Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Sci Rep ; 11(1): 21839, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34750429

ABSTRACT

The intimate association between obesity and type II diabetes urges for a deeper understanding of adipocyte function. We and others have previously delineated a role for the tumor suppressor p53 in adipocyte biology. Here, we show that mice haploinsufficient for MDM2, a key regulator of p53, in their adipose stores suffer from overt obesity, glucose intolerance, and hepatic steatosis. These mice had decreased levels of circulating palmitoleic acid [non-esterified fatty acid (NEFA) 16:1] concomitant with impaired visceral adipose tissue expression of Scd1 and Ffar4. A similar decrease in Scd and Ffar4 expression was found in in vitro differentiated adipocytes with perturbed MDM2 expression. Lowered MDM2 levels led to nuclear exclusion of the transcriptional cofactors, MORC2 and LIPIN1, and thereby possibly hampered adipocyte function by antagonizing LIPIN1-mediated PPARγ coactivation. Collectively, these data argue for a hitherto unknown interplay between MDM2 and MORC2/LIPIN1 involved in balancing adipocyte function.


Subject(s)
Adipose Tissue, White/metabolism , Insulin Resistance/physiology , Proto-Oncogene Proteins c-mdm2/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Diet, High-Fat/adverse effects , Fatty Acids, Monounsaturated/blood , Fatty Liver/genetics , Fatty Liver/metabolism , Female , Gene Regulatory Networks , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Haploinsufficiency/genetics , Haploinsufficiency/physiology , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , PPAR gamma/metabolism , Phosphatidate Phosphatase , Proto-Oncogene Proteins c-mdm2/deficiency , Proto-Oncogene Proteins c-mdm2/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
2.
Int J Mol Sci ; 22(18)2021 Sep 16.
Article in English | MEDLINE | ID: mdl-34576178

ABSTRACT

The transcription factor GATA2 regulates gene expression in several cells and tissues, including hematopoietic tissues and the central nervous system. Recent studies revealed that loss-of-function mutations in GATA2 are associated with hematological disorders. Our earlier in vitro studies showed that GATA2 plays an essential role in the hypothalamus-pituitary-thyroid axis (HPT axis) by regulating the genes encoding prepro-thyrotropin-releasing hormone (preproTRH) and thyroid-stimulating hormone ß (TSHß). However, the effect of GATA2 mutants on the transcriptional activity of their promoters remains unelucidated. In this study, we created five human GATA2 mutations (R308P, T354M, R396Q, R398W, and S447R) that were reported to be associated with hematological disorders and analyzed their functional properties, including transactivation potential and DNA-binding capacity toward the preproTRH and the TSHß promoters. Three mutations (T354M, R396Q, and R398W) within the C-terminal zinc-finger domain reduced the basal GATA2 transcriptional activity on both the preproTRH and the TSHß promoters with a significant loss of DNA binding affinity. Interestingly, only the R398W mutation reduced the GATA2 protein expression. Subsequent analysis demonstrated that the R398W mutation possibly facilitated the GATA2 degradation process. R308P and S447R mutants exhibited decreased transcriptional activity under protein kinase C compared to the wild-type protein. In conclusion, we demonstrated that naturally occurring GATA2 mutations impair the HPT axis through differential functional mechanisms in vitro.


Subject(s)
GATA2 Transcription Factor/genetics , Hypothalamus/metabolism , Mutation/genetics , Pituitary Gland/metabolism , Thyroid Gland/metabolism , Blotting, Western , Haploinsufficiency/genetics , Haploinsufficiency/physiology , Humans , Hypothyroidism/genetics , Promoter Regions, Genetic/genetics , Thyrotropin, beta Subunit/genetics , Thyrotropin, beta Subunit/metabolism , Transcriptional Activation/genetics , Transcriptional Activation/physiology
3.
BMC Neurosci ; 22(1): 32, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33933000

ABSTRACT

BACKGROUND: Autism spectrum disorder (ASD) is characterized by the core symptoms of impaired social interactions. Increasing evidence suggests that ASD has a strong genetic link with mutations in chromodomain helicase DNA binding protein 8 (CHD8), a gene encoding a chromatin remodeler. It has previously been shown that Chd8 haplodeficient male mice manifest ASD-like behavioral characteristics such as anxiety and altered social behavior. Along with that, oxytocin (OT) is one of the main neuropeptides involved in social behavior. Administration of OT has shown improvement of social behavior in genetic animal models of ASD. The present study was undertaken to further explore behavioral abnormalities of Chd8 haplodeficient mice of both sexes, their link with OT, and possible effects of OT administration. First, we performed a battery of behavioral tests on wild-type and Chd8+/∆SL female and male mice. Next, we measured plasma OT levels and finally studied the effects of intraperitoneal OT injection on observed behavioral deficits. RESULTS: We showed general anxiety phenotype in Chd8+/∆SL mice regardless of sex, the depressive phenotype in Chd8+/∆SL female mice only and bidirectional social deficit in female and male mice. We observed decreased level of OT in Chd+/∆SL mice, possibly driven by males. Mice injected by OT demonstrated recovery of social behavior, while reduced anxiety was observed only in male mice. CONCLUSIONS: Here, we demonstrated that abnormal social behaviors were observed in both male and female Chd8+/∆SL mice. The ability of peripheral OT administration to affect such behaviors along with altered plasma OT levels indicated a possible link between Chd8 + /∆SL and OT in the pathogenesis of ASD as well as the possible usefulness of OT as a therapeutic tool for ASD patients with CHD8 mutations.


Subject(s)
Autistic Disorder/drug therapy , Autistic Disorder/genetics , DNA-Binding Proteins/genetics , Haploinsufficiency/drug effects , Oxytocin/therapeutic use , Social Behavior , Animals , Autistic Disorder/metabolism , DNA-Binding Proteins/deficiency , Female , Haploinsufficiency/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Oxytocin/pharmacology
4.
J Mol Cell Biol ; 13(5): 374-382, 2021 08 18.
Article in English | MEDLINE | ID: mdl-33386850

ABSTRACT

TDP43 pathology is seen in a large majority of amyotrophic lateral sclerosis (ALS) cases, suggesting a central pathogenic role of this regulatory protein. Clarifying the molecular mechanism controlling TDP43 stability and subcellular location might provide important insights into ALS therapy. The ubiquitin E3 ligase RNF220 is involved in different neural developmental processes through various molecular targets in the mouse. Here, we report that the RNF220+/- mice showed progressively decreasing mobility to different extents, some of which developed typical ALS pathological characteristics in spinal motor neurons, including TDP43 cytoplasmic accumulation, atrocytosis, muscle denervation, and atrophy. Mechanistically, RNF220 interacts with TDP43 in vitro and in vivo and promotes its polyubiquitination and proteasomal degradation. In conclusion, we propose that RNF220 might be a modifier of TDP43 function in vivo and contribute to TDP43 pathology in neurodegenerative disease like ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/metabolism , Haploinsufficiency/physiology , Motor Neurons/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism , Animals , Cell Line , Disease Models, Animal , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitination/physiology
5.
Hum Mol Genet ; 28(24): 4113-4131, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31628460

ABSTRACT

Genetic and environmental influences are thought to interact in their contribution to the etiology of major neuropsychiatric disorders. One of the best replicated findings obtained in genome-wide association studies are genetic variants in the CACNA1C gene. Here, we used our constitutive heterozygous Cacna1c rat model in combination with a 4-week exposure to either post-weaning social isolation, standard housing or social and physical environmental enrichment during the critical juvenile developmental period to observe their long-term interactive effects with Cacna1c haploinsufficiency. Our study provides evidence for a gene × environment interaction, i.e. an interplay between Cacna1c haploinsufficiency and environment during juvenile development, on object recognition, spatial memory and reversal learning capabilities. Social and physical enrichment had a positive influence on Cacna1c+/- rats and Cacna1c+/+ littermate controls on spatial and reversal learning, while post-weaning social isolation negatively affected novel object recognition in both genotypes. Despite intact spatial learning and re-learning abilities in all groups, slight but consistent deficits were evident in Cacna1c+/- rats previously housed under standard conditions particularly during reversal learning but not Cacna1c+/- rats previously exposed to social and physical enrichment. Together, this supports the notion that Cacna1c interacts with the environment to shape disease vulnerability and associated alterations in cognitive functioning.


Subject(s)
Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Gene-Environment Interaction , Animals , Behavior, Animal , Cognition , Environment , Female , Genome-Wide Association Study , Haploinsufficiency/physiology , Hippocampus/metabolism , Male , Rats , Rats, Sprague-Dawley , Reversal Learning/physiology , Social Isolation , Spatial Memory/physiology
6.
Int J Mol Sci ; 20(17)2019 Aug 21.
Article in English | MEDLINE | ID: mdl-31438494

ABSTRACT

Human variants in plakophilin-2 (PKP2) associate with most cases of familial arrhythmogenic cardiomyopathy (ACM). Recent studies show that PKP2 not only maintains intercellular coupling, but also regulates transcription of genes involved in Ca2+ cycling and cardiac rhythm. ACM penetrance is low and it remains uncertain, which genetic and environmental modifiers are crucial for developing the cardiomyopathy. In this study, heterozygous PKP2 knock-out mice (PKP2-Hz) were used to investigate the influence of exercise, pressure overload, and inflammation on a PKP2-related disease progression. In PKP2-Hz mice, protein levels of Ca2+-handling proteins were reduced compared to wildtype (WT). PKP2-Hz hearts exposed to voluntary exercise training showed right ventricular lateral connexin43 expression, right ventricular conduction slowing, and a higher susceptibility towards arrhythmias. Pressure overload increased levels of fibrosis in PKP2-Hz hearts, without affecting the susceptibility towards arrhythmias. Experimental autoimmune myocarditis caused more severe subepicardial fibrosis, cell death, and inflammatory infiltrates in PKP2-Hz hearts than in WT. To conclude, PKP2 haploinsufficiency in the murine heart modulates the cardiac response to environmental modifiers via different mechanisms. Exercise upon PKP2 deficiency induces a pro-arrhythmic cardiac remodeling, likely based on impaired Ca2+ cycling and electrical conduction, versus structural remodeling. Pathophysiological stimuli mainly exaggerate the fibrotic and inflammatory response.


Subject(s)
Calcium/metabolism , Cardiomyopathies/metabolism , Haploinsufficiency/physiology , Nervous System Autoimmune Disease, Experimental/etiology , Nervous System Autoimmune Disease, Experimental/metabolism , Plakophilins/metabolism , Animals , Blotting, Western , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Echocardiography , Electrocardiography , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Haploinsufficiency/genetics , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System Autoimmune Disease, Experimental/pathology , Plakophilins/genetics , Polymerase Chain Reaction
7.
Nat Commun ; 10(1): 3455, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31371763

ABSTRACT

The biological basis of the increased risk for psychiatric disorders seen in 15q11.2 copy number deletion is unknown. Previous work has shown disturbances in white matter tracts in human carriers of the deletion. Here, in a novel rat model, we recapitulated low dosage of the candidate risk gene CYFIP1 present within the 15q11.2 interval. Using diffusion tensor imaging, we first showed extensive white matter changes in Cyfip1 mutant rats, which were most pronounced in the corpus callosum and external capsule. Transmission electron microscopy showed that these changes were associated with thinning of the myelin sheath in the corpus callosum. Myelin thinning was independent of changes in axon number or diameter but was associated with effects on mature oligodendrocytes, including aberrant intracellular distribution of myelin basic protein. Finally, we demonstrated effects on cognitive phenotypes sensitive to both disruptions in myelin and callosal circuitry.


Subject(s)
Haploinsufficiency/physiology , Myelin Sheath/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , White Matter/metabolism , Adaptor Proteins, Signal Transducing , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Axons/metabolism , Axons/pathology , Behavior, Animal , Corpus Callosum/metabolism , Corpus Callosum/pathology , Diffusion Tensor Imaging , Disease Models, Animal , Gene Knockout Techniques , Humans , Male , Myelin Basic Protein/metabolism , Myelin Sheath/pathology , Rats
8.
Am J Physiol Cell Physiol ; 317(3): C576-C583, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31291141

ABSTRACT

A murine line haploinsufficient in the cardiac sodium channel has been used to model human Brugada syndrome: a disease causing sudden cardiac death due to lethal ventricular arrhythmias. We explored the effects of cholinergic tone on electrophysiological parameters in wild-type and genetically modified, heterozygous, Scn5a+/- knockout mice. Scn5a+/- ventricular slices showed longer refractory periods than wild-type both at baseline and during isoprenaline challenge. Scn5a+/- hearts also showed lower conduction velocities and increased mean increase in delay than did littermate controls at baseline and blunted responses to isoprenaline challenge. Carbachol exerted limited effects but reversed the effects of isoprenaline with coapplication. Scn5a+/- mice showed a reduction in conduction reserve in that isoprenaline no longer increased conduction velocity, and this was not antagonized by muscarinic agonists.


Subject(s)
Brugada Syndrome/metabolism , Haploinsufficiency/physiology , Isolated Heart Preparation , Myocardial Contraction/physiology , NAV1.5 Voltage-Gated Sodium Channel/deficiency , Animals , Brugada Syndrome/genetics , Brugada Syndrome/physiopathology , Female , Isolated Heart Preparation/methods , Male , Mice , Mice, Knockout , NAV1.5 Voltage-Gated Sodium Channel/genetics , Sodium Channels/deficiency , Sodium Channels/genetics
9.
Stem Cell Res ; 39: 101488, 2019 08.
Article in English | MEDLINE | ID: mdl-31272037

ABSTRACT

Autism spectrum disorders (ASDs) are a group of diseases that affect social interaction, communication and behavior. Molecular mechanisms involved in the pathogenesis of ASDs are complex due to genetic heterogeneity. Recently, pathogenic variants of SCN2A have been strongly associated with ASDs. Here, we generated iPSCs from a patient with ASD and a heterozygous nonsense mutation in SCN2A, by reprogramming mesenchymal stromal cells with non-integrating vectors. The generated iPSC line expresses pluripotency markers, presents a normal karyotype and is able to differentiate into the three germ layers. This iPSC line is a useful tool for modeling ASD and drug screening studies.


Subject(s)
Autism Spectrum Disorder/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , NAV1.2 Voltage-Gated Sodium Channel/genetics , Autism Spectrum Disorder/genetics , Cell Line , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Flow Cytometry , Haploinsufficiency/genetics , Haploinsufficiency/physiology , Humans , Karyotype , Microsatellite Repeats/genetics , Mutation/genetics , Real-Time Polymerase Chain Reaction
10.
Biol Rev Camb Philos Soc ; 94(5): 1774-1785, 2019 10.
Article in English | MEDLINE | ID: mdl-31149781

ABSTRACT

Haploinsufficiency is a form of genetic dominance and is the underlying mechanism of numerous human inherited conditions in which the causal genes are sensitive to altered dosage. This review examines the poorly understood relationships between haploinsufficiency, dosage sensitivity and genetic dominance, whose common theme is the existence of nonlinear relationships between genotype and phenotype. We present an up-to-date account of the bases of haploinsufficiency from the perspective of theoretical and experimental models. We also discuss human conditions caused by haploinsufficiency, including developmental syndromes and cancer. Connections between the understanding of these conditions' genetic mechanisms and advances in treatments are also described.


Subject(s)
Haploinsufficiency/physiology , Chromosome Disorders/genetics , Gene Dosage , Gene Expression , Genes, Tumor Suppressor , Haploinsufficiency/genetics , Humans , Mutation , Neoplasms/genetics , Stochastic Processes
11.
Diabetes ; 68(8): 1635-1648, 2019 08.
Article in English | MEDLINE | ID: mdl-31092480

ABSTRACT

Hyperglycemia-triggered vascular abnormalities are the most serious complications of diabetes mellitus (DM). The major cause of vascular dysfunction in DM is endothelial injury and dysfunction associated with the reduced number and dysfunction of endothelial progenitor cells (EPCs). A major challenge is to identify key regulators of EPCs to restore DM-associated vascular dysfunction. We show that EPCs from heterozygous knockout Aggf1+/- mice presented with impairment of proliferation, migration, angiogenesis, and transendothelial migration as in hyperglycemic mice fed a high-fat diet (HFD) or db/db mice. The number of EPCs from Aggf1+/- mice was significantly reduced. Ex vivo, AGGF1 protein can fully reverse all damaging effects of hyperglycemia on EPCs. In vivo, transplantation of AGGF1-primed EPCs successfully restores blood flow and blocks tissue necrosis and ambulatory impairment in HFD-induced hyperglycemic mice or db/db mice with diabetic hindlimb ischemia. Mechanistically, AGGF1 activates AKT, reduces nuclear localization of Fyn, which increases the nuclear level of Nrf2 and expression of antioxidative genes, and inhibits reactive oxygen species generation. These results suggest that Aggf1 is required for essential function of EPCs, AGGF1 fully reverses the damaging effects of hyperglycemia on EPCs, and AGGF1 priming of EPCs is a novel treatment modality for vascular complications in DM.


Subject(s)
Angiogenic Proteins/metabolism , Bone Marrow Cells/metabolism , Muscle, Skeletal/metabolism , Angiogenic Proteins/genetics , Animals , Cell- and Tissue-Based Therapy , Cells, Cultured , Diet, High-Fat , HEK293 Cells , Haploinsufficiency/genetics , Haploinsufficiency/physiology , Humans , Hyperglycemia/genetics , Hyperglycemia/physiopathology , Lentivirus/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
12.
Mol Neurobiol ; 56(7): 5157-5166, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30519817

ABSTRACT

The canonical Wnt pathway is critical for both the development and adulthood survival and homeostatic maintenance of the midbrain dopaminergic (DA) neurons. Expanding evidence has demonstrated that genetic factors associated with familial Parkinson disease (PD) deregulate this important pathway, suggesting that a disturbed canonical Wnt pathway is likely involved in PD pathogenesis; yet, the specific role of this pathway in sporadic PD remains unclear. In this study, we aimed to determine the effects of specific inhibition of the canonical pathway by hemizygous knockout of ß-catenin, the obligatory component of the canonical Wnt pathway, on paraquat (PQ)-induced DA neuronal degeneration in the substantia nigra in vivo. We found that while hemizygous conditional knockout of ß-catenin in DA neurons did not cause any significant TH+ neuronal loss in the substantia nigra at basal level, it triggered elevated oxidative stress at basal level and further enhanced PQ-induced oxidative damage and loss of TH+ neurons in the substantia nigra and axonal termini in the striatum that manifested as exacerbated motor deficits. These data support the notion that reduced Wnt/ß-catenin signaling in sporadic PD likely contributes to DA neuronal loss through an enhanced oxidative stress-response pathway.


Subject(s)
Dopaminergic Neurons/physiology , Haploinsufficiency/physiology , Paraquat/toxicity , Parkinsonian Disorders/genetics , beta Catenin/deficiency , beta Catenin/genetics , Animals , Dopaminergic Neurons/drug effects , Female , Haploinsufficiency/drug effects , Male , Mice , Mice, Knockout , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism
13.
Int J Mol Sci ; 19(12)2018 Nov 30.
Article in English | MEDLINE | ID: mdl-30513694

ABSTRACT

CCCTC-binding factor (CTCF) is a conserved transcription factor that performs diverse roles in transcriptional regulation and chromatin architecture. Cancer genome sequencing reveals diverse acquired mutations in CTCF, which we have shown functions as a tumour suppressor gene. While CTCF is essential for embryonic development, little is known of its absolute requirement in somatic cells and the consequences of CTCF haploinsufficiency. We examined the consequences of CTCF depletion in immortalised human and mouse cells using shRNA knockdown and CRISPR/Cas9 genome editing as well as examined the growth and development of heterozygous Ctcf (Ctcf+/-) mice. We also analysed the impact of CTCF haploinsufficiency by examining gene expression changes in CTCF-altered endometrial carcinoma. Knockdown and CRISPR/Cas9-mediated editing of CTCF reduced the cellular growth and colony-forming ability of K562 cells. CTCF knockdown also induced cell cycle arrest and a pro-survival response to apoptotic insult. However, in p53 shRNA-immortalised Ctcf+/- MEFs we observed the opposite: increased cellular proliferation, colony formation, cell cycle progression, and decreased survival after apoptotic insult compared to wild-type MEFs. CRISPR/Cas9-mediated targeting in Ctcf+/- MEFs revealed a predominance of in-frame microdeletions in Ctcf in surviving clones, however protein expression could not be ablated. Examination of CTCF mutations in endometrial cancers showed locus-specific alterations in gene expression due to CTCF haploinsufficiency, in concert with downregulation of tumour suppressor genes and upregulation of estrogen-responsive genes. Depletion of CTCF expression imparts a dramatic negative effect on normal cell function. However, CTCF haploinsufficiency can have growth-promoting effects consistent with known cancer hallmarks in the presence of additional genetic hits. Our results confirm the absolute requirement for CTCF expression in somatic cells and provide definitive evidence of CTCF's role as a haploinsufficient tumour suppressor gene. CTCF genetic alterations in endometrial cancer indicate that gene dysregulation is a likely consequence of CTCF loss, contributing to, but not solely driving cancer growth.


Subject(s)
CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Survival/physiology , Endometrial Neoplasms/genetics , Gene Editing , Animals , CRISPR-Cas Systems , Cell Proliferation/genetics , Cell Proliferation/physiology , Cell Survival/genetics , Female , Haploinsufficiency/genetics , Haploinsufficiency/physiology , Humans , K562 Cells , Mice , RNA, Small Interfering/genetics
14.
Nat Commun ; 9(1): 3440, 2018 08 24.
Article in English | MEDLINE | ID: mdl-30143629

ABSTRACT

While multiple mechanisms of BRAFV600-mutant melanoma resistance to targeted MAPK signaling inhibitors (MAPKi) have been reported, the epigenetic regulation of this process remains undetermined. Here, using a CRISPR-Cas9 screen targeting chromatin regulators, we discover that haploinsufficiency of the histone deacetylase SIRT6 allows melanoma cell persistence in the presence of MAPKi. Haploinsufficiency, but not complete loss of SIRT6 promotes IGFBP2 expression via increased chromatin accessibility, H3K56 acetylation at the IGFBP2 locus, and consequent activation of the IGF-1 receptor (IGF-1R) and downstream AKT signaling. Combining a clinically applicable IGF-1Ri with BRAFi overcomes resistance of SIRT6 haploinsufficient melanoma cells in vitro and in vivo. Using matched melanoma samples derived from patients receiving dabrafenib + trametinib, we identify IGFBP2 as a potential biomarker for MAPKi resistance. Our study has not only identified an epigenetic mechanism of drug resistance, but also provides insights into a combinatorial therapy that may overcome resistance to standard-of-care therapy for BRAFV600-mutant melanoma patients.


Subject(s)
Haploinsufficiency/physiology , Melanoma/drug therapy , Melanoma/metabolism , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/metabolism , Receptor, IGF Type 1/metabolism , Sirtuins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromatin Immunoprecipitation , DNA Damage/drug effects , DNA Damage/genetics , Female , Haploinsufficiency/genetics , Humans , Melanoma/genetics , Mice, Nude , Proto-Oncogene Proteins B-raf/genetics , Receptor, IGF Type 1/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Sirtuins/genetics
15.
J Immunol ; 200(6): 2129-2139, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29427417

ABSTRACT

Emergency (stress) granulopoiesis is an episodic process for the production of granulocytes in response to infectious challenge. We previously determined that Fanconi C, a component of the Fanconi DNA-repair pathway, is necessary for successful emergency granulopoiesis. Fanconi anemia results from mutation of any gene in this pathway and is characterized by bone marrow failure (BMF) in childhood and clonal progression in adolescence. Although murine Fanconi anemia models exhibit relatively normal steady-state hematopoiesis, FANCC-/- mice are unable to mount an emergency granulopoiesis response. Instead, these mice develop BMF and die during repeated unsuccessful emergency granulopoiesis attempts. In FANCC-/- mice, BMF is associated with extensive apoptosis of hematopoietic stem and progenitor cells through an undefined mechanism. In this study, we find that TP53 haploinsufficiency completely rescues emergency granulopoiesis in FANCC-/- mice and protects them from BMF during repeated emergency granulopoiesis episodes. Instead, such recurrent challenges accelerated clonal progression in FANCC-/-TP53+/- mice. In FANCC-/- mice, BMF during multiple emergency granulopoiesis attempts was associated with increased ataxia telangiectasia and Rad3-related protein (Atr) and p53 activation with each attempt. In contrast, we found progressive attenuation of expression and activity of Atr, and consequent p53 activation and apoptosis, in the bone marrow of FANCC-/-TP53+/- mice during this process. Therefore, activation of Atr-with consequent Fanconi-mediated DNA repair or p53-dependent apoptosis-is an essential component of emergency granulopoiesis and it protects the bone marrow from genotoxic stress during this process.


Subject(s)
Fanconi Anemia Complementation Group C Protein/metabolism , Granulocytes/metabolism , Haploinsufficiency/physiology , Leukopoiesis/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/physiology , Ataxia Telangiectasia Mutated Proteins/metabolism , Bone Marrow/metabolism , DNA Damage/physiology , DNA Repair/physiology , Fanconi Anemia/metabolism , Mice
16.
Hum Mol Genet ; 27(4): 706-715, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29315381

ABSTRACT

Mutations in the mitochondrially located protein CHCHD10 cause motoneuron disease by an unknown mechanism. In this study, we investigate the mutations p.R15L and p.G66V in comparison to wild-type CHCHD10 and the non-pathogenic variant p.P34S in vitro, in patient cells as well as in the vertebrate in vivo model zebrafish. We demonstrate a reduction of CHCHD10 protein levels in p.R15L and p.G66V mutant patient cells to approximately 50%. Quantitative real-time PCR revealed that expression of CHCHD10 p.R15L, but not of CHCHD10 p.G66V, is already abrogated at the mRNA level. Altered secondary structure and rapid protein degradation are observed with regard to the CHCHD10 p.G66V mutant. In contrast, no significant differences in expression, degradation rate or secondary structure of non-pathogenic CHCHD10 p.P34S are detected when compared with wild-type protein. Knockdown of CHCHD10 expression in zebrafish to about 50% causes motoneuron pathology, abnormal myofibrillar structure and motility deficits in vivo. Thus, our data show that the CHCHD10 mutations p.R15L and p.G66V cause motoneuron disease primarily based on haploinsufficiency of CHCHD10.


Subject(s)
Haploinsufficiency/physiology , Mitochondrial Proteins/metabolism , Motor Neuron Disease/metabolism , Animals , DNA, Complementary/genetics , DNA, Complementary/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Haploinsufficiency/genetics , Humans , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Motor Neuron Disease/genetics , Mutation/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
17.
Biol Res Nurs ; 20(2): 237-243, 2018 03.
Article in English | MEDLINE | ID: mdl-29343077

ABSTRACT

OBJECTIVE: PAX6 haploinsufficiency ( +/-) can occur due to mutations involving only PAX6 in patients with isolated aniridia or as contiguous gene deletions in patients with Wilms tumor, aniridia, genitourinary anomalies, and range of developmental and intellectual disabilities syndrome. Given the role of PAX6 in pineal development and circadian regulation, adolescents with PAX6+/- may experience sleep-wake disturbances. The purpose of this observational study was to explore sleep-related phenotypes in adolescents with PAX6+/-. METHODS: This study compared sleep phenotypes of nine subjects with PAX6+/- (aged 10-19 years) with previously published data on healthy adolescents ( n = 25, aged 10-18 years). Subjects completed the Cleveland Adolescent Sleepiness Questionnaire (CASQ), Patient Reported Outcomes Measurement Information System (PROMIS) Sleep Disturbance (v. 1.0; 8a), and PROMIS Sleep-Related Impairment (v. 1.0; 8b) Questionnaires and wore actigraphs for seven nights to record sleep patterns. RESULTS: Total CASQ, PROMIS sleep-related impairment, and PROMIS sleep disturbance scores were not statistically different between the groups ( ps > .15). Actigraph data for lights off to sleep-onset time were found to be significantly higher in subjects with PAX6+/- versus the healthy comparison group (adjusted mean [95% confidence interval]: 20.1 min [8.1, 49.8] vs. 6.2 min [3.7, 10.4], respectively, p = .04). CONCLUSION: Both adolescents with PAX6+/- and the healthy comparison group on average slept less than 8 hr/night, and overall sleep deprivation in adolescents may have masked differences between groups. This study used rare genetic disorders with biological vulnerability to sleep problems as a genotype-phenotype model. Knowledge of sleep-related phenotypes will assist in designing studies to manage sleep-related symptoms in adolescents.


Subject(s)
Haploinsufficiency/genetics , Haploinsufficiency/physiology , PAX6 Transcription Factor/genetics , Sleep Wake Disorders/genetics , Sleep Wake Disorders/physiopathology , Sleep/genetics , Sleep/physiology , Adolescent , Child , Female , Humans , Male , Mutation , Phenotype , Surveys and Questionnaires
18.
Eur J Neurosci ; 48(8): 2816-2825, 2018 10.
Article in English | MEDLINE | ID: mdl-28925523

ABSTRACT

Schizophrenia is a complex psychiatric disorder with a heterogeneous aetiology involving genetic and environmental factors. Deficiencies in both brain-derived neurotrophic factor (BDNF) and NMDA receptor function have been implicated in the disorder and may play causal and synergistic roles. Perturbations in the regulation of electrophysiological signals, including high-frequency (γ: 30-80 Hz and ß: 20-30 Hz) neuronal oscillations, are also associated with the disorder. This study investigated the influence of BDNF deficiency and NMDA receptor hypofunction on electrophysiological responses to brief acoustic stimuli. Adult BDNF heterozygote (BDNF+/- ) and wild-type littermate C57Bl/6J mice were surgically implanted with EEG recording electrodes. All mice underwent EEG recording sessions to measure ongoing and auditory-evoked electrophysiological responses following treatment with MK-801 (0.3 mg/kg ip) or vehicle. Western blotting on post-mortem cortical tissue assessed parvalbumin and GAD67 expression - markers of interneurons which are involved in the generation of gamma oscillations. Compared with wild-type controls, BDNF+/- mice exhibited markedly dampened electrophysiological responses to auditory stimuli, including reductions in the amplitude of multiple components of the event-related potential and auditory-evoked oscillations, as well as reduced ongoing cortical gamma oscillations. MK-801 elevated ongoing gamma power but suppressed evoked gamma power, and this was observed equally across genotypes. BDNF+/- mice also displayed reductions in parvalbumin, but not GAD67 expression. We conclude that reduced BDNF expression leads to impairments in the generation of high-frequency neural oscillations, but this is not synergistic with NMDA receptor hypofunction. Reduced parvalbumin expression associated with BDNF haploinsufficiency may provide a molecular explanation for these electrophysiological deficits.


Subject(s)
Brain Waves/physiology , Brain-Derived Neurotrophic Factor/biosynthesis , Haploinsufficiency/physiology , Prefrontal Cortex/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
19.
Hum Mol Genet ; 27(4): 716-731, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29281018

ABSTRACT

In humans, a copy of the DUX4 retrogene is located in each unit of the D4Z4 macrosatellite repeat that normally comprises 8-100 units. The D4Z4 repeat has heterochromatic features and does not express DUX4 in somatic cells. Individuals with facioscapulohumeral muscular dystrophy (FSHD) have a partial failure of somatic DUX4 repression resulting in the presence of DUX4 protein in sporadic muscle nuclei. Somatic DUX4 derepression is caused by contraction of the D4Z4 repeat to 1-10 units (FSHD1) or by heterozygous mutations in genes responsible for maintaining the D4Z4 chromatin structure in a repressive state (FSHD2). One of the FSHD2 genes is the structural maintenance of chromosomes hinge domain 1 (SMCHD1) gene. SMCHD1 mutations have also been identified in FSHD1; patients carrying a contracted D4Z4 repeat and a SMCHD1 mutation are more severely affected than relatives with only a contracted repeat or a SMCHD1 mutation. To evaluate the modifier role of SMCHD1, we crossbred mice carrying a contracted D4Z4 repeat (D4Z4-2.5 mice) with mice that are haploinsufficient for Smchd1 (Smchd1MommeD1 mice). D4Z4-2.5/Smchd1MommeD1 mice presented with a significantly reduced body weight and developed skin lesions. The same skin lesions, albeit in a milder form, were also observed in D4Z4-2.5 mice, suggesting that reduced Smchd1 levels aggravate disease in the D4Z4-2.5 mouse model. Our study emphasizes the evolutionary conservation of the SMCHD1-dependent epigenetic regulation of the D4Z4 repeat array and further suggests that the D4Z4-2.5/Smchd1MommeD1 mouse model may be used to unravel the function of DUX4 in non-muscle tissues like the skin.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Haploinsufficiency/physiology , Animals , Blotting, Western , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , DNA Methylation/genetics , DNA Methylation/physiology , Fibroblasts/metabolism , Flow Cytometry , Haploinsufficiency/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Keratinocytes/metabolism , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Skin , Thymocytes
20.
Orphanet J Rare Dis ; 12(1): 183, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29258554

ABSTRACT

CLINICAL DESCRIPTION: KBG syndrome is characterized by macrodontia of upper central incisors, distinctive craniofacial features such as triangular face, prominent nasal bridge, thin upper lip and synophrys; skeletal findings including short stature, delayed bone age, and costovertebral anomalies; and developmental delay/intellectual disability sometimes associated with seizures and EEG abnormalities. The condition was named KBG syndrome after the initials of the last names of three original families reported in 1975. EPIDEMIOLOGY: The prevalence of KBG syndrome is not established. There are over 100 patients reported in the literature. It is likely that KBG syndrome is underreported due to incomplete recognition and very mild presentations of the disorder in some individuals. KBG syndrome is typically milder in females. ETIOLOGY: Causative variants in ANKRD11 have been identified in affected individuals. The vast majority of identified variants are loss of function, which include nonsense and frameshift variants and larger deletions at 16q24.3. Haploinsufficiency appears to be the mechanism of pathogenicity. GENETIC COUNSELING: Familial and de novo cases have been reported. Causative de novo variants occur approximately one third of the time. Transmission follows an autosomal dominant pattern. The syndrome displays inter- and intra-familial variability.


Subject(s)
Abnormalities, Multiple/metabolism , Bone Diseases, Developmental/metabolism , Intellectual Disability/metabolism , Tooth Abnormalities/metabolism , Abnormalities, Multiple/genetics , Animals , Bone Diseases, Developmental/genetics , Chromosome Deletion , Comparative Genomic Hybridization , Facies , Female , Haploinsufficiency/genetics , Haploinsufficiency/physiology , Humans , Intellectual Disability/genetics , Male , Repressor Proteins/genetics , Repressor Proteins/metabolism , Seizures/genetics , Seizures/metabolism , Tooth Abnormalities/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...