Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 55(12): 2336-2351.e12, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36462502

RESUMEN

Therapeutic promotion of intestinal regeneration holds great promise, but defining the cellular mechanisms that influence tissue regeneration remains an unmet challenge. To gain insight into the process of mucosal healing, we longitudinally examined the immune cell composition during intestinal damage and regeneration. B cells were the dominant cell type in the healing colon, and single-cell RNA sequencing (scRNA-seq) revealed expansion of an IFN-induced B cell subset during experimental mucosal healing that predominantly located in damaged areas and associated with colitis severity. B cell depletion accelerated recovery upon injury, decreased epithelial ulceration, and enhanced gene expression programs associated with tissue remodeling. scRNA-seq from the epithelial and stromal compartments combined with spatial transcriptomics and multiplex immunostaining showed that B cells decreased interactions between stromal and epithelial cells during mucosal healing. Activated B cells disrupted the epithelial-stromal cross talk required for organoid survival. Thus, B cell expansion during injury impairs epithelial-stromal cell interactions required for mucosal healing, with implications for the treatment of IBD.


Asunto(s)
Colitis , Mucosa Intestinal , Animales , Cicatrización de Heridas , Células Epiteliales/metabolismo , Epitelio , Modelos Animales de Enfermedad
2.
Hum Mol Genet ; 30(21): 2012-2026, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34169315

RESUMEN

Deoxyguanosine kinase (DGUOK) deficiency causes mtDNA depletion and mitochondrial dysfunction. We reported long survival of DGUOK knockout (Dguok-/-) mice despite low (<5%) mtDNA content in liver tissue. However, the molecular mechanisms enabling the extended survival remain unknown. Using transcriptomics, proteomics and metabolomics followed by in vitro assays, we aimed to identify the molecular pathways involved in the extended survival of the Dguok-/- mice. At the early stage, the serine synthesis and folate cycle were activated but declined later. Increased activity of the mitochondrial citric acid cycle (TCA cycle) and the urea cycle and degradation of branched chain amino acids were hallmarks of the extended lifespan in DGUOK deficiency. Furthermore, the increased synthesis of TCA cycle intermediates was supported by coordination of two pyruvate kinase genes, PKLR and PKM, indicating a central coordinating role of pyruvate kinases to support the long-term survival in mitochondrial dysfunction.


Asunto(s)
Adaptación Biológica , Metabolismo Energético , Mitocondrias/genética , Mitocondrias/metabolismo , Piruvato Quinasa/genética , Piruvato Quinasa/metabolismo , Aminoácidos/metabolismo , Animales , Supervivencia Celular/genética , Ciclo del Ácido Cítrico , Ciclooxigenasa 1 , ADN Mitocondrial/genética , Metabolismo de los Lípidos , Hígado/metabolismo , Proteínas de la Membrana , Redes y Vías Metabólicas , Ratones , Ratones Noqueados , Fosforilación Oxidativa
3.
Hum Mol Genet ; 28(17): 2874-2884, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31127938

RESUMEN

Deoxyguanosine kinase (DGUOK) provides guanosine and adenosine nucleotides for mitochondrial DNA (mtDNA) replication, and its deficiency in humans leads to hepatocerebral mtDNA depletion syndrome or to isolated hepatic disease. There are poor treatment options for DGUOK deficiency and the aim of this study was to generate a model for further studies of the disease that could reveal novel treatment strategies. We report a Dguok-deficient mouse strain that, similar to humans, is most severely affected in the liver. The Dguok complete knockout mice (Dguok-/-) were born normal, but began to lose weight at week 6. A change of fur color from black to blueish grey started at week 16 and was complete at week 20. The movements and behavior were indistinguishable compared to wild-type (wt) mice. A decrease of mtDNA copy number occurred in multiple tissues, with the liver being the most severely affected. The mtDNA-encoded protein cytochrome c oxidase was much lower in Dguok-/- liver tissue than in the wt, whereas the expression of the nuclear-encoded succinate dehydrogenase complex subunit A was unaffected. Histopathology showed severe alterations and immunohistochemistry showed signs of both oxidative stress and regeneration in Dguok-/- liver. The subcutaneous fat layer was undetectable in Dguok-/-, which, in addition to gene expression analysis, indicated an altered lipid metabolism. We conclude that Dguok has a major role for the synthesis of deoxyribonucleotides for mtDNA replication particularly in the liver, similar to the human disorder. Our data also show a catabolic lipid metabolism in liver tissue of Dguok-/-.


Asunto(s)
ADN Mitocondrial , Metabolismo de los Lípidos , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Animales , Biomarcadores , Modelos Animales de Enfermedad , Femenino , Dosificación de Gen , Perfilación de la Expresión Génica , Marcación de Gen , Sitios Genéticos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Fenotipo , Grasa Subcutánea/metabolismo , Transcriptoma
4.
Exp Cell Res ; 395(1): 112156, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32707133

RESUMEN

Hyperactivated Notch signalling has been implicated in breast cancer, but how elevated levels of Notch signalling contribute to mammary dysplasia and tumorigenesis is not fully understood. In this study, we express an activated form of Notch1 in the mouse mammary luminal lineage and analyse the consequences for tumour formation and the transcriptomic landscape in the luminal lineage. Simultaneous conditional activation of a Notch1 intracellular domain (Notch1 ICD) and EGFP in the luminal lineage was achieved by removal of a stop cassette by CRE-recombinase expression from the whey acidic protein (WAP) promoter. Mice in which Notch1 ICD was activated in the luminal lineage (WAP-CRE;R26-N1ICD mice) exhibit ductal hyperplasia after lactation with an increase in branching frequency and in the number of side-branch ends in the ductal tree. A subset of the mice developed mammary tumours and the majority of the tumour cells expressed EGFP (as a proxy for Notch1 ICD), indicating that the tumours originate from the Notch1 ICD-expressing cells. Single-cell transcriptome analysis of the EGFP-positive mammary cells identified six subtypes of luminal cells. The same six subtypes were found in control mice (WAP-CRE;R26-tdTomato mice expressing the tdTomato reporter from WAP-CRE-mediated activation), but the proportion of cells in the various subtypes differed between the WAP-CRE;R26-N1ICD and control WAP-CRE;R26-tdTomato mice. In conclusion, we show that Notch1 ICD expression in the luminal lineage produces a ductal hyperplasia and branching phenotype accompanied by altered luminal cell subtype partitioning.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Hiperplasia/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Regiones Promotoras Genéticas/genética , Animales , Femenino , Glándulas Mamarias Animales/citología , Neoplasias Mamarias Animales/patología , Ratones Transgénicos , Fenobarbital/metabolismo , Transducción de Señal/fisiología
5.
Int J Cancer ; 146(4): 1125-1138, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31219615

RESUMEN

Increased expression of GLI1, the main Hedgehog signalling pathway effector, is related to unfavourable prognosis and progressive disease of certain breast cancer subtypes. We used conditional transgenic mice induced to overexpress GLI1 in the mammary epithelium either alone or in combination with deletion of one Trp53 allele to address the role of elevated GLI1 expression in breast tumour initiation and progression. Induced GLI1 expression facilitates mammary gland tumour formation and this was further increased upon heterozygous deletion of Trp53. The GLI1-induced primary tumours were of different murine molecular subtypes, including Normal-likeEx , Class8Ex , Claudin-LowEx and Erbb2-likeEx . The gene expression profiles of some of the tumours correlated well with the PAM50 subtypes for human breast cancer. Whole-exome sequencing revealed somatic mutation profiles with only little overlap between the primary tumours. Orthotopically serially transplanted GLI1-induced tumours maintained the main morphological characteristics of the primary tumours for ≥10 generations. Independent of Trp53 status and molecular subtype, the serially transplanted GLI1-induced tumours were able to grow both in the absence of transgenic GLI1 expression and in the presence of the GLI1 inhibitor GANT61. These data suggest that elevated GLI1 expression has a determinant role in tumour initiation; however, additional genetic events are required for tumour progression.


Asunto(s)
Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Proteína con Dedos de Zinc GLI1/genética , Animales , Femenino , Expresión Génica , Heterogeneidad Genética , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Proteína con Dedos de Zinc GLI1/biosíntesis
6.
Breast Cancer Res ; 21(1): 85, 2019 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-31366361

RESUMEN

BACKGROUND: Claudin-low breast cancer is a molecular subtype associated with poor prognosis and without targeted treatment options. The claudin-low subtype is defined by certain biological characteristics, some of which may be clinically actionable, such as high immunogenicity. In mice, the medroxyprogesterone acetate (MPA) and 7,12-dimethylbenzanthracene (DMBA)-induced mammary tumor model yields a heterogeneous set of tumors, a subset of which display claudin-low features. Neither the genomic characteristics of MPA/DMBA-induced claudin-low tumors nor those of human claudin-low breast tumors have been thoroughly explored. METHODS: The transcriptomic characteristics and subtypes of MPA/DMBA-induced mouse mammary tumors were determined using gene expression microarrays. Somatic mutations and copy number aberrations in MPA/DMBA-induced tumors were identified from whole exome sequencing data. A publicly available dataset was queried to explore the genomic characteristics of human claudin-low breast cancer and to validate findings in the murine tumors. RESULTS: Half of MPA/DMBA-induced tumors showed a claudin-low-like subtype. All tumors carried mutations in known driver genes. While the specific genes carrying mutations varied between tumors, there was a consistent mutational signature with an overweight of T>A transversions in TG dinucleotides. Most tumors carried copy number aberrations with a potential oncogenic driver effect. Overall, several genomic events were observed recurrently; however, none accurately delineated claudin-low-like tumors. Human claudin-low breast cancers carried a distinct set of genomic characteristics, in particular a relatively low burden of mutations and copy number aberrations. The gene expression characteristics of claudin-low-like MPA/DMBA-induced tumors accurately reflected those of human claudin-low tumors, including epithelial-mesenchymal transition phenotype, high level of immune activation, and low degree of differentiation. There was an elevated expression of the immunosuppressive genes PTGS2 (encoding COX-2) and CD274 (encoding PD-L1) in human and murine claudin-low tumors. CONCLUSIONS: Our findings show that the claudin-low breast cancer subtype is not demarcated by specific genomic aberrations, but carries potentially targetable characteristics warranting further research.


Asunto(s)
Claudinas/metabolismo , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Oncogenes , Transcriptoma , Animales , Biopsia , Variaciones en el Número de Copia de ADN , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Neoplasias Mamarias Animales/patología , Ratones , Ratones Transgénicos , Mutación
7.
Dev Biol ; 429(1): 132-146, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28688895

RESUMEN

Suppressor of Fused (SUFU) is an essential negative regulator of the Hedgehog (HH) pathway and involved in GLI transcription factor regulation. Due to early embryonic lethality of Sufu-/- mice, investigations of SUFU's role later in development are limited to conditional, tissue-specific knockout models. In this study we developed a mouse model (SufuEx456(fl)/Ex456(fl)) with hypomorphic features where embryos were viable up to E18.5, although with a spectrum of developmental defects of varying severity, including polydactyly, exencephaly and omphalocele. Development of certain tissues, like the skeleton, was more affected than that of others such as skin, which remained largely normal. Interestingly, no apparent changes in the dorso-ventral patterning of the neural tube at E9.0 could be seen. Thus, this model provides an opportunity to globally study SUFU's molecular function in organogenesis beyond E9.5. Molecularly, SufuEx456(fl)/Ex456(fl) embryos displayed aberrant mRNA splicing and drastically reduced levels of Sufu wild-type mRNA and SUFU protein in all tissues. As a consequence, at E9.5 the levels of all three different GLI proteins were reduced. Interestingly, despite the reduction of GLI3 protein levels, the critical ratio of the GLI3 full-length transcriptional activator versus GLI3 truncated repressor remained unchanged compared to wild-type embryos. This suggests that the limited amount of SUFU protein present is sufficient for GLI processing but not for stabilization. Our data demonstrate that tissue development is differentially affected in response to the reduced SUFU levels, providing novel insight regarding the requirements of different levels of SUFU for proper organogenesis.


Asunto(s)
Organogénesis , Proteínas Represoras/metabolismo , Alelos , Animales , Tipificación del Cuerpo/genética , Embrión de Mamíferos/metabolismo , Exones/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Homocigoto , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Modelos Animales , Tubo Neural/embriología , Tubo Neural/metabolismo , Organogénesis/genética , Mutación Puntual/genética , Sitios de Empalme de ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética
8.
Biol Res ; 51(1): 3, 2018 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-29316982

RESUMEN

BACKGROUND: The WNT pathway regulates intestinal stem cells and is frequently disrupted in intestinal adenomas. The pathway contains several potential biotargets for interference, including the poly-ADP ribosyltransferase enzymes tankyrase1 and 2. LGR5 is a known WNT pathway target gene and marker of intestinal stem cells. The LGR5+ stem cells are located in the crypt base and capable of regenerating all intestinal epithelial cell lineages. RESULTS: We treated Lgr5-EGFP-Ires-CreERT2;R26R-Confetti mice with the tankyrase inhibitor G007-LK for up to 3 weeks to assess the effect on duodenal stem cell homeostasis and on the integrity of intestinal epithelium. At the administered doses, G007-LK treatment inhibited WNT signalling in LGR5+ stem cells and reduced the number and distribution of cells traced from duodenal LGR5+ stem cells. However, the gross morphology of the duodenum remained unaltered and G007-LK-treated mice showed no signs of weight loss or any other visible morphological changes. The inhibitory effect on LGR5+ stem cell proliferation was reversible. CONCLUSION: We show that the tankyrase inhibitor G007-LK is well tolerated by the mice, although proliferation of the LGR5+ intestinal stem cells was inhibited. Our observations suggest the presence of a tankyrase inhibitor-resistant cell population in the duodenum, able to rescue tissue integrity in the presence of G007-LK-mediated inhibition of the WNT signalling dependent LGR5+ intestinal epithelial stem cells.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Duodeno/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Células Madre/efectos de los fármacos , Sulfonas/farmacología , Tanquirasas/antagonistas & inhibidores , Triazoles/farmacología , Animales , Duodeno/citología , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Microscopía Confocal , Receptores Acoplados a Proteínas G/genética , Sulfonas/farmacocinética , Tanquirasas/farmacocinética , Tanquirasas/farmacología , Triazoles/farmacocinética
9.
Exp Mol Pathol ; 102(1): 128-132, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28038939

RESUMEN

Mus pahari is a wild-derived, inbred mouse strain. M. pahari colony managers observed fragility of this strain's skin resulting in separation of tail skin from the mouse if handled incorrectly. Tail skin tension testing of M. pahari resulted in significantly lowered force threshold for caudal skin rupture and loss in comparison to closely related inbred mouse species and subspecies and even more than a model for junctional epidermolysis bullosa. Histologically, the tail skin separated at the subdermal level with the dermis firmly attached to the epidermis, excluding the epidermolysis bullosa complex of diseases. The dermal collagen bundles were abnormally thickened and branched. Elastin fiber deposition was focally altered in the dermis adjacent to the hair follicle. Collagens present in the skin could not be differentiated between the species in protein gels following digestion with pepsin. Together these data suggest that M. pahari have altered extracellular matrix development resulting in separation of the skin below the level of the dermis with moderate force similar to the African spiny mouse (Acomys spp.).


Asunto(s)
Piel/metabolismo , Piel/fisiopatología , Cola (estructura animal) , Animales , Colágeno/metabolismo , Dermis/metabolismo , Dermis/patología , Dermis/fisiopatología , Elasticidad , Elastina/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos , Piel/patología , Especificidad de la Especie
10.
Dev Biol ; 404(2): 35-48, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26003047

RESUMEN

LGR5 is a known marker of embryonic and adult stem cells in several tissues. In a mouse model, Lgr5+ cells have shown tumour-initiating properties, while in human cancers, such as basal cell carcinoma and colon cancer, LGR5 expression levels are increased: however, the effect of increased LGR5 expression is not fully understood. To study the effects of elevated LGR5 expression levels we generated a novel tetracycline-responsive, conditional transgenic mouse line expressing human LGR5, designated TRELGR5. In this transgenic line, LGR5 expression can be induced in any tissue depending on the expression pattern of the chosen transcriptional regulator. For the current study, we used transgenic mice with a tetracycline-regulated transcriptional transactivator linked to the bovine keratin 5 promoter (K5tTA) to drive expression of LGR5 in the epidermis. As expected, expression of human LGR5 was induced in the skin of double transgenic mice (K5tTA;TRELGR5). Inducing LGR5 expression during embryogenesis and early development resulted in macroscopically and microscopically detectable phenotypic changes, including kink tail, sparse fur coat and enlarged sebaceous glands. The fur and sebaceous gland phenotypes were reversible upon discontinued expression of transgenic LGR5, but this was not observed for the kink tail phenotype. There were no apparent phenotypic changes if LGR5 expression was induced at three weeks of age. The results demonstrate that increased expression of LGR5 during embryogenesis and the neonatal period alter skin development and homeostasis.


Asunto(s)
Epidermis/embriología , Folículo Piloso/embriología , Receptores Acoplados a Proteínas G/genética , Glándulas Sebáceas/embriología , Animales , Biomarcadores , Bovinos , Diferenciación Celular , Línea Celular , Regulación de la Expresión Génica , Humanos , Queratina-15/genética , Queratina-6/biosíntesis , Ratones , Ratones Transgénicos , Neoplasias/genética , Permeabilidad , Regiones Promotoras Genéticas , Receptores Acoplados a Proteínas G/biosíntesis , Elementos de Respuesta/genética , Células Madre/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt-5a
11.
J Biol Chem ; 289(47): 32835-44, 2014 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-25296759

RESUMEN

Mitochondrial DNA depletion caused by thymidine kinase 2 (TK2) deficiency can be compensated by a nucleoside kinase from Drosophila melanogaster (Dm-dNK) in mice. We show that transgene expression of Dm-dNK in Tk2 knock-out (Tk2(-/-)) mice extended the life span of Tk2(-/-) mice from 3 weeks to at least 20 months. The Dm-dNK(+/-)Tk2(-/-) mice maintained normal mitochondrial DNA levels throughout the observation time. A significant difference in total body weight due to the reduction of subcutaneous and visceral fat in the Dm-dNK(+/-)Tk2(-/-) mice was the only visible difference compared with control mice. This indicates an effect on fat metabolism mediated through residual Tk2 deficiency because Dm-dNK expression was low in both liver and fat tissues. Dm-dNK expression led to increased dNTP pools and an increase in the catabolism of purine and pyrimidine nucleotides but these alterations did not apparently affect the mice during the 20 months of observation. In conclusion, Dm-dNK expression in the cell nucleus expanded the total dNTP pools to levels required for efficient mitochondrial DNA synthesis, thereby compensated the Tk2 deficiency, during a normal life span of the mice. The Dm-dNK(+/-) mouse serves as a model for nucleoside gene or enzyme substitutions, nucleotide imbalances, and dNTP alterations in different tissues.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Nucleótidos/metabolismo , Fosfotransferasas/metabolismo , Timidina Quinasa/metabolismo , Tejido Adiposo/metabolismo , Animales , Western Blotting , Peso Corporal/genética , ADN Mitocondrial/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulación Enzimológica de la Expresión Génica , Inmunohistoquímica , Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Mitocondrias/genética , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Mutación , Nucleótidos/genética , Fosfotransferasas/genética , Análisis de Supervivencia , Timidina Quinasa/deficiencia , Timidina Quinasa/genética , Factores de Tiempo
12.
Biochem Biophys Res Commun ; 464(3): 737-42, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26187672

RESUMEN

Air-filled polyvinyl alcohol microbubbles (PVA-MBs) were recently introduced as a contrast agent for ultrasound imaging. In the present study, we explore the possibility of extending their application in multimodal imaging by labeling them with a near infrared (NIR) fluorophore, VivoTag-680. PVA-MBs were injected intravenously into FVB/N female mice and their dynamic biodistribution over 24 h was determined by 3D-fluorescence imaging co-registered with 3D-µCT imaging, to verify the anatomic location. To further confirm the biodistribution results from in vivo imaging, organs were removed and examined histologically using bright field and fluorescence microscopy. Fluorescence imaging detected PVA-MB accumulation in the lungs within the first 30 min post-injection. Redistribution to a low extent was observed in liver and kidneys at 4 h, and to a high extent mainly in the liver and spleen at 24 h. Histology confirmed PVA-MB localization in lung capillaries and macrophages. In the liver, they were associated with Kupffer cells; in the spleen, they were located mostly within the marginal-zone. Occasional MBs were observed in the kidney glomeruli and interstitium. The potential application of PVA-MBs as a contrast agent was also studied using ultrasound (US) imaging in subcutaneous and orthotopic pancreatic cancer mouse models, to visualize blood flow within the tumor mass. In conclusion, this study showed that PVA-MBs are useful as a contrast agent for multimodal imaging.


Asunto(s)
Medios de Contraste , Colorantes Fluorescentes , Microburbujas , Imagen Multimodal/métodos , Animales , Línea Celular Tumoral , Femenino , Xenoinjertos , Humanos , Imagenología Tridimensional , Ratones , Ratones Endogámicos C57BL , Imagen Óptica , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/diagnóstico por imagen , Ultrasonografía , Microtomografía por Rayos X
13.
J Biol Chem ; 288(7): 5072-9, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23288848

RESUMEN

A strategy to reverse the symptoms of thymidine kinase 2 (TK2) deficiency in a mouse model was investigated. The nucleoside kinase from Drosophila melanogaster (Dm-dNK) was expressed in TK2-deficient mice that have been shown to present with a severe phenotype caused by mitochondrial DNA depletion. The Dm-dNK(+/-) transgenic mice were shown to be able to rescue the TK2-deficient mice. The Dm-dNK(+/-)TK2(-/-) mice were normal as judged by growth and behavior during the observation time of 6 months. The Dm-dNK-expressing mice showed a substantial increase in thymidine-phosphorylating activity in investigated tissues. The Dm-dNK expression also resulted in highly elevated dTTP pools. The dTTP pool alterations did not cause specific mitochondrial DNA mutations or deletions when 6-month-old mice were analyzed. The mitochondrial DNA was also detected at normal levels. In conclusion, the Dm-dNK(+/-)TK2(-/-) mouse model illustrates how dTMP synthesized in the cell nucleus can compensate for loss of intramitochondrial dTMP synthesis in differentiated tissue. The data presented open new possibilities to treat the severe symptoms of TK2 deficiency.


Asunto(s)
Drosophila melanogaster/metabolismo , Regulación Enzimológica de la Expresión Génica , Mitocondrias/enzimología , Fosfotransferasas/metabolismo , Timidina Quinasa/metabolismo , Transgenes , Animales , Animales Modificados Genéticamente , ADN Mitocondrial/metabolismo , Genotipo , Ratones , Ratones Transgénicos , Nucleótidos/genética , Mutación Puntual
14.
Toxicol Appl Pharmacol ; 266(2): 289-97, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23153559

RESUMEN

An accurate assessment of the carcinogenic potential of chemicals and pharmaceutical drugs is essential to protect humans and the environment. Therefore, substances are extensively tested before they are marketed to the public. Currently, the rodent two-year bioassay is still routinely used to assess the carcinogenic potential of substances. However, over time it has become clear that this assay yields false positive results and also has several economic and ethical drawbacks including the use of large numbers of animals, the long duration, and the high cost. The need for a suitable alternative assay is therefore high. Previously, we have proposed the Xpa*p53 mouse model as a very suitable alternative to the two-year bioassay. We now show that the Xpc*p53 mouse model preserves all the beneficial traits of the Xpa*p53 model for sub-chronic carcinogen identification and can identify both genotoxic and non-genotoxic carcinogens. Moreover, Xpc*p53 mice appear to be more responsive than Xpa*p53 mice towards several genotoxic and non-genotoxic carcinogens. Furthermore, Xpc*p53 mice are far less sensitive than Xpa*p53 mice for the toxic activity of DNA damaging agents and as such clearly respond in a similar way as wild type mice do. These advantageous traits of the Xpc*p53 model make it a better alternative for in vivo carcinogen testing than Xpa*p53. This pilot study suggests that Xpc*p53 mice are suited for routine sub-chronic testing of both genotoxic and non-genotoxic carcinogens and as such represent a suitable alternative to possibly replace the murine life time cancer bioassay.


Asunto(s)
Carcinógenos/toxicidad , Proteínas de Unión al ADN/genética , Genes p53/genética , Mutágenos/toxicidad , Proteína de la Xerodermia Pigmentosa del Grupo A/genética , Animales , Pruebas de Carcinogenicidad/métodos , Daño del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas de Mutagenicidad/métodos , Proyectos Piloto
15.
PLoS One ; 18(10): e0285242, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37796969

RESUMEN

Deficiency in thymidine kinase 2 (TK2) causes mitochondrial DNA depletion. Liver mitochondria are severely affected in Tk2 complete knockout models and have been suggested to play a role in the pathogenesis of the Tk2 knockout phenotype, characterized by loss of hypodermal fat tissue, growth retardation and reduced life span. Here we report a liver specific Tk2 knockout (KO) model to further study mechanisms contributing to the phenotypic changes associated with Tk2 deficiency. Interestingly, the liver specific Tk2 KO mice had a normal life span despite a much lower mtDNA level in liver tissue. Mitochondrial DNA encoded peptide COXI did not differ between the Tk2 KO and control mice. However, the relative liver weight was significantly increased in the male Tk2 KO mouse model. Histology analysis indicated an increased lipid accumulation. We conclude that other enzyme activities can partly compensate Tk2 deficiency to maintain mtDNA at a low but stable level throughout the life span of the liver specific Tk2 KO mice. The lower level of mtDNA was sufficient for survival but led to an abnormal lipid accumulation in liver tissue.


Asunto(s)
ADN Mitocondrial , Timidina Quinasa , Ratones , Masculino , Animales , Timidina Quinasa/genética , ADN Mitocondrial/genética , Ratones Noqueados , Hígado , Lípidos
16.
Viruses ; 15(12)2023 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-38140691

RESUMEN

Salmonid alphavirus strain 3 is responsible for outbreaks of pancreas disease in salmon and rainbow trout in Norway. Although the extensive amount of research on SAV3 focused mainly on the heart and pancreas (of clinical importance), tropism and pathogenesis studies of the virus in other salmon tissues are limited. Here, we used a combination of RT-qPCR (Q_nsp1 gene) and in situ hybridization (RNAscope®) to demonstrate the tropism of SAV3 in situ in tissues of Atlantic salmon, employing a challenge model (by cohabitation). In addition, as previous results suggested that the pseudobranch may harbor the virus, the change in the expression of different immune genes upon SAV3 infection (RT-qPCR) was focused on the pseudobranch in this study. In situ hybridization detected SAV3 in different tissues of Atlantic salmon during the acute phase of the infection, with the heart ventricle showing the most extensive infection. Furthermore, the detection of the virus in different adipose tissues associated with the internal organs of the salmon suggests a specific affinity of SAV3 to adipocyte components. The inconsistent immune response to SAV3 in the pseudobranch after infection did not mitigate the infection in that tissue and is probably responsible for the persistent low infection at 4 weeks post-challenge. The early detection of SAV3 in the pseudobranch after infection, along with the persistent low infection over the experimental infection course, suggests a pivotal role of the pseudobranch in SAV3 pathogenesis in Atlantic salmon.


Asunto(s)
Infecciones por Alphavirus , Alphavirus , Enfermedades de los Peces , Salmo salar , Animales , Alphavirus/genética , Corazón
17.
Front Immunol ; 14: 1158077, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37180109

RESUMEN

Many sialic acid-binding viruses express a receptor-destroying enzyme (RDE) that removes the virus-targeted receptor and limits viral interactions with the host cell surface. Despite a growing appreciation of how the viral RDE promotes viral fitness, little is known about its direct effects on the host. Infectious salmon anemia virus (ISAV) attaches to 4-O-acetylated sialic acids on Atlantic salmon epithelial, endothelial, and red blood cell surfaces. ISAV receptor binding and destruction are effectuated by the same molecule, the haemagglutinin esterase (HE). We recently discovered a global loss of vascular 4-O-acetylated sialic acids in ISAV-infected fish. The loss correlated with the expression of viral proteins, giving rise to the hypothesis that it was mediated by the HE. Here, we report that the ISAV receptor is also progressively lost from circulating erythrocytes in infected fish. Furthermore, salmon erythrocytes exposed to ISAV ex vivo lost their capacity to bind new ISAV particles. The loss of ISAV binding was not associated with receptor saturation. Moreover, upon loss of the ISAV receptor, erythrocyte surfaces became more available to the lectin wheat germ agglutinin, suggesting a potential to alter interactions with endogenous lectins of similar specificity. The pruning of erythrocyte surfaces was inhibited by an antibody that prevented ISAV attachment. Furthermore, recombinant HE, but not an esterase-silenced mutant, was sufficient to induce the observed surface modulation. This links the ISAV-induced erythrocyte modulation to the hydrolytic activity of the HE and shows that the observed effects are not mediated by endogenous esterases. Our findings are the first to directly link a viral RDE to extensive cell surface modulation in infected individuals. This raises the questions of whether other sialic acid-binding viruses that express RDEs affect host cells to a similar extent, and if such RDE-mediated cell surface modulation influences host biological functions with relevance to viral disease.


Asunto(s)
Isavirus , Salmo salar , Animales , Isavirus/fisiología , Ácidos Siálicos , Ácido N-Acetilneuramínico , Esterasas , Eritrocitos
18.
Proteomics ; 12(13): 2149-57, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22807454

RESUMEN

Identification of biomarkers for early breast cancer detection in blood is a challenging task, since breast cancer is a heterogeneous disease with a wide range of tumor subtypes. This is envisioned to result in differences in serum protein levels. The p53(R270H/+) WAPCre mouse model is unique in that these mice spontaneously develop both ER- and ER+ tumors, in proportions comparable to humans. Therefore, these mice provide a well-suited model system to identify human relevant biomarkers for early breast cancer detection that are additionally specific for different tumor subtypes. Mammary gland tumors were obtained from p53(R270H/+) WAPCre mice and cellular origin, ER, and HER2 status were characterized. We compared gene expression profiles for tumors with different characteristics versus control tissue, and determined genes differentially expressed across tumor subtypes. By using literature data (Gene Ontology, UniProt, and Human Plasma Proteome), we further identified protein candidate biomarkers for blood-based detection of breast cancer. Functional overrepresentation analysis (using Gene Ontology, MSigDB, BioGPS, Cancer GeneSigDB, and proteomics literature data) showed enrichment for several processes relevant for human breast cancer. Finally, Human Protein Atlas data were used to obtain a prioritized list of 16 potential biomarkers that should facilitate further studies on blood-based breast cancer detection in humans.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/sangre , Neoplasias de la Mama/genética , Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Genómica/métodos , Proteínas/genética , Animales , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/sangre , Proteínas Sanguíneas/análisis , Proteínas Sanguíneas/genética , Mama/patología , Neoplasias de la Mama/patología , Femenino , Humanos , Ratones , Proteínas/análisis , Transcriptoma/métodos
19.
Am J Pathol ; 179(4): 1616-22, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21854749

RESUMEN

The TP53 tumor suppressor gene is mutated in the majority of human cancers. Inactivation of p53 in a variety of animal models results in early-onset tumorigenesis, reflecting the importance of p53 as a gatekeeper tumor suppressor. We generated a mutant Tp53 allele in the rat using a target-selected mutagenesis approach. Here, we report that homozygosity for this allele results in complete loss of p53 function. Homozygous mutant rats predominantly develop sarcomas with an onset of 4 months of age with a high occurrence of pulmonary metastases. Heterozygous rats develop sarcomas starting at 8 months of age. Molecular analysis revealed that these tumors exhibit a loss-of-heterozygosity of the wild-type Tp53 allele. These unique features make this rat highly complementary to other rodent p53 knockout models and a versatile tool for investigating tumorigenesis processes as well as genotoxic studies.


Asunto(s)
Técnicas de Inactivación de Genes , Heterocigoto , Homocigoto , Sarcoma/genética , Sarcoma/patología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Femenino , Genoma/genética , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Metástasis de la Neoplasia , Ratas , Ratas Mutantes , Análisis de Supervivencia , Proteína p53 Supresora de Tumor/química
20.
Proc Natl Acad Sci U S A ; 106(40): 17111-6, 2009 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-19805189

RESUMEN

The CHEK2 kinase (Chk2 in mouse) is a member of a DNA damage response pathway that regulates cell cycle arrest at cell cycle checkpoints and facilitates the repair of dsDNA breaks by a recombination-mediated mechanism. There are numerous variants of the CHEK2 gene, at least one of which, CHEK2*1100delC (SNP), associates with breast cancer. A mouse model in which the wild-type Chk2 has been replaced by a Chk2*1100delC allele was tested for elevated risk of spontaneous cancer and increased sensitivity to challenge by a carcinogenic compound. Mice homozygous for Chk2*1100delC produced more tumors than wild-type mice, whereas heterozygous mice were not statistically different. When fractionated by gender, however, homozygous and heterozygous mice developed spontaneous tumors more rapidly and to a far greater extent than wild-type mice, indicative of a marked gender bias in mice harboring the variant allele. Consistent with our previous data showing elevated genomic instability in mouse embryonic fibroblasts (MEFs) derived from mice homozygous for Chk2*1100delC, the level of Cdc25A was elevated in heterozygous and homozygous MEFs and tumors. When challenged with the carcinogen 7,12-dimethylbenz[a]anthracene, all mice, regardless of genotype, had a reduced lifespan. Latency for mammary tumorigenesis was reduced significantly in mice homozygous for Chk2*1100delC but unexpectedly increased for the development of lymphomas. An implication from this study is that individuals who harbor the variant CHEK2*1100delC allele not only are at an elevated risk for the development of cancer but also that this risk can be further increased as a result of environmental exposure.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Neoplasias/genética , Polimorfismo de Nucleótido Simple , Proteínas Serina-Treonina Quinasas/genética , 9,10-Dimetil-1,2-benzantraceno , Animales , Western Blotting , Quinasa de Punto de Control 2 , Femenino , Fibroblastos/metabolismo , Eliminación de Gen , Genotipo , Inmunohistoquímica , Masculino , Ratones , Neoplasias/inducido químicamente , Neoplasias/patología , Fosforilación , Factores de Riesgo , Factores Sexuales , Factores de Tiempo , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA