Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(50): e2304074120, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38051767

ABSTRACT

Severity of neurobehavioral deficits in children born from adverse pregnancies, such as maternal alcohol consumption and diabetes, does not always correlate with the adversity's duration and intensity. Therefore, biological signatures for accurate prediction of the severity of neurobehavioral deficits, and robust tools for reliable identification of such biomarkers, have an urgent clinical need. Here, we demonstrate that significant changes in the alternative splicing (AS) pattern of offspring lymphocyte RNA can function as accurate peripheral biomarkers for motor learning deficits in mouse models of prenatal alcohol exposure (PAE) and offspring of mother with diabetes (OMD). An aptly trained deep-learning model identified 29 AS events common to PAE and OMD as superior predictors of motor learning deficits than AS events specific to PAE or OMD. Shapley-value analysis, a game-theory algorithm, deciphered the trained deep-learning model's learnt associations between its input, AS events, and output, motor learning performance. Shapley values of the deep-learning model's input identified the relative contribution of the 29 common AS events to the motor learning deficit. Gene ontology and predictive structure-function analyses, using Alphafold2 algorithm, supported existing evidence on the critical roles of these molecules in early brain development and function. The direction of most AS events was opposite in PAE and OMD, potentially from differential expression of RNA binding proteins in PAE and OMD. Altogether, this study posits that AS of lymphocyte RNA is a rich resource, and deep-learning is an effective tool, for discovery of peripheral biomarkers of neurobehavioral deficits in children of diverse adverse pregnancies.


Subject(s)
Diabetes Mellitus , Fetal Alcohol Spectrum Disorders , Prenatal Exposure Delayed Effects , Mice , Animals , Child , Humans , Pregnancy , Female , Alternative Splicing , Prenatal Exposure Delayed Effects/chemically induced , Ethanol , Diabetes Mellitus/chemically induced , Biomarkers/metabolism , RNA/metabolism , Fetal Alcohol Spectrum Disorders/genetics
2.
J Neurosci ; 41(31): 6775-6792, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34193554

ABSTRACT

Epigenetic modifiers are increasingly being investigated as potential therapeutics to modify and overcome disease phenotypes. Diseases of the nervous system present a particular problem as neurons are postmitotic and demonstrate relatively stable gene expression patterns and chromatin organization. We have explored the ability of epigenetic modifiers to prevent degeneration of rod photoreceptors in a mouse model of retinitis pigmentosa (RP), using rd10 mice of both sexes. The histone modification eraser enzymes lysine demethylase 1 (LSD1) and histone deacetylase 1 (HDAC1) are known to have dramatic effects on the development of rod photoreceptors. In the RP mouse model, inhibitors of these enzymes blocked rod degeneration, preserved vision, and affected the expression of multiple genes including maintenance of rod-specific transcripts and downregulation of those involved in inflammation, gliosis, and cell death. The neuroprotective activity of LSD1 inhibitors includes two pathways. First, through targeting histone modifications, they increase accessibility of chromatin and upregulate neuroprotective genes, such as from the Wnt pathway. We propose that this process is going in rod photoreceptors. Second, through nonhistone targets, they inhibit transcription of inflammatory genes and inflammation. This process is going in microglia, and lack of inflammation keeps rod photoreceptors alive.SIGNIFICANCE STATEMENT Retinal degenerations are a leading cause of vision loss. RP is genetically very heterogeneous, and the multiple pathways leading to cell death are one reason for the slow progress in identifying suitable treatments for patients. Here we demonstrate that inhibition of LSD1and HDAC1 in a mouse model of RP leads to preservation of rod photoreceptors and visual function, retaining of expression of rod-specific genes, and with decreased inflammation, cell death, and Müller cell gliosis. We propose that these epigenetic inhibitors cause more open and accessible chromatin, allowing expression of neuroprotective genes. A second mechanism that allows rod photoreceptor survival is suppression of inflammation by epigenetic inhibitors in microglia. Manipulation of epigenetic modifiers is a new strategy to fight neurodegeneration in RP.


Subject(s)
Histone Deacetylase 1/antagonists & inhibitors , Histone Demethylases/antagonists & inhibitors , Nerve Degeneration/pathology , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/metabolism , Animals , Cell Death/drug effects , Disease Models, Animal , Epigenesis, Genetic/drug effects , Female , Histone Deacetylase Inhibitors/pharmacology , Male , Mice , Mice, Inbred C57BL , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/pathology , Tranylcypromine/pharmacology
3.
Blood ; 136(9): 1067-1079, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32396937

ABSTRACT

FLT3 is a frequently mutated gene that is highly associated with a poor prognosis in acute myeloid leukemia (AML). Despite initially responding to FLT3 inhibitors, most patients eventually relapse with drug resistance. The mechanism by which resistance arises and the initial response to drug treatment that promotes cell survival is unknown. Recent studies show that a transiently maintained subpopulation of drug-sensitive cells, so-called drug-tolerant "persisters" (DTPs), can survive cytotoxic drug exposure despite lacking resistance-conferring mutations. Using RNA sequencing and drug screening, we find that treatment of FLT3 internal tandem duplication AML cells with quizartinib, a selective FLT3 inhibitor, upregulates inflammatory genes in DTPs and thereby confers susceptibility to anti-inflammatory glucocorticoids (GCs). Mechanistically, the combination of FLT3 inhibitors and GCs enhances cell death of FLT3 mutant, but not wild-type, cells through GC-receptor-dependent upregulation of the proapoptotic protein BIM and proteasomal degradation of the antiapoptotic protein MCL-1. Moreover, the enhanced antileukemic activity by quizartinib and dexamethasone combination has been validated using primary AML patient samples and xenograft mouse models. Collectively, our study indicates that the combination of FLT3 inhibitors and GCs has the potential to eliminate DTPs and therefore prevent minimal residual disease, mutational drug resistance, and relapse in FLT3-mutant AML.


Subject(s)
Antineoplastic Agents/therapeutic use , Glucocorticoids/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11/biosynthesis , Bcl-2-Like Protein 11/genetics , Benzothiazoles/pharmacology , Benzothiazoles/therapeutic use , Computer Simulation , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Drug Resistance, Neoplasm , Drug Synergism , Gene Expression Regulation, Leukemic/drug effects , Glucocorticoids/pharmacology , Humans , Inflammation/genetics , Mice , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplastic Stem Cells/drug effects , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Protein Kinase Inhibitors/pharmacology , Selection, Genetic , Transcriptome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , fms-Like Tyrosine Kinase 3/genetics
4.
Mol Psychiatry ; 25(6): 1159-1174, 2020 06.
Article in English | MEDLINE | ID: mdl-31439936

ABSTRACT

Exposure to stress during early life (infancy/childhood) has long-term effects on the structure and function of the prefrontal cortex (PFC), and increases the risk for adult depression and anxiety disorders. However, little is known about the molecular and cellular mechanisms of these effects. Here, we focused on changes induced by chronic maternal separation during the first 2 weeks of postnatal life. Unbiased mRNA expression profiling in the medial PFC (mPFC) of maternally separated (MS) pups identified an increased expression of myelin-related genes and a decreased expression of immediate early genes. Oligodendrocyte lineage markers and birthdating experiments indicated a precocious oligodendrocyte differentiation in the mPFC at P15, leading to a depletion of the oligodendrocyte progenitor pool in MS adults. We tested the role of neuronal activity in oligodendrogenesis, using designed receptors exclusively activated by designed drugs (DREADDs) techniques. hM4Di or hM3Dq constructs were transfected into mPFC neurons using fast-acting AAV8 viruses. Reduction of mPFC neuron excitability during the first 2 postnatal weeks caused a premature differentiation of oligodendrocytes similar to the MS pups, while chemogenetic activation normalised it in the MS animals. Bidirectional manipulation of neuron excitability in the mPFC during the P2-P14 period had long lasting effects on adult emotional behaviours and on temporal object recognition: hM4Di mimicked MS effects, while hM3Dq prevented the pro-depressive effects and short-term memory impairment of MS. Thus, our results identify neuronal activity as a critical target of early-life stress and demonstrate its function in controlling both postnatal oligodendrogenesis and adult mPFC-related behaviours.


