismo , Péptidos Natriuréticos/metabolismo , Obesidad/metabolismo , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Receptores de Péptidos/metabolismo , Animales , Células CACO-2 , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Genotipo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/patología , Comunicación Paracrina , Receptores de Enterotoxina , Factores de Riesgo , Transducción de Señal
8.
Cancer Epidemiol Biomarkers Prev ; 23(11): 2328-37, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25304930

RESUMEN

BACKGROUND: Although colorectal cancer is a disease characterized by sequential accumulation of mutations in epithelial cells, mechanisms leading to genomic vulnerability contributing to tumor initiation remain undefined. GUCY2C has emerged as an intestine-specific tumor suppressor controlling epithelial homeostasis through circuits canonically disrupted in cancer. Surprisingly, the GUCY2C tumor suppressor is universally overexpressed by human colorectal cancer cells. This apparent paradox likely reflects silencing of GUCY2C through loss of its paracrine hormone guanylin. Here, we quantified expression of guanylin mRNA and protein in tumors and normal epithelia from patients with colorectal cancer. METHODS: Guanylin mRNA was quantified in tumors and normal adjacent epithelia from 281 patients by the reverse transcriptase-polymerase chain reaction. Separately, the guanylin protein was quantified by immunohistochemistry in 54 colorectal tumors and 30 specimens of normal intestinal epithelium. RESULTS: Guanylin mRNA in colorectum varied more than a 100-fold across the population. Guanylin mRNA was reduced 100- to 1,000-fold in >85% of tumors compared with matched normal adjacent mucosa (P < 0.001). Loss of guanylin mRNA was greatest in tumors from patients <50 years old (P < 0.005) and with the highest expression in normal adjacent mucosa (Spearman correlation coefficient = 0.61; P < 0.001). In a separate validation cohort, guanylin protein was detected in all 30 normal colorectal mucosa specimens, but in none of 54 colorectal tumors. CONCLUSIONS: Colorectal cancer may initiate as a disease of paracrine hormone insufficiency through loss of guanylin expression, silencing the GUCY2C tumor suppressor and disrupting homeostatic mechanisms regulating colorectal epithelia cells. IMPACT: Intestinal tumorigenesis may be prevented by oral GUCY2C hormone replacement therapy.


Asunto(s)
Neoplasias Colorrectales/química , Hormonas Gastrointestinales/análisis , Hormonas Gastrointestinales/genética , Mucosa Intestinal/química , Péptidos Natriuréticos/análisis , Péptidos Natriuréticos/genética , ARN Mensajero/análisis , Factores de Edad , Anciano , Colon/química , Neoplasias Colorrectales/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Comunicación Paracrina , Estudios Prospectivos , Receptores de Enterotoxina , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Receptores de Péptidos/metabolismo , Recto/química
9.
PLoS One ; 9(8): e104293, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25101906

RESUMEN

Gene expression is a dynamic and coordinated process coupling transcription with pre-mRNA processing. This regulation enables tissue-specific transcription factors to induce expression of specific transcripts that are subsequently amplified by alternative splicing allowing for increased proteome complexity and functional diversity. The intestine-specific transcription factor CDX2 regulates development and maintenance of the intestinal epithelium by inducing expression of genes characteristic of the mature enterocyte phenotype. Here, sequence analysis of CDX2 mRNA from colonic mucosa-derived tissues revealed an alternatively spliced transcript (CDX2/AS) that encodes a protein with a truncated homeodomain and a novel carboxy-terminal domain enriched in serine and arginine residues (RS domain). CDX2 and CDX2/AS exhibited distinct nuclear expression patterns with minimal areas of co-localization. CDX2/AS did not activate the CDX2-dependent promoter of guanylyl cyclase C nor inhibit transcriptional activity of CDX2. Unlike CDX2, CDX2/AS co-localized with the putative splicing factors ASF/SF2 and SC35. CDX2/AS altered splicing patterns of CD44v5 and Tra2-ß1 minigenes in Lovo colon cancer cells independent of CDX2 expression. These data demonstrate unique dual functions of the CDX2 gene enabling it to regulate gene expression through both transcription (CDX2) and pre-mRNA processing (CDX2/AS).


Asunto(s)
Empalme Alternativo/fisiología , Colon/metabolismo , Enterocitos/metabolismo , Proteínas de Homeodominio/biosíntesis , Animales , Factor de Transcripción CDX2 , Línea Celular Tumoral , Colon/citología , Enterocitos/citología , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Receptores de Hialuranos/biosíntesis , Receptores de Hialuranos/genética , Proteína Cofactora de Membrana/biosíntesis , Proteína Cofactora de Membrana/genética , Ratones , Ratones Endogámicos NOD , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo
10.
Cancer Res ; 73(22): 6654-66, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24085786

RESUMEN

Tumorigenesis is a multistep process that reflects intimate reciprocal interactions between epithelia and underlying stroma. However, tumor-initiating mechanisms coordinating transformation of both epithelial and stromal components are not defined. In humans and mice, initiation of colorectal cancer is universally associated with loss of guanylin and uroguanylin, the endogenous ligands for the tumor suppressor guanylyl cyclase C (GUCY2C), disrupting a network of homeostatic mechanisms along the crypt-surface axis. Here, we reveal that silencing GUCY2C in human colon cancer cells increases Akt-dependent TGF-ß secretion, activating fibroblasts through TGF-ß type I receptors and Smad3 phosphorylation. In turn, activating TGF-ß signaling induces fibroblasts to secrete hepatocyte growth factor (HGF), reciprocally driving colon cancer cell proliferation through cMET-dependent signaling. Elimination of GUCY2C signaling in mice (Gucy2c(-/-)) produces intestinal desmoplasia, with increased reactive myofibroblasts, which is suppressed by anti-TGF-ß antibodies or genetic silencing of Akt. Thus, GUCY2C coordinates intestinal epithelial-mesenchymal homeostasis through reciprocal paracrine circuits mediated by TGF-ß and HGF. In that context, GUCY2C signaling constitutes a direct link between the initiation of colorectal cancer and the induction of its associated desmoplastic stromal niche. The recent regulatory approval of oral GUCY2C ligands to treat chronic gastrointestinal disorders underscores the potential therapeutic opportunity for oral GUCY2C hormone replacement to prevent remodeling of the microenvironment essential for colorectal tumorigenesis.


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/patología , Intestinos/patología , Receptores Acoplados a la Guanilato-Ciclasa/fisiología , Receptores de Péptidos/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Células CACO-2 , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Regulación hacia Abajo , Transición Epitelial-Mesenquimal/genética , Fibrosis , Células HCT116 , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Enterotoxina , Nicho de Células Madre/genética
11.
Expert Rev Clin Pharmacol ; 6(5): 557-64, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23971873

RESUMEN

Colorectal cancer (CRC) is a major public health concern, ranking among the leading causes of cancer death in both men and women. Because of this continued burden there is a clear need for improved treatment, and more importantly prevention of this disease. In recent years there is significant evidence to support the hypothesis that guanylyl cyclase C (GCY2C) is a tumor suppressor in the intestine, and that the loss of hormone ligands for this receptor is an important step in the disease process. Thus, ligand replacement therapy has been proposed as a strategy to prevent CRC. Until recently this strategy was not clinically plausible; however, the recent regulatory approval of linaclotide (LINZESS™, Forest Laboratories and Ironwood Pharmaceuticals, Inc.), an oral GUCY2C ligand, has raised the possibility of utilizing this strategy clinically to prevent CRC.


Asunto(s)
Anticarcinógenos/uso terapéutico , Neoplasias Colorrectales/prevención & control , Péptidos/uso terapéutico , Receptores Acoplados a la Guanilato-Ciclasa/agonistas , Receptores de Péptidos/agonistas , Animales , Anticarcinógenos/administración & dosificación , Anticarcinógenos/farmacología , Neoplasias Colorrectales/enzimología , Descubrimiento de Drogas , Humanos , Ligandos , Péptidos/administración & dosificación , Péptidos/farmacología , Receptores de Enterotoxina
12.
Cancer Res ; 73(14): 4548-58, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23576566

RESUMEN

Hepatic metastasis is responsible for the majority of colorectal cancer (CRC)-related mortalities. Although Gankyrin (PSMD10) has been implicated in cancer metastasis, its exact role and underlying mechanisms of CRC hepatic metastasis remain largely unknown. Herein, we showed that the expression of Gankyrin was higher in primary CRC with hepatic metastasis compared with CRC without metastasis. RNAi-mediated silencing of Gankyrin expression in highly metastatic human CRC cells impaired their migratory and metastatic capacity in vivo. Genome-wide transcriptome profiling revealed activation of the interleukin (IL)-8 signaling pathway by Gankyrin. Protein levels of IL-8 and cyclin D1 (CCND1), the two important molecules in the IL-8 pathway, were positively correlated with Gankyrin expression in human CRC specimens. Furthermore, genetic deletion of cyclin D1 showed its requirement in Gankyrin-mediated cell migration. Finally, administration of recombinant IL-8 rescued the migratory defect of CRC cells where Gankyrin expression was silenced. Together, our findings highlight the importance of Gankyrin in hepatic metastasis of CRC and point out its candidature as a potential prognostic marker and therapeutic target to improve the clinical management of metastatic CRC.


Asunto(s)
Neoplasias Colorrectales/patología , Interleucina-8/metabolismo , Neoplasias Hepáticas/secundario , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Células 3T3 , Animales , Línea Celular , Movimiento Celular/fisiología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Células HEK293 , Humanos , Interleucina-8/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Metástasis de la Neoplasia , Complejo de la Endopetidasa Proteasomal/genética , Proteínas Proto-Oncogénicas/genética , Transducción de Señal
13.
Expert Opin Drug Discov ; 8(6): 655-71, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23621300

RESUMEN

INTRODUCTION: Obesity is a worldwide pandemic. Obesity-related health and economic costs are staggering. Existing strategies to combat obesity through lifestyle improvements and medical intervention have had limited success. Pharmacotherapy, in combination with lifestyle modification, may play a vital role in reversing the disease burden. However, past and current weight-loss medications have had serious safety risks, notably cardiovascular and psychiatric events. AREAS COVERED: The authors review the strategies for designing new anti-obesity drugs by describing those currently in development. They describe their target, mechanism of action and developmental or regulatory status. Furthermore, they discuss the problem of weight regain following weight loss, and its relevance to the long-term success of anti-obesity pharmacotherapy. EXPERT OPINION: For weight management drugs to achieve the safety and efficacy required to be impactful, current studies are uncovering and characterizing new targets, including new signaling circuits and hormones regulating appetite and metabolism, and re-evaluating the role of pharmacotherapy in weight management. To avoid the safety failures of many past weight-loss drugs, the models and strategies covered in this article incorporate recent advances in knowledge and technology. We discuss the emergence of cGMP signaling as a potentially transformative target in weight management. Modulating cGMP signaling may represent an ideal goal for an anti-obesity pharmacotherapy, reflecting some of the major themes described in the present review: targeting pathways that are newly realized as relevant for weight management; promoting safety by re-purposing drugs that are safe, proven, and approved for clinical use; and having a synergistic effect on multiple, reinforcing pathways.


Asunto(s)
Fármacos Antiobesidad/farmacología , Depresores del Apetito/farmacología , GMP Cíclico/metabolismo , Obesidad/tratamiento farmacológico , Aumento de Peso/efectos de los fármacos , Pérdida de Peso/efectos de los fármacos , Fármacos Antiobesidad/uso terapéutico , Depresores del Apetito/uso terapéutico , Diseño de Fármacos , Humanos , Estilo de Vida , Modelos Teóricos
14.
Trends Endocrinol Metab ; 24(4): 165-73, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23375388

RESUMEN

Guanylyl cyclase C (GUCY2C) has canonical centrality in defense of key intestinal homeostatic mechanisms, encompassing fluid and electrolyte balance, epithelial dynamics, antitumorigenesis, and intestinal barrier function. Recent discoveries expand the homeostatic role of GUCY2C to reveal a novel gut-brain endocrine axis regulating appetite, anchored by hypothalamic GUCY2C which is responsive to intestine-derived uroguanylin. Thus, GUCY2C may represent a new target for anti-obesity pharmacotherapy. Moreover, the coincident regulation of energy balance and tumor suppression by a single hormone receptor system suggests that the GUCY2C axis might contribute to the established relationship between obesity and colorectal cancer. This confluence suggests that hormone supplementation to reconstitute GUCY2C signaling may be an elegant strategy to reverse both pathophysiologic processes.


Asunto(s)
Neoplasias/metabolismo , Obesidad/metabolismo , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Receptores de Péptidos/metabolismo , Humanos , Neoplasias/genética , Obesidad/genética , Receptores de Enterotoxina , Receptores Acoplados a la Guanilato-Ciclasa/genética , Receptores de Péptidos/genética
15.
Mol Aspects Med ; 34(1): 71-83, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23103610

RESUMEN

The increase in obesity in the Unites States and around the world in the last decade is overwhelming. The number of overweight adults in the world surpassed 1 billion in 2008. Health hazards associated with obesity are serious and include heart disease, sleep apnea, diabetes, and cancer. Although lifestyle modifications are the most straightforward way to control weight, a large portion of the population may not be able to rely on this modality alone. Thus, the development of anti-obesity therapeutics represents a major unmet medical need. Historically, anti-obesity pharmacotherapies have been unsafe and minimally efficacious. A better understanding of the biology of appetite and metabolism provides an opportunity to develop drugs that may offer safer and more effective alternatives for weight management. This review discusses drugs that are currently on the market and in development as anti-obesity therapeutics based on their target and mechanism of action. It should serve as a roadmap to establish expectations for the near future for anti-obesity drug development.


Asunto(s)
Fármacos Antiobesidad/farmacología , Obesidad/tratamiento farmacológico , Apetito/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Humanos , Estilo de Vida , Lipasa/antagonistas & inhibidores , Lipasa/metabolismo , Neuropéptidos/metabolismo , Hormonas Pancreáticas/metabolismo , Transmisión Sináptica/efectos de los fármacos , Pérdida de Peso/efectos de los fármacos
16.
J Clin Invest ; 121(9): 3578-88, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21865642

RESUMEN

Intestinal enteroendocrine cells are critical to central regulation of caloric consumption, since they activate hypothalamic circuits that decrease appetite and thereby restrict meal size by secreting hormones in response to nutrients in the gut. Although guanylyl cyclase and downstream cGMP are essential regulators of centrally regulated feeding behavior in invertebrates, the role of this primordial signaling mechanism in mammalian appetite regulation has eluded definition. In intestinal epithelial cells, guanylyl cyclase 2C (GUCY2C) is a transmembrane receptor that makes cGMP in response to the paracrine hormones guanylin and uroguanylin, which regulate epithelial cell dynamics along the crypt-villus axis. Here, we show that silencing of GUCY2C in mice disrupts satiation, resulting in hyperphagia and subsequent obesity and metabolic syndrome. This defined an appetite-regulating uroguanylin-GUCY2C endocrine axis, which we confirmed by showing that nutrient intake induces intestinal prouroguanylin secretion into the circulation. The prohormone signal is selectively decoded in the hypothalamus by proteolytic liberation of uroguanylin, inducing GUCY2C signaling and consequent activation of downstream anorexigenic pathways. Thus, evolutionary diversification of primitive guanylyl cyclase signaling pathways allows GUCY2C to coordinate endocrine regulation of central food acquisition pathways with paracrine control of intestinal homeostasis. Moreover, the uroguanylin-GUCY2C endocrine axis may provide a therapeutic target to control appetite, obesity, and metabolic syndrome.


Asunto(s)
Conducta Animal/fisiología , Ingestión de Alimentos , Sistema Endocrino/metabolismo , Conducta Alimentaria/fisiología , Péptidos Natriuréticos/metabolismo , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Receptores de Péptidos/metabolismo , Sistemas de Mensajero Secundario/fisiología , Animales , Composición Corporal , Peso Corporal , GMP Cíclico/metabolismo , Sistema Endocrino/citología , Células Epiteliales/citología , Células Epiteliales/inmunología , Femenino , Hipotálamo/metabolismo , Insulina/sangre , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos Natriuréticos/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Receptores de Enterotoxina , Receptores Acoplados a la Guanilato-Ciclasa/genética , Receptores de Péptidos/genética , Saciedad
17.
Expert Rev Clin Pharmacol ; 4(2): 243-59, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21666781

RESUMEN

Obesity has escalated into a pandemic over the past few decades. In turn, research efforts have sought to elucidate the molecular mechanisms underlying the regulation of energy balance. A host of endogenous mediators regulate appetite and metabolism, and thereby control both short- and long-term energy balance. These mediators, which include gut, pancreatic and adipose neuropeptides, have been targeted in the development of anti-obesity pharmacotherapy, with the goal of amplifying anorexigenic and lipolytic signaling or blocking orexigenic and lipogenic signaling. This article presents the efficacy and safety of these anti-obesity drugs.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Regulación del Apetito/efectos de los fármacos , Regulación del Apetito/fisiología , Obesidad/tratamiento farmacológico , Animales , Fármacos Antiobesidad/metabolismo , Fármacos Antiobesidad/farmacología , Apetito/efectos de los fármacos , Apetito/fisiología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Humanos , Obesidad/metabolismo , Resultado del Tratamiento
18.
Expert Rev Endocrinol Metab ; 5(5): 765-783, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21297878

RESUMEN

With the eruption of the obesity pandemic over the past few decades, much research has been devoted to understanding the molecular mechanisms by which the human body regulates energy balance. These studies have revealed several mediators, including gut/pancreatic/adipose hormones and neuropeptides that control both short- and long-term energy balance by regulating appetite and/or metabolism. These endogenous mediators of energy balance have been the focus of many anti-obesity drug-development programs aimed at either amplifying endogenous anorexigenic/lipolytic signaling or blocking endogenous orexigenic/lipogenic signaling. Here, we discuss the efficacy and safety of targeting these pathways for the pharmacologic treatment of obesity.

19.
Toxins (Basel) ; 2(8): 2028-54, 2010 08.
Artículo en Inglés | MEDLINE | ID: mdl-22069671

RESUMEN

Heat-stable toxins (STs) produced by enterotoxigenic bacteria cause endemic and traveler's diarrhea by binding to and activating the intestinal receptor guanylyl cyclase C (GC-C). Advances in understanding the biology of GC-C have extended ST from a diarrheagenic peptide to a novel therapeutic agent. Here, we summarize the physiological and pathophysiological role of GC-C in fluid-electrolyte regulation and intestinal crypt-villus homeostasis, as well as describe translational opportunities offered by STs, reflecting the unique characteristics of GC-C, in treating irritable bowel syndrome and chronic constipation, and in preventing and treating colorectal cancer.


Asunto(s)
Toxinas Bacterianas/uso terapéutico , Enterotoxinas/uso terapéutico , Receptores Acoplados a la Guanilato-Ciclasa/fisiología , Receptores de Péptidos/fisiología , Secuencia de Aminoácidos , Animales , Toxinas Bacterianas/toxicidad , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Estreñimiento/tratamiento farmacológico , Enterotoxinas/toxicidad , Proteínas de Escherichia coli , Humanos , Síndrome del Colon Irritable/tratamiento farmacológico , Síndrome del Colon Irritable/etiología , Datos de Secuencia Molecular , Estadificación de Neoplasias , Péptidos/uso terapéutico , Receptores de Enterotoxina , Receptores Acoplados a la Guanilato-Ciclasa/agonistas , Receptores Acoplados a la Guanilato-Ciclasa/genética , Receptores de Péptidos/agonistas , Receptores de Péptidos/genética , Transducción de Señal
20.
Future Oncol ; 5(4): 509-22, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19450179

RESUMEN

Colorectal cancer is one of the leading causes of tumor-related morbidity and mortality worldwide. While mechanisms underlying this disease have been elucidated over the past two decades, these molecular insights have failed to translate into efficacious therapy. The oncogenomic view of cancer suggests that terminal transformation reflects the sequential corruption of signal transduction circuits regulating key homeostatic mechanisms, whose multiplicity underlies the therapeutic resistance of most tumors to interventions targeting individual pathways. Conversely, the paucity of mechanistic insights into proximal pathophysiological processes that initiate and amplify oncogenic circuits preceding accumulation of mutations and transformation impedes development of effective prevention and therapy. In that context, guanylyl cyclase C (GCC), the intestinal receptor for the paracrine hormones guanylin and uroguanylin, whose early loss characterizes colorectal transformation, has emerged as a component of lineage-specific homeostatic programs organizing spatiotemporal patterning along the crypt-surface axis. Dysregulation of GCC signaling, reflecting hormone loss, promotes tumorigenesis through reprogramming of replicative and bioenergetic circuits and genomic instability. Compensatory upregulation of GCC in response to hormone loss provides a unique translational opportunity for prevention and treatment of colorectal tumors by hormone-replacement therapy.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/genética , Predisposición Genética a la Enfermedad , Guanilato Ciclasa/genética , Receptores de Péptidos/genética , Animales , Humanos , Receptores de Enterotoxina , Receptores Acoplados a la Guanilato-Ciclasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...