Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 43(7): 114347, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38941190

RESUMO

Our skin provides a protective barrier that shields us from our environment. Barrier function is typically associated with the interfollicular epidermis; however, whether hair follicles influence this process remains unclear. Here, we utilize a potent genetic tool to probe barrier function by conditionally ablating a quintessential epidermal barrier gene, Abca12, which is mutated in the most severe skin barrier disease, harlequin ichthyosis. With this tool, we deduced 4 ways by which hair follicles modulate skin barrier function. First, the upper hair follicle (uHF) forms a functioning barrier. Second, barrier disruption in the uHF elicits non-cell-autonomous responses in the epidermis. Third, deleting Abca12 in the uHF impairs desquamation and blocks sebum release. Finally, barrier perturbation causes uHF cells to move into the epidermis. Neutralizing IL-17a, whose expression is enriched in the uHF, partially alleviated some disease phenotypes. Altogether, our findings implicate hair follicles as multi-faceted regulators of skin barrier function.

2.
bioRxiv ; 2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38712094

RESUMO

Our skin provides a protective barrier that shields us from our environment. Barrier function is typically associated with interfollicular epidermis; however, whether hair follicles influence this process remains unclear. Here, we utilize a potent genetic tool to probe barrier function by conditionally ablating a quintessential epidermal barrier gene, Abca12, which is mutated in the most severe skin barrier disease, harlequin ichthyosis. With this tool, we deduced 4 ways by which hair follicles modulate skin barrier function. First, the upper hair follicle (uHF) forms a functioning barrier. Second, barrier disruption in the uHF elicits non-cell autonomous responses in the epidermis. Third, deleting Abca12 in the uHF impairs desquamation and blocks sebum release. Finally, barrier perturbation causes uHF cells to move into the epidermis. Neutralizing Il17a, whose expression is enriched in the uHF, partially alleviated some disease phenotypes. Altogether, our findings implicate hair follicles as multi-faceted regulators of skin barrier function.

3.
Cell Rep ; 42(9): 113121, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37715952

RESUMO

Sebaceous glands (SGs) release oils that protect our skin, but how these glands respond to injury has not been previously examined. Here, we report that SGs are largely self-renewed by dedicated stem cell pools during homeostasis. Using targeted single-cell RNA sequencing, we uncovered both direct and indirect paths by which resident SG progenitors ordinarily differentiate into sebocytes, including transit through a Krt5+PPARγ+ transitional basal cell state. Upon skin injury, however, SG progenitors depart their niche, reepithelialize the wound, and are replaced by hair-follicle-derived stem cells. Furthermore, following targeted genetic ablation of >99% of SGs from dorsal skin, these glands unexpectedly regenerate within weeks. This regenerative process is mediated by alternative stem cells originating from the hair follicle bulge, is dependent upon FGFR2 signaling, and can be accelerated by inducing hair growth. Altogether, our studies demonstrate that stem cell plasticity promotes SG durability following injury.


Assuntos
Glândulas Sebáceas , Pele , Diferenciação Celular , Folículo Piloso , Células Epiteliais
4.
bioRxiv ; 2023 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-37205445

RESUMO

Sebaceous glands (SGs) release oils that protect our skin, but how these glands respond to injury has not been previously examined. Here, we report that SGs are largely self-renewed by dedicated stem cell pools during homeostasis. Using targeted single cell RNA-sequencing, we uncovered both direct and indirect paths by which these resident SG progenitors ordinarily differentiate into sebocytes, including transit through a PPARγ+Krt5+ transitional cell state. Upon skin injury, however, SG progenitors depart their niche, reepithelialize the wound, and are replaced by hair follicle-derived stem cells. Furthermore, following targeted genetic ablation of >99% of SGs from dorsal skin, these glands unexpectedly regenerate within weeks. This regenerative process is mediated by alternative stem cells originating from the hair follicle bulge, is dependent upon FGFR signaling, and can be accelerated by inducing hair growth. Altogether, our studies demonstrate that stem cell plasticity promotes SG durability following injury.

5.
Cell Rep ; 39(5): 110779, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35508126

RESUMO

Basal cell carcinomas (BCCs) frequently possess immense mutational burdens; however, the functional significance of most of these mutations remains unclear. Here, we report that loss of Ptch1, the most common mutation that activates upstream Hedgehog (Hh) signaling, initiates the formation of nascent BCC-like tumors that eventually enter into a dormant state. However, rare tumors that overcome dormancy acquire the ability to hyperactivate downstream Hh signaling through a variety of mechanisms, including amplification of Gli1/2 and upregulation of Mycn. Furthermore, we demonstrate that MYCN overexpression promotes the progression of tumors induced by loss of Ptch1. These findings suggest that canonical mutations that activate upstream Hh signaling are necessary, but not sufficient, for BCC to fully progress. Rather, tumors likely acquire secondary mutations that further hyperactivate downstream Hh signaling in order to escape dormancy and enter a trajectory of uncontrolled expansion.


Assuntos
Carcinoma Basocelular , Neoplasias Cutâneas , Carcinoma Basocelular/genética , Carcinoma Basocelular/patologia , Proteínas Hedgehog/genética , Humanos , Mutação/genética , Proteína Proto-Oncogênica N-Myc/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Proteína GLI1 em Dedos de Zinco/genética
6.
Dev Cell ; 51(3): 326-340.e4, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31564613

RESUMO

Oil-secreting sebaceous glands (SGs) are critical for proper skin function; however, it remains unclear how different factors act together to modulate SG stem cells. Here, we provide functional evidence that each SG lobe is serviced by its own dedicated stem cell population. Upon ablating Notch signaling in different skin subcompartments, we find that this pathway exerts dual counteracting effects on SGs. Suppressing Notch in SG progenitors traps them in a hybrid state where stem and differentiation features become intermingled. In contrast, ablating Notch outside of the SG stem cell compartment indirectly drives SG expansion. Finally, we report that a K14:K5→K14:K79 keratin shift occurs during SG differentiation. Deleting K79 destabilizes K14 in sebocytes, and attenuates SGs and eyelid meibomian glands, leading to corneal ulceration. Altogether, our findings demonstrate that SGs integrate diverse signals from different niches and suggest that mutations incurred within one stem cell compartment can indirectly influence another.


Assuntos
Glândulas Sebáceas/citologia , Pele/citologia , Nicho de Células-Tronco , Células-Tronco/citologia , Animais , Diferenciação Celular , Feminino , Proteínas Hedgehog/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Queratinas/metabolismo , Masculino , Glândulas Tarsais/metabolismo , Camundongos Knockout , Mutação/genética , Receptores Notch/genética
7.
Exp Dermatol ; 28(4): 345-349, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30033638

RESUMO

The uppermost aspect of the hair follicle, known as the infundibulum or hair canal, provides a passageway for hair shaft egress and sebum secretion. Recent studies have indicated that the infundibulum and sebaceous ducts are lined by molecularly distinct differentiated cells expressing markers including Keratin 79 and Gata6. Here, we ablated Gata6 from the skin and observed dilation of both the hair canal and sebaceous ducts, independent of gender and hair cycle stage. Constitutive loss of Gata6 yielded only a mild delay in depilation-induced entry into anagen, while unperturbed mutant mice possessed overtly normal skin and hair. Furthermore, we noted that Keratin 79 and Gata6 expression and localization did not depend upon each other. Our findings implicate Gata6 in maintaining the upper hair follicle and suggest that regulation of this transcription factor may be compromised in pathologies such as acne or infundibular cystic diseases that are characterized by abnormal expansion of this follicular domain.


Assuntos
Fator de Transcrição GATA6/genética , Folículo Piloso/patologia , Glândulas Sebáceas/patologia , Animais , Núcleo Celular/metabolismo , Dilatação Patológica/genética , Feminino , Fator de Transcrição GATA6/metabolismo , Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/metabolismo , Queratinas/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Regeneração/genética , Glândulas Sebáceas/metabolismo
8.
Cell Rep ; 19(4): 809-821, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28445731

RESUMO

During development and regeneration, matrix progenitors undergo terminal differentiation to form the concentric layers of the hair follicle. These differentiation events are thought to require signals from the mesenchymal dermal papilla (DP); however, it remains unclear how DP-progenitor cell interactions govern specific cell fate decisions. Here, we show that the hair follicle differentiated layers are specified asynchronously, with early matrix progenitors initiating differentiation before surrounding the DP. Furthermore, these early matrix cells can undergo terminal differentiation in the absence of Shh, BMP signaling, and DP maturation. Whereas early matrix progenitors form the hair follicle companion layer, later matrix populations progressively form the inner root sheath and hair shaft. Altogether, our findings characterize some of the earliest terminal differentiation events in the hair follicle and reveal that the matrix progenitor pool can be divided into early and late phases based on distinct temporal, molecular, and functional characteristics.


Assuntos
Diferenciação Celular/fisiologia , Folículo Piloso/citologia , Células-Tronco Mesenquimais/citologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Derme/citologia , Fator de Transcrição GATA3/metabolismo , Folículo Piloso/metabolismo , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Queratina-6/metabolismo , Queratinas/genética , Queratinas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
9.
Cell Stem Cell ; 16(4): 400-12, 2015 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-25842978

RESUMO

Basal cell carcinoma (BCC) is characterized by frequent loss of PTCH1, leading to constitutive activation of the Hedgehog pathway. Although the requirement for Hedgehog in BCC is well established, the identity of disease-initiating cells and the compartments in which they reside remain controversial. By using several inducible Cre drivers to delete Ptch1 in different cell compartments in mice, we show here that multiple hair follicle stem cell populations readily develop BCC-like tumors. In contrast, stem cells within the interfollicular epidermis do not efficiently form tumors. Notably, we observed that innervated Gli1-expressing progenitors within mechanosensory touch dome epithelia are highly tumorigenic. Sensory nerves activate Hedgehog signaling in normal touch domes, while denervation attenuates touch dome-derived tumors. Together, our studies identify varying tumor susceptibilities among different stem cell populations in the skin, highlight touch dome epithelia as "hot spots" for tumor formation, and implicate cutaneous nerves as mediators of tumorigenesis.


Assuntos
Carcinoma Basocelular/metabolismo , Células Epiteliais/fisiologia , Folículo Piloso/fisiologia , Células de Merkel/fisiologia , Nervo Musculocutâneo/fisiologia , Células-Tronco Pluripotentes/fisiologia , Receptores de Superfície Celular/metabolismo , Neoplasias Cutâneas/metabolismo , Animais , Carcinogênese/genética , Carcinoma Basocelular/patologia , Denervação , Folículo Piloso/inervação , Proteínas Hedgehog/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Mecanorreceptores/metabolismo , Mecanotransdução Celular/genética , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Camundongos Transgênicos , Nervo Musculocutâneo/cirurgia , Receptores Patched , Receptor Patched-1 , Receptores de Superfície Celular/genética , Neoplasias Cutâneas/patologia , Proteína GLI1 em Dedos de Zinco
10.
Development ; 140(24): 4870-80, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24198274

RESUMO

The formation of epithelial tubes underlies the development of diverse organs. In the skin, hair follicles resemble tube-like structures with lumens that are generated through poorly understood cellular rearrangements. Here, we show that creation of the hair follicle lumen is mediated by early outward movement of keratinocytes from within the cores of developing hair buds. These migratory keratinocytes express keratin 79 (K79) and stream out of the hair germ and into the epidermis prior to lumen formation in the embryo. Remarkably, this process is recapitulated during hair regeneration in the adult mouse, when K79(+) cells migrate out of the reactivated secondary hair germ prior to formation of a new hair canal. During homeostasis, K79(+) cells line the hair follicle infundibulum, a domain we show to be multilayered, biochemically distinct and maintained by Lrig1(+) stem cell-derived progeny. Upward movement of these cells sustains the infundibulum, while perturbation of this domain during acne progression is often accompanied by loss of K79. Our findings uncover previously unappreciated long-distance cell movements throughout the life cycle of the hair follicle, and suggest a novel mechanism by which the follicle generates its hollow core through outward cell migration.


Assuntos
Acne Vulgar/metabolismo , Folículo Piloso/embriologia , Queratinócitos/metabolismo , Queratinas/metabolismo , Regeneração , Animais , Linhagem Celular , Movimento Celular , Células HEK293 , Cabelo/embriologia , Folículo Piloso/metabolismo , Humanos , Queratinas/genética , Queratinas Específicas do Cabelo , Queratinas Tipo II , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Morfogênese , Proteínas do Tecido Nervoso/metabolismo
11.
Gastroenterology ; 144(7): 1466-77, 1477.e1-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23395646

RESUMO

BACKGROUND & AIMS: ZBP-89 (also ZNF148 or Zfp148) is a butyrate-inducible zinc finger transcription factor that binds to GC-rich DNA elements. Deletion of the N-terminal domain is sufficient to increase mucosal susceptibility to chemical injury and inflammation. We investigated whether conditional deletion of ZBP-89 from the intestinal and colonic epithelium of mice increases their susceptibility to pathogens such as Salmonella typhimurium. METHODS: We generated mice with a conditional null allele of Zfp148 (ZBP-89(FL/FL)) using homologous recombination to flank Zfp148 with LoxP sites (ZBP-89(FL/FL)), and then bred the resulting mice with those that express VillinCre. We used microarray analysis to compare gene expression patterns in colonic mucosa between ZBP-89(ΔInt) and C57BL/6 wild-type mice (controls). Mice were gavaged with 2 isogenic strains of S. typhimurium after administration of streptomycin. RESULTS: Microarray analysis revealed that the colonic mucosa of ZBP-89(ΔInt) mice had reduced levels of tryptophan hydroxylase 1 (Tph1) messenger RNA, encoding the rate-limiting enzyme in enterochromaffin cell serotonin (5-hydroxytryptamine [5HT]) biosynthesis. DNA affinity precipitation demonstrated direct binding of ZBP-89 to the mouse Tph1 promoter, which was required for its basal and butyrate-inducible expression. ZBP-89(ΔInt) mice did not increase mucosal levels of 5HT in response to S. typhimurium infection, and succumbed to the infection 2 days before control mice. The ΔhilA isogenic mutant of S. typhimurium lacks this butyrate-regulated locus and stimulated, rather than suppressed, expression of Tph1 approximately 50-fold in control, but not ZBP-89(ΔInt), mice, correlating with fecal levels of butyrate. CONCLUSIONS: ZBP-89 is required for butyrate-induced expression of the Tph1 gene and subsequent production of 5HT in response to bacterial infection in mice. Reductions in epithelial ZBP-89 increase susceptibility to colitis and sepsis after infection with S. typhimurium, partly because of reduced induction of 5HT production in response to butyrate and decreased secretion of antimicrobial peptides.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Mucosa Intestinal/imunologia , RNA Mensageiro/análise , Infecções por Salmonella/imunologia , Serotonina/biossíntese , Fatores de Transcrição/fisiologia , Triptofano Hidroxilase/fisiologia , Animais , Butiratos/imunologia , Colite/imunologia , Proteínas de Ligação a DNA/genética , Células Enterocromafins/imunologia , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Salmonella typhimurium , Serotonina/imunologia , Fatores de Transcrição/genética
12.
PLoS One ; 7(10): e48039, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23110168

RESUMO

Chronic inflammation in the stomach can lead to gastric cancer. We previously reported that gastrin-deficient (Gast⁻/⁻) mice develop bacterial overgrowth, inflammatory infiltrate, increased Il-1ß expression, antral hyperplasia and eventually antral tumors. Since Hedgehog (Hh) signaling is active in gastric cancers but its role in precursor lesions is poorly understood, we examined the role of inflammation and Hh signaling in antral hyperplasia. LacZ reporter mice for Sonic hedgehog (Shh), Gli1, and Gli2 expression bred onto the Gast⁻/⁻ background revealed reduced Shh and Gli1 expression in the antra compared to wild type controls (WT). Gli2 expression in the Gast⁻/⁻ corpus was unchanged. However in the hyperplastic Gast⁻/⁻ antra, Gli2 expression increased in both the mesenchyme and epithelium, whereas expression in WT mice remained exclusively mesenchymal. These observations suggested that Gli2 is differentially regulated in the hyperplastic Gast⁻/⁻ antrum versus the corpus and by a Shh ligand-independent mechanism. Moreover, the proinflammatory cytokines Il-1ß and Il-11, which promote gastric epithelial proliferation, were increased in the Gast⁻/⁻ stomach along with Infγ. To test if inflammation could account for elevated epithelial Gli2 expression in the Gast⁻/⁻ antra, the human gastric cell line AGS was treated with IL-1ß and was found to increase GLI2 but decrease GLI1 levels. IL-1ß also repressed human GAST gene expression. Indeed, GLI2 but not GLI1 or GLI3 expression repressed gastrin luciferase reporter activity by ∼50 percent. Moreover, chromatin immunoprecipitation of GLI2 in AGS cells confirmed that GLI2 directly binds to the GAST promoter. Using a mouse model of constitutively active epithelial GLI2 expression, we found that activated GLI2 repressed Gast expression but induced Il-1ß gene expression and proliferation in the gastric antrum, along with a reduction of the number of G-cells. In summary, epithelial Gli2 expression was sufficient to stimulate Il-1ß expression, repress Gast gene expression and increase proliferation, leading to antral hyperplasia.


Assuntos
Gastrinas/genética , Regulação da Expressão Gênica , Inflamação/genética , Fatores de Transcrição Kruppel-Like/genética , Antro Pilórico/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Epitélio/metabolismo , Feminino , Gastrinas/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Hiperplasia , Imuno-Histoquímica , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Antro Pilórico/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Proteína Gli2 com Dedos de Zinco
13.
Am J Physiol Gastrointest Liver Physiol ; 303(12): G1384-92, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23086919

RESUMO

The gut microbiota is essential for the maintenance of intestinal immune homeostasis and is responsible for breaking down dietary fiber into short-chain fatty acids (SCFAs). Butyrate, the most abundant bioactive SCFA in the gut, is a histone deacetylase inhibitor (HDACi), a class of drug that has potent immunomodulatory properties. This characteristic of butyrate, along with our previous discovery that conventional dendritic cells (DCs) are required for the development of experimental colitis, led us to speculate that butyrate may modulate DC function to regulate gut mucosal homeostasis. We found that butyrate, in addition to suppressing LPS-induced bone marrow-derived DC maturation and inhibiting DC IL-12 production, significantly induced IL-23 expression. The upregulation of mRNA subunit IL-23p19 at the pretranslational level was consistent with the role of HDACi on the epigenetic modification of gene expression. Furthermore, the mechanism of IL-23p19 upregulation was independent of Stat3 and ZBP89. Coculture of splenocytes with LPS-stimulated DCs pretreated with or without butyrate was performed and showed a significant induction of IL-17 and IL-10. We demonstrated further the effect of butyrate in vivo using dextran sulfate sodium (DSS)-induced colitis and found that the addition of butyrate in the drinking water of mice worsened DSS-colitis. This is in contrast to the daily intraperitoneal butyrate injection of DSS-treated mice, which mildly improved disease severity. Our study highlights a novel effect of butyrate in upregulating IL-23 production of activated DCs and demonstrates a difference in the host response to the oral vs. systemic route of butyrate administration.


Assuntos
Butiratos/farmacologia , Colite/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Interleucina-23/metabolismo , Animais , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL
14.
Am J Physiol Gastrointest Liver Physiol ; 303(6): G752-64, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22766853

RESUMO

Antral gastrin is the hormone known to stimulate acid secretion and proliferation of the gastric corpus epithelium. Patients with mutations in the multiple endocrine neoplasia type 1 (MEN1) locus, which encodes the protein menin, develop pituitary hyperplasia, insulinomas, and gastrinomas in the duodenum. We previously hypothesized that loss of menin leads to derepression of the gastrin gene and hypergastrinemia. Indeed, we show that menin represses JunD induction of gastrin in vitro. Therefore, we examined whether conditional deletion of Men1 (Villin-Cre and Lgr5-EGFP-IRES-CreERT2), with subsequent loss of menin from the gastrointestinal epithelium, increases gastrin expression. We found that epithelium-specific deletion of Men1 using Villin-Cre increased plasma gastrin, antral G cell numbers, and gastrin expression in the antrum, but not the duodenum. Moreover, the mice were hypochlorhydric by 12 mo of age, and gastric somatostatin mRNA levels were reduced. However, duodenal mRNA levels of the cyclin-dependent kinase inhibitor p27(Kip1) were decreased, and cell proliferation determined by Ki67 staining was increased. About 11% of the menin-deficient mice developed antral tumors that were negative for gastrin; however, gastrinomas were not observed, even at 12 mo of age. No gastrinomas were observed with conditional deletion of Men1 in the Lgr5 stem cells 5 mo after Cre induction. In summary, epithelium-specific deletion of the Men1 locus resulted in hypergastrinemia due to antral G cell hyperplasia and a hyperproliferative epithelium, but no gastrinomas. This result suggests that additional mutations in gene targets other than the Men1 locus are required to produce gastrin-secreting tumors.


Assuntos
Células Secretoras de Gastrina/fisiologia , Hiperplasia/patologia , Proteínas Proto-Oncogênicas/metabolismo , Antro Pilórico/citologia , Animais , Trato Gastrointestinal/metabolismo , Deleção de Genes , Regulação da Expressão Gênica/fisiologia , Genótipo , Hiperplasia/genética , Hiperplasia/metabolismo , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética , Somatostatina/metabolismo
15.
Nature ; 482(7386): 542-6, 2012 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-22327296

RESUMO

Menin is a tumour suppressor protein whose loss or inactivation causes multiple endocrine neoplasia 1 (MEN1), a hereditary autosomal dominant tumour syndrome that is characterized by tumorigenesis in multiple endocrine organs. Menin interacts with many proteins and is involved in a variety of cellular processes. Menin binds the JUN family transcription factor JUND and inhibits its transcriptional activity. Several MEN1 missense mutations disrupt the menin-JUND interaction, suggesting a correlation between the tumour-suppressor function of menin and its suppression of JUND-activated transcription. Menin also interacts with mixed lineage leukaemia protein 1 (MLL1), a histone H3 lysine 4 methyltransferase, and functions as an oncogenic cofactor to upregulate gene transcription and promote MLL1-fusion-protein-induced leukaemogenesis. A recent report on the tethering of MLL1 to chromatin binding factor lens epithelium-derived growth factor (LEDGF) by menin indicates that menin is a molecular adaptor coordinating the functions of multiple proteins. Despite its importance, how menin interacts with many distinct partners and regulates their functions remains poorly understood. Here we present the crystal structures of human menin in its free form and in complexes with MLL1 or with JUND, or with an MLL1-LEDGF heterodimer. These structures show that menin contains a deep pocket that binds short peptides of MLL1 or JUND in the same manner, but that it can have opposite effects on transcription. The menin-JUND interaction blocks JUN N-terminal kinase (JNK)-mediated JUND phosphorylation and suppresses JUND-induced transcription. In contrast, menin promotes gene transcription by binding the transcription activator MLL1 through the peptide pocket while still interacting with the chromatin-anchoring protein LEDGF at a distinct surface formed by both menin and MLL1.


Assuntos
Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Transcrição Gênica , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cromatina/metabolismo , Cristalografia por Raios X , Fibroblastos , Células HEK293 , Histona-Lisina N-Metiltransferase , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Proteína de Leucina Linfoide-Mieloide/química , Fosforilação , Ligação Proteica , Multimerização Proteica , Proteínas Proto-Oncogênicas c-jun/química , Relação Estrutura-Atividade
16.
Gene ; 487(2): 113-7, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21855615

RESUMO

Many genes of small RNAs and short interspersed elements (SINEs) are transcribed by RNA polymerase III due to an internal promoter that is composed of two boxes (A and B) spaced by 30-45bp. Rodent SINE B1 originated from 7SL RNA, and a 29-bp tandem duplication took place in B1 at an early stage of its evolution. As a result of this duplication, an additional box B (named B') located at a distance of 79-82bp from box A arose in SINE B1. Here we have shown that despite the unusually large distance between boxes A and B', they can form an active promoter. In chinchillas, guinea pigs, and other rodents belonging to clade Ctenohystrica, structure of the B' box was well preserved and closely resembles the canonical B box. One may suggest therefore, that box B' can functionally replace box B in those copies of B1 where the latter has lost activity due to mutations.


Assuntos
Regiões Promotoras Genéticas/fisiologia , RNA Polimerase III/genética , Elementos Nucleotídeos Curtos e Dispersos/fisiologia , Animais , Sequência de Bases , Chinchila , Mapeamento Cromossômico , Cobaias , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Homologia de Sequência do Ácido Nucleico , Elementos Nucleotídeos Curtos e Dispersos/genética
17.
Am J Physiol Gastrointest Liver Physiol ; 301(5): G783-90, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21852362

RESUMO

Mutations in the MEN1 gene correlate with multiple endocrine neoplasia I (MEN1). Gastrinomas are the most malignant of the neuroendocrine tumors associated with MEN1. Because menin and JunD proteins interact, we examined whether JunD binds to and regulates the gastrin gene promoter. Both menin and JunD are ubiquitous nuclear proteins that we showed colocalize in the gastrin-expressing G cells of the mouse antrum. Transfection with a JunD expression vector alone induced endogenous gastrin mRNA in AGS human gastric cells, and the induction was blocked by menin overexpression. We mapped repression by menin to both a nonconsensus AP-1 site and proximal GC-rich elements within the human gastrin promoter. Chromatin immunoprecipitation assays, EMSAs, and DNA affinity precipitation assays documented that JunD and Sp1 proteins bind these two elements and are both targets for menin regulation. Consistent with menin forming a complex with histone deacetylases, we found that repression of gastrin gene expression by menin was reversed by trichostatin A. In conclusion, proximal DNA elements within the human gastrin gene promoter mediate interactions between JunD, which induces gastrin gene expression and menin, which suppresses JunD-mediated activation.


Assuntos
DNA/genética , Gastrinas/genética , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas/genética , Células Cultivadas , DNA/metabolismo , Gastrinas/metabolismo , Regulação da Expressão Gênica , Humanos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo
18.
Gene ; 460(1-2): 30-8, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20399257

RESUMO

Until recently, zokors (Myospalacinae) were assigned to the Cricetidae family. However, analysis of mitochondrial and nuclear genes suggests a sister relationship between zokors and subterranean rodents of the Spalacidae family, namely blind mole rats (Spalacinae) and bamboo rats (Rhizomyinae). Here, we cloned and sequenced copies of the B1 short interspersed element (SINE) from the genome of zokor Myospalax psilurus. The consensus nucleotide sequence of zokor B1 was very similar to spalacids and rhizomyids, but not cricetids. Similar to spalacids (Spalax microphthalmus) and rhizomyids (Tachyoryctes splendens), zokor contained two variants of the 4.5S(I) small nuclear RNA. The longer variant (L-variant, 104 nucleotides) was found only in zokor, spalacids and rhizomyids. The short, or S-variant (98 nucleotides), had a wider distribution; however, analysis of the nucleotide sequences of S-variants of 4.5S(I) RNA confirmed that zokors are closely related to spalacids and rhizomyids, but not to cricetids. The evolution of the 4.5S(I) RNA genes and pseudogenes is discussed.


Assuntos
Evolução Molecular , Ratos-Toupeira/genética , RNA Nuclear Pequeno , Ratos/genética , Animais , Sequência Consenso , Dados de Sequência Molecular , Filogenia , Retroelementos , Alinhamento de Sequência , Elementos Nucleotídeos Curtos e Dispersos
19.
Genomics ; 89(6): 678-86, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17433864

RESUMO

B1 SINEs were studied in 22 families covering all major rodent lineages. The number of B1 copies considerably varies, from 1 x 10(4) in Geomyidae to 1 x 10(6) in Myodonta. B1 sequences can be divided into three main structural variants: B1 with a 20-bp tandem duplication (found in Gliridae, Sciuridae, and Aplodontidae), B1 with a 29-bp duplication (found in other families), and proto-B1 without duplication (pB1). These variants can be further subdivided according to their characters, including specific 7-, 9-, or 10-bp deletions. Different B1 subfamilies predominate in different rodent families. The analysis of B1 variants allowed us to propose possible pathways for the evolution of this SINE in the context of rodent evolution.


Assuntos
Roedores/genética , Elementos Nucleotídeos Curtos e Dispersos , Animais , Sequência de Bases , Evolução Molecular , Modelos Genéticos , Dados de Sequência Molecular , Hibridização de Ácido Nucleico , Roedores/classificação , Deleção de Sequência , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...