Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Dev Dyn ; 251(7): 1175-1195, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35048440

RESUMO

BACKGROUND: Hedgehog (HH) signaling is essential for homeostasis in gustatory fungiform papillae (FP) and taste buds. However, activities of HH antagonists in these tissues remain unexplored. We investigated a potential role for HH-interacting protein (HHIP), an endogenous pathway antagonist, in regulating HH signaling during taste organ homeostasis. We found a restricted pattern of Hhip-expressing cells in the anterior epithelium of each nongustatory filiform papilla (FILIF) only. To test for roles in antagonism of HH signaling, we investigated HHIP after pathway inhibition with SMO inhibition via sonidegib and Smo deletion, Gli2 deletion/suppression, or with chorda tympani/lingual nerve cut. RESULTS: In all approaches, the HHIP expression pattern was retained in FILIF suggesting HH-independent regulation of HHIP. Remarkably, after pathway inhibition, HHIP expression was detected also in the conical, FILIF-like atypical FP. We found a close association of de novo expression of HHIP in atypical FP with loss of Gli1+, HH-responding cells. Further, we report that PTCH1 is another potential HH antagonist in FILIF that co-localizes with HHIP. CONCLUSIONS: After HH pathway inhibition the ectopic expression of HHIP correlates with a FILIF-like morphology in atypical FP and we propose that localized expression of the HH antagonist HHIP regulates pathway inhibition to maintain FILIF during tongue homeostasis.


Assuntos
Papilas Gustativas , Expressão Ectópica do Gene , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Homeostase , Papilas Gustativas/metabolismo , Língua
2.
Proc Natl Acad Sci U S A ; 114(48): E10369-E10378, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29133390

RESUMO

Striking taste disturbances are reported in cancer patients treated with Hedgehog (HH)-pathway inhibitor drugs, including sonidegib (LDE225), which block the HH pathway effector Smoothened (SMO). We tested the potential for molecular, cellular, and functional recovery in mice from the severe disruption of taste-organ biology and taste sensation that follows HH/SMO signaling inhibition. Sonidegib treatment led to rapid loss of taste buds (TB) in both fungiform and circumvallate papillae, including disruption of TB progenitor-cell proliferation and differentiation. Effects were selective, sparing nontaste papillae. To confirm that taste-organ effects of sonidegib treatment result from HH/SMO signaling inhibition, we studied mice with conditional global or epithelium-specific Smo deletions and observed similar effects. During sonidegib treatment, chorda tympani nerve responses to lingual chemical stimulation were maintained at 10 d but were eliminated after 16 d, associated with nearly complete TB loss. Notably, responses to tactile or cold stimulus modalities were retained. Further, innervation, which was maintained in the papilla core throughout treatment, was not sufficient to sustain TB during HH/SMO inhibition. Importantly, treatment cessation led to rapid and complete restoration of taste responses within 14 d associated with morphologic recovery in about 55% of TB. However, although taste nerve responses were sustained, TB were not restored in all fungiform papillae even with prolonged recovery for several months. This study establishes a physiologic, selective requirement for HH/SMO signaling in taste homeostasis that includes potential for sensory restoration and can explain the temporal recovery after taste dysgeusia in patients treated with HH/SMO inhibitors.


Assuntos
Antineoplásicos/efeitos adversos , Compostos de Bifenilo/efeitos adversos , Disgeusia/fisiopatologia , Piridinas/efeitos adversos , Transdução de Sinais/efeitos dos fármacos , Paladar/efeitos dos fármacos , Língua/fisiopatologia , Animais , Carcinoma Basocelular/tratamento farmacológico , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Nervo da Corda do Tímpano/efeitos dos fármacos , Nervo da Corda do Tímpano/fisiopatologia , Modelos Animais de Doenças , Disgeusia/induzido quimicamente , Disgeusia/patologia , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Hedgehog/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Recuperação de Função Fisiológica , Neoplasias Cutâneas/tratamento farmacológico , Receptor Smoothened/antagonistas & inibidores , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Células-Tronco/efeitos dos fármacos , Paladar/fisiologia , Papilas Gustativas/citologia , Papilas Gustativas/efeitos dos fármacos , Papilas Gustativas/patologia , Papilas Gustativas/fisiopatologia , Língua/efeitos dos fármacos , Língua/inervação
3.
PLoS Genet ; 12(11): e1006442, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27893742

RESUMO

For homeostasis, lingual taste papilla organs require regulation of epithelial cell survival and renewal, with sustained innervation and stromal interactions. To investigate a role for Hedgehog/GLI signaling in adult taste organs we used a panel of conditional mouse models to manipulate GLI activity within epithelial cells of the fungiform and circumvallate papillae. Hedgehog signaling suppression rapidly led to taste bud loss, papilla disruption, and decreased proliferation in domains of papilla epithelium that contribute to taste cells. Hedgehog responding cells were eliminated from the epithelium but retained in the papilla stromal core. Despite papilla disruption and loss of taste buds that are a major source of Hedgehog ligand, innervation to taste papillae was maintained, and not misdirected, even after prolonged GLI blockade. Further, vimentin-positive fibroblasts remained in the papilla core. However, retained innervation and stromal cells were not sufficient to maintain taste bud cells in the context of compromised epithelial Hedgehog signaling. Importantly taste organ disruption after GLI blockade was reversible in papillae that retained some taste bud cell remnants where reactivation of Hedgehog signaling led to regeneration of papilla epithelium and taste buds. Therefore, taste bud progenitors were either retained during epithelial GLI blockade or readily repopulated during recovery, and were poised to regenerate taste buds once Hedgehog signaling was restored, with innervation and papilla connective tissue elements in place. Our data argue that Hedgehog signaling is essential for adult tongue tissue maintenance and that taste papilla epithelial cells represent the key targets for physiologic Hedgehog-dependent regulation of taste organ homeostasis. Because disruption of GLI transcriptional activity in taste papilla epithelium is sufficient to drive taste organ loss, similar to pharmacologic Hedgehog pathway inhibition, the findings suggest that taste alterations in cancer patients using systemic Hedgehog pathway inhibitors result principally from interruption of signaling activity in taste papillae.


Assuntos
Proteínas Hedgehog/genética , Papilas Gustativas/metabolismo , Paladar/genética , Língua/metabolismo , Animais , Células Epiteliais/metabolismo , Epitélio/metabolismo , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Hedgehog/metabolismo , Camundongos , Fibras Nervosas/metabolismo , Transdução de Sinais , Células Estromais/metabolismo , Papilas Gustativas/crescimento & desenvolvimento , Percepção Gustatória/genética
4.
J Neurophysiol ; 113(3): 1034-40, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25392175

RESUMO

Taste sensation on the anterior tongue requires chorda tympani nerve function and connections with continuously renewing taste receptor cells. However, it is unclear which signaling pathways regulate the receptor cells to maintain chorda tympani sensation. Hedgehog (HH) signaling controls cell proliferation and differentiation in numerous tissues and is active in taste papillae and taste buds. In contrast, uncontrolled HH signaling drives tumorigenesis, including the common skin cancer, basal cell carcinoma. Systemic HH pathway inhibitors (HPIs) lead to basal cell carcinoma regression, but these drugs cause severe taste disturbances. We tested the hypothesis that taste disruption by HPIs reflects a direct requirement for HH signaling in maintaining taste organs and gustatory sensation. In mice treated with the HPI LDE225 up to 28 days, HH-responding cells were lost in fungiform papilla epithelium, and papillae acquired a conical apex. Taste buds were either absent or severely reduced in size in more than 90% of aberrant papillae. Taste bud remnants expressed the taste cell marker keratin 8, and papillae retained expression of nerve markers, neurofilament and P2X3. Chorda tympani nerve responses to taste stimuli were markedly reduced or absent in LDE225-treated mice. Responses to touch were retained, however, whereas cold responses were retained after 16 days of treatment but lost after 28 days. These data identify a critical, modality-specific requirement for HH signaling in maintaining taste papillae, taste buds and neurophysiological taste function, supporting the proposition that taste disturbances in HPI-treated patients are an on-target response to HH pathway blockade in taste organs.


Assuntos
Antineoplásicos/farmacologia , Compostos de Bifenilo/farmacologia , Proteínas Hedgehog/metabolismo , Piridinas/farmacologia , Papilas Gustativas/efeitos dos fármacos , Paladar , Animais , Feminino , Camundongos , Transdução de Sinais , Papilas Gustativas/metabolismo , Papilas Gustativas/fisiologia , Tato
5.
Mol Cancer Res ; 17(5): 1024-1035, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30683671

RESUMO

Aberrant activation of the Hedgehog signaling pathway has been linked to the formation of numerous cancer types, including the myogenic soft tissue sarcoma, embryonal rhabdomyosarcoma (eRMS). Here, we report PCG2, a novel mouse model in which human GLI2A, a constitutive activator of Hedgehog signaling, induced undifferentiated sarcomas that were phenotypically divergent from eRMS. Rather, sarcomas arising in PCG2 mice featured some characteristics that were reminiscent of Ewing sarcoma. Even though it is widely understood that Ewing sarcoma formation is driven by EWS-ETS gene fusions, a genetically defined mouse model is not well-established. While EWS-ETS gene fusions were not present in PCG2 sarcomas, precluding their designation as Ewing sarcoma, we did find that GLI2A induced expression of known EWS-ETS gene targets essential to Ewing pathogenesis, most notably, Nkx2.2. Moreover, we found that naïve mesenchymal progenitors originate tumors in PCG2 mice. Altogether, our work provides a novel genetic mouse model, which directly connects oncogenic Hedgehog activity to the etiology of undifferentiated soft tissue sarcomas for the first time. IMPLICATIONS: The finding that activation of Gli2 transcription factor is sufficient to induce Ewing-like sarcomas provides a direct transformative role of the Hedgehog signaling pathway in undifferentiated soft tissue sarcoma.


Assuntos
Proteínas de Homeodomínio/metabolismo , Proteínas do Tecido Nervoso/genética , Sarcoma de Ewing/patologia , Proteínas de Peixe-Zebra/metabolismo , Proteína Gli3 com Dedos de Zinco/genética , Animais , Regulação Neoplásica da Expressão Gênica , Proteínas Hedgehog/metabolismo , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Transplante de Neoplasias , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Transdução de Sinais , Fatores de Transcrição , Proteínas de Peixe-Zebra/genética , Proteína Gli3 com Dedos de Zinco/metabolismo
6.
J Invest Dermatol ; 135(5): 1415-1424, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25313532

RESUMO

Merkel cell carcinoma (MCC) is a rare and deadly neuroendocrine skin tumor frequently associated with clonal integration of a polyomavirus, Merkel cell polyomavirus (MCPyV), and MCC tumor cells express putative polyomavirus oncoprotein small T antigen (sTAg) and truncated large T antigen. Here, we show robust transforming activity of sTAg in vivo in a panel of transgenic mouse models. Epithelia of preterm sTAg-expressing embryos exhibited hyperplasia, impaired differentiation, increased proliferation, and apoptosis, and activation of a DNA damage response. Epithelial transformation did not require sTAg interaction with the protein phosphatase 2A protein complex, a tumor suppressor in some other polyomavirus transformation models, but was strictly dependent on a recently described sTAg domain that binds Fbxw7, the substrate-binding component of the Skp1/Cullin1/F-box protein ubiquitin ligase complex. Postnatal induction of sTAg using a Cre-inducible transgene also led to epithelial transformation with development of lesions resembling squamous cell carcinoma in situ and elevated expression of Fbxw7 target proteins. Our data establish that expression of MCPyV sTAg alone is sufficient for rapid neoplastic transformation in vivo, implicating sTAg as an oncogenic driver in MCC and perhaps other human malignancies. Moreover, the loss of transforming activity following mutation of the sTAg Fbxw7 binding domain identifies this domain as crucial for in vivo transformation.


Assuntos
Antígenos Virais de Tumores/fisiologia , Carcinogênese/patologia , Carcinoma de Célula de Merkel/fisiopatologia , Poliomavírus das Células de Merkel/imunologia , Infecções por Polyomavirus/imunologia , Neoplasias Cutâneas/fisiopatologia , Infecções Tumorais por Vírus/imunologia , Animais , Antígenos Virais de Tumores/imunologia , Apoptose/fisiologia , Carcinogênese/imunologia , Carcinoma de Célula de Merkel/imunologia , Carcinoma de Célula de Merkel/patologia , Proliferação de Células/fisiologia , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/patologia , Dano ao DNA/fisiologia , Modelos Animais de Doenças , Células de Merkel/imunologia , Células de Merkel/patologia , Camundongos , Camundongos Transgênicos , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia
7.
EMBO Mol Med ; 4(3): 218-33, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22294553

RESUMO

Inhibition of Hedgehog (HH)/GLI signalling in cancer is a promising therapeutic approach. Interactions between HH/GLI and other oncogenic pathways affect the strength and tumourigenicity of HH/GLI. Cooperation of HH/GLI with epidermal growth factor receptor (EGFR) signalling promotes transformation and cancer cell proliferation in vitro. However, the in vivo relevance of HH-EGFR signal integration and the critical downstream mediators are largely undefined. In this report we show that genetic and pharmacologic inhibition of EGFR signalling reduces tumour growth in mouse models of HH/GLI driven basal cell carcinoma (BCC). We describe HH-EGFR cooperation response genes including SOX2, SOX9, JUN, CXCR4 and FGF19 that are synergistically activated by HH-EGFR signal integration and required for in vivo growth of BCC cells and tumour-initiating pancreatic cancer cells. The data validate EGFR signalling as drug target in HH/GLI driven cancers and shed light on the molecular processes controlled by HH-EGFR signal cooperation, providing new therapeutic strategies based on combined targeting of HH-EGFR signalling and selected downstream target genes.


Assuntos
Carcinoma Basocelular/metabolismo , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas Hedgehog/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Carcinoma Basocelular/genética , Carcinoma Basocelular/patologia , Linhagem Celular Tumoral , Receptores ErbB/genética , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Hedgehog/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Fenótipo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Carga Tumoral , Proteína GLI1 em Dedos de Zinco
8.
J Clin Invest ; 121(5): 1768-81, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21519145

RESUMO

Uncontrolled Hedgehog (Hh) signaling leads to the development of basal cell carcinoma (BCC), the most common human cancer, but the cell of origin for BCC is unclear. While Hh pathway dysregulation is common to essentially all BCCs, there exist multiple histological subtypes, including superficial and nodular variants, raising the possibility that morphologically distinct BCCs may arise from different cellular compartments in skin. Here we have shown that induction of a major mediator of Hh signaling, GLI2 activator (GLI2ΔN), selectively in stem cells of resting hair follicles in mice, induced nodular BCC development from a small subset of cells in the lower bulge and secondary hair germ compartments. Tumorigenesis was markedly accelerated when GLI2ΔN was induced in growing hair follicles. In contrast, induction of GLI2ΔN in epidermis led to the formation of superficial BCCs. Expression of GLI2ΔN at reduced levels in mice yielded lesions resembling basaloid follicular hamartomas, which have previously been linked to low-level Hh signaling in both mice and humans. Our data show that the cell of origin, tissue context (quiescent versus growing hair follicles), and level of oncogenic signaling can determine the phenotype of Hh/Gli-driven skin tumors, with high-level signaling required for development of superficial BCC-like tumors from interfollicular epidermis and nodular BCC-like tumors from hair follicle stem cells.


Assuntos
Carcinoma Basocelular/metabolismo , Células Epiteliais/citologia , Folículo Piloso/metabolismo , Neoplasias Cutâneas/metabolismo , Células-Tronco/citologia , Alelos , Animais , Epiderme/metabolismo , Hamartoma/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Hiperplasia , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Estrutura Terciária de Proteína , Transdução de Sinais , Proteína Gli2 com Dedos de Zinco
9.
Nat Med ; 15(9): 1055-61, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19701205

RESUMO

Primary cilia are present on most mammalian cells and are implicated in transducing Hedgehog (Hh) signals during development; however, the prevalence of cilia on human tumors remains unclear, and the role of cilia in cancer has not been examined. Here we show that human basal cell carcinomas (BCCs) are frequently ciliated, and we test the role of cilia in BCC by conditionally deleting Kif3a (encoding kinesin family member 3A) or Ift88 (encoding intraflagellar transport protein 88), genes required for ciliogenesis, in two Hh pathway-dependent mouse tumor models. Ciliary ablation strongly inhibited BCC-like tumors induced by an activated form of Smoothened. In contrast, removal of cilia accelerated tumors induced by activated Gli2, a transcriptional effector of Hh signaling. These seemingly paradoxical effects are consistent with a dual role for cilia in mediating both the activation and the repression of the Hh signaling pathway. Our findings demonstrate that cilia function as unique signaling organelles that can either mediate or suppress tumorigenesis depending on the nature of the oncogenic initiating event.


Assuntos
Carcinoma Basocelular/etiologia , Carcinoma Basocelular/fisiopatologia , Cílios/fisiologia , Proteínas Hedgehog/fisiologia , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/fisiopatologia , Animais , Carcinoma Basocelular/patologia , Cílios/patologia , Humanos , Cinesinas/deficiência , Cinesinas/genética , Cinesinas/fisiologia , Fatores de Transcrição Kruppel-Like/fisiologia , Camundongos , Camundongos Knockout , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais , Neoplasias Cutâneas/patologia , Receptor Smoothened , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia , Proteína Gli2 com Dedos de Zinco
10.
Neoplasia ; 10(12): 1343-9, 5p following 1349, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19048113

RESUMO

Inappropriate Hedgehog (Hh) signaling underlies development of a subset of medulloblastomas, and tumors with elevated HH signaling activity express the stem cell self-renewal gene BMI1. To test whether Bmi1 is required for Hh-driven medulloblastoma development, we varied Bmi1 gene dosage in transgenic mice expressing an oncogenic Hh effector, SmoA1, driven by a glial fibrillary acidic protein (GFAP) promoter. Whereas 100% of SmoA1; Bmi1(+/+) or SmoA1;Bmi1(+/-) mice examined between postnatal (P) days 14 and 26 had typical medulloblastomas (N = 29), tumors were not detected in any of the SmoA1;Bmi1(-/-) animals examined (N = 6). Instead, small ectopic collections of cells were present in the region of greatest tumor load in SmoA1 animals, suggesting that medulloblastomas were initiated but failed to undergo expansion into frank tumors. Cells within these Bmi1(-/-) lesions expressed SmoA1 but were largely nonproliferative, in contrast to cells in Bmi1(+/+) tumors (6.2% vs 81.9% PCNA-positive, respectively). Ectopic cells were negative for the progenitor marker nestin, strongly GFAP-positive, and highly apoptotic, relative to Bmi1(+/+) tumor cells (29.6% vs 6.3% TUNEL-positive). The alterations in proliferation and apoptosis in SmoA1;Bmi1(-/-) ectopic cells are associated with reduced levels of Cyclin D1 and elevated expression of cyclin-dependent kinase inhibitor p19(Arf), two inversely regulated downstream targets of Bmi1. These data provide the first demonstration that Bmi1 is required for spontaneous de novo development of a solid tumor arising in the brain, suggest a crucial role for Bmi1-dependent, nestin-expressing progenitor cells in medulloblastoma expansion, and implicate Bmi1 as a key factor required for Hh pathway-driven tumorigenesis.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas Hedgehog/metabolismo , Meduloblastoma/metabolismo , Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Repressoras/fisiologia , Animais , Neoplasias Encefálicas/metabolismo , Proliferação de Células , Cerebelo/metabolismo , Ciclina D1/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Genótipo , Proteína Glial Fibrilar Ácida , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 1 , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo
11.
Hum Mol Genet ; 14(15): 2181-8, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15994174

RESUMO

Zinc finger-containing Gli proteins mediate responsiveness to Hedgehog (Hh) signaling, with Gli2 acting as the major transcriptional activator in this pathway in mice. The discovery of disease-associated mutations points to a critical role for GLI2 in human Hh signaling as well. Here, we show that human GLI2 contains previously undescribed 5' sequence, extending the amino-terminus an additional 328 amino acids. In vitro, transcriptional activity of full-length GLI2 is up to 30 times lower than that of GLI2DeltaN (previously thought to represent the entire GLI2 protein), revealing the presence of an amino-terminal repressor domain in the full-length protein. GLI2DeltaN also exhibits potent transcriptional activity in vivo: overexpression in mouse skin leads to the formation of Hh-independent epithelial downgrowths resembling basal cell carcinomas, which in humans are associated with constitutive Hh signaling. The discovery of this additional, functionally relevant GLI2 sequence led us to re-examine several pathogenic human GLI2 mutants, now containing the entire amino-terminal domain. On the basis of the functional domains affected by the mutations, mutant GLI2 proteins exhibited either loss-of-function or dominant-negative activity. Moreover, deletion of the amino-terminus abrogated dominant-negative activity of mutant GLI2, revealing that this domain is required for transcriptional repressor activity of pathogenic GLI2. Our results establish the presence of an amino-terminal transcriptional repressor domain that plays a critical role in modulating the function of wild-type GLI2 and is essential for dominant-negative activity of a GLI2 mutant associated with human disease.


Assuntos
Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Nucleares/metabolismo , Ativação Transcricional , Animais , Células Cultivadas , DNA Complementar/isolamento & purificação , Feminino , Humanos , Hipopituitarismo/genética , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Transgênicos , Mutação , Proteínas Nucleares/genética , Linhagem , Polidactilia/genética , Estrutura Terciária de Proteína , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transativadores/genética , Transativadores/metabolismo , Proteína Gli2 com Dedos de Zinco , Dedos de Zinco
12.
Mol Cell Biochem ; 230(1-2): 139-47, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11952089

RESUMO

Cyclic GMP is essential for the ability of rods and cones to respond to the light stimuli. Light triggers hydrolysis of cGMP and stops the influx of sodium and calcium through the cGMP-gated ion channels. The consequence of this event is 2-fold: first, the decrease in the inward sodium current plays the major role in an abrupt hyperpolarization of the cellular membrane; secondly, the decrease in the Ca2+ influx diminishes the free intracellular Ca2+ concentration. While the former constitutes the essence of the phototransduction pathway in rods and cones, the latter gives rise to a potent feedback mechanism that accelerates photoreceptor recovery and adaptation to background light. One of the most important events by which Ca2+ feedback controls recovery and light adaptation is synthesis of cGMP by guanylyl cyclase. Two isozymes of membrane photoreceptor guanylyl cyclase (retGC) have been identified in rods and cones that are regulated by Ca2+-binding proteins, GCAPs. At low intracellular concentrations of Ca2+ typical for light-adapted rods and cones GCAPs activate RetGC, but concentrations above 500 nM typical for dark-adapted photoreceptors turn them into inhibitors of retGC. A variety of mutations found in GCAP and retGC genes have been linked to several forms of human congenital retinal diseases, such as dominant cone degeneration, cone-rod dystrophy and Leber congenital amaurosis.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , GMP Cíclico/biossíntese , Guanilato Ciclase/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Degeneração Retiniana/enzimologia , Degeneração Retiniana/genética , Animais , Proteínas de Ligação ao Cálcio/genética , Guanilato Ciclase/genética , Proteínas Ativadoras de Guanilato Ciclase , Humanos , Mutação , Células Fotorreceptoras de Vertebrados/enzimologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa