Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Hum Mol Genet ; 32(15): 2485-2501, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37171606

RESUMEN

ATRX is a chromatin remodelling ATPase that is involved in transcriptional regulation, DNA damage repair and heterochromatin maintenance. It has been widely studied for its role in ALT-positive cancers, but its role in neurological function remains elusive. Hypomorphic mutations in the X-linked ATRX gene cause a rare form of intellectual disability combined with alpha-thalassemia called ATR-X syndrome in hemizygous males. Clinical features also include facial dysmorphism, microcephaly, short stature, musculoskeletal defects and genital abnormalities. As complete deletion of ATRX in mice results in early embryonic lethality, the field has largely relied on conditional knockout models to assess the role of ATRX in multiple tissues. Given that null alleles are not found in patients, a more patient-relevant model was needed. Here, we have produced and characterized the first patient mutation knock-in model of ATR-X syndrome, carrying the most common causative mutation, R246C. This is one of a cluster of missense mutations located in the chromatin-binding domain and disrupts its function. The knock-in mice recapitulate several aspects of the patient disorder, including craniofacial defects, microcephaly, reduced body size and impaired neurological function. They provide a powerful model for understanding the molecular mechanisms underlying ATR-X syndrome and testing potential therapeutic strategies.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X , Microcefalia , Talasemia alfa , Animales , Masculino , Ratones , Talasemia alfa/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Microcefalia/genética , Mutación , Proteínas Nucleares/genética , Proteína Nuclear Ligada al Cromosoma X/genética , Humanos
2.
Blood Adv ; 5(20): 4167-4178, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34464976

RESUMEN

Ribosome dysfunction is implicated in multiple abnormal developmental and disease states in humans. Heterozygous germline mutations in genes encoding ribosomal proteins are found in most individuals with Diamond-Blackfan anemia (DBA), whereas somatic mutations have been implicated in a variety of cancers and other disorders. Ribosomal protein-deficient animal models show variable phenotypes and penetrance, similar to human patients with DBA. In this study, we characterized a novel ENU mouse mutant (Skax23m1Jus) with growth and skeletal defects, cardiac malformations, and increased mortality. After genetic mapping and whole-exome sequencing, we identified an intronic Rpl5 mutation, which segregated with all affected mice. This mutation was associated with decreased ribosome generation, consistent with Rpl5 haploinsufficiency. Rpl5Skax23-Jus/+ animals had a profound delay in erythroid maturation and increased mortality at embryonic day (E) 12.5, which improved by E14.5. Surviving mutant animals had macrocytic anemia at birth, as well as evidence of ventricular septal defect (VSD). Surviving adult and aged mice exhibited no hematopoietic defect or VSD. We propose that this novel Rpl5Skax23-Jus/+ mutant mouse will be useful in studying the factors influencing the variable penetrance that is observed in DBA.


Asunto(s)
Anemia de Diamond-Blackfan , Anemia de Diamond-Blackfan/genética , Animales , Haploinsuficiencia , Humanos , Ratones , Mutación , Proteínas Ribosómicas/genética
3.
Genome Res ; 30(4): 540-552, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32317254

RESUMEN

Mutations in X-linked methyl-CpG-binding protein 2 (MECP2) cause Rett syndrome (RTT). To identify functional pathways that could inform therapeutic entry points, we carried out a genetic screen for secondary mutations that improved phenotypes in Mecp2/Y mice after mutagenesis with N-ethyl-N-nitrosourea (ENU). Here, we report the isolation of 106 founder animals that show suppression of Mecp2-null traits from screening 3177 Mecp2/Y genomes. Whole-exome sequencing, genetic crosses, and association analysis identified 22 candidate genes. Additional lesions in these candidate genes or pathway components associate variant alleles with phenotypic improvement in 30 lines. A network analysis shows that 63% of the genes cluster into the functional categories of transcriptional repression, chromatin modification, or DNA repair, delineating a pathway relationship with MECP2. Many mutations lie in genes that modulate synaptic signaling or lipid homeostasis. Mutations in genes that function in the DNA damage response (DDR) also improve phenotypes in Mecp2/Y mice. Association analysis was successful in resolving combinatorial effects of multiple loci. One line, which carries a suppressor mutation in a gene required for cholesterol synthesis, Sqle, carries a second mutation in retinoblastoma binding protein 8, endonuclease (Rbbp8, also known as CtIP), which regulates a DDR choice in double-stranded break (DSB) repair. Cells from Mecp2/Y mice have increased DSBs, so this finding suggests that the balance between homology-directed repair and nonhomologous end joining is important for neuronal cells. In this and other lines, two suppressor mutations confer greater improvement than one alone, suggesting that combination therapies could be effective in RTT.


Asunto(s)
Daño del ADN , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Proteína 2 de Unión a Metil-CpG/genética , Síndrome de Rett/diagnóstico , Síndrome de Rett/genética , Supresión Genética , Alelos , Animales , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Genotipo , Homocigoto , Metabolismo de los Lípidos , Masculino , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Ratones Noqueados , Mutación , Fenotipo , Síndrome de Rett/metabolismo , Transducción de Señal , Secuenciación del Exoma
4.
Trends Genet ; 35(7): 489-500, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31130394

RESUMEN

Despite advances in chemotherapies that improve cancer survival, most patients who relapse succumb to the disease due to the presence of cancer stem cells (CSCs), which are highly chemoresistant. The pluripotency factor PR domain 14 (PRDM14) has a key role in initiating many types of cancer. Normally, PRDM14 uses epigenetic mechanisms to establish and maintain the pluripotency of embryonic cells, and its role in cancer is similar. This important link between cancer and induced pluripotency is a key revelation for how CSCs may form: pluripotency genes, such as PRDM14, can expand stem-like cells as they promote ongoing DNA damage. PRDM14 and its protein-binding partners, the ETO/CBFA2T family, are ideal candidates for eliminating CSCs from relevant cancers, preventing relapse and improving long-term survival.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Neoplasias/genética , Células Madre Pluripotentes/patología , Proteínas de Unión al ARN/fisiología , Factores de Transcripción/fisiología , Daño del ADN , Epigénesis Genética , Inestabilidad Genómica , Humanos , Neoplasias/patología
5.
Mol Cancer Res ; 17(7): 1468-1479, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31015254

RESUMEN

PR domain-containing 14 (Prdm14) is a pluripotency regulator central to embryonic stem cell identity and primordial germ cell specification. Genomic regions containing PRDM14 are often amplified leading to misexpression in human cancer. Prdm14 expression in mouse hematopoietic stem cells (HSC) leads to progenitor cell expansion prior to the development of T-cell acute lymphoblastic leukemia (T-ALL), consistent with PRDM14's role in cancer initiation. Here, we demonstrate mechanistic insight into PRDM14-driven leukemias in vivo. Mass spectrometry revealed novel PRDM14-protein interactions including histone H1, RNA-binding proteins, and the master hematopoietic regulator CBFA2T3. In mouse leukemic cells, CBFA2T3 and PRDM14 associate independently of the related ETO family member CBFA2T2, PRDM14's primary protein partner in pluripotent cells. CBFA2T3 plays crucial roles in HSC self-renewal and lineage commitment, and participates in oncogenic translocations in acute myeloid leukemia. These results suggest a model whereby PRDM14 recruits CBFA2T3 to DNA, leading to gene misregulation causing progenitor cell expansion and lineage perturbations preceding T-ALL development. Strikingly, Prdm14-induced T-ALL does not occur in mice deficient for Cbfa2t3, demonstrating that Cbfa2t3 is required for leukemogenesis. Moreover, T-ALL develops in Cbfa2t3 heterozygotes with a significantly longer latency, suggesting that PRDM14-associated T-ALL is sensitive to Cbfa2t3 levels. Our study highlights how an oncogenic protein uses a native protein in progenitor cells to initiate leukemia, providing insight into PRDM14-driven oncogenesis in other cell types. IMPLICATIONS: The pluripotency regulator PRDM14 requires the master hematopoietic regulator CBFA2T3 to initiate leukemia in progenitor cells, demonstrating an oncogenic role for CBFA2T3 and providing an avenue for targeting cancer-initiating cells.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Neoplásicas/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Animales , Metilación de ADN/genética , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología
6.
Emerg Top Life Sci ; 3(1): 1-10, 2019 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-33523198

RESUMEN

Model systems have played a large role in understanding human diseases and are instrumental in taking basic research findings to the clinic; however, for rare diseases, model systems play an even larger role. Here, we outline how model organisms are crucial for confirming causal associations, understanding functional mechanisms and developing therapies for disease. As diseases that have been studied extensively through genetics and molecular biology, cystic fibrosis and Rett syndrome are portrayed as primary examples of how genetic diagnosis, model organism development and therapies have led to improved patient health. Considering which model to use, yeast, worms, flies, fish, mice or larger animals requires a careful evaluation of experimental genetic tools and gene pathway conservation. Recent advances in genome editing will aid in confirming diagnoses and developing model systems for rare disease. Genetic or chemical screening for disease suppression may reveal functional pathway members and provide candidate entry points for developing therapies. Model organisms may also be used in drug discovery and as preclinical models as a prelude to testing treatments in patient populations. Now, model organisms will increasingly be used as platforms for understanding variation in rare disease severity and onset, thereby informing therapeutic intervention.

7.
Nat Genet ; 49(8): 1231-1238, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28650483

RESUMEN

Although next-generation sequencing has revolutionized the ability to associate variants with human diseases, diagnostic rates and development of new therapies are still limited by a lack of knowledge of the functions and pathobiological mechanisms of most genes. To address this challenge, the International Mouse Phenotyping Consortium is creating a genome- and phenome-wide catalog of gene function by characterizing new knockout-mouse strains across diverse biological systems through a broad set of standardized phenotyping tests. All mice will be readily available to the biomedical community. Analyzing the first 3,328 genes identified models for 360 diseases, including the first models, to our knowledge, for type C Bernard-Soulier, Bardet-Biedl-5 and Gordon Holmes syndromes. 90% of our phenotype annotations were novel, providing functional evidence for 1,092 genes and candidates in genetically uncharacterized diseases including arrhythmogenic right ventricular dysplasia 3. Finally, we describe our role in variant functional validation with The 100,000 Genomes Project and others.


Asunto(s)
Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Animales , Femenino , Enfermedades Genéticas Congénitas , Predisposición Genética a la Enfermedad , Humanos , Masculino , Ratones , Ratones Noqueados , Fenotipo
8.
Biol Open ; 5(11): 1595-1606, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27638768

RESUMEN

FOXF1 heterozygous point mutations and genomic deletions have been reported in newborns with the neonatally lethal lung developmental disorder, alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV). However, no gain-of-function mutations in FOXF1 have been identified yet in any human disease conditions. To study the effects of FOXF1 overexpression in lung development, we generated a Foxf1 overexpression mouse model by knocking-in a Cre-inducible Foxf1 allele into the ROSA26 (R26) locus. The mice were phenotyped using micro-computed tomography (micro-CT), head-out plethysmography, ChIP-seq and transcriptome analyses, immunohistochemistry, and lung histopathology. Thirty-five percent of heterozygous R26-Lox-Stop-Lox (LSL)-Foxf1 embryonic day (E)15.5 embryos exhibit subcutaneous edema, hemorrhages and die perinatally when bred to Tie2-cre mice, which targets Foxf1 overexpression to endothelial and hematopoietic cells. Histopathological and micro-CT evaluations revealed that R26Foxf1; Tie2-cre embryos have immature lungs with a diminished vascular network. Neonates exhibited respiratory deficits verified by detailed plethysmography studies. ChIP-seq and transcriptome analyses in E18.5 lungs identified Sox11, Ghr, Ednrb, and Slit2 as potential downstream targets of FOXF1. Our study shows that overexpression of the highly dosage-sensitive Foxf1 impairs lung development and causes vascular abnormalities. This has important clinical implications when considering potential gene therapy approaches to treat disorders of FOXF1 abnormal dosage, such as ACDMPV.

9.
Biol Open ; 5(5): 645-53, 2016 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-27106930

RESUMEN

PRDM14 is an epigenetic regulator known for maintaining embryonic stem cell identity and resetting potency in primordial germ cells. However, hematopoietic expression of Prdm14 at supraphysiological levels results in fully penetrant and rapid-onset T-cell acute lymphoblastic leukemia (T-ALL) in the mouse. Here, we show that PRDM14-induced T-ALLs are driven by NOTCH1, a frequently mutated driver of human T-ALL. Notch1 is activated in this murine model via RAG-dependent promoter deletions and subsequent production of truncated, ligand-independent protein from downstream regions of the Notch1 locus. These T-ALLs also have focal changes in H3K4me3 deposition at the Notch1 locus and global increases in both H3K4me1 and H3K4me3. Using a PRDM14-FLAG mouse model, we show that PRDM14 binds within an intron of Notch1 prior to leukemia development. Our data support the idea that PRDM14 binding promotes a chromatin state that allows access of the RAG recombinase complex to cryptic RAG signal sequences embedded at the Notch1 locus. Indeed, breeding into a RAG recombination-deficient background abrogates T-ALL development and prevents Notch1 deletions, while allowing for transient hematopoietic stem cell (HSC)-like pre-leukemia cell expansion. Together, our data suggest that PRDM14 expands a progenitor cell population while promoting a permissive epigenetic state for the creation of driver mutations (here, in Notch1), enabling cancer development through the misappropriation of endogenous cellular DNA recombination machinery.

10.
Curr Protoc Mouse Biol ; 5(2): 187-204, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26069083

RESUMEN

Cre-inducible mouse models are often utilized for the spatial and temporal expression of oncogenes. With the wide number of Cre recombinase lines available, inducible transgenesis represents a tractable approach to achieve discrete oncogene expression. Here, we describe a protocol for targeting Cre-inducible genes to the ubiquitously expressed ROSA26 locus. Gene targeting provides several advantages over standard transgenic techniques, including a known site of integration and previously characterized pattern of expression. Historically, an inherent instability of ROSA26 targeting vectors has hampered the efficiency of developing ROSA26 knock-in lines. In this protocol, we provide individual steps for utilizing Gateway recombination for cloning as well as detailed instructions for screening targeted ES cell clones. By following this protocol, one can achieve germline transmission of a ROSA26 knock-in line within several months.


Asunto(s)
Regulación de la Expresión Génica , Técnicas de Sustitución del Gen/métodos , Marcación de Gen/métodos , Integrasas/metabolismo , Ratones/genética , Proteínas Oncogénicas/genética , Animales , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Ratones/metabolismo , Ratones Transgénicos , Proteínas Oncogénicas/metabolismo , Especificidad de Órganos
11.
PLoS One ; 9(4): e94390, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24722050

RESUMEN

Alveolar Capillary Dysplasia with Misalignment of Pulmonary Veins (ACDMPV) is a developmental disorder of the lungs, primarily affecting their vasculature. FOXF1 haploinsufficiency due to heterozygous genomic deletions and point mutations have been reported in most patients with ACDMPV. The majority of mice with heterozygous loss-of-function of Foxf1 exhibit neonatal lethality with evidence of pulmonary hemorrhage in some of them. By comparing transcriptomes of human ACDMPV lungs with control lungs using expression arrays, we found that several genes and pathways involved in lung development, angiogenesis, and in pulmonary hypertension development, were deregulated. Similar transcriptional changes were found in lungs of the postnatal day 0.5 Foxf1+/- mice when compared to their wildtype littermate controls; 14 genes, COL15A1, COL18A1, COL6A2, ESM1, FSCN1, GRINA, IGFBP3, IL1B, MALL, NOS3, RASL11B, MATN2, PRKCDBP, and SIRPA, were found common to both ACDMPV and Foxf1 heterozygous lungs. Our results advance knowledge toward understanding of the molecular mechanism of ACDMPV, lung development, and its vasculature pathology. These data may also be useful for understanding etiologies of other lung disorders, e.g. pulmonary hypertension, bronchopulmonary dysplasia, or cancer.


Asunto(s)
Factores de Transcripción Forkhead/genética , Genes Letales , Pulmón/metabolismo , Síndrome de Circulación Fetal Persistente/genética , Alveolos Pulmonares/anomalías , Venas Pulmonares/metabolismo , Transcriptoma , Animales , Animales Recién Nacidos , Femenino , Factores de Transcripción Forkhead/deficiencia , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Heterocigoto , Humanos , Recién Nacido , Pulmón/anomalías , Pulmón/irrigación sanguínea , Masculino , Redes y Vías Metabólicas , Ratones , Ratones Noqueados , Síndrome de Circulación Fetal Persistente/metabolismo , Alveolos Pulmonares/irrigación sanguínea , Alveolos Pulmonares/metabolismo , Venas Pulmonares/anomalías
12.
Hum Mol Genet ; 23(7): 1687-99, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24203697

RESUMEN

Wnt signaling has been classified as canonical Wnt/ß-catenin-dependent or non-canonical planar cell polarity (PCP) pathway. Misregulation of either pathway is linked mainly to cancer or neural tube defects (NTDs), respectively. Both pathways seem to antagonize each other, and recent studies have implicated a number of molecular switches that activate one pathway while simultaneously inhibiting the other thereby partially mediating this antagonism. The lipoprotein receptor-related protein Lrp6 is crucial for the activation of the Wnt/ß-catenin pathway, but its function in Wnt/PCP signaling remains largely unknown. In this study, we investigate the role of Lrp6 as a molecular switch between both Wnt pathways in a novel ENU mouse mutant of Lrp6 (Skax26(m1Jus)) and in human NTDs. We demonstrate that Skax26(m1Jus) represents a hypermorphic allele of Lrp6 with increased Wnt canonical and abolished PCP-induced JNK activities. We also show that Lrp6(Skax26-Jus) genetically interacts with a PCP mutant (Vangl2(Lp)) where double heterozygotes showed an increased frequency of NTDs and defects in cochlear hair cells' polarity. Importantly, our study also demonstrates the association of rare and novel missense mutations in LRP6 that is an inhibitor rather than an activator of the PCP pathway with human NTDs. We show that three LRP6 mutations in NTDs led to a reduced Wnt canonical activity and enhanced PCP signaling. Our data confirm an inhibitory role of Lrp6 in PCP signaling in neurulation and indicate the importance of a tightly regulated and highly dosage-sensitive antagonism between both Wnt pathways in this process.


Asunto(s)
Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Defectos del Tubo Neural/genética , Proteínas Wnt/genética , Adolescente , Adulto , Animales , Secuencia de Bases , Polaridad Celular/genética , Niño , Cóclea/citología , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación Missense , Neurulación/genética , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN , Vía de Señalización Wnt , Adulto Joven , beta Catenina/genética
13.
Dis Model Mech ; 6(6): 1494-506, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24046360

RESUMEN

PRDM14 functions in embryonic stem cell (ESC) maintenance to promote the expression of pluripotency-associated genes while suppressing differentiation genes. Expression of PRDM14 is tightly regulated and typically limited to ESCs and primordial germ cells; however, aberrant expression is associated with tumor initiation in a wide variety of human cancers, including breast cancer and leukemia. Here, we describe the generation of a Cre-recombinase-inducible mouse model for the spatial and temporal control of Prdm14 misexpression [ROSA26 floxed-stop Prdm14 (R26PR)]. When R26PR is mated to either of two Cre lines, Mx1-cre or MMTV-cre, mice develop early-onset T-cell acute lymphoblastic leukemia (T-ALL) with median overall survival of 41 and 64 days for R26PR;Mx1-cre and R26PR;MMTV-cre, respectively. T-ALL is characterized by the accumulation of immature single-positive CD8 cells and their widespread infiltration. Leukemia is preceded by a dramatic expansion of cells resembling hematopoietic stem cells and lymphoid-committed progenitors prior to disease onset, accompanied by a blockage in B-cell differentiation at the early pro-B stage. Rapid-onset PRDM14-induced T-ALL requires factors that are present in stem and progenitor cells: R26PR;dLck-cre animals, which express Prdm14 starting at the double-positive stage of thymocyte development, do not develop disease. PRDM14-induced leukemic cells contain high levels of activated NOTCH1 and downstream NOTCH1 targets, including MYC and HES1, and are sensitive to pharmacological inhibition of NOTCH1 with the γ-secretase inhibitor DAPT. Greater than 50% of human T-ALLs harbor activating mutations in NOTCH1; thus, our model carries clinically relevant molecular aberrations. The penetrance, short latency and involvement of the NOTCH1 pathway will make this hematopoietic R26PR mouse model ideal for future studies on disease initiation, relapse and novel therapeutic drug combinations. Furthermore, breeding R26PR to additional Cre lines will allow for the continued development of novel cancer models.


Asunto(s)
Modelos Animales de Enfermedad , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Factores de Transcripción/genética , Animales , Diferenciación Celular , Proteínas de Unión al ADN , Ratones , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Proteínas de Unión al ARN , Factores de Transcripción/metabolismo
14.
BMC Dev Biol ; 13: 9, 2013 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-23521771

RESUMEN

BACKGROUND: Tenm4 is a mouse homolog of the Drosophila gene Tenascin-m (Ten-m (Odd oz)), which functions in motor neuron routing. Recently, a genome-wide association analysis for bipolar disorder identified a new susceptibility locus at TENM4 increasing the importance of understanding Tenm4. A series of Tenm4 mouse alleles showing a broad range of phenotypes were isolated after ENU mutagenesis. Here, we examine the timing and features of gastrulation failure in a loss of function allele. RESULTS: Embryonic mesoderm did not form in loss of function Tenm4m1/m1 mutant embryos. Genes normally expressed in embryonic mesoderm were not expressed in the mutant, the primitive streak did not form, and markers of the anteroposterior axis were not expressed or were mislocalized. The lack of embryonic mesoderm could not be attributed to poor proliferation of the epiblast, as normal numbers of dividing cells were observed. Epiblast cells maintained expression of Pou5f1 suggesting that they remain pluripotent, but they did not have the capacity to form any germ layer derivatives in teratomas, showing that the inability to induce mesoderm is cell autonomous. Misexpression of E-cadherin and N-cadherin suggest that the embryos did not undergo an epithelial-to-mesenchymal transition. In addition, Wnt signaling did not occur in the mutants, as assessed by the TOPGAL reporter assay, while a GSK3ß inhibitor partially rescued the mutant embryos, and rescued TOPGAL reporter expression. CONCLUSIONS: These data demonstrate that Tenm4 mutants fail to form a primitive streak and to induce embryonic mesoderm. Markers of anterior posterior patterning fail to be expressed or are mislocalized. Further, Tenm4 mutants lack the ability to differentiate in a cell autonomous manner. Together, our data suggest that embryos become impaired prior to E6.5 and as a result, Wnt signaling fails to occur; however, the involvement of other signaling pathways remains to be examined.


Asunto(s)
Proteínas de la Membrana/fisiología , Mesodermo/crecimiento & desarrollo , Alelos , Animales , Apoptosis , Cadherinas/metabolismo , Proliferación Celular , Gastrulación , Hibridación in Situ , Ratones , Transducción de Señal , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo
15.
Cancers (Basel) ; 4(4): 1050-1066, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23487523

RESUMEN

Aberrant expression and activation of oncogenes in somatic cells has been associated with cancer initiation. Required for reacquisition of pluripotency in the developing germ cell, PRDM14 initiates lymphoblastic leukemia when misexpressed in murine bone marrow. Activation of pluripotency in somatic cells can lead to aneuploidy and copy number alterations during iPS cell generation, and we hypothesized that PRDM14-induced lymphoblastic leukemias would demonstrate significant chromosomal damage. High-resolution oligo array comparative genomic hybridization demonstrated infrequent aneuploidy but frequent amplification and deletion, with amplifications occurring in a 5:1 ratio with deletions. Many deletions (i.e., Cdkn2a, Ebf1, Pax5, Ikzf1) involved B-cell development genes and tumor suppressor genes, recapitulating deletions occurring in human leukemia. Pathways opposing senescence were frequently deactivated via Cdkn2a deletion or Tbx2 amplification, with corollary gene expression. Additionally, gene expression studies of abnormal pre-leukemic B-precursors showed downregulation of genes involved in chromosomal stability (i.e., Xrcc6) and failure to upregulate DNA repair pathways. We propose a model of leukemogenesis, triggered by pluripotency genes like Prdm14, which involves ongoing DNA damage and failure to activate non-homologous end-joining secondary to aberrant gene expression.

16.
J Pathol ; 224(4): 540-52, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21674502

RESUMEN

Ribosomal stress is an important, yet poorly understood, mechanism that results in activation of the p53 tumour suppressor. We present a mutation in the ribosomal protein Rpl27a gene (sooty foot ataxia mice), isolated through a sensitized N-ethyl-N-nitrosourea (ENU) mutagenesis screen for p53 pathway defects, that shares striking phenotypic similarities with high p53 mouse models, including cerebellar ataxia, pancytopenia and epidermal hyperpigmentation. This phenocopy is rescued in a haploinsufficient p53 background. A detailed examination of the bone marrow in these mice identified reduced numbers of haematopoietic stem cells and a p53-dependent c-Kit down-regulation. These studies suggest that reduced Rpl27a increases p53 activity in vivo, further evident with a delay in tumorigenesis in mutant mice. Taken together, these data demonstrate that Rpl27a plays a crucial role in multiple tissues and that disruption of this ribosomal protein affects both development and transformation.


Asunto(s)
Ataxia Cerebelosa/genética , Proteínas Ribosómicas/genética , Proteína p53 Supresora de Tumor/metabolismo , Anemia/genética , Anemia/metabolismo , Animales , Peso Corporal/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Ataxia Cerebelosa/metabolismo , Ataxia Cerebelosa/patología , Modelos Animales de Enfermedad , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/metabolismo , Haploinsuficiencia/genética , Células Madre Hematopoyéticas/patología , Hiperpigmentación/genética , Hiperpigmentación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Pruebas de Mutagenicidad , Fenotipo , Proteínas Ribosómicas/metabolismo , Proteínas Ribosómicas/fisiología , Transducción de Señal/fisiología
17.
PLoS Genet ; 6(6): e1000985, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20548961

RESUMEN

Protein palmitoylation has emerged as an important mechanism for regulating protein trafficking, stability, and protein-protein interactions; however, its relevance to disease processes is not clear. Using a genome-wide, phenotype driven N-ethyl-N-nitrosourea-mediated mutagenesis screen, we identified mice with failure to thrive, shortened life span, skin and hair abnormalities including alopecia, severe osteoporosis, and systemic amyloidosis (both AA and AL amyloids depositions). Whole-genome homozygosity mapping with 295 SNP markers and fine mapping with an additional 50 SNPs localized the disease gene to chromosome 7 between 53.9 and 56.3 Mb. A nonsense mutation (c.1273A>T) was located in exon 12 of the Zdhhc13 gene (Zinc finger, DHHC domain containing 13), a gene coding for palmitoyl transferase. The mutation predicted a truncated protein (R425X), and real-time PCR showed markedly reduced Zdhhc13 mRNA. A second gene trap allele of Zdhhc13 has the same phenotypes, suggesting that this is a loss of function allele. This is the first report that palmitoyl transferase deficiency causes a severe phenotype, and it establishes a direct link between protein palmitoylation and regulation of diverse physiologic functions where its absence can result in profound disease pathology. This mouse model can be used to investigate mechanisms where improper palmitoylation leads to disease processes and to understand molecular mechanisms underlying human alopecia, osteoporosis, and amyloidosis and many other neurodegenerative diseases caused by protein misfolding and amyloidosis.


Asunto(s)
Aciltransferasas/genética , Alopecia/genética , Amiloidosis/genética , Mutación , Osteoporosis/genética , Aciltransferasas/metabolismo , Envejecimiento , Alopecia/metabolismo , Alopecia/patología , Amiloidosis/metabolismo , Amiloidosis/patología , Animales , Secuencia de Bases , Regulación de la Expresión Génica , Inmunohistoquímica , Ratones , Especificidad de Órganos , Osteoporosis/metabolismo , Osteoporosis/patología , Fenotipo
18.
PLoS One ; 3(11): e3823, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19043588

RESUMEN

BACKGROUND: AKXD recombinant inbred strains of mice have proven to be very useful in the identification of potential oncogenes and tumor suppressors involved in the development of lymphoid and myeloid malignancies. In these tumors, the hematopoietic insertion of an active AKV murine leukemia virus (MuLV) is associated with the onset of disease. Common sites of retroviral insertion (CIS) identify genes causally associated with the development or initiation of lymphoma. METHODOLOGY: In the present study, we analyzed a previously uncharacterized CIS, Ecotropic Viral Integration Site 32 (Evi32), which is located on mouse chromosome 1. We analyzed candidate genes in the region to identify those involved in Evi32 mediated oncogenesis. RESULTS: Here we show that proviral insertion at Evi32 correlates with significant overexpression of a putative transcription factor, PR-domain containing 14 (Prdm14). Tumors with insertions at Evi32 are consistently lymphoid in nature. Prdm14 is normally expressed early in embryonic development with the highest expression in undifferentiated embryonic stem (ES) cells. This study implicates Prdm14 as a proto-oncogene involved in lymphoblastic lymphoma formation.


Asunto(s)
Virus de la Leucemia Murina/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proto-Oncogenes , Factores de Transcripción/genética , Integración Viral , Animales , Cromosomas , Proteínas de Unión al ADN , Regulación Neoplásica de la Expresión Génica , Ratones , Mutagénesis Insercional , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/virología , Proto-Oncogenes Mas , Proteínas de Unión al ARN , Retroviridae/genética , Dedos de Zinc
19.
Blood ; 112(10): 4308-13, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18780836

RESUMEN

Mouse models have proven invaluable for understanding erythropoiesis. Here, we describe an autosomal recessive, inherited anemia in the mouse mutant hem6. Hematologic and transplantation analyses reveal a mild, congenital, hypochromic, microcytic anemia intrinsic to the hematopoietic system that is associated with a decreased red blood cell zinc protoporphyrin to heme ratio, indicative of porphyrin insufficiency. Intercross matings show that hem6 can suppress the porphyric phenotype of mice with erythropoietic protoporphyria (EPP). Furthermore, iron uptake studies in hem6 reticulocytes demonstrate defective incorporation of iron into heme that can be partially corrected by the addition of porphyrin precursors. Gene expression and enzymatic assays indicate that erythroid 5-aminolevulinic acid synthase (Alas2) is decreased in hem6 animals, suggesting a mechanism that could account for the anemia. Overall, these data lead to the hypothesis that hem6 encodes a protein that directly or indirectly regulates the expression of Alas2.


Asunto(s)
5-Aminolevulinato Sintetasa/biosíntesis , Anemia Hipocrómica/enzimología , Regulación Enzimológica de la Expresión Génica , Hematopoyesis , Protoporfiria Eritropoyética/enzimología , Reticulocitos/enzimología , 5-Aminolevulinato Sintetasa/genética , Anemia Hipocrómica/genética , Anemia Hipocrómica/patología , Animales , Cruzamientos Genéticos , Femenino , Regulación Enzimológica de la Expresión Génica/genética , Hematopoyesis/genética , Hemo/biosíntesis , Hemo/genética , Hierro/metabolismo , Masculino , Ratones , Ratones Mutantes , Protoporfiria Eritropoyética/genética , Protoporfiria Eritropoyética/patología , Reticulocitos/patología
20.
Cancer Res ; 67(21): 10138-47, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17974955

RESUMEN

Proper chromosome segregation in eukaryotes is driven by a complex superstructure called the mitotic spindle. Assembly, maintenance, and function of the spindle depend on centrosome migration, organization of microtubule arrays, and force generation by microtubule motors. Spindle pole migration and elongation are controlled by the unique balance of forces generated by antagonistic molecular motors that act upon microtubules of the mitotic spindle. Defects in components of this complex structure have been shown to lead to chromosome missegregation and genomic instability. Here, we show that overexpression of Eg5, a member of the Bim-C class of kinesin-related proteins, leads to disruption of normal spindle development, as we observe both monopolar and multipolar spindles in Eg5 transgenic mice. Our findings show that perturbation of the mitotic spindle leads to chromosomal missegregation and the accumulation of tetraploid cells. Aging of these mice revealed a higher incidence of tumor formation with a mixed array of tumor types appearing in mice ages 3 to 30 months with the mean age of 20 months. Analysis of the tumors revealed widespread aneuploidy and genetic instability, both hallmarks of nearly all solid tumors. Together with previous findings, our results indicate that Eg5 overexpression disrupts the unique balance of forces associated with normal spindle assembly and function, and thereby leads to the development of spindle defects, genetic instability, and tumors.


Asunto(s)
Inestabilidad Genómica , Cinesinas/fisiología , Neoplasias Experimentales/etiología , Animales , Segregación Cromosómica , ADN de Neoplasias/análisis , Cinesinas/genética , Ratones , Ratones Transgénicos , Neoplasias Experimentales/genética , Proteínas Proto-Oncogénicas c-pim-1/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Huso Acromático/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA