Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 113(23): E3250-9, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27226307

RESUMEN

Growth hormone (GH) excess in acromegaly is associated with increased precancerous colon polyps and soft tissue adenomas, whereas short-stature humans harboring an inactivating GH receptor mutation do not develop cancer. We show that locally expressed colon GH is abundant in conditions predisposing to colon cancer and in colon adenocarcinoma-associated stromal fibroblasts. Administration of a GH receptor (GHR) blocker in acromegaly patients induced colon p53 and adenomatous polyposis coli (APC), reversing progrowth GH signals. p53 was also induced in skin fibroblasts derived from short-statured humans with mutant GHR. GH-deficient prophet of pituitary-specific positive transcription factor 1 (Prop1)(-/-) mice exhibited induced colon p53 levels, and cross-breeding them with Apc(min+/-) mice that normally develop intestinal and colon tumors resulted in GH-deficient double mutants with markedly decreased tumor number and size. We also demonstrate that GH suppresses p53 and reduces apoptosis in human colon cell lines as well as in induced human pluripotent stem cell-derived intestinal organoids, and confirm in vivo that GH suppresses colon mucosal p53/p21. GH excess leads to decreased colon cell phosphatase and tensin homolog deleted on chromosome 10 (PTEN), increased cell survival with down-regulated APC, nuclear ß-catenin accumulation, and increased epithelial-mesenchymal transition factors and colon cell motility. We propose that GH is a molecular component of the "field change" milieu permissive for neoplastic colon growth.


Asunto(s)
Neoplasias del Colon/metabolismo , Hormona del Crecimiento/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acromegalia/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Colon/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Fibroblastos/metabolismo , Humanos , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Mutación , Fosfohidrolasa PTEN/metabolismo , Receptores de Somatotropina/genética , Piel/citología , Proteína p53 Supresora de Tumor/genética , Adulto Joven , beta Catenina/metabolismo
2.
J Neuroinflammation ; 15(1): 260, 2018 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-30201019

RESUMEN

BACKGROUND: Inflammation has been proposed to contribute to the decline in adult hippocampal neurogenesis. Proinflammatory cytokines activate transcription of chemokine growth-regulated oncogene α (Gro1) in human and murine hippocampal neuronal progenitor cells (NPC). The goal of this study was to investigate the effects of Gro1 on hippocampal neurogenesis in the presence of inflammation. METHODS: Human hippocampal NPC were transfected with lentivirus expressing Gro1, and murine NPC and hippocampal neuronal HT-22 cells were treated with Gro1 protein. A plasmid expressing mGro1 was electroporated in the hippocampus of newborn mice that were sacrificed 10 days later. Adult male and female mice were injected with lipopolysaccharide (LPS; 1 mg/kg, i.p in five daily injections) or normal saline. Adult male mice were implanted with pellets releasing 17-ß estradiol (E2; 2.5 mg/pellet, 41.666 µg/day release) or placebo for 6 weeks and challenged with LPS or normal saline as above. In both experiments, mice were sacrificed 3 h after the last injection. Hippocampal markers of neurogenesis were assessed in vitro and in vivo by Western blot, real-time PCR, and immunohisto/cytochemistry. RESULTS: Gro1 induced premature senescence in NPC and HT-22 cells, activating senescence-associated ß-galactosidase and the cell cycle inhibitor p16 and suppressing neuroblast proliferation and expression of doublecortin (DCX) and neuron-specific class III beta-tubulin (Tuj-1), both neuroblast markers, while promoting proliferation of neural glial antigen 2 (Ng2)-positive oligodendrocytes. Gro1 overexpression in the hippocampus of newborn mice resulted in decreased neuroblast development, as evidenced by decreased DCX expression and increased expression of platelet-derived growth factor α receptor (PDGFαR), a marker of oligodendrocyte precursors. In adult mice, Gro1 was induced in response to LPS treatment in male but not in female hippocampus, with a subsequent decrease in neurogenesis and activation of oligodendrocyte progenitors. No changes in neurogenesis were observed in females. Treatment with E2 blunted LPS-induced Gro1 in the male hippocampus. CONCLUSIONS: Inflammation-induced Gro1 triggers neuroblast senescence, thus suppressing new neuron development in the hippocampus. Sex-dependent differences in Gro1 response are attributed to estradiol, which blunts these changes, protecting the female hippocampus from the deleterious effects of inflammation-induced Gro1 on neurogenesis.


Asunto(s)
Quimiocina CXCL1/metabolismo , Citocinas/metabolismo , Estradiol/farmacología , Estrógenos/farmacología , Inflamación/inducido químicamente , Células-Madre Neurales/efectos de los fármacos , Adulto , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Quimiocina CXCL1/genética , Citocinas/genética , Proteína Doblecortina , Epilepsia/patología , Femenino , Galactosa/genética , Galactosa/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Células-Madre Neurales/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
3.
Brain Behav Immun ; 58: 1-8, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26802985

RESUMEN

Adult hippocampal neurogenesis is involved in memory and learning, and disrupted neurogenesis is implicated in cognitive impairment and mood disorders, including anxiety and depression. Some long-term peripheral illnesses and metabolic disorders, as well as normal aging, create a state of chronic peripheral inflammation. These conditions are associated with behavioral disturbances linked to disrupted adult hippocampal neurogenesis, such as cognitive impairment, deficits in learning and memory, and depression and anxiety. Pro-inflammatory cytokines released in the periphery are involved in peripheral immune system-to-brain communication by activating resident microglia in the brain. Activated microglia reduce neurogenesis by suppressing neuronal stem cell proliferation, increasing apoptosis of neuronal progenitor cells, and decreasing survival of newly developing neurons and their integration into existing neuronal circuits. In this review, we summarize evolving evidence that the state of chronic peripheral inflammation reduces adult hippocampal neurogenesis, which, in turn, produces the behavioral disturbances observed in chronic inflammatory disorders. As there are no data available on neurogenesis in humans with chronic peripheral inflammatory disease, we focus on animal models and, in parallel, consider the evidence of cognitive disturbance and mood disorders in human patients.


Asunto(s)
Hipocampo/inmunología , Hipocampo/fisiopatología , Inflamación/inmunología , Inflamación/psicología , Neurogénesis , Envejecimiento , Animales , Complicaciones de la Diabetes , Diabetes Mellitus/inmunología , Enterocolitis/complicaciones , Enterocolitis/inmunología , Humanos , Inflamación/complicaciones , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/inmunología , Obesidad/complicaciones , Obesidad/inmunología
4.
Proc Natl Acad Sci U S A ; 110(35): E3331-9, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23940366

RESUMEN

Premature proliferative arrest in benign or early-stage tumors induced by oncoproteins, chromosomal instability, or DNA damage is associated with p53/p21 activation, culminating in either senescence or apoptosis, depending on cell context. Growth hormone (GH) elicits direct peripheral metabolic actions as well as growth effects mediated by insulin-like growth factor 1 (IGF1). Locally produced peripheral tissue GH, in contrast to circulating pituitary-derived endocrine GH, has been proposed to be both proapoptotic and prooncogenic. Pituitary adenomas expressing and secreting GH are invariably benign and exhibit DNA damage and a senescent phenotype. We therefore tested effects of nutlin-induced p53-mediated senescence in rat and human pituitary cells. We show that DNA damage senescence induced by nutlin triggers the p53/p21 senescent pathway, with subsequent marked induction of intracellular pituitary GH in vitro. In contrast, GH is not induced in cells devoid of p53. Furthermore we show that p53 binds specific GH promoter motifs and enhances GH transcription and secretion in senescent pituitary adenoma cells and also in nonpituitary (human breast and colon) cells. In vivo, treatment with nutlin results in up-regulation of both p53 and GH in the pituitary gland, as well as increased GH expression in nonpituitary tissues (lung and liver). Intracrine GH acts in pituitary cells as an apoptosis switch for p53-mediated senescence, likely protecting the pituitary adenoma from progression to malignancy. Unlike in the pituitary, in nonpituitary cells GH exerts antiapoptotic properties. Thus, the results show that GH is a direct p53 transcriptional target and fulfills criteria as a p53 target gene. Induced GH is a readily measurable cell marker for p53-mediated cellular senescence.


Asunto(s)
Senescencia Celular/fisiología , Hormona del Crecimiento/fisiología , Hipófisis/metabolismo , Adenoma/metabolismo , Adenoma/patología , Animales , Línea Celular Tumoral , Células Cultivadas , Daño del ADN , Etopósido/farmacología , Hormona del Crecimiento/genética , Humanos , Imidazoles/farmacología , Ratones , Ratones Endogámicos C57BL , Piperazinas/farmacología , Hipófisis/citología , Hipófisis/efectos de los fármacos , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Regiones Promotoras Genéticas , Ratas , Transcripción Genética/fisiología , Proteína p53 Supresora de Tumor/fisiología
5.
J Neuroinflammation ; 12: 65, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25889852

RESUMEN

BACKGROUND: Adult neurogenesis in the subgranular zone of the hippocampus is involved in learning, memory, and mood control. Decreased hippocampal neurogenesis elicits significant behavioral changes, including cognitive impairment and depression. Inflammatory bowel disease (IBD) is a group of chronic inflammatory conditions of the intestinal tract, and cognitive dysfunction and depression frequently occur in patients suffering from this disorder. We therefore tested the effects of chronic intestinal inflammation on hippocampal neurogenesis. METHODS: The dextran sodium sulfate (DSS) mouse model of IBD was used. Mice were treated with multiple-cycle administration of 3% wt/vol DSS in drinking water on days 1 to 5, 8 to 12, 15 to 19, and 22 to 26. Mice were sacrificed on day 7 (acute phase of inflammation) or day 29 (chronic phase of inflammation) after the beginning of the treatment. RESULTS: During the acute phase of inflammation, we found increased plasma levels of IL-6 and TNF-α and increased expression of Iba1, a marker of activated microglia, accompanied by induced IL-6 and IL-1ß, and the cyclin-dependent kinase inhibitor p21(Cip1) (p21) in hippocampus. During the chronic phase of inflammation, plasma levels of IL-6 were elevated. In the hippocampus, p21 protein levels were continued to be induced. Furthermore, markers of stem/early progenitor cells, including nestin and brain lipid binding protein (BLBP), and neuronal marker doublecortin (DCX) were all down-regulated, whereas glial fibrillary acidic protein (GFAP), a marker for astroglia, was induced. In addition, the number of proliferating precursors of neuronal lineage assessed by double Ki67 and DCX staining was significantly diminished in the hippocampus of DSS-treated animals, indicating decreased production of new neurons. CONCLUSIONS: We show for the first time that chronic intestinal inflammation alters hippocampal neurogenesis. As p21 arrests early neuronal progenitor proliferation, it is likely that p21 induction during acute phase of inflammation resulted in the reduction of hippocampal neurogenesis observed later, on day 29, after the beginning of DSS treatment. The reduction in hippocampal neurogenesis might underlie the behavioral manifestations that occur in patients with IBD.


Asunto(s)
Citocinas/metabolismo , Gastroenteritis/patología , Hipocampo/fisiopatología , Neurogénesis/fisiología , Animales , Células Cultivadas , Enfermedad Crónica , Citocinas/genética , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Proteína Doblecortina , Ensayo de Inmunoadsorción Enzimática , Femenino , Gastroenteritis/inducido químicamente , Hipocampo/efectos de los fármacos , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , ARN Mensajero/metabolismo , Estadísticas no Paramétricas , Factores de Tiempo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
6.
Aging Cell ; : e14193, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724466

RESUMEN

DNA damage-induced senescence is initially sustained by p53. Senescent cells produce a senescence-associated secretory phenotype (SASP) that impacts the aging microenvironment, often promoting cell transformation. Employing normal non-tumorous human colon cells (hNCC) derived from surgical biopsies and three-dimensional human intestinal organoids, we show that local non-pituitary growth hormone (npGH) induced in senescent cells is a SASP component acting to suppress p53. npGH autocrine/paracrine suppression of p53 results in senescence evasion and cell-cycle reentry, as evidenced by increased Ki67 and BrdU incorporation. Post-senescent cells exhibit activated epithelial-to-mesenchymal transition (EMT), and increased cell motility. Nu/J mice harboring GH-secreting HCT116 xenografts with resultant high GH levels and injected intrasplenic with post-senescent hNCC developed fourfold more metastases than did mice harboring control xenografts, suggesting that paracrine npGH enables post-senescent cell transformation. By contrast, senescent cells with suppressed npGH exhibit downregulated Ki67 and decreased soft agar colony formation. Mechanisms underlying these observations include npGH induction by the SASP chemokine CXCL1, which attracts immune effectors to eliminate senescent cells; GH, in turn, suppresses CXCL1, likely by inhibiting phospho-NFκB, resulting in SASP cytokine downregulation. Consistent with these findings, GH-receptor knockout mice exhibited increased colon phospho-NFκB and CXCL1, while GH excess decreased colon CXCL1. The results elucidate mechanisms for local hormonal regulation of microenvironmental changes in DNA-damaged non-tumorous epithelial cells and portray a heretofore unappreciated GH action favoring age-associated epithelial cell transformation.

7.
Hippocampus ; 23(12): 1383-94, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23966332

RESUMEN

Altered neurogenesis in adult hippocampus is implicated in cognition impairment and depression. Inflammation is a potent inhibitor of neurogenesis. The cyclin-dependent kinase inhibitor p21(Cip1) (p21) restrains cell cycle progression and arrests the cell in the G1 phase. We recently showed that p21 is expressed in neuronal progenitors and regulates proliferation of these cells in the subgranular zone of the dentate gyrus of hippocampus where adult neurogenesis occurs. The current study suggests that p21 is induced in vivo in the hippocampus of WT mice in response to acute systemic inflammation caused by LPS injections, restrains neuronal progenitor proliferation and protects these cells from inflammation-induced apoptosis. In intact p21-/- hippocampus, neuronal progenitors proliferate more actively as assessed by BrdU incorporation, and give rise to increased number of DCX positive neuroblasts. However, when mice were treated with LPS, the number of neuroblasts decreased due to induced subgranular zone apoptosis. In vitro, differentiating Tuj-1 positive neuroblasts isolated from p21-/- hippocampus exhibited increased proliferation rate, measured by Ki-67 staining, as compared to WT cells (p<0.05). In WT neuronal progenitors treated with IL-6, the number of p21-positive cells was increased (p<0.05), and this led to Tuj-1(+) cell proliferation restraint, whereas the number of proliferating GFAP(+) astrocytes was increased ~ 2-fold. Thus, when p21 is intact, inflammation might divert neuronal progenitors towards astrogliogenesis by inducing p21. At the same time, when p21 is lacking, no effects of IL-6 on proliferation of Tuj-1(+) cells or GFAP(+) cells are detected in differentiating p21-/- neuronal progenitors. These results underscore the important role of p21 controlling hippocampal neuronal differentiation during inflammation.


Asunto(s)
Apoptosis/genética , Hipocampo/fisiopatología , Inflamación/patología , Células-Madre Neurales/fisiología , Neurogénesis/genética , Quinasas p21 Activadas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Proteína Doblecortina , Inflamación/inducido químicamente , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/efectos de los fármacos , Quinasas p21 Activadas/deficiencia
8.
Endocr Relat Cancer ; 30(7)2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37066857

RESUMEN

Non-pituitary growth hormone (npGH) expression is well established in extrapituitary tissues, but an understanding of the physiological role of npGH remains rather limited. Pro-tumorigenic npGH impacting the tumor microenvironment has been reviewed. We focus here on autocrine/paracrine npGH effects in non-tumorous tissues and discuss its mechanisms of action in the normal tissue microenvironment. We address the tissue-specific effects of npGH in regulating stem, endothelial, immune, and epithelial cells and highlight the related role of npGH-associated changes in tissue aging.


Asunto(s)
Hormona de Crecimiento Humana , Hipófisis , Humanos , Hipófisis/metabolismo , Hormona de Crecimiento Humana/metabolismo , Hormona del Crecimiento/metabolismo
9.
iScience ; 26(11): 108117, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37876819

RESUMEN

DNA damage repair (DDR) is mediated by phosphorylating effectors ATM kinase, CHK2, p53, and γH2AX. We showed earlier that GH suppresses DDR by suppressing pATM, resulting in DNA damage accumulation. Here, we show GH acting through GH receptor (GHR) inducing wild-type p53-inducible phosphatase 1 (WIP1), which dephosphorylated ATM and its effectors in normal human colon cells and three-dimensional human intestinal organoids. Mice bearing GH-secreting xenografts exhibited induced colon WIP1 with suppressed pATM and γH2AX. WIP1 was also induced in buffy coats derived from patients with elevated GH from somatotroph adenomas. In contrast, decreased colon WIP1 was observed in GHR-/- mice. WIP1 inhibition restored ATM phosphorylation and reversed GH-induced DNA damage. We elucidated a novel GH signaling pathway activating Src/AMPK to trigger HIPK2 nuclear-cytoplasmic relocation and suppressing WIP1 ubiquitination. Concordantly, blocking either AMPK or Src abolished GH-induced WIP1. We identify WIP1 as a specific target for GH-mediated epithelial DNA damage accumulation.

10.
Cells ; 11(2)2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35053333

RESUMEN

Over the past two decades, interest in the role of the somatotroph growth hormone/insulin-like growth factor (GH/IGF1) axis in multiple aspects of physiology and pathology has grown exponentially [...].


Asunto(s)
Hormona del Crecimiento/metabolismo , Transducción de Señal , Somatomedinas/metabolismo , Envejecimiento/metabolismo , Animales , Genómica , Humanos
11.
J Endocr Soc ; 6(1): bvab177, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-34909519

RESUMEN

Replicative senescence occurs due to an inability to repair DNA damage and activation of p53/p21 and p16INK4 pathways. It is considered a preventive mechanism for arresting proliferation of DNA-damaged cells. Stably senescent cells are characterized by a senescence-associated secretory phenotype (SASP), which produces and secretes cytokines, chemokines, and/or matrix metalloproteinases depending on the cell type. SASP proteins may increase cell proliferation, facilitating conversion of premalignant to malignant tumor cells, triggering DNA damage, and altering the tissue microenvironment. Further, senescent cells accumulate with age, thereby aggravating age-related tissue damage. Here, we review a heretofore unappreciated role for growth hormone (GH) as a SASP component, acting in an autocrine and paracrine fashion. In senescent cells, GH is activated by DNA-damage-induced p53 and inhibits phosphorylation of DNA repair proteins ATM, Chk2, p53, and H2AX. Somatotroph adenomas containing abundant intracellular GH exhibit increased somatic copy number alterations, indicative of DNA damage, and are associated with induced p53/p21. As this pathway restrains proliferation of DNA-damaged cells, these mechanisms may underlie the senescent phenotype and benign nature of slowly proliferating pituitary somatotroph adenomas. In highly proliferative cells, such as colon epithelial cells, GH induced in response to DNA damage suppresses p53, thereby triggering senescent cell proliferation. As senescent cells harbor unrepaired DNA damage, GH may enable senescent cells to evade senescence and reenter the cell cycle, resulting in acquisition of harmful mutations. These mechanisms, at least in part, may underlie pro-aging effects of GH observed in animal models and in patients with chronically elevated GH levels.

12.
Proc Natl Acad Sci U S A ; 105(4): 1358-63, 2008 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-18172194

RESUMEN

The subgranular zone (SGZ) of the dentate gyrus of the hippocampus is a brain region where robust neurogenesis continues throughout adulthood. Cyclin-dependent kinases (CDKs) have a primary role in controlling cell division and cellular proliferation. p21(Cip1) (p21) is a CDK inhibitor that restrains cell cycle progression. Confocal microscopy revealed that p21 is abundantly expressed in the nuclei of cells in the SGZ and is colocalized with NeuN, a marker for neurons. Doublecortin (DCX) is a cytoskeletal protein that is primarily expressed by neuroblasts. By using FACS analysis it was found that, among DCX-positive cells, 42.8% stained for p21, indicating that p21 is expressed in neuroblasts and in newly developing neurons. p21-null (p21(-/-)) mice were examined, and the rate of cellular proliferation, as measured by BrdU incorporation, was increased in the SGZ of p21(-/-) compared with WT mice. In addition, the levels of both DCX and NeuN protein were increased in p21(-/-) mice, further demonstrating increased hippocampal neuron proliferation. Chronic treatment with the tricyclic antidepressant imipramine (10 mg/kg per day i.p. for 21 days) markedly decreased hippocampal p21 mRNA and protein levels, produced antidepressant-like behavioral changes in the forced swim test, and stimulated neurogenesis in the hippocampus. These results suggest that p21 restrains neurogenesis in the SGZ and imipramine-induced stimulation of neurogenesis might be a consequence of decreased p21 expression and the subsequent release of neuronal progenitor cells from the blockade of proliferation. Because many antidepressants stimulate neurogenesis, it is possible that their shared common mechanism of action is suppression of p21.


Asunto(s)
Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Giro Dentado/citología , Giro Dentado/metabolismo , Inhibidores de Crecimiento/fisiología , Inhibición Neural/fisiología , Neuronas/citología , Neuronas/metabolismo , Animales , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteína Doblecortina , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos
13.
Proc Natl Acad Sci U S A ; 105(45): 17498-503, 2008 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-18981426

RESUMEN

As commonly encountered, pituitary adenomas are invariably benign. We therefore studied protective pituitary proliferative mechanisms. Pituitary tumor transforming gene (Pttg) deletion results in pituitary p21 induction and abrogates tumor development in Rb(+/-)Pttg(-/-) mice. p21 disruption restores attenuated Rb(+/-)Pttg(-/-) pituitary proliferation rates and enables high penetrance of pituitary, but not thyroid, tumor growth in triple mutant animals (88% of Rb(+/-) and 72% of Rb(+/-)Pttg(-/-)p21(-/-) vs. 30% of Rb(+/-)Pttg(-/-) mice developed pituitary tumors, P < 0.001). p21 deletion also accelerated S-phase entry and enhanced transformation rates in triple mutant MEFs. Intranuclear p21 accumulates in Pttg-null aneuploid GH-secreting cells, and GH(3) rat pituitary tumor cells overexpressing PTTG also exhibited increased levels of mRNA for both p21 (18-fold, P < 0.01) and ATM (9-fold, P < 0.01). PTTG is abundantly expressed in human pituitary tumors, and in 23 of 26 GH-producing pituitary adenomas with high PTTG levels, senescence was evidenced by increased p21 and SA-beta-galactosidase. Thus, either deletion or overexpression of Pttg promotes pituitary cell aneuploidy and p53/p21-dependent senescence, particularly in GH-secreting cells. Aneuploid pituitary cell p21 may constrain pituitary tumor growth, thus accounting for the very low incidence of pituitary carcinomas.


Asunto(s)
Transformación Celular Neoplásica/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Neoplasias/genética , Neoplasias Hipofisarias/metabolismo , Aneuploidia , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Inmunohistoquímica , Ratones , Ratones Noqueados , Microscopía Fluorescente , Proteínas Serina-Treonina Quinasas/metabolismo , Securina , Proteínas Supresoras de Tumor/metabolismo
14.
Cell Rep ; 37(11): 110068, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34910915

RESUMEN

Microenvironmental factors modulating age-related DNA damage are unclear. Non-pituitary growth hormone (npGH) is induced in human colon, non-transformed human colon cells, and fibroblasts, and in 3-dimensional intestinal organoids with age-associated DNA damage. Autocrine/paracrine npGH suppresses p53 and attenuates DNA damage response (DDR) by inducing TRIM29 and reducing ATM phosphorylation, leading to reduced DNA repair and DNA damage accumulation. Organoids cultured up to 4 months exhibit aging markers, p16, and SA-ß-galactosidase and decreased telomere length, as well as DNA damage accumulation, with increased npGH, suppressed p53, and attenuated DDR. Suppressing GH in aged organoids increases p53 and decreases DNA damage. WT mice exhibit age-dependent colon DNA damage accumulation, while in aged mice devoid of colon GH signaling, DNA damage remains low, with elevated p53. As age-associated npGH induction enables a pro-proliferative microenvironment, abrogating npGH signaling could be targeted as anti-aging therapy by impeding DNA damage and age-related pathologies.


Asunto(s)
Envejecimiento , Proteínas Portadoras/fisiología , Colon/patología , Daño del ADN , Fibroblastos/patología , Hormona de Crecimiento Humana/metabolismo , Mucosa Intestinal/patología , Animales , Colon/metabolismo , Reparación del ADN , Fibroblastos/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
15.
Front Horm Res ; 38: 7-14, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20616490

RESUMEN

As commonly encountered pituitary adenomas are invariably benign, we examined protective pituitary proliferative mechanisms. Cellular senescence is characterized by a largely irreversible cell cycle arrest and constitutes a strong anti-proliferative response, which can be triggered by DNA damage, chromosomal instability and aneuploidy, loss of tumor suppressive signaling or oncogene activation. Cellular senescence may prevent cells from undergoing transformation in vitro. Recently, evidence has accumulated that in vivo senescence is an important protective mechanisms against cancer. In this review we highlight an intrinsic predisposition of pituitary tumors to exhibit senescence-associated molecular pathways and show prospective mechanisms underlying the benign nature of these commonly encountered tumors.


Asunto(s)
Adenoma/patología , Senescencia Celular , Neoplasias Hipofisarias/patología , Animales , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Humanos , Proteínas de Neoplasias/fisiología , Securina
16.
Mol Endocrinol ; 23(3): 337-48, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19131507

RESUMEN

Somatostatin (SRIF) binds G protein-coupled SRIF receptor subtypes (SST1, -2, -3, -4, and -5) to regulate cell secretion and proliferation. Hypothalamic SRIF inhibits pituitary growth hormone, thyroid stimulating hormone, and ACTH secretion. We tested SRIF-independent constitutive SST activity in AtT20 mouse pituitary corticotroph cells in which ACTH secretion is highly sensitive to SRIF action. Stable transfectants expressing SST2 or SST5 were sensitized to selective agonist action, and constitutive SST receptor activity was demonstrated by forskolin and pertussis toxin cAMP cell responses. Persistent constitutive SST activity decreased cell ACTH responses to CRH through decreased expression of CRH receptor subtype 1. Decreased dopamine receptor type 1 expression was associated with attenuated dopamine agonist action, whereas responses to isoproterenol were enhanced through increased beta2-adrenoreceptor expression. Thus, integrated pituitary cell ACTH regulation is determined both by phasic SRIF action, as well as by tonic constitutive SST activity, independently of SRIF.


Asunto(s)
Células Endocrinas/fisiología , Hipófisis/fisiología , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/fisiología , Hormona Adrenocorticotrópica/metabolismo , Células Cultivadas , Células Endocrinas/efectos de los fármacos , Células Endocrinas/metabolismo , Humanos , Ligandos , Octreótido/farmacología , Péptidos/farmacología , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Receptores de Somatostatina/antagonistas & inhibidores , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Somatostatina/genética , Somatostatina/metabolismo , Somatostatina/farmacología , Especificidad por Sustrato , Transfección
17.
Endocr Rev ; 41(4)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32270196

RESUMEN

DNA damage response (DDR) and DNA repair pathways determine neoplastic cell transformation and therapeutic responses, as well as the aging process. Altered DDR functioning results in accumulation of unrepaired DNA damage, increased frequency of tumorigenic mutations, and premature aging. Recent evidence suggests that polypeptide hormones play a role in modulating DDR and DNA damage repair, while DNA damage accumulation may also affect hormonal status. We review the available reports elucidating involvement of insulin-like growth factor 1 (IGF1), growth hormone (GH), α-melanocyte stimulating hormone (αMSH), and gonadotropin-releasing hormone (GnRH)/gonadotropins in DDR and DNA repair as well as the current understanding of pathways enabling these actions. We discuss effects of DNA damage pathway mutations, including Fanconi anemia, on endocrine function and consider mechanisms underlying these phenotypes. (Endocrine Reviews 41: 1 - 19, 2020).


Asunto(s)
Daño del ADN/fisiología , Reparación del ADN/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Gonadotropinas/metabolismo , Hormona del Crecimiento/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Hormonas Peptídicas/fisiología , alfa-MSH/metabolismo , Animales , Humanos
18.
J Clin Invest ; 130(11): 5738-5755, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32673291

RESUMEN

Drivers of sporadic benign pituitary adenoma growth are largely unknown. Whole-exome sequencing of 159 prospectively resected pituitary adenomas showed that somatic copy number alteration (SCNA) rather than mutation is a hallmark of hormone-secreting adenomas and that SCNAs correlate with adenoma phenotype. Using single-gene SCNA pathway analysis, we observed that both cAMP and Fanconi anemia DNA damage repair pathways were affected by SCNAs in growth hormone-secreting (GH-secreting) somatotroph adenomas. As somatotroph differentiation and GH secretion are dependent on cAMP activation and we previously showed DNA damage, aneuploidy, and senescence in somatotroph adenomas, we studied links between cAMP signaling and DNA damage. Stimulation of cAMP in C57BL/6 mouse primary pituitary cultures using forskolin or a long-acting GH-releasing hormone (GHRH) analog increased GH production and DNA damage measured by H2AX phosphorylation and a comet assay. Octreotide, a somatostatin receptor ligand that targets somatotroph adenoma GH secretion in patients with acromegaly, inhibited cAMP and GH and reversed DNA damage induction. In vivo long-acting GHRH treatment also induced pituitary DNA damage in mice. We conclude that cAMP, which induces somatotroph proliferation and GH secretion, may concomitantly induce DNA damage, potentially linking hormone hypersecretion to SCNA and genome instability. These results elucidating somatotroph adenoma pathophysiology identify pathways for targeted treatment.


Asunto(s)
Adenoma , Daño del ADN , ADN de Neoplasias , Adenoma Hipofisario Secretor de Hormona del Crecimiento , Hormona de Crecimiento Humana , Proteínas de Neoplasias , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Animales , AMP Cíclico/genética , AMP Cíclico/metabolismo , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Femenino , Adenoma Hipofisario Secretor de Hormona del Crecimiento/genética , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Adenoma Hipofisario Secretor de Hormona del Crecimiento/patología , Hormona de Crecimiento Humana/genética , Hormona de Crecimiento Humana/metabolismo , Humanos , Masculino , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Sistemas de Mensajero Secundario/genética
19.
Horm Res ; 71 Suppl 2: 82-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19407503

RESUMEN

Pituitary tumours account for 15% of intracranial neoplasms and are benign monoclonal neoplasms that may be clinically silent or secrete hormones, including prolactin, growth hormone, adrenocorticotrophic hormone or, rarely, thyroid-stimulating hormone or gonadotrophins. These adenomas account for clinical infertility, growth disorders and hypercortisolism or metabolic dysfunctions associated with hypopituitarism. We explored the role of disordered pituitary cell proliferation control in the pathogenesis of these invariably benign adenomas, studying the mechanisms underlying pituitary aneuploidy, premature proliferative arrest (senescence), markers of cell proliferation and tumorigenesis in single, double or triply mutant transgenic mice with mutations of Rb, Pttg and/or p21. Our results provide further insights into the role of cell-cycle control and growth constraints on experimental and human pituitary tumours, which underlie their failure to progress to malignancy. These results improve our understanding of pituitary syndromes associated with infertility, growth disorders, hypercortisolism or adrenal, thyroid and gonadal failure due to abrogated pituitary function.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Ciclo Celular , Senescencia Celular , Mutación , Proteínas de Neoplasias/metabolismo , Neoplasias Hipofisarias/metabolismo , Hiperfunción de las Glándulas Suprarrenales/etiología , Hiperfunción de las Glándulas Suprarrenales/genética , Hiperfunción de las Glándulas Suprarrenales/metabolismo , Hiperfunción de las Glándulas Suprarrenales/terapia , Aneuploidia , Animales , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Trastornos del Crecimiento/etiología , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/metabolismo , Trastornos del Crecimiento/terapia , Humanos , Infertilidad/etiología , Infertilidad/genética , Infertilidad/metabolismo , Infertilidad/terapia , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Hormonas Hipofisarias/metabolismo , Neoplasias Hipofisarias/complicaciones , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/terapia , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Securina
20.
Arch Endocrinol Metab ; 63(6): 568-575, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31939481

RESUMEN

Tumor development is a multistep process whereby local mechanisms enable somatic mutations during preneoplastic stages. Once a tumor develops, it becomes a complex organ composed of multiple cell types. Interactions between malignant and non-transformed cells and tissues create a tumor microenvironment (TME) comprising epithelial cancer cells, cancer stem cells, non-tumorous cells, stromal cells, immune-inflammatory cells, blood and lymphatic vascular network, and extracellular matrix. We review reports and present a hypothesis that postulates the involvement of growth hormone (GH) in field cancerization. We discuss GH contribution to TME, promoting epithelial-to-mesenchymal transition, accumulation of unrepaired DNA damage, tumor vascularity, and resistance to therapy. Arch Endocrinol Metab. 2019;63(6):568-75.


Asunto(s)
Daño del ADN/fisiología , Resistencia a Antineoplásicos/fisiología , Transición Epitelial-Mesenquimal/fisiología , Hormona de Crecimiento Humana/fisiología , Neovascularización Patológica/fisiopatología , Microambiente Tumoral/fisiología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA