Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cell Death Discov ; 8(1): 387, 2022 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-36115851

RESUMO

Retinitis pigmentosa is a group of progressive inherited retinal dystrophies that may present clinically as part of a syndromic entity or as an isolated (nonsyndromic) manifestation. In an Indian family suffering from retinitis pigmentosa, we identified a missense variation in CNGA1 affecting the cyclic nucleotide binding domain (CNBD) and characterized a mouse model developed with mutated CNBD. A gene panel analysis comprising 105 known RP genes was used to analyze a family with autosomal-recessive retinitis pigmentosa (arRP) and revealed that CNGA1 was affected. From sperm samples of ENU mutagenesis derived F1 mice, we re-derived a mutant with a Cnga1 mutation. Homozygous mutant mice, developing retinal degeneration, were examined for morphological and functional consequences of the mutation. In the family, we identified a rare CNGA1 variant (NM_001379270.1) c.1525 G > A; (p.Gly509Arg), which co-segregated among the affected family members. Homozygous Cnga1 mice harboring a (ENSMUST00000087213.12) c.1526 A > G (p.Tyr509Cys) mutation showed progressive degeneration in the retinal photoreceptors from 8 weeks on. This study supports a role for CNGA1 as a disease gene for arRP and provides new insights on the pathobiology of cGMP-binding domain mutations in CNGA1-RP.

2.
Mol Metab ; 54: 101334, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34487921

RESUMO

OBJECTIVE: Protein disulfide isomerases (PDIs) are oxidoreductases that are involved in catalyzing the formation and rearrangement of disulfide bonds during protein folding. One of the PDI members is the PDI-associated 6 (PDIA6) protein, which has been shown to play a vital role in ß-cell dysfunction and diabetes. However, very little is known about the function of this protein in ß-cells in vivo. This study aimed to describe the consequences of a point mutation in Pdia6 on ß-cell development and function. METHODS: We generated an ENU mouse model carrying a missense mutation (Phe175Ser) in the second thioredoxin domain of the Pdia6 gene. Using biochemical and molecular tools, we determined the effects of the mutation on the ß-cell development at embryonic day (E)18.5 and ß-cell identity as well as function at postnatal stages. RESULTS: Mice homozygous for the Phe175Ser (F175S) mutation were mildly hyperglycemic at weaning and subsequently became hypoinsulinemic and overtly diabetic at the adult stage. Although no developmental phenotype was detected during embryogenesis, mutant mice displayed reduced insulin-expressing ß-cells at P14 and P21 without any changes in the rate of cell death and proliferation. Further analysis revealed an increase in BiP and the PDI family member PDIA4, but without any concomitant apoptosis and cell death. Instead, the expression of prominent markers of ß-cell maturation and function, such as Ins2, Mafa, and Slc2a2, along with increased expression of α-cell markers, Mafb, and glucagon was observed in adult mice, suggesting loss of ß-cell identity. CONCLUSIONS: The results demonstrate that a global Pdia6 mutation renders mice hypoinsulinemic and hyperglycemic. This occurs due to the loss of pancreatic ß-cell function and identity, suggesting a critical role of PDIA6 specifically for ß-cells.


Assuntos
Diabetes Mellitus/genética , Células Secretoras de Insulina/metabolismo , Isomerases de Dissulfetos de Proteínas/genética , Animais , Diabetes Mellitus/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Mutação Puntual , Isomerases de Dissulfetos de Proteínas/metabolismo
3.
Commun Biol ; 3(1): 628, 2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-33127955

RESUMO

The transcription factor PAX6 is involved in the development of the eye and pancreatic islets, besides being associated with sleep-wake cycles. Here, we investigated a point mutation in the RED subdomain of PAX6, previously described in a human patient, to present a comprehensive study of a homozygous Pax6 mutation in the context of adult mammalian metabolism and circadian rhythm. Pax6Leca2 mice lack appropriate retinal structures for light perception and do not display normal daily rhythmic changes in energy metabolism. Despite ß cell dysfunction and decreased insulin secretion, mutant mice have normal glucose tolerance. This is associated with reduced hepatic glucose production possibly due to altered circadian variation in expression of clock and metabolic genes, thereby evading hyperglycemia. Hence, our findings show that while the RED subdomain is important for ß cell functional maturity, the Leca2 mutation impacts peripheral metabolism via loss of circadian rhythm, thus revealing pleiotropic effects of PAX6.


Assuntos
Ritmo Circadiano/genética , Glucose/metabolismo , Secreção de Insulina/genética , Células Secretoras de Insulina/fisiologia , Fator de Transcrição PAX6/genética , Animais , Glicemia/genética , Ritmo Circadiano/fisiologia , Regulação da Expressão Gênica , Glucose/genética , Fígado/metabolismo , Fígado/fisiologia , Masculino , Camundongos Endogâmicos C3H , Camundongos Mutantes , Mutação , Nervo Óptico/anormalidades , Fator de Transcrição PAX6/metabolismo , Retina/ultraestrutura , Células Ganglionares da Retina/fisiologia
4.
Invest Ophthalmol Vis Sci ; 61(2): 44, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-32106289

RESUMO

Purpose: The clinical phenotype of retinal gliosis occurs in different forms; here, we characterize one novel genetic feature, (i.e., signaling via BMP-receptor 1b). Methods: Mouse mutants were generated within a recessive ENU mutagenesis screen; the underlying mutation was identified by linkage analysis and Sanger sequencing. The eye phenotype was characterized by fundoscopy, optical coherence tomography, optokinetic drum, electroretinography, and visual evoked potentials, by histology, immunohistology, and electron-microscopy. Results: The mutation affects intron 10 of the Bmpr1b gene, which is causative for skipping of exon 10. The expression levels of pSMAD1/5/8 were reduced in the mutant retina. The loss of BMPR1B-mediated signaling leads to optic nerve coloboma, gliosis in the optic nerve head and ventral retina, defective optic nerve axons, and irregular retinal vessels. The ventral retinal gliosis is proliferative and hypertrophic, which is concomitant with neuronal delamination and the reduction of retinal ganglion cells (RGCs); it is dominated by activated astrocytes overexpressing PAX2 and SOX2 but not PAX6, indicating that they may retain properties of gliogenic precursor cells. The expression pattern of PAX2 in the optic nerve head and ventral retina is altered during embryonic development. These events finally result in reduced electrical transmission of the retina and optic nerve and significantly reduced visual acuity. Conclusions: Our study demonstrates that BMPR1B is necessary for the development of the optic nerve and ventral retina. This study could also indicate a new mechanism in the formation of retinal gliosis; it opens new routes for its treatment eventually preventing scar formation in the retina.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Coloboma/genética , Gliose/genética , Mutação , Disco Óptico/anormalidades , Doenças Retinianas/genética , Animais , Camundongos , Disco Óptico/patologia
5.
Diabetes ; 69(5): 915-926, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32029480

RESUMO

Genes of the Notch signaling pathway are expressed in different cell types and organs at different time points during embryonic development and adulthood. The Notch ligand Delta-like 1 (DLL1) controls the decision between endocrine and exocrine fates of multipotent progenitors in the developing pancreas, and loss of Dll1 leads to premature endocrine differentiation. However, the role of Delta-Notch signaling in adult tissue homeostasis is not well understood. Here, we describe the spatial expression pattern of Notch pathway components in adult murine pancreatic islets and show that DLL1 and DLL4 are specifically expressed in ß-cells, whereas JAGGED1 is expressed in α-cells. We show that mice lacking both DLL1 and DLL4 in adult ß-cells display improved glucose tolerance, increased glucose-stimulated insulin secretion, and hyperglucagonemia. In contrast, overexpression of the intracellular domain of DLL1 in adult murine pancreatic ß-cells results in impaired glucose tolerance and reduced insulin secretion, both in vitro and in vivo. These results suggest that Notch ligands play specific roles in the adult pancreas and highlight a novel function of the Delta/Notch pathway in ß-cell insulin secretion.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Insulina/metabolismo , Pâncreas/metabolismo , Receptor Notch3/metabolismo , Receptor Notch4/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Adulto , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação ao Cálcio/genética , Regulação da Expressão Gênica/fisiologia , Glucagon/sangue , Células Secretoras de Glucagon/patologia , Células Secretoras de Glucagon/fisiologia , Glucose/genética , Glucose/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Receptor Notch3/genética , Receptor Notch4/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição HES-1/genética , Fatores de Transcrição HES-1/metabolismo
6.
7.
Mol Metab ; 6(10): 1304-1312, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29031729

RESUMO

OBJECTIVE: The fatty acid receptor 1 (FFAR1/GPR40) mediates fatty acid-dependent augmentation of glucose-induced insulin secretion (GIIS) in pancreatic ß-cells. Genetically engineered Ffar1-knockout/congenic mice univocally displayed impaired fatty acid-mediated insulin secretion, but in vivo experiments delivered controversial results regarding the function of FFAR1 in glucose homeostasis and liver steatosis. This study presents a new coisogenic mouse model carrying a point mutation in Ffar1 with functional consequence. These mice reflect the situations in humans in which point mutations can lead to protein malfunction and disease development. METHODS: The Munich N-ethyl-N-nitrosourea (ENU) mutagenesis-derived F1 archive containing over 16,800 sperms and corresponding DNA samples was screened for mutations in the coding region of Ffar1. Two missense mutations (R258W and T146S) in the extracellular domain of the protein were chosen and homozygote mice were generated. The functional consequence of these mutations was examined in vitro in isolated islets and in vivo in chow diet and high fat diet fed mice. RESULTS: Palmitate, 50 µM, and the FFAR1 agonist TUG-469, 3 µM, stimulated insulin secretion in islets of Ffar1T146S/T146S mutant mice and of wild-type littermates, while in islets of Ffar1R258W/R258W mutant mice, these stimulatory effects were abolished. Insulin content and mRNA levels of Ffar1, Glp1r, Ins2, Slc2a2, Ppara, and Ppard were not significantly different between wild-type and Ffar1R258W/R258W mouse islets. Palmitate exposure, 600 µM, significantly increased Ppara mRNA levels in wild-type but not in Ffar1R258W/R258W mouse islets. On the contrary, Slc2a2 mRNA levels were significantly reduced in both wild-type and Ffar1R258W/R258W mouse islets after palmitate treatment. HFD feeding induced glucose intolerance in wild-type mice. Ffar1R258W/R258W mutant mice remained glucose tolerant although their body weight gain, liver steatosis, insulin resistance, and plasma insulin levels were not different from those of wild-type littermates. Worth mentioning, fasting plasma insulin levels were lower in Ffar1R258W/R258W mice. CONCLUSION: A point mutation in Ffar1 abrogates the stimulatory effect of palmitate on GIIS, an effect that does not necessarily translate to HFD-induced glucose intolerance.


Assuntos
Secreção de Insulina/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Compostos de Anilina/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos/metabolismo , Fígado Gorduroso/metabolismo , Glucose/metabolismo , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Insulina/genética , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas , Camundongos , Palmitatos/metabolismo , Fenilpropionatos/metabolismo , Mutação Puntual/genética
8.
J Theor Biol ; 430: 32-44, 2017 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-28652000

RESUMO

During pancreas development, Neurog3 positive endocrine progenitors are specified by Delta/Notch (D/N) mediated lateral inhibition in the growing ducts. During neurogenesis, genes that determine the transition from the proneural state to neuronal or glial lineages are oscillating before their expression is sustained. Although the basic gene regulatory network is very similar, cycling gene expression in pancreatic development was not investigated yet, and previous simulations of lateral inhibition in pancreas development excluded by design the possibility of oscillations. To explore this possibility, we developed a dynamic model of a growing duct that results in an oscillatory phase before the determination of endocrine progenitors by lateral inhibition. The basic network (D/N + Hes1 + Neurog3) shows scattered, stable Neurog3 expression after displaying transient expression. Furthermore, we included the Hes1 negative feedback as previously discussed in neurogenesis and show the consequences for Neurog3 expression in pancreatic duct development. Interestingly, a weakened HES1 action on the Hes1 promoter allows the coexistence of stable patterning and oscillations. In conclusion, cycling gene expression and lateral inhibition are not mutually exclusive. In this way, we argue for a unified mode of D/N mediated lateral inhibition in neurogenic and pancreatic progenitor specification.


Assuntos
Modelos Biológicos , Neurogênese , Pâncreas/crescimento & desenvolvimento , Receptores Notch/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Padronização Corporal , Linhagem da Célula , Sistema Endócrino/citologia , Retroalimentação Fisiológica , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Proteínas do Tecido Nervoso/fisiologia , Oscilometria , Pâncreas/inervação , Fatores de Transcrição HES-1/fisiologia
9.
G3 (Bethesda) ; 6(12): 4035-4046, 2016 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-27815347

RESUMO

The vertebrate Scube (Signal peptide, CUB, and EGF-like domain-containing protein) family consists of three independent members, Scube1-3, which encode secreted cell surface-associated membrane glycoproteins. Limited information about the general function of this gene family is available, and their roles during adulthood. Here, we present the first Scube3 mutant mouse line (Scube3N294K/N294K), which clearly shows phenotypic alterations by carrying a missense mutation in exon 8, and thus contributes to our understanding of SCUBE3 functions. We performed a detailed phenotypic characterization in the German Mouse Clinic (GMC). Scube3N294K/N294K mutants showed morphological abnormalities of the skeleton, alterations of parameters relevant for bone metabolism, changes in renal function, and hearing impairments. These findings correlate with characteristics of the rare metabolic bone disorder Paget disease of bone (PDB), associated with the chromosomal region of human SCUBE3 In addition, alterations in energy metabolism, behavior, and neurological functions were detected in Scube3N294K/N294K mice. The Scube3N294K/N294K mutant mouse line may serve as a new model for further studying the effect of impaired SCUBE3 gene function.


Assuntos
Estudos de Associação Genética , Glicoproteínas/genética , Mutação , Fenótipo , Animais , Osso e Ossos/metabolismo , Proteínas de Ligação ao Cálcio , Mapeamento Cromossômico , Modelos Animais de Doenças , Metabolismo Energético/genética , Exoma , Feminino , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Rim/metabolismo , Rim/fisiopatologia , Testes de Função Renal , Masculino , Camundongos , Camundongos Knockout , Osteíte Deformante/genética , Osteíte Deformante/metabolismo , Osteíte Deformante/patologia , Esqueleto/anormalidades
10.
Diabetes ; 65(9): 2540-52, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27284107

RESUMO

Bezafibrate (BEZ), a pan activator of peroxisome proliferator-activated receptors (PPARs), has been generally used to treat hyperlipidemia for decades. Clinical trials with type 2 diabetes patients indicated that BEZ also has beneficial effects on glucose metabolism, although the underlying mechanisms of these effects remain elusive. Even less is known about a potential role for BEZ in treating type 1 diabetes. Here we show that BEZ markedly improves hyperglycemia and glucose and insulin tolerance in mice with streptozotocin (STZ)-induced diabetes, an insulin-deficient mouse model of type 1 diabetes. BEZ treatment of STZ mice significantly suppressed the hepatic expression of genes that are annotated in inflammatory processes, whereas the expression of PPAR and insulin target gene transcripts was increased. Furthermore, BEZ-treated mice also exhibited improved metabolic flexibility as well as an enhanced mitochondrial mass and function in the liver. Finally, we show that the number of pancreatic islets and the area of insulin-positive cells tended to be higher in BEZ-treated mice. Our data suggest that BEZ may improve impaired glucose metabolism by augmenting hepatic mitochondrial performance, suppressing hepatic inflammatory pathways, and improving insulin sensitivity and metabolic flexibility. Thus, BEZ treatment might also be useful for patients with impaired glucose tolerance or diabetes.


Assuntos
Bezafibrato/uso terapêutico , Diabetes Mellitus Experimental/tratamento farmacológico , Resistência à Insulina/fisiologia , Animais , Glicemia/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Teste de Tolerância a Glucose , Humanos , Hiperglicemia/tratamento farmacológico , Hiperglicemia/metabolismo , Hiperglicemia/fisiopatologia , Hipoglicemiantes/uso terapêutico , Hipolipemiantes/uso terapêutico , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Consumo de Oxigênio/efeitos dos fármacos , Receptores Ativados por Proliferador de Peroxissomo/antagonistas & inibidores
11.
Mamm Genome ; 27(3-4): 111-21, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26803617

RESUMO

We performed exome sequencing for mutation discovery of an ENU (N-ethyl-N-nitrosourea)-derived mouse model characterized by significant elevated plasma alkaline phosphatase (ALP) activities in female and male mutant mice, originally named BAP014 (bone screen alkaline phosphatase #14). We identified a novel loss-of-function mutation within the Fam46a (family with sequence similarity 46, member A) gene (NM_001160378.1:c.469G>T, NP_001153850.1:p.Glu157*). Heterozygous mice of this mouse line (renamed Fam46a (E157*Mhda)) had significantly high ALP activities and apparently no other differences in morphology compared to wild-type mice. In contrast, homozygous Fam46a (E157*Mhda) mice showed severe morphological and skeletal abnormalities including short stature along with limb, rib, pelvis, and skull deformities with minimal trabecular bone and reduced cortical bone thickness in long bones. ALP activities of homozygous mutants were almost two-fold higher than in heterozygous mice. Fam46a is weakly expressed in most adult and embryonic tissues with a strong expression in mineralized tissues as calvaria and femur. The FAM46A protein is computationally predicted as a new member of the superfamily of nucleotidyltransferase fold proteins, but little is known about its function. Fam46a (E157*Mhda) mice are the first mouse model for a mutation within the Fam46a gene.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/patologia , Códon sem Sentido , Exoma , Fosfatase Alcalina/metabolismo , Animais , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Análise Mutacional de DNA , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Expressão Gênica , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Camundongos , Camundongos Knockout , Fenótipo
12.
PLoS Comput Biol ; 10(10): e1003843, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25275459

RESUMO

While it is known that a large fraction of vertebrate genes are under the control of a gene regulatory network (GRN) forming a clock with circadian periodicity, shorter period oscillatory genes like the Hairy-enhancer-of split (Hes) genes are discussed mostly in connection with the embryonic process of somitogenesis. They form the core of the somitogenesis-clock, which orchestrates the periodic separation of somites from the presomitic mesoderm (PSM). The formation of sharp boundaries between the blocks of many cells works only when the oscillators in the cells forming the boundary are synchronized. It has been shown experimentally that Delta-Notch (D/N) signaling is responsible for this synchronization. This process has to happen rather fast as a cell experiences at most five oscillations from its 'birth' to its incorporation into a somite. Computer simulations describing synchronized oscillators with classical modes of D/N-interaction have difficulties to achieve synchronization in an appropriate time. One approach to solving this problem of modeling fast synchronization in the PSM was the consideration of cell movements. Here we show that fast synchronization of Hes-type oscillators can be achieved without cell movements by including D/N cis-inhibition, wherein the mutual interaction of DELTA and NOTCH in the same cell leads to a titration of ligand against receptor so that only one sort of molecule prevails. Consequently, the symmetry between sender and receiver is partially broken and one cell becomes preferentially sender or receiver at a given moment, which leads to faster entrainment of oscillators. Although not yet confirmed by experiment, the proposed mechanism of enhanced synchronization of mesenchymal cells in the PSM would be a new distinct developmental mechanism employing D/N cis-inhibition. Consequently, the way in which Delta-Notch signaling was modeled so far should be carefully reconsidered.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Relógios Biológicos/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Receptor Notch1/genética , Transdução de Sinais/genética , Animais , Biologia Computacional , Redes Reguladoras de Genes/genética , Camundongos , Modelos Genéticos , RNA Mensageiro
13.
PLoS One ; 8(11): e81833, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24278461

RESUMO

Members of the PRDM protein family have been shown to play important roles during embryonic development. Previous in vitro and in situ analyses indicated a function of Prdm6 in cells of the vascular system. To reveal physiological functions of Prdm6, we generated conditional Prdm6-deficient mice. Complete deletion of Prdm6 results in embryonic lethality due to cardiovascular defects associated with aberrations in vascular patterning. However, smooth muscle cells could be regularly differentiated from Prdm6-deficient embryonic stem cells and vascular smooth muscle cells were present and proliferated normally in Prdm6-deficient embryos. Conditional deletion of Prdm6 in the smooth muscle cell lineage using a SM22-Cre driver line resulted in perinatal lethality due to hemorrhage in the lungs. We thus identified Prdm6 as a factor that is essential for the physiological control of cardiovascular development.


Assuntos
Sistema Cardiovascular/embriologia , Proteínas Repressoras/fisiologia , Animais , Sequência de Bases , Northern Blotting , Southern Blotting , Padronização Corporal , Diferenciação Celular , Proliferação de Células , Primers do DNA , Camundongos , Camundongos Knockout , Músculo Liso/citologia , Neovascularização Fisiológica , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Proteínas Repressoras/genética
14.
J Bone Miner Metab ; 31(3): 293-303, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23371561

RESUMO

The mouse is a valuable model organism for studying bone biology and for unravelling pathological processes in skeletal disorders. In vivo methods like X-ray analysis, DXA measurements, pQCT and µCT are available to investigate the bone phenotype of mutant mice. However, the descriptive nature of such methods does not provide insights into the cellular and molecular bases of the observed bone alterations. Thus, first-line investigations might be complemented by cell culture-based methods to characterize the pathological processes at the cellular level independent from systemic influences. By combining well-established assays, we designed a comprehensive test system to investigate the cellular and molecular phenotype of primary calvarial osteoblasts in mutant mice compared to wild-type controls as a first-line phenotyping method. The compilation of 9 different quantifiable assays allows assessment of general properties of cell growth and investigation of bone-specific parameters at the functional, protein and RNA level in a kinetic fashion throughout a 3-week culture period, thus maximizing the chance to discover and explain new phenotypes in mutant mice. By analyzing mutant mouse lines for Col1a1 and Jag1 (Delta-Notch pathway) that both showed clear alterations in several bone-related parameters we could demonstrate the usefulness of our cell culture system to discriminate between primary (Col1a1) and secondary effects (Jag1) in osteoblasts.


Assuntos
Osso e Ossos/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Técnicas de Cultura de Células/métodos , Técnicas de Cultura de Células/normas , Colágeno Tipo I/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Osteoblastos/patologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Cadeia alfa 1 do Colágeno Tipo I , Fêmur/diagnóstico por imagem , Fêmur/patologia , Regulação da Expressão Gênica , Proteína Jagged-1 , Camundongos , Camundongos Mutantes , Osteoblastos/metabolismo , Fenótipo , Padrões de Referência , Reprodutibilidade dos Testes , Proteínas Serrate-Jagged , Tomografia Computadorizada por Raios X
15.
PLoS Comput Biol ; 8(6): e1002586, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761566

RESUMO

The segmentation of the vertebrate body is laid down during early embryogenesis. The formation of signaling gradients, the periodic expression of genes of the Notch-, Fgf- and Wnt-pathways and their interplay in the unsegmented presomitic mesoderm (PSM) precedes the rhythmic budding of nascent somites at its anterior end, which later develops into epithelialized structures, the somites. Although many in silico models describing partial aspects of somitogenesis already exist, simulations of a complete causal chain from gene expression in the growth zone via the interaction of multiple cells to segmentation are rare. Here, we present an enhanced gene regulatory network (GRN) for mice in a simulation program that models the growing PSM by many virtual cells and integrates WNT3A and FGF8 gradient formation, periodic gene expression and Delta/Notch signaling. Assuming Hes7 as core of the somitogenesis clock and LFNG as modulator, we postulate a negative feedback of HES7 on Dll1 leading to an oscillating Dll1 expression as seen in vivo. Furthermore, we are able to simulate the experimentally observed wave of activated NOTCH (NICD) as a result of the interactions in the GRN. We esteem our model as robust for a wide range of parameter values with the Hes7 mRNA and protein decays exerting a strong influence on the core oscillator. Moreover, our model predicts interference between Hes1 and HES7 oscillators when their intrinsic frequencies differ. In conclusion, we have built a comprehensive model of somitogenesis with HES7 as core oscillator that is able to reproduce many experimentally observed data in mice.


Assuntos
Padronização Corporal/genética , Padronização Corporal/fisiologia , Mesoderma/embriologia , Modelos Biológicos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Relógios Biológicos/genética , Relógios Biológicos/fisiologia , Biologia Computacional , Simulação por Computador , Retroalimentação Fisiológica , Fator 8 de Crescimento de Fibroblasto/genética , Fator 8 de Crescimento de Fibroblasto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Mesoderma/metabolismo , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Somitos/embriologia , Somitos/metabolismo , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo
16.
Hum Mol Genet ; 21(16): 3535-45, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22589248

RESUMO

Osteogenesis imperfecta (OI) is an inherited connective tissue disorder with skeletal dysplasia of varying severity, predominantly caused by mutations in the collagen I genes (COL1A1/COL1A2). Extraskeletal findings such as cardiac and pulmonary complications are generally considered to be significant secondary features. Aga2, a murine model for human OI, was systemically analyzed in the German Mouse Clinic by means of in vivo and in vitro examinations of the cardiopulmonary system, to identify novel mechanisms accounting for perinatal lethality. Pulmonary and, especially, cardiac fibroblast of perinatal lethal Aga2/+ animals display a strong down-regulation of Col1a1 transcripts in vivo and in vitro, resulting in a loss of extracellular matrix integrity. In addition, dysregulated gene expression of Nppa, different types of collagen and Agt in heart and lung tissue support a bone-independent vicious cycle of heart dysfunction, including hypertrophy, loss of myocardial matrix integrity, pulmonary hypertension, pneumonia and hypoxia leading to death in Aga2. These murine findings are corroborated by a pediatric OI cohort study, displaying significant progressive decline in pulmonary function and restrictive pulmonary disease independent of scoliosis. Most participants show mild cardiac valvular regurgitation, independent of pulmonary and skeletal findings. Data obtained from human OI patients and the mouse model Aga2 provide novel evidence for primary effects of type I collagen mutations on the heart and lung. The findings will have potential benefits of anticipatory clinical exams and early intervention in OI patients.


Assuntos
Sistema Cardiovascular/fisiopatologia , Colágeno Tipo I/genética , Pulmão/fisiopatologia , Osteogênese Imperfeita/fisiopatologia , Adolescente , Animais , Insuficiência da Valva Aórtica/fisiopatologia , Criança , Pré-Escolar , Cadeia alfa 1 do Colágeno Tipo I , Modelos Animais de Doenças , Expressão Gênica , Humanos , Camundongos , Miocárdio/metabolismo , Osteogênese Imperfeita/genética , Fenótipo , Insuficiência da Valva Pulmonar/fisiopatologia , Escoliose/etiologia , Adulto Jovem
17.
Gene Expr Patterns ; 10(6): 265-73, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20558326

RESUMO

MicroRNAs (miRNAs) have regulatory functions during vertebrate embryogenesis. They are short approximately 21bp long endogenously expressed single-stranded RNAs, which preferentially bind to complementary sequences in the 3' untranslated regions (UTR) of mRNAs and typically down-regulate the respective target mRNAs by translational repression or enhanced mRNA degradation. The Notch ligand Delta-like 1 (Dll1) is expressed in a highly dynamic pattern and has pleiotropic functions during embryogenesis and in adult tissues. Here, we report an interspecies in silico analysis to identify 16 miRNAs, which potentially bind to the mouse, human and chicken Dll1 3'UTRs. To analyze whether these miRNAs could regulate Dll1 gene expression during somitogenesis and neurogenesis, we performed a systematic whole mount in situ hybridisation screen, followed by radioactive in situ hybridisation on sections, using LNA modified DNA probes in mouse embryos. We find that 7 miRNAs (miR-34a, miR-103, miR-107, miR-130a, miR-130b, miR-449a and miR-449c) are expressed in developing somites, limbs, restricted regions of the brain and neural tube between 9.5 dpc and 12.5 dpc. This suggests that these miRNAs could possibly target the Dll1 3'UTR in these regions. The other miRNAs are not expressed or below the detection limit and thus are unlikely to regulate Dll1 at the analyzed embryonic stages.


Assuntos
Biologia Computacional , Desenvolvimento Embrionário/genética , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , MicroRNAs/isolamento & purificação , MicroRNAs/metabolismo , Regiões 3' não Traduzidas/genética , Animais , Proteínas de Ligação ao Cálcio , Embrião de Galinha , Galinhas , Biologia Computacional/métodos , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização In Situ/métodos , Camundongos , Ligação Proteica
18.
Development ; 137(3): 437-45, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20081190

RESUMO

Homeodomain (HD) transcription factors and components of the Notch pathway [Delta1 (Dll1), Jagged1 (Jag1) and the Fringe (Fng) proteins] are expressed in distinct progenitor domains along the dorsoventral (DV) axis of the developing spinal cord. However, the internal relationship between these two regulatory pathways has not been established. In this report we show that HD proteins act upstream of Notch signalling. Thus, HD proteins control the spatial distribution of Notch ligands and Fng proteins, whereas perturbation of the Notch pathway does not affect the regional expression of HD proteins. Loss of Dll1 or Jag1 leads to a domain-specific increase of neuronal differentiation but does not affect the establishment of progenitor domain boundaries. Moreover, gain-of-function experiments indicate that the ability of Dll1 and Jag1 to activate Notch is limited to progenitors endogenously expressing the respective ligand. Fng proteins enhance Dll1-activated Notch signalling and block Notch activation mediated by Jag1. This finding, combined with the overlapping expression of Fng with Dll1 but not with Jag1, is likely to explain the domain-specific activity of the Notch ligands. This outcome is opposite to the local regulation of Notch activity in most other systems, including the Drosophila wing, where Fng co-localizes with Jagged/Serrate rather than Dll/Delta, which facilitates Notch signalling at regional boundaries instead of within domains. The regulation of Notch activation in the spinal cord therefore appears to endow specific progenitor populations with a domain-wide autonomy in the control of neurogenesis and prevents any inadequate activation of Notch across progenitor domain boundaries.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Neurogênese , Receptores Notch/metabolismo , Animais , Padronização Corporal/genética , Embrião de Galinha , Peptídeos e Proteínas de Sinalização Intercelular , Ligantes , Camundongos , Medula Espinal , Fatores de Transcrição
19.
PLoS One ; 4(6): e6054, 2009 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-19562077

RESUMO

BACKGROUND: The Notch signaling pathway is an evolutionary conserved signal transduction pathway involved in embryonic patterning and regulation of cell fates during development and self-renewal. Recent studies have demonstrated that this pathway is integral to a complex system of interactions, involving as well other signal transduction pathways, and implicated in distinct human diseases. Delta-like 1 (Dll1) is one of the known ligands of the Notch receptors. The role of the Notch ligands is less well understood. Loss-of-function of Dll1 leads to embryonic lethality, but reduction of Delta-like 1 protein levels has not been studied in adult stage. METHODOLOGY/PRINCIPAL FINDINGS: Here we present the haploinsufficient phenotype of Dll1 and a missense mutant Dll1 allele (Dll1(C413Y)). Haploinsufficiency leads to a complex phenotype with several biological processes altered. These alterations reveal the importance of Dll1 mainly in metabolism, energy balance and in immunology. The animals are smaller, lighter, with altered fat to lean ratio and have increased blood pressure and a slight bradycardia. The animals have reduced cholesterol and triglyceride levels in blood. At the immunological level a subtle phenotype is observed due to the effect and fine-tuning of the signaling network at the different levels of differentiation, proliferation and function of lymphocytes. Moreover, the importance of the proteolytic regulation of the Notch signaling network emphasized. CONCLUSIONS/SIGNIFICANCE: In conclusion, slight alterations in one player of Notch signaling alter the entire organism, emphasizing the fine-tuning character of this pathway in a high number of processes.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Alelos , Sequência de Aminoácidos , Animais , Proteínas de Ligação ao Cálcio , Feminino , Ligantes , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Modelos Biológicos , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Receptores Notch/metabolismo , Transdução de Sinais
20.
PLoS Genet ; 4(2): e7, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18248096

RESUMO

Osteogenesis imperfecta is an inherited disorder characterized by increased bone fragility, fractures, and osteoporosis, and most cases are caused by mutations affecting the type I collagen genes. Here, we describe a new mouse model for Osteogenesis imperfecta termed Aga2 (abnormal gait 2) that was isolated from the Munich N-ethyl-N-nitrosourea mutagenesis program and exhibited phenotypic variability, including reduced bone mass, multiple fractures, and early lethality. The causal gene was mapped to Chromosome 11 by linkage analysis, and a C-terminal frameshift mutation was identified in the Col1a1 (procollagen type I, alpha 1) gene as the cause of the disorder. Aga2 heterozygous animals had markedly increased bone turnover and a disrupted native collagen network. Further studies showed that abnormal proalpha1(I) chains accumulated intracellularly in Aga2/+ dermal fibroblasts and were poorly secreted extracellularly. This was associated with the induction of an endoplasmic reticulum stress-specific unfolded protein response involving upregulation of BiP, Hsp47, and Gadd153 with caspases-12 and -3 activation and apoptosis of osteoblasts both in vitro and in vivo. These studies resulted in the identification of a new model for Osteogenesis imperfecta, and identified a role for intracellular modulation of the endoplasmic reticulum stress-associated unfolded protein response machinery toward osteoblast apoptosis during the pathogenesis of disease.


Assuntos
Osteogênese Imperfeita/genética , Sequência de Aminoácidos , Animais , Apoptose , Sequência de Bases , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , DNA/genética , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Feminino , Mutação da Fase de Leitura , Genes Letais , Heterozigoto , Humanos , Masculino , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteogênese Imperfeita/metabolismo , Osteogênese Imperfeita/patologia , Fenótipo , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...