Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hepatology ; 78(5): 1418-1432, 2023 11 01.
Article in English | MEDLINE | ID: mdl-36053190

ABSTRACT

BACKGROUND AND AIMS: The assembly and secretion of VLDL from the liver, a pathway that affects hepatic and plasma lipids, remains incompletely understood. We set out to identify players in the VLDL biogenesis pathway by identifying genes that are co-expressed with the MTTP gene that encodes for microsomal triglyceride transfer protein, key to the lipidation of apolipoprotein B, the core protein of VLDL. Using human and murine transcriptomic data sets, we identified small leucine-rich protein 1 ( SMLR1 ), encoding for small leucine-rich protein 1, a protein of unknown function that is exclusively expressed in liver and small intestine. APPROACH AND RESULTS: To assess the role of SMLR1 in the liver, we used somatic CRISPR/CRISPR-associated protein 9 gene editing to silence murine Smlr1 in hepatocytes ( Smlr1 -LKO). When fed a chow diet, male and female mice show hepatic steatosis, reduced plasma apolipoprotein B and triglycerides, and reduced VLDL secretion without affecting microsomal triglyceride transfer protein activity. Immunofluorescence studies show that SMLR1 is in the endoplasmic reticulum and Cis-Golgi complex. The loss of hepatic SMLR1 in female mice protects against diet-induced hyperlipidemia and atherosclerosis but causes NASH. On a high-fat, high-cholesterol diet, insulin and glucose tolerance tests did not reveal differences in male Smlr1 -LKO mice versus controls. CONCLUSIONS: We propose a role for SMLR1 in the trafficking of VLDL from the endoplasmic reticulum to the Cis-Golgi complex. While this study uncovers SMLR1 as a player in the VLDL assembly, trafficking, and secretion pathway, it also shows that NASH can occur with undisturbed glucose homeostasis and atheroprotection.


Subject(s)
Atherosclerosis , Lipoproteins, VLDL , Non-alcoholic Fatty Liver Disease , Small Leucine-Rich Proteoglycans , Animals , Female , Humans , Male , Mice , Apolipoproteins B/blood , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Leucine , Lipoproteins, VLDL/biosynthesis , Lipoproteins, VLDL/blood , Lipoproteins, VLDL/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Small Leucine-Rich Proteoglycans/genetics , Small Leucine-Rich Proteoglycans/metabolism , Triglycerides/blood
2.
J Physiol ; 601(10): 1761-1780, 2023 05.
Article in English | MEDLINE | ID: mdl-37010236

ABSTRACT

Hyperglycaemia in pregnancy (HIP) is a pregnancy complication characterized by mild to moderate hyperglycaemia that negatively impacts short- and long-term health of mother and child. However, relationships between severity and timing of pregnancy hyperglycaemia and postpartum outcomes have not been systemically investigated. We investigated the impact of hyperglycaemia developing during pregnancy (gestational diabetes mellitus, GDM) or already present pre-mating (pre-gestational diabetes mellitus, PDM) on maternal health and pregnancy outcomes. GDM and PDM were induced in C57BL/6NTac mice by combined 60% high fat diet (HF) and low dose streptozotocin (STZ). Animals were screened for PDM prior to mating, and all underwent an oral glucose tolerance test on gestational day (GD)15. Tissues were collected at GD18 or at postnatal day (PN)15. Among HFSTZ-treated dams, 34% developed PDM and 66% developed GDM, characterized by impaired glucose-induced insulin release and inadequate suppression of endogenous glucose production. No increased adiposity or overt insulin resistance was observed. Furthermore, markers of non-alcoholic fatty liver disease (NAFLD) were significantly increased in PDM at GD18 and were positively correlated with basal glucose levels at GD18 in GDM dams. By PN15, NAFLD markers were also increased in GDM dams. Only PDM affected pregnancy outcomes such as litter size. Our findings indicate that GDM and PDM, resulting in disturbances of maternal glucose homeostasis, increase the risk of postpartum NAFLD development, related to the onset and severity of pregnancy hyperglycaemia. These findings signal a need for earlier monitoring of maternal glycaemia and more rigorous follow-up of maternal health after GDM and PDM pregnancy in humans. KEY POINTS: We studied the impact of high-fat diet/streptozotocin induced hyperglycaemia in pregnancy in mice and found that this impaired glucose tolerance and insulin release. Litter size and embryo survival were compromised by pre-gestational, but not by gestational, diabetes. Despite postpartum recovery from hyperglycaemia in a majority of dams, liver disease markers were further elevated by postnatal day 15. Maternal liver disease markers were associated with the severity of hyperglycaemia at gestational day 18. The association between hyperglycaemic exposure and non-alcoholic fatty liver disease signals a need for more rigorous monitoring and follow-up of maternal glycaemia and health in diabetic pregnancy in humans.


Subject(s)
Diabetes, Gestational , Hyperglycemia , Non-alcoholic Fatty Liver Disease , Humans , Pregnancy , Female , Child , Mice , Animals , Hyperglycemia/complications , Pregnancy Outcome , Streptozocin/adverse effects , Mice, Inbred C57BL , Insulin , Glucose/metabolism , Lactation
3.
EMBO J ; 38(20): e101430, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31475738

ABSTRACT

E2F7 and E2F8 act as tumor suppressors via transcriptional repression of genes involved in S-phase entry and progression. Previously, we demonstrated that these atypical E2Fs are degraded by APC/CCdh1 during G1 phase of the cell cycle. However, the mechanism driving the downregulation of atypical E2Fs during G2 phase is unknown. Here, we show that E2F7 is targeted for degradation by the E3 ubiquitin ligase SCFcyclin F during G2. Cyclin F binds via its cyclin domain to a conserved C-terminal CY motif on E2F7. An E2F7 mutant unable to interact with SCFcyclin F remains stable during G2. Furthermore, SCFcyclin F can also interact and induce degradation of E2F8. However, this does not require the cyclin domain of SCFcyclin F nor the CY motifs in the C-terminus of E2F8, implying a different regulatory mechanism than for E2F7. Importantly, depletion of cyclin F causes an atypical-E2F-dependent delay of the G2/M transition, accompanied by reduced expression of E2F target genes involved in DNA repair. Live cell imaging of DNA damage revealed that cyclin F-dependent regulation of atypical E2Fs is critical for efficient DNA repair and cell cycle progression.


Subject(s)
Cyclins/metabolism , DNA Repair , E2F7 Transcription Factor/metabolism , G2 Phase/physiology , Proteolysis , Repressor Proteins/metabolism , Cell Cycle Checkpoints , Cyclins/genetics , DNA Damage , DNA Replication , E2F7 Transcription Factor/genetics , HeLa Cells , Humans , Protein Binding , Repressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
4.
EMBO J ; 38(18): e100849, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31424120

ABSTRACT

The placenta is an autonomous organ that maintains fetal growth and development. Its multinucleated syncytiotrophoblast layer, providing fetal nourishment during gestation, exhibits characteristics of cellular senescence. We show that in human placentas from pregnancies with intrauterine growth restriction, these characteristics are decreased. To elucidate the functions of pathways regulating senescence in syncytiotrophoblast, we used dynamic contrast-enhanced MRI in mice with attenuated senescence programs. This approach revealed an altered dynamics in placentas of p53-/- , Cdkn2a-/- , and Cdkn2a-/- ;p53-/- mice, accompanied by histopathological changes in placental labyrinths. Human primary syncytiotrophoblast upregulated senescence markers and molecular pathways associated with cell-cycle inhibition and senescence-associated secretory phenotype. The pathways and components of the secretory phenotype were compromised in mouse placentas with attenuated senescence and in human placentas from pregnancies with intrauterine growth restriction. We propose that molecular mediators of senescence regulate placental structure and function, through both cell-autonomous and non-autonomous mechanisms.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Fetal Growth Retardation/genetics , Gene Regulatory Networks , Placenta/diagnostic imaging , Tumor Suppressor Protein p53/genetics , Animals , Cellular Senescence , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Magnetic Resonance Imaging , Mice , Phenotype , Placenta/metabolism , Pregnancy , Signal Transduction , Trophoblasts/metabolism
5.
EMBO J ; 37(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29363506

ABSTRACT

The atypical E2Fs, E2F7 and E2F8, act as potent transcriptional repressors of DNA replication genes providing them with the ability to induce a permanent S-phase arrest and suppress tumorigenesis. Surprisingly in human cancer, transcript levels of atypical E2Fs are frequently elevated in proliferating cancer cells, suggesting that the tumor suppressor functions of atypical E2Fs might be inhibited through unknown post-translational mechanisms. Here, we show that atypical E2Fs can be directly phosphorylated by checkpoint kinase 1 (Chk1) to prevent a permanent cell cycle arrest. We found that 14-3-3 protein isoforms interact with both E2Fs in a Chk1-dependent manner. Strikingly, Chk1 phosphorylation and 14-3-3-binding did not relocate or degrade atypical E2Fs, but instead, 14-3-3 is recruited to E2F7/8 target gene promoters to possibly interfere with transcription. We observed that high levels of 14-3-3 strongly correlate with upregulated transcription of atypical E2F target genes in human cancer. Thus, we reveal that Chk1 and 14-3-3 proteins cooperate to inactivate the transcriptional repressor functions of atypical E2Fs. This mechanism might be of particular importance to cancer cells, since they are exposed frequently to DNA-damaging therapeutic reagents.


Subject(s)
14-3-3 Proteins/metabolism , Cell Cycle Checkpoints/physiology , Checkpoint Kinase 1/metabolism , E2F7 Transcription Factor/antagonists & inhibitors , Neoplasms/pathology , Repressor Proteins/antagonists & inhibitors , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/genetics , DNA Replication/genetics , E2F7 Transcription Factor/metabolism , HEK293 Cells , HeLa Cells , Humans , Phosphorylation , Promoter Regions, Genetic/genetics , Protein Binding , Protein Biosynthesis/genetics , Repressor Proteins/metabolism
6.
Hepatology ; 73(1): 303-317, 2021 01.
Article in English | MEDLINE | ID: mdl-32259305

ABSTRACT

BACKGROUND AND AIMS: Up-regulation of the E2F-dependent transcriptional network has been identified in nearly every human malignancy and is an important driver of tumorigenesis. Two members of the E2F family, E2F7 and E2F8, are potent repressors of E2F-dependent transcription. They are atypical in that they do not bind to dimerization partner proteins and are not controlled by retinoblastoma protein. The physiological relevance of E2F7 and E2F8 remains incompletely understood, largely because tools to manipulate their activity in vivo have been lacking. APPROACH AND RESULTS: Here, we generated transgenic mice with doxycycline-controlled transcriptional activation of E2f7 and E2f8 and induced their expression during postnatal development, in adulthood, and in the context of cancer. Systemic induction of E2f7 and, to lesser extent, E2f8 transgenes in juvenile mice impaired cell proliferation, caused replication stress, DNA damage, and apoptosis, and inhibited animal growth. In adult mice, however, E2F7 and E2F8 induction was well tolerated, yet profoundly interfered with DNA replication, DNA integrity, and cell proliferation in diethylnitrosamine-induced liver tumors. CONCLUSION: Collectively, our findings demonstrate that atypical E2Fs can override cell-cycle entry and progression governed by other E2F family members and suggest that this property can be exploited to inhibit proliferation of neoplastic hepatocytes when growth and development have subsided during adulthood.


Subject(s)
Cell Proliferation , E2F7 Transcription Factor/physiology , Hepatocytes/metabolism , Liver Neoplasms/pathology , Repressor Proteins/physiology , Animals , Apoptosis/physiology , Cell Cycle/physiology , DNA Damage , E2F7 Transcription Factor/deficiency , E2F7 Transcription Factor/genetics , HeLa Cells , Humans , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/deficiency , Repressor Proteins/genetics , Transcriptional Activation
7.
Hepatology ; 74(5): 2491-2507, 2021 11.
Article in English | MEDLINE | ID: mdl-34157136

ABSTRACT

BACKGROUND AND AIMS: Patients with glycogen storage disease type 1a (GSD-1a) primarily present with life-threatening hypoglycemia and display severe liver disease characterized by hepatomegaly. Despite strict dietary management, long-term complications still occur, such as liver tumor development. Variations in residual glucose-6-phosphatase (G6PC1) activity likely contribute to phenotypic heterogeneity in biochemical symptoms and complications between patients. However, lack of insight into the relationship between G6PC1 activity and symptoms/complications and poor understanding of the underlying disease mechanisms pose major challenges to provide optimal health care and quality of life for GSD-1a patients. Currently available GSD-1a animal models are not suitable to systematically investigate the relationship between hepatic G6PC activity and phenotypic heterogeneity or the contribution of gene-gene interactions (GGIs) in the liver. APPROACH AND RESULTS: To meet these needs, we generated and characterized a hepatocyte-specific GSD-1a mouse model using somatic CRISPR/CRISPR-associated protein 9 (Cas9)-mediated gene editing. Hepatic G6pc editing reduced hepatic G6PC activity up to 98% and resulted in failure to thrive, fasting hypoglycemia, hypertriglyceridemia, hepatomegaly, hepatic steatosis (HS), and increased liver tumor incidence. This approach was furthermore successful in simultaneously modulating hepatic G6PC and carbohydrate response element-binding protein, a transcription factor that is activated in GSD-1a and protects against HS under these conditions. Importantly, it also allowed for the modeling of a spectrum of GSD-1a phenotypes in terms of hepatic G6PC activity, fasting hypoglycemia, hypertriglyceridemia, hepatomegaly and HS. CONCLUSIONS: In conclusion, we show that somatic CRISPR/Cas9-mediated gene editing allows for the modeling of a spectrum of hepatocyte-borne GSD-1a disease symptoms in mice and to efficiently study GGIs in the liver. This approach opens perspectives for translational research and will likely contribute to personalized treatments for GSD-1a and other genetic liver diseases.


Subject(s)
CRISPR-Associated Protein 9/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Disease Models, Animal , Gene Editing/methods , Genetic Heterogeneity , Glycogen Storage Disease Type I/genetics , Phenotype , Animals , Genetic Vectors , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Hepatocytes/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
Liver Int ; 42(11): 2442-2452, 2022 11.
Article in English | MEDLINE | ID: mdl-35924448

ABSTRACT

The tumour suppressor PTEN is a negative regulator of the PI3K/AKT signalling pathway. Liver-specific deletion of Pten in mice results in the hyper-activation PI3K/AKT signalling accompanied by enhanced genome duplication (polyploidization), marked lipid accumulation (steatosis) and formation of hepatocellular carcinomas. However, it is unknown whether polyploidization in this model has an impact on the development of steatosis and the progression towards liver cancer. Here, we used a liver-specific conditional knockout approach to delete Pten in combination with deletion of E2f7/8, known key inducers of polyploidization. As expected, Pten deletion caused severe steatosis and liver tumours accompanied by enhanced polyploidization. Additional deletion of E2f7/8 inhibited polyploidization, alleviated Pten-induced steatosis without affecting lipid species composition and accelerated liver tumour progression. Global transcriptomic analysis showed that inhibition of polyploidization in Pten-deficient livers resulted in reduced expression of genes involved in energy metabolism, including PPAR-gamma signalling. However, we find no evidence that deregulated genes in Pten-deficient livers are direct transcriptional targets of E2F7/8, supporting that reduction in steatosis and progression towards liver cancer are likely consequences of inhibiting polyploidization. Lastly, flow cytometry and image analysis on isolated primary wildtype mouse hepatocytes provided further support that polyploid cells can accumulate more lipid droplets than diploid hepatocytes. Collectively, we show that polyploidization promotes steatosis and function as an important barrier against liver tumour progression in Pten-deficient livers.


Subject(s)
Fatty Liver , Liver Neoplasms , Animals , Fatty Liver/pathology , Hepatocytes/metabolism , Lipids , Liver/pathology , Liver Neoplasms/pathology , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt
9.
EMBO Rep ; 21(12): e50893, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33225610

ABSTRACT

Polyploidization frequently precedes tumorigenesis but also occurs during normal development in several tissues. Hepatocyte ploidy is controlled by the PIDDosome during development and regeneration. This multi-protein complex is activated by supernumerary centrosomes to induce p53 and restrict proliferation of polyploid cells, otherwise prone for chromosomal instability. PIDDosome deficiency in the liver results in drastically increased polyploidy. To investigate PIDDosome-induced p53-activation in the pathogenesis of liver cancer, we chemically induced hepatocellular carcinoma (HCC) in mice. Strikingly, PIDDosome deficiency reduced tumor number and burden, despite the inability to activate p53 in polyploid cells. Liver tumors arise primarily from cells with low ploidy, indicating an intrinsic pro-tumorigenic effect of PIDDosome-mediated ploidy restriction. These data suggest that hyperpolyploidization caused by PIDDosome deficiency protects from HCC. Moreover, high tumor cell density, as a surrogate marker of low ploidy, predicts poor survival of HCC patients receiving liver transplantation. Together, we show that the PIDDosome is a potential therapeutic target to manipulate hepatocyte polyploidization for HCC prevention and that tumor cell density may serve as a novel prognostic marker for recurrence-free survival in HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Humans , Liver Neoplasms/genetics , Mice , Ploidies , Tumor Suppressor Protein p53/genetics
10.
Hepatology ; 72(5): 1638-1653, 2020 11.
Article in English | MEDLINE | ID: mdl-32083759

ABSTRACT

BACKGROUND AND AIMS: Glycogen storage disease (GSD) type 1a is an inborn error of metabolism caused by defective glucose-6-phosphatase catalytic subunit (G6PC) activity. Patients with GSD 1a exhibit severe hepatomegaly due to glycogen and triglyceride (TG) accumulation in the liver. We have shown that the activity of carbohydrate response element binding protein (ChREBP), a key regulator of glycolysis and de novo lipogenesis, is increased in GSD 1a. In the current study, we assessed the contribution of ChREBP to nonalcoholic fatty liver disease (NAFLD) development in a mouse model for hepatic GSD 1a. APPROACH AND RESULTS: Liver-specific G6pc-knockout (L-G6pc-/- ) mice were treated with adeno-associated viruses (AAVs) 2 or 8 directed against short hairpin ChREBP to normalize hepatic ChREBP activity to levels observed in wild-type mice receiving AAV8-scrambled short hairpin RNA (shSCR). Hepatic ChREBP knockdown markedly increased liver weight and hepatocyte size in L-G6pc-/- mice. This was associated with hepatic accumulation of G6P, glycogen, and lipids, whereas the expression of glycolytic and lipogenic genes was reduced. Enzyme activities, flux measurements, hepatic metabolite analysis and very low density lipoprotein (VLDL)-TG secretion assays revealed that hepatic ChREBP knockdown reduced downstream glycolysis and de novo lipogenesis but also strongly suppressed hepatic VLDL lipidation, hence promoting the storage of "old fat." Interestingly, enhanced VLDL-TG secretion in shSCR-treated L-G6pc-/- mice associated with a ChREBP-dependent induction of the VLDL lipidation proteins microsomal TG transfer protein and transmembrane 6 superfamily member 2 (TM6SF2), the latter being confirmed by ChIP-qPCR. CONCLUSIONS: Attenuation of hepatic ChREBP induction in GSD 1a liver aggravates hepatomegaly because of further accumulation of glycogen and lipids as a result of reduced glycolysis and suppressed VLDL-TG secretion. TM6SF2, critical for VLDL formation, was identified as a ChREBP target in mouse liver. Altogether, our data show that enhanced ChREBP activity limits NAFLD development in GSD 1a by balancing hepatic TG production and secretion.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Glycogen Storage Disease Type I/complications , Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Adipose Tissue, White/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Dependovirus/genetics , Disease Models, Animal , Gene Knockdown Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Glucose-6-Phosphatase/genetics , Glycogen/metabolism , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/metabolism , Glycolysis , Hepatocytes , Humans , Lipogenesis , Lipoproteins, VLDL/metabolism , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , RNA, Small Interfering/genetics , Triglycerides/metabolism
11.
Circ Res ; 124(1): 94-100, 2019 01 04.
Article in English | MEDLINE | ID: mdl-30582442

ABSTRACT

RATIONALE: Several studies have suggested a role for the gut microbiota in inflammation and atherogenesis. A causal relation relationship between gut microbiota, inflammation, and atherosclerosis has not been explored previously. OBJECTIVE: Here, we investigated whether a proinflammatory microbiota from Caspase1-/- ( Casp1-/-) mice accelerates atherogenesis in Ldlr-/- mice. METHOD AND RESULTS: We treated female Ldlr-/- mice with antibiotics and subsequently transplanted them with fecal microbiota from Casp1-/- mice based on a cohousing approach. Autologous transplantation of fecal microbiota of Ldlr-/- mice served as control. Mice were cohoused for 8 or 13 weeks and fed chow or high-fat cholesterol-rich diet. Fecal samples were collected, and factors related to inflammation, metabolism, intestinal health, and atherosclerotic phenotypes were measured. Unweighted Unifrac distances of 16S rDNA (ribosomal DNA) sequences confirmed the introduction of the Casp1-/- and Ldlr-/- microbiota into Ldlr-/- mice (referred to as Ldlr-/-( Casp1-/-) or Ldlr-/-( Ldlr-/-) mice). Analysis of atherosclerotic lesion size in the aortic root demonstrated a significant 29% increase in plaque size in 13-week high-fat cholesterol-fed Ldlr-/-( Casp1-/-) mice compared with Ldlr-/-( Ldlr-/-) mice. We found increased numbers of circulating monocytes and neutrophils and elevated proinflammatory cytokine levels in plasma in high-fat cholesterol-fed Ldlr-/-( Casp1-/-) compared with Ldlr-/-( Ldlr-/-) mice. Neutrophil accumulation in the aortic root of Ldlr-/-( Casp1-/-) mice was enhanced compared with Ldlr-/-( Ldlr-/-) mice. 16S-rDNA-encoding sequence analysis in feces identified a significant reduction in the short-chain fatty acid-producing taxonomies Akkermansia, Christensenellaceae, Clostridium, and Odoribacter in Ldlr-/-( Casp1-/-) mice. Consistent with these findings, cumulative concentrations of the anti-inflammatory short-chain fatty acids propionate, acetate and butyrate in the cecum were significantly reduced in 13-week high-fat cholesterol-fed Ldlr-/-( Casp1-/-) compared with Ldlr-/-( Ldlr-/-) mice. CONCLUSIONS: Introduction of the proinflammatory Casp1-/- microbiota into Ldlr-/- mice enhances systemic inflammation and accelerates atherogenesis.


Subject(s)
Aorta/metabolism , Aortic Diseases/microbiology , Atherosclerosis/microbiology , Bacteria/metabolism , Cytokines/metabolism , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Inflammation Mediators/metabolism , Inflammation/microbiology , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Caspase 1/genetics , Caspase 1/metabolism , Disease Models, Animal , Disease Progression , Dysbiosis , Fatty Acids/metabolism , Female , Host-Pathogen Interactions , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice, Knockout , Plaque, Atherosclerotic , Receptors, LDL/genetics , Receptors, LDL/metabolism , Time Factors
12.
Vet Pathol ; 58(3): 453-471, 2021 05.
Article in English | MEDLINE | ID: mdl-33813952

ABSTRACT

With a size range from 30 to 1000 nm, extracellular vesicles (EVs) are one of the smallest cell components able to transport biologically active molecules. They mediate intercellular communications and play a fundamental role in the maintenance of tissue homeostasis and pathogenesis in several types of diseases. In particular, EVs actively contribute to cancer initiation and progression, and there is emerging understanding of their role in creation of the metastatic niche. This fact underlies the recent exponential growth in EV research, which has improved our understanding of their specific roles in disease and their potential applications in diagnosis and therapy. EVs and their biomolecular cargo reflect the state of the diseased donor cells, and can be detected in body fluids and exploited as biomarkers in cancer and other diseases. Relatively few studies have been published on EVs in the veterinary field. This review provides an overview of the features and biology of EVs as well as recent developments in EV research including techniques for isolation and analysis, and will address the way in which the EVs released by diseased tissues can be studied and exploited in the field of veterinary pathology. Uniquely, this review emphasizes the important contribution that pathologists can make to the field of EV research: pathologists can help EV scientists in studying and confirming the role of EVs and their molecular cargo in diseased tissues and as biomarkers in liquid biopsies.


Subject(s)
Extracellular Vesicles , Neoplasms , Animals , Biomarkers , Neoplasms/diagnosis , Neoplasms/veterinary
13.
Carcinogenesis ; 41(2): 171-181, 2020 04 22.
Article in English | MEDLINE | ID: mdl-30770929

ABSTRACT

In Western countries, a rising incidence of obesity and type 2 diabetes correlates with an increase of non-alcoholic steatohepatitis (NASH)-a major risk factor for liver cirrhosis and hepatocellular carcinoma (HCC). NASH is associated with chronic liver injury, triggering hepatocyte death and enhanced translocation of intestinal bacteria, leading to persistent liver inflammation through activation of Toll-like receptors and their adapter protein myeloid differentiation factor 88 (MyD88). Therefore, we investigated the role of MyD88 during progression from NASH to HCC using a mouse model of chronic liver injury (hepatocyte-specific deletion of nuclear factor κB essential modulator, Nemo; NemoΔhepa). NemoΔhepa; NemoΔhepa/MyD88-/- and NemoΔhepa/MyD88Δhepa were generated and the impact on liver disease progression was investigated. Ubiquitous MyD88 ablation (NemoΔhepa/MyD88-/-) aggravated the degree of liver damage, accompanied by an overall decrease in inflammation, whereas infiltrating macrophages and natural killer cells were elevated. At a later stage, MyD88 deficiency impaired HCC formation. In contrast, hepatocyte-specific MyD88 deletion (NemoΔhepa/MyD88Δhepa) did not affect disease progression. These results suggest that signaling of Toll-like receptors through MyD88 in non-parenchymal liver cells is required for carcinogenesis during chronic liver injury. Hence, blocking MyD88 signaling may offer a therapeutic option to prevent HCC formation in patients with NASH.


Subject(s)
Carcinogenesis/pathology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Myeloid Differentiation Factor 88/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Animals , Carcinoma, Hepatocellular/prevention & control , Disease Models, Animal , Disease Progression , Hepatocytes/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Liver/cytology , Liver/pathology , Liver Neoplasms/prevention & control , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/genetics , Signal Transduction/drug effects
14.
EMBO J ; 35(9): 942-60, 2016 05 02.
Article in English | MEDLINE | ID: mdl-26933123

ABSTRACT

The generation of multiciliated cells (MCCs) is required for the proper function of many tissues, including the respiratory tract, brain, and germline. Defects in MCC development have been demonstrated to cause a subclass of mucociliary clearance disorders termed reduced generation of multiple motile cilia (RGMC). To date, only two genes, Multicilin (MCIDAS) and cyclin O (CCNO) have been identified in this disorder in humans. Here, we describe mice lacking GEMC1 (GMNC), a protein with a similar domain organization as Multicilin that has been implicated in DNA replication control. We have found that GEMC1-deficient mice are growth impaired, develop hydrocephaly with a high penetrance, and are infertile, due to defects in the formation of MCCs in the brain, respiratory tract, and germline. Our data demonstrate that GEMC1 is a critical regulator of MCC differentiation and a candidate gene for human RGMC or related disorders.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation , Cilia/genetics , Cilia/physiology , Growth Disorders/genetics , Growth Disorders/pathology , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Mice , Mice, Knockout
15.
Am J Hum Genet ; 101(3): 391-403, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28886341

ABSTRACT

In five separate families, we identified nine individuals affected by a previously unidentified syndrome characterized by growth retardation, spine malformation, facial dysmorphisms, and developmental delays. Using homozygosity mapping, array CGH, and exome sequencing, we uncovered bi-allelic loss-of-function CDK10 mutations segregating with this disease. CDK10 is a protein kinase that partners with cyclin M to phosphorylate substrates such as ETS2 and PKN2 in order to modulate cellular growth. To validate and model the pathogenicity of these CDK10 germline mutations, we generated conditional-knockout mice. Homozygous Cdk10-knockout mice died postnatally with severe growth retardation, skeletal defects, and kidney and lung abnormalities, symptoms that partly resemble the disease's effect in humans. Fibroblasts derived from affected individuals and Cdk10-knockout mouse embryonic fibroblasts (MEFs) proliferated normally; however, Cdk10-knockout MEFs developed longer cilia. Comparative transcriptomic analysis of mutant and wild-type mouse organs revealed lipid metabolic changes consistent with growth impairment and altered ciliogenesis in the absence of CDK10. Our results document the CDK10 loss-of-function phenotype and point to a function for CDK10 in transducing signals received at the primary cilia to sustain embryonic and postnatal development.


Subject(s)
Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/physiology , Developmental Disabilities/genetics , Growth Disorders/genetics , Mutation , Spine/abnormalities , Spine/pathology , Animals , Cell Cycle , Cell Proliferation , Cells, Cultured , Child , Child, Preschool , Cilia/metabolism , Cilia/pathology , Developmental Disabilities/pathology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Growth Disorders/pathology , Humans , Infant , Male , Mice , Mice, Knockout , Pedigree , Phosphorylation , Signal Transduction , Spine/metabolism
16.
Gastroenterology ; 157(1): 210-226.e12, 2019 07.
Article in English | MEDLINE | ID: mdl-30878468

ABSTRACT

BACKGROUND & AIMS: The CCNE1 locus, which encodes cyclin E1, is amplified in many types of cancer cells and is activated in hepatocellular carcinomas (HCCs) from patients infected with hepatitis B virus or adeno-associated virus type 2, due to integration of the virus nearby. We investigated cell-cycle and oncogenic effects of cyclin E1 overexpression in tissues of mice. METHODS: We generated mice with doxycycline-inducible expression of Ccne1 (Ccne1T mice) and activated overexpression of cyclin E1 from age 3 weeks onward. At 14 months of age, livers were collected from mice that overexpress cyclin E1 and nontransgenic mice (controls) and analyzed for tumor burden and by histology. Mouse embryonic fibroblasts (MEFs) and hepatocytes from Ccne1T and control mice were analyzed to determine the extent to which cyclin E1 overexpression perturbs S-phase entry, DNA replication, and numbers and structures of chromosomes. Tissues from 4-month-old Ccne1T and control mice (at that age were free of tumors) were analyzed for chromosome alterations, to investigate the mechanisms by which cyclin E1 predisposes hepatocytes to transformation. RESULTS: Ccne1T mice developed more hepatocellular adenomas and HCCs than control mice. Tumors developed only in livers of Ccne1T mice, despite high levels of cyclin E1 in other tissues. Ccne1T MEFs had defects that promoted chromosome missegregation and aneuploidy, including incomplete replication of DNA, centrosome amplification, and formation of nonperpendicular mitotic spindles. Whereas Ccne1T mice accumulated near-diploid aneuploid cells in multiple tissues and organs, polyploidization was observed only in hepatocytes, with losses and gains of whole chromosomes, DNA damage, and oxidative stress. CONCLUSIONS: Livers, but not other tissues of mice with inducible overexpression of cyclin E1, develop tumors. More hepatocytes from the cyclin E1-overexpressing mice were polyploid than from control mice, and had losses or gains of whole chromosomes, DNA damage, and oxidative stress; all of these have been observed in human HCC cells. The increased risk of HCC in patients with hepatitis B virus or adeno-associated virus type 2 infection might involve activation of cyclin E1 and its effects on chromosomes and genomes of liver cells.


Subject(s)
Adenoma, Liver Cell/genetics , Carcinoma, Hepatocellular/genetics , Chromosomal Instability/genetics , Cyclin E/genetics , Liver Neoplasms/genetics , Liver/metabolism , Oncogene Proteins/genetics , Adenoma, Liver Cell/pathology , Adenoma, Liver Cell/virology , Animals , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Chromosome Structures , DNA Damage/genetics , DNA Replication , Dependovirus , Fibroblasts , Hepatitis B, Chronic , Hepatocytes , Liver/pathology , Liver Neoplasms/pathology , Liver Neoplasms/virology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Mice , Oxidative Stress/genetics , Parvoviridae Infections , Parvovirinae , Polyploidy , S Phase Cell Cycle Checkpoints
17.
Clin Sci (Lond) ; 134(7): 727-746, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32202295

ABSTRACT

We previously identified genomic instability as a causative factor for vascular aging. In the present study, we determined which vascular aging outcomes are due to local endothelial DNA damage, which was accomplished by genetic removal of ERCC1 (excision repair cross-complementation group 1) DNA repair in mice (EC-knockout (EC-KO) mice). EC-KO showed a progressive decrease in microvascular dilation of the skin, increased microvascular leakage in the kidney, decreased lung perfusion, and increased aortic stiffness compared with wild-type (WT). EC-KO showed expression of DNA damage and potential senescence marker p21 exclusively in the endothelium, as demonstrated in aorta. Also the kidney showed p21-positive cells. Vasodilator responses measured in organ baths were decreased in aorta, iliac and coronary artery EC-KO compared with WT, of which coronary artery was the earliest to be affected. Nitric oxide-mediated endothelium-dependent vasodilation was abolished in aorta and coronary artery, whereas endothelium-derived hyperpolarization and responses to exogenous nitric oxide (NO) were intact. EC-KO showed increased superoxide production compared with WT, as measured in lung tissue, rich in endothelial cells (ECs). Arterial systolic blood pressure (BP) was increased at 3 months, but normal at 5 months, at which age cardiac output (CO) was decreased. Since no further signs of cardiac dysfunction were detected, this decrease might be an adaptation to prevent an increase in BP. In summary, a selective DNA repair defect in the endothelium produces features of age-related endothelial dysfunction, largely attributed to loss of endothelium-derived NO. Increased superoxide generation might contribute to the observed changes affecting end organ perfusion, as demonstrated in kidney and lung.


Subject(s)
Aging/genetics , Cellular Senescence/genetics , DNA Damage , DNA Repair , DNA-Binding Proteins/deficiency , Endonucleases/deficiency , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Age Factors , Aging/metabolism , Aging/pathology , Animals , Capillary Permeability , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/genetics , Endonucleases/genetics , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Superoxides/metabolism , Vascular Stiffness , Vasodilation
19.
Pediatr Res ; 85(7): 1041-1047, 2019 06.
Article in English | MEDLINE | ID: mdl-30851724

ABSTRACT

BACKGROUND: Phototherapy (PT) is the standard treatment of neonatal unconjugated hyperbilirubinemia. Fluorescent tube (FT)-emitted PT light is known to induce oxidative DNA damage in neonates. Nowadays, however, FTs have largely been replaced by light-emitting diodes (LEDs) for delivering PT. Until now, it is unknown whether LED-PT causes oxidative DNA damage. We aim to determine whether LED-PT induces oxidative DNA damage in hyperbilirubinemic rats. METHODS: Adult Gunn rats, with genetically unconjugated hyperbilirubinemia, received LED-PT in the clinically relevant doses of 10 or 30 µW/cm2/nm. Urine was collected at 0, 24, and 48 h of PT. A group of young Gunn rats received intensive LED-PT of 100 µW/cm2/nm for 24 h. Urine was collected every 8 h and analyzed for the levels of oxidative DNA damage marker 8-hydroxy-2'deoxyguanosine (8-OHdG) and creatinine. DNA damage was evaluated by immunohistochemistry (γH2AX) of skin and spleen samples. RESULTS: LED-PT of 10 and 30 µW/cm2/nm did not affect urinary concentrations of 8-OHdG and creatinine or the 8-OHdG/creatinine ratio. Likewise, intensive LED-PT did not affect the 8-OHdG/creatinine ratio or the number of γH2AX-positive cells in the skin or spleen. CONCLUSIONS: Our results show that LED-PT does not induce oxidative DNA damage in hyperbilirubinemic Gunn rats either at clinically relevant or intensive dosages.


Subject(s)
DNA Damage , Oxidative Stress , Phototherapy/methods , Animals , Hyperbilirubinemia, Neonatal , Rats , Rats, Gunn
20.
Br J Nutr ; 122(12): 1321-1328, 2019 12 28.
Article in English | MEDLINE | ID: mdl-31439052

ABSTRACT

Breast-feeding is associated with a lower risk of developing obesity during childhood and adulthood compared with feeding infant milk formula (IMF). Previous studies have shown that an experimental IMF (eIMF; comprising Nuturis®) programmed mouse pups for a lower body weight and fat mass gain in adulthood when challenged with a high-fat diet (HFD) compared with a control IMF (cIMF). Nuturis has a lipid composition and structure more similar to breast milk. Here, the long-term effects were tested of a similar eIMF, but with an adapted lipid composition and a cIMF, on body weight, glucose homoeostasis, liver and adipose tissue. Nutrient composition was similar for the eIMF and cIMF; the lipid fractions comprised approximately 50 % milk fat. C57BL/6JOlaHsd mice were fed cIMF or eIMF from postnatal day (PN) 16-42 followed by an HFD until PN168. Feeding eIMF v. cIMF in early life resulted in a lower body weight (-9 %) and body fat deposition (-14 %) in adulthood (PN105). The effect appeared transient, as from PN126 onwards, after 12 weeks' HFD, eIMF-fed mice caught up on controls and body and fat weights became comparable between groups. Glucose and energy metabolism were similar between groups. At dissection (PN168), eIMF-fed mice showed larger (+27 %) epididymal fat depots and a lower (-26 %) liver weight without clear morphological aberrations. Our data suggest the size and coating but not the lipid composition of IMF fat globules underlie the programming effect observed. Prolonged exposure to an HFD challenge partly overrules the programming effect of early diet.


Subject(s)
Diet, High-Fat , Diet , Dietary Fats/metabolism , Food, Formulated , Glycolipids/chemistry , Glycoproteins/chemistry , Phospholipids/chemistry , Adipose Tissue/metabolism , Animal Feed , Animals , Body Composition , Body Weight , Female , Gene Expression Profiling , Glucose/metabolism , Homeostasis , Lipid Droplets , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Phenotype , Pyruvic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL