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










Base de dados
Intervalo de ano de publicação
1.
Birth Defects Res A Clin Mol Teratol ; 103(7): 573-82, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26178632

RESUMO

BACKGROUND: Congenital cytomegalovirus infection is the major nongenetic cause of sensorineural hearing loss at birth and beyond. Among other pathologies, there is a striking dysplasia/hyperplasia of organ of Corti hair and supporting cells. METHODS: Using an in vitro embryonic mouse model of cytomegalovirus-induced cochlear teratogenesis that mimics the known human pathology, and functional signaling network modeling, we tested the hypothesis that cytomegalovirus disrupts the highly ordered organ of Corti hair and supporting cells pattern by dysregulating Notch and Fgfr3, their cognate ligands and downstream effectors. RESULTS: Several novel emergent properties of the critical lateral inhibition subnetwork became apparent. The subnetwork has classic small-world properties such as short paths between most gene pairs, few long-distance links, and considerable clustering. Concomitantly, the calculated probability that our specific gene expression dataset is from dysplastic organs of Corti is highly significant (p < 1 × 10(-12) ). Furthermore, we determined that the subnetwork has a highly heterogeneous scale-free topology in which the highly linked genes (hubs), Notch and Fgfr3, play a central role in mediating interactions among the less linked genes. CONCLUSION: This phenomenon has important biologic and therapeutic implications.


Assuntos
Citomegalovirus/patogenicidade , Órgão Espiral/patologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
2.
Exp Mol Pathol ; 97(1): 154-65, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24984270

RESUMO

Mucoepidermoid carcinoma (MEC) is the most prevalent malignant tumor in major and minor salivary glands (SGs). We have recently identified human cytomegalovirus (hCMV) as a principle component in the multifactorial causation of SG-MEC. This finding is corroborated by the ability of the purified mouse CMV (mCMV) to induce malignant transformation of SG cells in a three-dimensional in vitro mouse model, using a similar oncogenic signaling pathway. Our prior studies indicate that the core tumor microenvironment (TME) is a key regulator of pathologic progression, particularly the cancer-associated fibroblast (CAF) component. Studies of early CAFs immunodetect aberrant expression of ECM components, as well as multiple growth factors, cytokines and transcription factors. Here we present the mechanistic insight derived from a mathematical structure ("wiring diagram") used to model complex relationships between a highly relevant (p=9.43×10(-12)) global "cancer network" of 32 genes and their known links. Detailed characterization of the functional architecture of the examined "cancer network" exposes the critical crosstalk and compensatory pathways that limit the efficacy of targeted anti-kinase therapies.


Assuntos
Citomegalovirus/patogenicidade , Regulação da Expressão Gênica , Modelos Teóricos , Glândulas Salivares/patologia , Glândulas Salivares/virologia , Animais , Carcinoma Mucoepidermoide/genética , Carcinoma Mucoepidermoide/patologia , Carcinoma Mucoepidermoide/virologia , Infecções por Citomegalovirus/genética , Epistasia Genética , Feminino , Gefitinibe , Redes Reguladoras de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Neoplasias das Glândulas Salivares/genética , Neoplasias das Glândulas Salivares/patologia , Neoplasias das Glândulas Salivares/virologia , Glândulas Salivares/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral
3.
Exp Mol Pathol ; 94(2): 386-97, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23399805

RESUMO

Mucoepidermoid carcinoma (MEC) is the most common malignant tumor originating in major and minor salivary glands (SGs). Although the precise multifactorial etiology of human SG-MEC is largely unknown, we have recently shown that cytomegalovirus (CMV) is an important component of MEC tumorigenesis. Despite the well-documented overexpression of the EGFR → ERK signaling pathway in SG-MEC, there has been limited to no clinical success with inhibition of this pathway. Using our previously characterized mouse model of CMV-induced SG dysplasia/neoplasia, we report that inhibitors of the EGFR → ERK pathway do not ameliorate or rescue well-established pathology, either singly or in combination, but they do inhibit the evolution of progressive pathogenesis ("disease tolerance") in the face of mounting CMV burden. Failure to rescue SG pathology, suggested a possible increase in the ligand levels of alternative pathways that share cell proliferation and survival effectors (e.g. ERK and PI3K). Here we present evidence of a highly significant upregulation of ligands for the EGFR, FGFR, IL-6R, and TNFR signaling pathways, all of which converge upon the Raf/MEK/ERK amplifier module. This explains our finding that even in the presence of the highest nontoxic dose of an ERK phosphorylation inhibitor, pERK is undiminished. Given the considerable pathway crosstalk, a deep understanding of subversion and dysregulation of the SG interactome by CMV is a priori quite daunting. Circumventing this dilemma, we present evidence that concurrent inhibition of ERK phosphorylation (U0126) and CMV replication (acyclovir) obviates progressive pathogenesis and results in complete SG rescue (tumor regression). These findings provide a mechanistic foundation for potential clinical trials that utilize similar concurrent treatment with extant FDA-approved drugs.


Assuntos
Carcinoma Mucoepidermoide/virologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Infecções por Herpesviridae/patologia , Muromegalovirus/fisiologia , Neoplasias da Glândula Submandibular/virologia , Glândula Submandibular/patologia , Glândula Submandibular/virologia , Aciclovir/farmacologia , Anfirregulina , Animais , Carcinoma Mucoepidermoide/metabolismo , Carcinoma Mucoepidermoide/patologia , Efeito Citopatogênico Viral , Modelos Animais de Doenças , Família de Proteínas EGF , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Feminino , Fator 8 de Crescimento de Fibroblasto/metabolismo , Glicoproteínas/metabolismo , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-6/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Muromegalovirus/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Fosforilação , Glândula Submandibular/metabolismo , Neoplasias da Glândula Submandibular/metabolismo , Neoplasias da Glândula Submandibular/patologia , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
4.
Birth Defects Res A Clin Mol Teratol ; 97(2): 69-78, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23281115

RESUMO

Congenital human cytomegalovirus (CMV) infection is the leading nongenetic etiology of sensorineural hearing loss (SNHL) at birth and prelingual SNHL not expressed at birth. The paucity of temporal bone autopsy specimens from infants with congenital CMV infection has hindered the critical correlation of histopathology with pathogenesis. Here, we present an in vitro embryonic mouse model of CMV-infected cochleas that mimics the human sites of viral infection and associated pathology. There is a striking dysplasia/hyperplasia in mouse CMV-infected cochlear epithelium and mesenchyme, including organ of Corti hair and supporting cells and stria vascularis. This is concomitant with significant dysregulation of p19, p21, p27, and Pcna gene expression, as well as proliferating cell nuclear antigen (PCNA) protein expression. Other pathologies similar to those arising from known deafness gene mutations include downregulation of KCNQ1 protein expression in the stria vascularis, as well as hypoplastic and dysmorphic melanocytes. Thus, this model provides a relevant and reliable platform within which the detailed cell and molecular biology of CMV-induced deafness may be studied.


Assuntos
Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/patologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/patologia , Camundongos , Órgão Espiral/anormalidades , Estria Vascular/patologia , Animais , Inibidor de Quinase Dependente de Ciclina p19/genética , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Modelos Animais de Doenças , Feto , Regulação da Expressão Gênica no Desenvolvimento , Perda Auditiva Neurossensorial/congênito , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Humanos , Lactente , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Melanócitos/metabolismo , Melanócitos/patologia , Muromegalovirus/fisiologia , Técnicas de Cultura de Órgãos , Órgão Espiral/metabolismo , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Estria Vascular/metabolismo
5.
Herpesviridae ; 4(1): 1, 2013 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-23342981

RESUMO

BACKGROUND: Recently we identified a relationship between human cytomegalovirus (hCMV) and human salivary gland (SG) mucoepidermoid carcinoma (MEC) in over 90% of cases; tumorigenesis in these cases uniformly correlated with active hCMV protein expression and an upregulation of the EGFR → ERK pathway. Our previously characterized, novel mouse organ culture model of mouse CMV (mCMV)-induced tumorigenesis displays a number of histologic and molecular characteristics similar to human MEC. METHODS: Newborn mouse submandibular glands (SMGs) were incubated with 1 × 105 PFU/ml of lacZ-tagged mCMV RM427+ on day 0 for 24 hours and then cultured in virus-free media for a total of 6 or 12 days with or without EGFR/ERK inhibitors and/or aciclovir. SMGs were collected for histology, immunolocalization (pERK, FN, IL-6), viral distribution, or Western blot analysis (pERK). RESULTS: Here we report: (1) mouse SMG tumors soon exhibit an acquired resistance to EGFR/ERK pathway kinase inhibitors, alone or in combination; (2) long term tumor regression can only be sustained by concurrent inhibitor and antiviral treatment; (3) CMV-dependent, kinase inhibitor resistance is associated with overexpression of fibronectin and IL-6 proteins in abnormal stromal cells. CONCLUSIONS: Acquired resistance to kinase inhibitors is dependent upon CMV dysregulation of alternative pathways with downstream effectors common with the targeted pathway, a phenomenon with important therapeutic implications for human MEC of salivary glands.

6.
Exp Mol Pathol ; 92(1): 118-25, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22101257

RESUMO

Human cytomegalovirus (hCMV) infection is common. Although still controversial, there is growing evidence that active hCMV infection is associated with a variety of malignancies, including brain, breast, lung, colon, and prostate. Given that hCMV is frequently resident in salivary gland (SG) ductal epithelium, we hypothesized that hCMV would be important to the pathogenesis of SG mucoepidermoid carcinoma (MEC). This was initially supported by our finding that purified CMV induces malignant transformation in SG cells in an in vitro mouse model, and utilizes a pathogenic pathway previously reported for human MEC. Here we present the histologic and molecular characterizations of 39 human SG MECs selected randomly from a repository of cases spanning 2004-2011. Serial sections were obtained from formalin-fixed, paraffin embedded, tissue blocks from previous incisional or excisional biopsies. Immunohistochemical assays were performed for active hCMV proteins (IE1 and pp65) and the activated COX/AREG/EGFR/ERK signaling pathway. All four prospective causal criteria for viruses and cancer are fully satisfied: (1) protein markers for active hCMV are present in 97% of MECs; (2) markers of active hCMV are absent in non-neoplastic SG tissues; (3) hCMV-specific proteins (IE1, pp65) are in specific cell types and expression is positively correlated with severity; (4) hCMV correlates and colocalizes with an upregulation and activation of an established oncogenic signaling pathway (COX/AREG/EGFR/ERK). Thus, the evidential support reported here and previously in a mouse model is strongly confirmatory of a causal relationship between hCMV and SG mucoepidermoid carcinoma. To our knowledge, this is the first demonstration of hCMV's role in human oncogenesis that fully responds to all of Koch's Postulates as revised for viruses and cancer. In the absence of any contrary evidence, hCMV can reasonably be designated an "oncovirus."


Assuntos
Carcinoma Mucoepidermoide/virologia , Infecções por Citomegalovirus/complicações , Citomegalovirus/metabolismo , Neoplasias das Glândulas Salivares/virologia , Proteínas Virais/análise , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Anfirregulina , Animais , Antígenos Virais/análise , Antígenos Virais/metabolismo , Carcinoma Mucoepidermoide/patologia , Ciclo-Oxigenase 2/análise , Ciclo-Oxigenase 2/metabolismo , Citomegalovirus/isolamento & purificação , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/patologia , Família de Proteínas EGF , Receptores ErbB/análise , Receptores ErbB/metabolismo , Feminino , Glicoproteínas/análise , Glicoproteínas/metabolismo , Humanos , Proteínas Imediatamente Precoces/análise , Proteínas Imediatamente Precoces/metabolismo , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/análise , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Pessoa de Meia-Idade , Fosfoproteínas/análise , Fosfoproteínas/metabolismo , Neoplasias das Glândulas Salivares/patologia , Glândulas Salivares/patologia , Glândulas Salivares/virologia , Proteínas da Matriz Viral/análise , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/metabolismo , Adulto Jovem
7.
Front Physiol ; 2: 59, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21941513

RESUMO

Twisted gastrulation (TWSG1) is a conserved, secreted glycoprotein that modulates signaling of bone morphogenetic proteins (BMPs) in the extracellular space. Deletion of exon 4 of mouse Twsg1 (mTwsg1) is associated with significant craniofacial defects. However, little is understood about the biochemical properties of the corresponding region of the protein. We have uncovered a significant role for exon 4 sequences as encoding the only two glycosylation sites of the mTWSG1 protein. Deletion of the entire exon 4 or mutation of both glycosylation sites within exon 4 abolishes glycosylation of mTWSG1. Importantly, we find that constructs with mutated glycosylation sites have significantly reduced BMP binding activity. We further show that glycosylation and activity of TWSG1 recombinant proteins vary markedly by cellular source. Non-glycosylated mTWSG1 made in E. coli has both reduced affinity for BMPs, as shown by surface plasmon resonance analysis, and reduced BMP inhibitory activity in a mandibular explant culture system compared to glycosylated proteins made in insect cells or murine myeloma cells. This study highlights an essential role for glycosylation in Twisted gastrulation action.

8.
Exp Mol Pathol ; 91(1): 400-10, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21565184

RESUMO

As with other herpesviruses, human cytomegalovirus (hCMV) has the ability to establish lifelong persistence and latent infection following primary exposure, salivary glands (SMGs) being the primary site of both. In the immunocompromised patient, hCMV is a common cause of opportunistic infections, and subsequent morbidity and mortality. Elucidating the molecular pathogenesis of CMV-induced disease is critical to the development of more effective and safer drug therapies. In the present study, we used a novel mouse postnatal SMG organ culture model of mCMV-induced dysplasia to investigate a candidate signaling network suggested by our prior studies (COX-2/AREG/EGFR/ERK). The objective was to employ small molecule inhibitors to target several key steps in the autocrine loop, and in this way ameliorate pathology. Our results indicate that upregulation of ERK phosphorylation is necessary for initial mCMV-induced pathogenesis, and that ErbB receptor family phosphorylation and downstream signaling are highly relevant targets for drug discovery.


Assuntos
Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Glicoproteínas/antagonistas & inibidores , Infecções por Herpesviridae/prevenção & controle , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Muromegalovirus/patogenicidade , Anfirregulina , Animais , Animais Recém-Nascidos , Western Blotting , Butadienos/farmacologia , Proliferação de Células , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , Diclofenaco/farmacologia , Modelos Animais de Doenças , Família de Proteínas EGF , Receptores ErbB/metabolismo , Feminino , Glicoproteínas/metabolismo , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Interações Hospedeiro-Patógeno , Técnicas Imunoenzimáticas , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Nitrilas/farmacologia , Técnicas de Cultura de Órgãos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Glândula Submandibular/virologia
9.
Gene Expr Patterns ; 11(1-2): 57-63, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20837164

RESUMO

Dysregulation of the transcription factor CRTC1 by a t(11;19) chromosomal rearrangement mediates the formation of mucoepidermoid salivary gland carcinoma (MEC). Although the CRTC1 promoter is consistently active in fusion-positive MEC and low levels of CRTC1 transcripts have been reported in normal adult salivary glands, the distribution of CRTC1 protein in the normal salivary gland is not known. The aim of this study was to determine if CRTC1, like many known oncogenes, is expressed during early submandibular salivary gland (SMG) development and re-expressed in an experimental tumor model. Our results indicate that CRTC1 protein is expressed in SMG epithelia during early stages of morphogenesis, disappears with differentiation, and reappears in initial tumor-like pathology. This stage-dependent expression pattern suggests that CRTC1 may play a role during embryonic SMG branching morphogenesis but not for pro-acinar/acinar differentiation, supporting a precursor cell origin for MEC tumorigenesis. Moreover, the coincident expression of CRTC1 protein and cell proliferation markers in tumor-like histopathology suggests that CRTC1-mediated cell proliferation may contribute, in part, to initial tumor formation.


Assuntos
Carcinoma Mucoepidermoide/genética , Carcinoma Mucoepidermoide/patologia , Neoplasias das Glândulas Salivares/genética , Neoplasias das Glândulas Salivares/patologia , Fatores de Transcrição/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos
10.
Cells Tissues Organs ; 192(4): 221-39, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20484882

RESUMO

Of the approximately 8,400 children born each year in the US with cytomegalovirus (CMV)-induced birth defects, more than one third exhibit hypoplasia and hypocalcification of tooth enamel. Our prior studies indicated that CMV severely delayed, but did not completely interrupt, early mouse mandibular first molar morphogenesis in vitro. The aim of the present study was to examine the effects of CMV infection on progressive tooth differentiation and amelogenesis. Since initial CMV infection in human fetuses can occur at different developmental times, we varied the stage of initial viral infection (that is, Cap stage, Early Bell stage and Bell stage), as well as the duration of infection. CMV infection of embryonic mouse mandibular first molars in vitro induces tooth dysmorphogenesis and enamel defects in a developmental stage- and duration-dependent manner. Cap stage- and Early Bell stage-infected molars exhibit enamel agenesis and Bell stage-infected molars exhibit enamel hypoplasia. This viral-induced pathology is coincident with stage-dependent changes in Amelx, Enam and Dspp gene expression, distribution of amelogenin, enamelin and DSP proteins, cell proliferation localization and dedifferentiation of secretory ameloblasts. Importantly, our data indicate that specific levels of Amelx and Dspp gene expression define whether mouse CMV induces enamel agenesis or hypoplasia.


Assuntos
Amelogênese , Amelogenina/genética , Citomegalovirus/patogenicidade , Esmalte Dentário/anormalidades , Dente Molar/embriologia , Dente Molar/virologia , Ameloblastos/citologia , Amelogênese Imperfeita/embriologia , Amelogênese Imperfeita/virologia , Amelogenina/biossíntese , Animais , Desdiferenciação Celular , Diferenciação Celular , Proliferação de Células , Hipoplasia do Esmalte Dentário/embriologia , Hipoplasia do Esmalte Dentário/virologia , Proteínas do Esmalte Dentário/biossíntese , Proteínas do Esmalte Dentário/genética , Proteínas da Matriz Extracelular/biossíntese , Proteínas da Matriz Extracelular/genética , Expressão Gênica , Perfilação da Expressão Gênica , Camundongos , Dente Molar/fisiologia , Odontoblastos/citologia , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Sialoglicoproteínas/biossíntese , Sialoglicoproteínas/genética , Técnicas de Cultura de Tecidos
11.
BMC Dev Biol ; 9: 32, 2009 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-19500387

RESUMO

BACKGROUND: Ectodysplasin-A appears to be a critical component of branching morphogenesis. Mutations in mouse Eda or human EDA are associated with absent or hypoplastic sweat glands, sebaceous glands, lacrimal glands, salivary glands (SMGs), mammary glands and/or nipples, and mucous glands of the bronchial, esophageal and colonic mucosa. In this study, we utilized EdaTa (Tabby) mutant mice to investigate how a marked reduction in functional Eda propagates with time through a defined genetic subcircuit and to test the proposition that canonical NFkappaB signaling is sufficient to account for the differential expression of developmentally regulated genes in the context of Eda polymorphism. RESULTS: The quantitative systems analyses do not support the stated hypothesis. For most NFkappaB-regulated genes, the observed time course of gene expression is nearly unchanged in Tabby (EdaTa) as compared to wildtype mice, as is NFkappaB itself. Importantly, a subset of genes is dramatically differentially expressed in Tabby (Edar, Fgf8, Shh, Egf, Tgfa, Egfr), strongly suggesting the existence of an alternative Eda-mediated transcriptional pathway pivotal for SMG ontogeny. Experimental and in silico investigations have identified C/EBPalpha as a promising candidate. CONCLUSION: In Tabby SMGs, upregulation of the Egf/Tgfalpha/Egfr pathway appears to mitigate the potentially severe abnormal phenotype predicted by the downregulation of Fgf8 and Shh. Others have suggested that the buffering of the phenotypic outcome that is coincident with variant Eda signaling could be a common mechanism that permits viable and diverse phenotypes, normal and abnormal. Our results support this proposition. Further, if branching epithelia use variations of a canonical developmental program, our results are likely applicable to understanding the phenotypes of other branching organs affected by Eda (EDA) mutation.


Assuntos
Ectodisplasinas/genética , Receptor Edar/genética , NF-kappa B/metabolismo , Glândulas Salivares/embriologia , Transdução de Sinais/genética , Animais , Ectodisplasinas/metabolismo , Receptor Edar/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos , Modelos Biológicos , Morfogênese/fisiologia , NF-kappa B/genética , Fenótipo , Glândulas Salivares/crescimento & desenvolvimento
12.
BMC Dev Biol ; 8: 33, 2008 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-18371224

RESUMO

BACKGROUND: Human clinical studies and mouse models clearly demonstrate that cytomegalovirus (CMV) disrupts normal organ and tissue development. Although CMV is one of the most common causes of major birth defects in humans, little is presently known about the mechanism(s) underlying CMV-induced congenital malformations. Our prior studies have demonstrated that CMV infection of first branchial arch derivatives (salivary glands and teeth) induced severely abnormal phenotypes and that CMV has a particular tropism for neural crest-derived mesenchyme (NCM). Since early embryos are barely susceptible to CMV infection, and the extant evidence suggests that the differentiation program needs to be well underway for embryonic tissues to be susceptible to viral infection and viral-induced pathology, the aim of this study was to determine if first branchial arch NCM cells are susceptible to mCMV infection prior to differentiation of NCM derivatives. RESULTS: E11 mouse mandibular processes (MANs) were infected with mouse CMV (mCMV) for up to 16 days in vitro. mCMV infection of undifferentiated embryonic mouse MANs induced micrognathia consequent to decreased Meckel's cartilage chondrogenesis and mandibular osteogenesis. Specifically, mCMV infection resulted in aberrant stromal cellularity, a smaller, misshapen Meckel's cartilage, and mandibular bone and condylar dysmorphogenesis. Analysis of viral distribution indicates that mCMV primarily infects NCM cells and derivatives. Initial localization studies indicate that mCMV infection changed the cell-specific expression of FN, NF-kappaB2, RelA, RelB, and Shh and Smad7 proteins. CONCLUSION: Our results indicate that mCMV dysregulation of key signaling pathways in primarily NCM cells and their derivatives severely disrupts mandibular morphogenesis and skeletogenesis. The pathogenesis appears to be centered around the canonical and noncanonical NF-kappaB pathways, and there is unusual juxtaposition of abnormal stromal cells and surrounding matrix. Moreover, since it is critically important that signaling molecules are expressed in appropriate cell populations during development, the aberrant localization of components of relevant signaling pathways may reveal the pathogenic mechanism underlying mandibular malformations.


Assuntos
Condrogênese/genética , Infecções por Citomegalovirus/embriologia , Mandíbula/embriologia , Muromegalovirus , Osteogênese/genética , Animais , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/fisiopatologia , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Camundongos , NF-kappa B/genética , Gravidez , Transdução de Sinais , Fator de Crescimento Transformador beta/genética
13.
Arch Oral Biol ; 53(5): 405-15, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18201685

RESUMO

OBJECTIVE: Cytomegalovirus (CMV) is one of the most common causes of major birth defects in humans. Of the approximately 8400 children born each year in the U.S. with CMV-induced birth defects, more than 1/3 of these children exhibit hypoplasia and hypocalcification of tooth enamel. Our objective was to initiate the investigation of the pathogenesis of CMV-induced tooth defects. DESIGN: Mouse Cap stage mandibular first molars were infected with mouse CMV (mCMV) in vitro in a chemically-defined organ culture system and analysed utilising histological and immunolocalisation methodologies. The antiviral, acyclovir, was used to inhibit mCMV replication and comparisons made between mCMV-infected and acyclovir-treated, mCMV-infected teeth. RESULTS: Active infection of Cap stage molars for up to 15 days in vitro results in smaller, developmentally-delayed and dysmorphic molars characterised by shallow, broad and misshapen cusps, infected and affected dental papilla mesenchyme, poorly differentiated odontoblasts and ameloblasts, and no dentin matrix. Initial protein localisation studies suggest that the pathogenesis is mediated through NF-kappaB signaling and that there appears to be an unusual interaction between abnormal mesenchymal cells and surrounding matrix. Rescue with acyclovir indicates that mCMV replication is necessary to initiate and sustain progressive tooth dysmorphogenesis. CONCLUSIONS: Our results indicate that mCMV-induced changes in signaling pathways severely delays, but does not completely interrupt, tooth morphogenesis. Importantly, our results demonstrate that this well-defined embryonic mouse organ culture system can be utilised to delineate the molecular mechanism underlying the CMV-induced tooth defects that characterise the amelogenesis imperfecta phenocopy seen in many CMV-infected children.


Assuntos
Amelogênese Imperfeita/embriologia , Modelos Animais de Doenças , Infecções por Herpesviridae , Muromegalovirus , Germe de Dente/virologia , Aciclovir/uso terapêutico , Ameloblastos , Amelogênese Imperfeita/virologia , Animais , Antivirais/uso terapêutico , Proliferação de Células , Células Epiteliais/fisiologia , Humanos , Camundongos , NF-kappa B/metabolismo , Odontoblastos , Odontogênese , Dente/embriologia , Dente/metabolismo , Germe de Dente/embriologia
14.
BMC Dev Biol ; 6: 42, 2006 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-16959038

RESUMO

BACKGROUND: Human studies suggest, and mouse models clearly demonstrate, that cytomegalovirus (CMV) is dysmorphic to early organ and tissue development. CMV has a particular tropism for embryonic salivary gland and other head mesenchyme. CMV has evolved to co-opt cell signaling networks so to optimize replication and survival, to the detriment of infected tissues. It has been postulated that mesenchymal infection is the critical step in disrupting organogenesis. If so, organogenesis dependent on epithelial-mesenchymal interactions would be particularly vulnerable. In this study, we chose to model the vulnerability by investigating the cell and molecular pathogenesis of CMV infected mouse embryonic submandibular salivary glands (SMGs). RESULTS: We infected E15 SMG explants with mouse CMV (mCMV). Active infection for up to 12 days in vitro results in a remarkable cell and molecular pathology characterized by atypical ductal epithelial hyperplasia, apparent epitheliomesenchymal transformation, oncocytic-like stromal metaplasia, beta-catenin nuclear localization, and upregulation of Nfkb2, Relb, Il6, Stat3, and Cox2. Rescue with an antiviral nucleoside analogue indicates that mCMV replication is necessary to initiate and maintain SMG dysmorphogenesis. CONCLUSION: mCMV infection of embryonic mouse explants results in dysplasia, metaplasia, and, possibly, anaplasia. The molecular pathogenesis appears to center around the activation of canonical and, perhaps more importantly, noncanonical NFkappaB. Further, COX-2 and IL-6 are important downstream effectors of embryopathology. At the cellular level, there appears to be a consequential interplay between the transformed SMG cells and the surrounding extracellular matrix, resulting in the nuclear translocation of beta-catenin. From these studies, a tentative framework has emerged within which additional studies may be planned and performed.


Assuntos
Infecções por Citomegalovirus/patologia , Citomegalovirus , Modelos Animais de Doenças , Mesoderma/patologia , Glândula Submandibular/embriologia , Glândula Submandibular/patologia , Animais , Células Cultivadas , Infecções por Citomegalovirus/virologia , Feminino , Mesoderma/virologia , Camundongos , Gravidez , Glândulas Salivares/embriologia , Glândulas Salivares/patologia , Glândulas Salivares/virologia , Glândula Submandibular/virologia , Fatores de Tempo
15.
Arch Oral Biol ; 51(5): 433-8, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16289463

RESUMO

OBJECTIVE: Mouse Twisted gastrulation gene (Twsg1) expression is found throughout embryonic development, including substantial levels in the first branchial arch that gives rise to the submandibular salivary gland (SMG). We addressed the proposition that normal Twsg1 expression is critical to normal SMG ontogenesis. DESIGN: Utilizing C57BL/6 embryos that were Twsg1-/- homozygotes, as well as wild type and heterozygote littermates, we investigated SMG development from gestational day 13 to newborn. RESULTS: Twsg1 protein is immunodetected in epithelia throughout SMG development. Twsg1-/- embryos display widely variable craniofacial phenotypes that range from normal to severe holoprosencephaly/agnathia with no mandibular arch or stomodeum. The SMG phenotypes are correlated with the external craniofacial phenotype, ranging from normal to agenesis/aplasia. CONCLUSIONS: It is evident that normal Twsg1 expression is critical for normal mouse SMG ontogenesis. Twsg1 loss of function is ultimately epistatic to the epigenome under normal physiologic conditions, but not always so. The reduced penetrance and variable expressivity seen in the SMGs of Twsg1-/- embryos is a challenging enigma.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas/genética , Glândulas Salivares/anormalidades , Glândulas Salivares/embriologia , Animais , Cruzamento , Feminino , Homozigoto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Gravidez , Proteínas/análise , Proteínas/fisiologia
16.
Cells Tissues Organs ; 179(4): 146-57, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16046861

RESUMO

The hedgehog (Hh) signaling pathway has been shown to be essential for craniofacial development. Although mandibular arch derivatives are largely absent in Shh null mice, little is known about the role of Hh signaling during Meckel's cartilage development per se. Mandible development is dependent on the morphogenesis of Meckel's cartilage, which then serves as a template for subsequent skeletal differentiation. In this study, we examine the biological function of Hh signaling during Meckel's cartilage development in vivo and in vitro. E13.5 Shh null mice present a small mesenchymal condensation in the region of a presumptive Meckel's cartilage in the hypoplastic mandibular arch. By E15.5, the Shh mutant exhibits a mere remnant of the mandibular arch, without evidence of Meckel's cartilage differentiation. Further, wild-type embryonic (E11 or E12) mandibular explants cultured for up to 5 days in the presence of cyclopamine, a steroidal alkaloid that specifically disrupts the Hh signaling pathway, exhibit a stage-dependent inhibition of Meckel's cartilage chondroblast differentiation to mature chondrocytes. This phenotype can be rescued by exogenous FGF8, a downstream effector of Hh signaling. Taken together, our results indicate that the Hh signaling pathway is critical to Meckel's cartilage ontogenesis and the rate of chondrogenesis, but not to initial primordium formation. The reliance on Hh signaling is stage dependent.


Assuntos
Cartilagem/crescimento & desenvolvimento , Mandíbula/embriologia , Transdução de Sinais , Transativadores/fisiologia , Animais , Cartilagem/embriologia , Diferenciação Celular/fisiologia , Proliferação de Células , Condrogênese/efeitos dos fármacos , Feminino , Fator 8 de Crescimento de Fibroblasto , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Hedgehog , Masculino , Mesoderma/fisiologia , Camundongos , Morfogênese , Osteogênese/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Transativadores/antagonistas & inibidores , Alcaloides de Veratrum/farmacologia
17.
BMC Dev Biol ; 5: 11, 2005 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-15972105

RESUMO

BACKGROUND: Analyses of Fgf10 and Fgfr2b mutant mice, as well as human studies, suggest that FGF10/FGFR2b signaling may play an essential, nonredundant role during embryonic SMG development. To address this question, we have analyzed the SMG phenotype in Fgf10 and Fgfr2b heterozygous and null mutant mice. In addition, although previous studies suggest that the FGF10/FGFR2b and FGF8/FGFR2c signaling pathways are functionally interrelated, little is known about the functional relationship between these two pathways during SMG development. We have designed in vivo and in vitro experiments to address this question. RESULTS: We analyzed Fgf10 and Fgfr2b heterozygous mutant and null mice and demonstrate dose-dependent SMG phenotypic differences. Hypoplastic SMGs are seen in Fgf10 and Fgfr2b heterozygotes whereas SMG aplasia is seen in Fgf10 and Fgfr2b null embryos. Complementary in vitro studies further indicate that FGF10/FGFR2b signaling regulates SMG epithelial branching and cell proliferation. To delineate the functional relationship between the FGF10/FGFR2b and FGF8/FGFR2c pathways, we compared the SMG phenotype in Fgfr2c+/Delta/Fgf10+/- double heterozygous mice to that seen in wildtype, Fgf10+/- (Fgfr2c+/+/Fgf10+/-) and Fgfr2c+/Delta (Fgfr2c+/Delta/Fgf10+/+) single heterozygous mutant littermates and demonstrate genotype-specific SMG phenotypes. In addition, exogenous FGF8 was able to rescue the abnormal SMG phenotype associated with abrogated FGFR2b signaling in vitro and restore branching to normal levels. CONCLUSION: Our data indicates that FGF10/FGFR2b signaling is essential for the SMG epithelial branching and histodifferentiation, but not earliest initial bud formation. The functional presence of other endogenous signaling pathways could not prevent complete death of embryonic SMG cells in Fgf10 and Fgfr2b null mice. Though we were able to rescue the abnormal phenotype associated with reduced in vitro FGF10/FGFR2b signaling with exogenous FGF8 supplementation, our results indicate that the FGF10/FGFR2b and FGF8/FGFR2c are nonredundant signaling pathways essential for in vivo embryonic SMG development. What remains to be determined is the in vivo functional relationship between the FGF10/FGFR2b signal transduction pathway and other key signaling pathways, and how these pathways are integrated during embryonic SMG development to compose the functional epigenome.


Assuntos
Fator 10 de Crescimento de Fibroblastos/fisiologia , Morfogênese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais/fisiologia , Glândula Submandibular/embriologia , Animais , Proliferação de Células , Embrião de Mamíferos , Células Epiteliais/citologia , Fator 10 de Crescimento de Fibroblastos/genética , Fator 8 de Crescimento de Fibroblasto/fisiologia , Genótipo , Camundongos , Camundongos Knockout , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Glândula Submandibular/citologia , Glândula Submandibular/crescimento & desenvolvimento
18.
Dev Biol ; 268(2): 457-69, 2004 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15063181

RESUMO

FGF8 has been shown to play important morphoregulatory roles during embryonic development. The observation that craniofacial, cardiovascular, pharyngeal, and neural phenotypes vary with Fgf8 gene dosage suggests that FGF8 signaling induces differences in downstream responses in a dose-dependent manner. In this study, we investigated if FGF8 plays a dose-dependent regulatory role during embryonic submandibular salivary gland (SMG) morphogenesis. We evaluated SMG phenotypes of Fgf8 hypomorphic mice, which have decreased Fgf8 gene function throughout embryogenesis. We also evaluated SMG phenotypes of Fgf8 conditional mutants in which Fgf8 function has been completely ablated in its expression domain in the first pharyngeal arch ectoderm from the time of arch formation. Fgf8 hypomorphs have hypoplastic SMGs, whereas conditional mutant SMGs exhibit ontogenic arrest followed by involution and are absent by E18.5. SMG aplasia in Fgf8 ectoderm conditional mutants indicates that FGF8 signaling is essential for the morphogenesis and survival of Pseudoglandular Stage and older SMGs. Equally important, the presence of an initial SMG bud in Fgf8 conditional mutants indicates that initial bud formation is FGF8 independent. Mice heterozygous for either the Fgf8 null allele (Fgf8(+/N)) or the hypomorphic allele (Fgf8(+/H)) have SMGs that are indistinguishable from wild-type (Fgf8(+/+)) mice which suggest that there is not only an FGF8 dose-dependent phenotypic response, but a nonlinear, threshold-like, epistatic response as well. We also found that enhanced FGF8 signaling induced, and abrogated FGF8 signaling decreased, SMG branching morphogenesis in vitro. Furthermore, since FGF10 and Shh expression is modulated by Fgf8 levels, we postulated that exogenous FGF10, Shh, or FGF10 + Shh peptide supplementation in vitro would largely "rescue" the abnormal SMG phenotype associated with decreased FGF8 signaling. This is as expected, though there is no synergistic effect with FGF10 + Shh peptide supplementation. These in vitro experiments model the principle that mutations have different effects in the context of different epigenotypes.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Glândula Submandibular/embriologia , Animais , Feminino , Fator 8 de Crescimento de Fibroblasto , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia , Glândula Submandibular/metabolismo , Glândula Submandibular/patologia
19.
Anat Rec A Discov Mol Cell Evol Biol ; 271(2): 322-31, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12629675

RESUMO

Hypohidrotic (anhidrotic) ectodermal dysplasia (HED), the most common of the approximately 150 described ectodermal dysplasias, is a disorder characterized by abnormal hair, teeth, sweat glands, and salivary glands. Mutations in the EDA (ectodysplasin-A) and EDAR (ectodysplasin-A receptor) genes are responsible for X-linked and autosomal HED, respectively. Abnormal phenotypes similar to HED are seen in Tabby (Eda(Ta)) and downless (Edar(dl)) mutant mice. Although recent studies have focused on the role of Eda/Edar signaling during hair and tooth development, very little is known about its role during embryonic submandibular salivary gland (SMG) development. To this end, we analyzed the SMG phenotypes in Tabby (Ta) and downless (dl) mutant mice and determined that Ta SMGs are hypoplastic, whereas dl SMGs are severely dysplastic. The absence of SMG ducts and acini in dl SMGs suggests that Eda/Edar signaling is essential for lumina formation and glandular histodifferentiation. Our localization of Eda and Edar proteins at sites of lumen and acini formation supports this conclusion. Moreover, the presence of SMGs in both Ta and dl mutant mice, as well as the absence of immunodetectable Eda and Edar protein in Initial Bud and Early Pseudoglandular stage SMGs, indicate that Eda/Edar-mediated signaling is important for branching morphogenesis and histodifferentiation, but not for initial gland formation. To initially delineate the morphoregulatory role of Eda/Edar-mediated signaling during embryonic SMG development, we cultured embryonic day 14 SMGs with enhanced or abrogated Eda/Edar signaling. Eda supplementation induced a significant increase in SMG branching, and enhanced activation of NF-kappaB. Abrogating Eda/Edar signaling by adding the soluble form of Edar to bind endogenous ligand in embryonic SMGs results in a significant dose-dependent decrease in branching morphogenesis. Taken together, our results suggest that the Eda/Edar/NF-kappaB pathway exerts its effect on SMG epithelial cell proliferation, lumina formation, and histodifferentiation.


Assuntos
Proteínas de Peixes/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais , Glândula Submandibular/embriologia , Glândula Submandibular/metabolismo , Animais , Diferenciação Celular , Displasia Ectodérmica/genética , Displasia Ectodérmica/metabolismo , Receptor Edar , Desenvolvimento Embrionário e Fetal , Feminino , Proteínas de Peixes/genética , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Mutantes , NF-kappa B/metabolismo , Técnicas de Cultura de Órgãos , Fragmentos de Peptídeos/imunologia , Fenótipo , Coelhos , Receptores da Ectodisplasina , Receptores do Fator de Necrose Tumoral , Glândula Submandibular/crescimento & desenvolvimento
20.
Cells Tissues Organs ; 170(2-3): 83-98, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11731698

RESUMO

Embryonic submandibular salivary gland (SMG) initiation and branching morphogenesis are dependent on cell-cell communications between and within epithelium and mesenchyme. Such communications are typically mediated in other organs (teeth, lung, lacrimal glands) by growth factors in such a way as to translate autocrine, juxtacrine and paracrine signals into specific gene responses regulating cell division and histodifferentiation. Using Wnt1-Cre/R26R transgenic mice, we demonstrate that embryonic SMG mesenchyme is derived exclusively from cranial neural crest. This origin contrasts to that known for tooth mesenchyme, previously shown to be derived from both neural crest and nonneural crest cells. Thus, although both SMGs and teeth are mandibular derivatives, we can expect overlap and differences in the details of their early inductive interactions. In addition, since embryonic SMG branching morphogenesis is analogous to that seen in other branching organs, we also expect similarities of expression regarding those molecules known to be ubiquitous regulators of morphogenesis. In this study, we performed an analysis of the distribution of specific fibroblast growth factors (FGFs), FGF receptors, bone morphogenetic proteins (BMPs) and Pax transcription factors, previously shown to be important for tooth development and/or branching morphogenesis, from the time of initiation of embryonic SMG development until early branching morphogenesis. In addition, we report abnormal SMG phenotypes in FgfR2- IIIc(+/Delta), BMP7(-/-) and Pax6(-/-) mice. Our results, in comparison with functional studies in other systems, suggest that FGF-2/FGFR-1, FGF-8/FGFR-2(IIIc) and FGF-10/FGFR-2(IIIb) signaling have different paracrine and juxtacrine functions during SMG initial bud formation and branching. Finally, our observations of abnormal SMGs in BMP7(-/-) and Pax6(-/-) indicate that both BMP7 and Pax6 play important roles during embryonic SMG branching morphogenesis.


Assuntos
Proteínas Morfogenéticas Ósseas/análise , Proteínas de Ligação a DNA/análise , Fatores de Crescimento de Fibroblastos/análise , Proteínas de Homeodomínio/análise , Glândula Submandibular/embriologia , Fatores de Transcrição/análise , Animais , Diferenciação Celular , Epitélio/fisiologia , Proteínas do Olho , Feminino , Idade Gestacional , Mesoderma/fisiologia , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Morfogênese , Fator de Transcrição PAX6 , Fator de Transcrição PAX9 , Fatores de Transcrição Box Pareados , Proteínas Repressoras , Glândula Submandibular/anormalidades
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...