Subject(s)
Maternal Deprivation , Oligodendroglia/pathology , Stress, Psychological , Animals , Behavior, Animal , Cell Proliferation , Emotions , Female , Male , Mice , Prefrontal Cortex/pathology , Prefrontal Cortex/physiopathology , Pregnancy
5.
J Biol Chem ; 294(14): 5508-5520, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30733333

ABSTRACT

Diabetes promotes the posttranslational modification of proteins by O-linked addition of GlcNAc (O-GlcNAcylation) to Ser/Thr residues of proteins and thereby contributes to diabetic complications. In the retina of diabetic mice, the repressor of mRNA translation, eIF4E-binding protein 1 (4E-BP1), is O-GlcNAcylated, and sequestration of the cap-binding protein eukaryotic translation initiation factor (eIF4E) is enhanced. O-GlcNAcylation has also been detected on several eukaryotic translation initiation factors and ribosomal proteins. However, the functional consequence of this modification is unknown. Here, using ribosome profiling, we evaluated the effect of enhanced O-GlcNAcylation on retinal gene expression. Mice receiving thiamet G (TMG), an inhibitor of the O-GlcNAc hydrolase O-GlcNAcase, exhibited enhanced retinal protein O-GlcNAcylation. The principal effect of TMG on retinal gene expression was observed in ribosome-associated mRNAs (i.e. mRNAs undergoing translation), as less than 1% of mRNAs exhibited changes in abundance. Remarkably, ∼19% of the transcriptome exhibited TMG-induced changes in ribosome occupancy, with 1912 mRNAs having reduced and 1683 mRNAs having increased translational rates. In the retina, the effect of O-GlcNAcase inhibition on translation of specific mitochondrial proteins, including superoxide dismutase 2 (SOD2), depended on 4E-BP1/2. O-GlcNAcylation enhanced cellular respiration and promoted mitochondrial superoxide levels in WT cells, and 4E-BP1/2 deletion prevented O-GlcNAcylation-induced mitochondrial superoxide in cells in culture and in the retina. The retina of diabetic WT mice exhibited increased reactive oxygen species levels, an effect not observed in diabetic 4E-BP1/2-deficient mice. These findings provide evidence for a mechanism whereby diabetes-induced O-GlcNAcylation promotes oxidative stress in the retina by altering the selection of mRNAs for translation.


Subject(s)
Carrier Proteins/metabolism , Diabetic Retinopathy/metabolism , Eye Proteins/metabolism , Mitochondria/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Retina/metabolism , Acylation , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , Eukaryotic Initiation Factors , Eye Proteins/genetics , Female , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Oxygen Consumption/drug effects , Phosphoproteins/genetics , Pyrans/pharmacology , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Retina/pathology , Thiazoles/pharmacology
6.
Nucleic Acids Res ; 46(17): 8908-8916, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30032250

ABSTRACT

During carcinogenesis, cells are exposed to increased replication stress due to replication fork arrest at sites of DNA lesions and difficult to replicate genomic regions. Efficient fork restart and DNA repair are important for cancer cell proliferation. We previously showed that the ADP-ribosyltransferase PARP10 interacts with the replication protein proliferating cell nuclear antigen and promotes lesion bypass by recruiting specialized, non-replicative DNA polymerases. Here, we show that PARP10 is overexpressed in a large proportion of human tumors. To understand the role of PARP10 in cellular transformation, we inactivated PARP10 in HeLa cancer cells by CRISPR/Cas9-mediated gene knockout, and overexpressed it in non-transformed RPE-1 cells. We found that PARP10 promotes cellular proliferation, and its overexpression alleviates cellular sensitivity to replication stress and fosters the restart of stalled replication forks. Importantly, mouse xenograft studies showed that loss of PARP10 reduces the tumorigenesis activity of HeLa cells, while its overexpression results in tumor formation by non-transformed RPE-1 cells. Our findings indicate that PARP10 promotes cellular transformation, potentially by alleviating replication stress and suggest that targeting PARP10 may represent a novel therapeutic approach.


Subject(s)
Carcinogenesis/genetics , Neoplasm Proteins/physiology , Poly(ADP-ribose) Polymerases/physiology , Proto-Oncogene Proteins/physiology , Animals , CRISPR-Cas Systems , Cell Division , Cell Line, Transformed , DNA Damage , DNA Replication , Female , Gene Knockout Techniques , HeLa Cells , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Proteins/deficiency , Poly(ADP-ribose) Polymerases/deficiency , Proto-Oncogene Proteins/deficiency , Retinal Pigment Epithelium/cytology , Up-Regulation
7.
Int J Mol Sci ; 21(8)2020 Apr 13.
Article in English | MEDLINE | ID: mdl-32294904

ABSTRACT

Osteochondromas are cartilage-capped growths located proximate to the physis that can cause skeletal deformities, pain, limited motion, and neurovascular impingement. Previous studies have demonstrated retinoic acid receptor gamma (RARγ) agonists to inhibit ectopic endochondral ossification, therefore we hypothesize that RARγ agonists can target on established osteochondromas. The purpose of this study was to examine the action of RARγ agonist in human osteochondromas. Osteochondroma specimens were obtained during surgery, subjected to explant culture and were treated with RARγ agonists or vehicles. Gene expression analysis confirmed the up-regulation of RARγ target genes in the explants treated with NRX 204647 and Palovarotene and revealed strong inhibition of cartilage matrix and increased extracellular matrix proteases gene expression. In addition, immunohistochemical staining for the neoepitope of protease-cleaved aggrecan indicated that RARγ agonist treatment stimulated cartilage matrix degradation. Interestingly, cell survival studies demonstrated that RARγ agonist treatment stimulated cell death. Moreover, RNA sequencing analysis indicates changes in multiple molecular pathways due to RARγ agonists treatment, showing similarly to human growth plate chondrocytes. Together, these findings suggest that RARγ agonist may exert anti-tumor function on osteochondromas by inhibiting matrix synthesis, promoting cartilage matrix degradation and stimulating cell death.


Subject(s)
Bone Neoplasms/metabolism , Osteochondroma/metabolism , Receptors, Retinoic Acid/agonists , Animals , Biomarkers , Bone Neoplasms/drug therapy , Bone Neoplasms/etiology , Bone Neoplasms/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Computational Biology/methods , Gene Expression Profiling , Gene Ontology , Growth Plate/metabolism , Growth Plate/pathology , Humans , Molecular Sequence Annotation , Osteochondroma/drug therapy , Osteochondroma/etiology , Osteochondroma/pathology , Signal Transduction , Tissue Culture Techniques , Transcriptome , Retinoic Acid Receptor gamma
8.
EMBO Rep ; 17(6): 874-86, 2016 06.
Article in English | MEDLINE | ID: mdl-27146073

ABSTRACT

Defects in DNA replication, DNA damage response, and DNA repair compromise genomic stability and promote cancer development. In particular, unrepaired DNA lesions can arrest the progression of the DNA replication machinery during S-phase, causing replication stress, mutations, and DNA breaks. HUWE1 is a HECT-type ubiquitin ligase that targets proteins involved in cell fate, survival, and differentiation. Here, we report that HUWE1 is essential for genomic stability, by promoting replication of damaged DNA We show that HUWE1-knockout cells are unable to mitigate replication stress, resulting in replication defects and DNA breakage. Importantly, we find that this novel role of HUWE1 requires its interaction with the replication factor PCNA, a master regulator of replication fork restart, at stalled replication forks. Finally, we provide evidence that HUWE1 mono-ubiquitinates H2AX to promote signaling at stalled forks. Altogether, our work identifies HUWE1 as a novel regulator of the replication stress response.


Subject(s)
DNA Replication , Proliferating Cell Nuclear Antigen/metabolism , Stress, Physiological , Ubiquitin-Protein Ligases/metabolism , Cell Line , DNA Damage , DNA Repair , Gene Knockout Techniques , Genomic Instability , Histones/metabolism , Humans , Phenotype , Protein Binding , Protein Processing, Post-Translational , Protein Transport , Tumor Suppressor Proteins , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
9.
Clin Transplant ; 32(9): e13358, 2018 09.
Article in English | MEDLINE | ID: mdl-30044009

ABSTRACT

Kidney transplant from donors with hepatitis C virus (HCV) antibody has been limited to HCV viremic recipients only, due to concern of the HCV transmission. However, the new antiviral medications provide an opportunity to expand the utilization of these donors. To study the risk of HCV transmission in kidney transplantation, we used discarded donor kidneys and determined HCV RNA levels by quantitative real-time PCR in bilateral (right and left) kidney biopsies and plasma from 14 HCV antibody-positive donors (sensitivity: 15 international unit (IU)/mL plasma; 1.8 IU/50 nL kidney). In three NAT-negative donors, HCV RNA was negative in plasma and kidney. In all 11 NAT-positive donors, HCV RNA was positive in plasma (range: 5807-19 134 177 IU/mL) but negative in six kidneys from four donors with plasma HCV RNA <1.5 million IU/µL. HCV RNA correlated between right and left kidneys (P = 0.75) and between kidney and plasma (r = 0.86). When normalized by volume, HCV RNA median (range) was 49 (0-957) IU/50 nL plasma and 1.0 (0-103) IU/50 nL kidney, significantly lower in kidney (P = 0.005) than in plasma (14-fold). Plasma HCV RNA can be used to predict the kidney HCV load. Future studies are needed if plasma/kidney HCV levels can be used to stratify donors for transmission risk and recipients for post-transplant management in extended utilization of HCV antibody-positive donors.


Subject(s)
Hepacivirus/genetics , Hepatitis C Antibodies/blood , Hepatitis C/diagnosis , Kidney/metabolism , RNA, Viral/genetics , Tissue Donors/supply & distribution , Tissue and Organ Harvesting/statistics & numerical data , Hepatitis C/genetics , Hepatitis C/transmission , Humans , Kidney/virology
10.
Int J Mol Sci ; 18(12)2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29232918

ABSTRACT

The contribution of low-frequency and damaging genetic variants associated with platelet function to ischemic stroke (IS) susceptibility remains unknown. We employed a deep re-sequencing approach in Polish patients in order to investigate the contribution of rare variants (minor allele frequency, MAF < 1%) to the IS genetic susceptibility in this population. The genes selected for re-sequencing consisted of 26 genes coding for proteins associated with the surface membrane of platelets. Targeted pooled re-sequencing (Illumina HiSeq 2500) was performed on genomic DNA of 500 cases (patients with history of clinically proven diagnosis of large-vessel IS) and 500 controls. After quality control and prioritization based on allele frequency and damaging probability, follow-up individual genotyping of deleterious rare variants was performed in patients from the original cohort. Gene-based analyses identified an association between IS and 6 rare functional and damaging variants in the purinergic genes (P2RY1 and P2RY12 locus). The predicted properties of the most damaging rare variants in P2RY1 and P2RY12 were confirmed by using mouse fibroblast cell cultures transfected with plasmid constructs containing cDNA of mutated variants (FLIPR on FlexStation3). This study identified a putative role for rare variants in P2RY1 and P2RY12 genes involved in platelet reactivity on large-vessel IS susceptibility in a Polish population.


Subject(s)
Brain Ischemia/complications , Genetic Association Studies/methods , Polymorphism, Single Nucleotide , Receptors, Purinergic P2Y12/genetics , Receptors, Purinergic P2Y1/genetics , Stroke/genetics , Animals , Brain Ischemia/genetics , Cell Line , Female , Gene Frequency , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Male , Mice , Poland , Sequence Analysis, DNA , Sequence Deletion
11.
J Clin Densitom ; 18(1): 30-6, 2015.
Article in English | MEDLINE | ID: mdl-25220887

ABSTRACT

To investigate the association between abdominal obesity and metabolic syndrome (MetS) burden in a population-based sample of adolescents, we used data from 421 adolescents who completed the follow-up examination in the Penn State Children Cohort study. Dual-energy x-ray absorptiometry (DXA) was used to assess abdominal obesity, as measured by android/gynoid fat ratio (A/G ratio), android/whole body fat proportion (A/W proportion), visceral (VAT) and subcutaneous fat (SAT) areas. Continuous metabolic syndrome score (cMetS), calculated as the sum of the age and sex-adjusted standardized residual (Z-score) of five established MetS components, was used to assess the MetS burden. Linear regression models were used to analyze the impact of DXA measures on cMetS components. All models were adjusted for age, race, sex, and general obesity. We found abdominal obesity is significantly associated with increased cMetS. With 1 standard deviation (SD) increase in A/G ratio, A/W proportion, VAT area, and SAT area, cMetS increased by 1.34 (SE=0.17), 1.25 (SE=0.19), 1.67 (SE=0.17), and 1.84 (SE=0.20) units, respectively. At individual component level, strongest association was observed between abdominal obesity and insulin resistance (IR) than lipid-based or blood pressure-based components. VAT and SAT had a stronger impact on IR than android ratio-based DXA measurements. In conclusion, abdominal obesity is associated with higher MetS burden in adolescent population. The association between abdominal obesity and IR measure is the strongest, suggesting the key impact of abdominal obesity on IR in adolescents MetS burden.


Subject(s)
Metabolic Syndrome , Obesity, Abdominal , Absorptiometry, Photon/methods , Adolescent , Blood Glucose/analysis , Body Fat Distribution/methods , Cohort Studies , Cost of Illness , Female , Humans , Insulin Resistance , Intra-Abdominal Fat/metabolism , Linear Models , Lipoproteins, HDL/blood , Male , Metabolic Syndrome/epidemiology , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Obesity, Abdominal/complications , Obesity, Abdominal/diagnosis , Obesity, Abdominal/epidemiology , Obesity, Abdominal/metabolism , Risk Assessment , Risk Factors , Statistics as Topic , Triglycerides/blood , United States/epidemiology
12.
Front Cell Dev Biol ; 11: 1236356, 2023.
Article in English | MEDLINE | ID: mdl-37829185

ABSTRACT

Introduction: Wnt/ß-catenin signaling controls cell division and lineage specification during embryonic development, and is crucial for stem cells maintenance and gut tissue regeneration in adults. Aberrant activation of Wnt/ß-catenin signaling is also essential for the pathogenesis of a variety of malignancies. The RNA-binding protein IGF2BP1 is a transcriptional target of Wnt/ß-catenin signaling, normally expressed during development and often reactivated in cancer cells, where it regulates the stability of oncogenic mRNA. Methods: In this study, we employed iCLIP and RNA sequencing techniques to investigate the role of IGF2BP1 in the post-transcriptional regulation of Wnt/ß-catenin-induced genes at a global level within colorectal cancer (CRC) cells characterized by constitutively active Wnt/ß-catenin signaling. Results and Discussion: In our study, we show that, in contrast to normal cells, CRC cells exhibit a much stronger dependency on IGF2BP1 expression for Wnt/ß-catenin-regulated genes. We show that both untransformed and CRC cells have their unique subsets of Wnt/ß-catenin-regulated genes that IGF2BP1 directly controls through binding to their mRNA. Our iCLIP analysis revealed a significant change in the IGF2BP1-binding sites throughout the target transcriptomes and a significant change in the enrichment of 6-mer motifs associated with IGF2BP1 binding in response to Wnt/ß-catenin signaling. Our study also revealed a signature of IGF2BP1-regulated genes that are significantly associated with colon cancer-free survival in humans, as well as potential targets for CRC treatment. Overall, this study highlights the complex and context-dependent regulation of Wnt/ß-catenin signaling target genes by IGF2BP1 in non-transformed and CRC cells and identifies potential targets for colon cancer treatment.

13.
Bone Res ; 11(1): 20, 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37080994

ABSTRACT

Longitudinal bone growth relies on endochondral ossification in the cartilaginous growth plate, where chondrocytes accumulate and synthesize the matrix scaffold that is replaced by bone. The chondroprogenitors in the resting zone maintain the continuous turnover of chondrocytes in the growth plate. Malnutrition is a leading cause of growth retardation in children; however, after recovery from nutrient deprivation, bone growth is accelerated beyond the normal rate, a phenomenon termed catch-up growth. Although nutritional status is a known regulator of long bone growth, it is largely unknown whether and how chondroprogenitor cells respond to deviations in nutrient availability. Here, using fate-mapping analysis in Axin2CreERT2 mice, we showed that dietary restriction increased the number of Axin2+ chondroprogenitors in the resting zone and simultaneously inhibited their differentiation. Once nutrient deficiency was resolved, the accumulated chondroprogenitor cells immediately restarted differentiation and formed chondrocyte columns, contributing to accelerated growth. Furthermore, we showed that nutrient deprivation reduced the level of phosphorylated Akt in the resting zone and that exogenous IGF-1 restored the phosphorylated Akt level and stimulated differentiation of the pooled chondroprogenitors, decreasing their numbers. Our study of Axin2CreERT2 revealed that nutrient availability regulates the balance between accumulation and differentiation of chondroprogenitors in the growth plate and further demonstrated that IGF-1 partially mediates this regulation by promoting the committed differentiation of chondroprogenitor cells.

14.
CNS Neurosci Ther ; 28(6): 922-931, 2022 06.
Article in English | MEDLINE | ID: mdl-35238164

ABSTRACT

AIMS: The molecular genetic mechanisms underlying postoperative nausea and vomiting (PONV) in the brain have not been fully elucidated. This study aimed to determine the changes in whole transcriptome in the nucleus of the solitary tract (NTS) in an animal model of PONV, to screen a drug candidate and to elucidate the molecular genetic mechanisms of PONV development. METHODS: Twenty-one female musk shrews were assigned into three groups: the Surgery group (shrew PONV model, n = 9), the Sham group (n = 6), and the Naïve group (n = 6). In behavioral studies, the main outcome was the number of emetic episodes. In genetic experiments, changes in the transcriptome in the NTS were measured. In a separate study, 12 shrews were used to verify the candidate mechanism underlying PONV. RESULTS: A median of six emetic episodes occurred in both the Sham and Surgery groups. Whole-transcriptome analysis indicated the inhibition of the GABAB receptor-mediated signaling pathway in the PONV model. Baclofen (GABAB receptor agonist) administration eliminated emetic behaviors in the shrew PONV model. CONCLUSIONS: Our findings suggest that the GABAB receptor-mediated signaling pathway is involved in emesis and that baclofen may be a novel therapeutic or prophylactic agent for PONV.


Subject(s)
Antiemetics , Animals , Antiemetics/therapeutic use , Baclofen/pharmacology , Baclofen/therapeutic use , Emetics , Female , Gene Expression Profiling , Postoperative Nausea and Vomiting/drug therapy , Shrews/physiology , Solitary Nucleus , Vomiting/drug therapy , Vomiting/prevention & control
15.
Genes Brain Behav ; : e12759, 2021 Jun 10.
Article in English | MEDLINE | ID: mdl-34114352

ABSTRACT

The genetic mechanisms of postoperative nausea and vomiting (PONV) and the involvement of the catecholamine system in the brain have not been elucidated. Eating kaolin clay as a type of pica has been examined as an alternative behavior to emesis. Here, we evaluated changes in whole-transcriptome analysis in the nucleus of the solitary tract (NTS) in a rat pica model as a surrogate behavior of PONV to elucidate the molecular genetic mechanisms of the development of PONV and the involvement of the catecholamine system in the NTS. First, kaolin pica behaviors were investigated in 71 female Wistar rats following isoflurane anesthesia, surgical insult or morphine administration. Multiple linear regression analysis showed that 3 mg/kg morphine increased kaolin intake by 2.8 g (P = 0.0002). Next, total RNA and protein were extracted from the dissected NTS, and whole-transcriptome sequencing (RNA-seq) was performed to identify PONV-associated genes and to verify the involvement of the catecholamine system. The gene expression levels of tyrosine hydroxylase and dopamine beta-hydroxylase in the catecholamine biosynthesis pathway decreased significantly in the PONV model. Release of noradrenaline, a catecholamine pathway end product, may have increased at the synaptic terminal of the NTS neuron after pica behavior. Systematic administration of α2 adrenergic receptor agonists after surgery reduced kaolin intake from 3.2 g (control) to 1.0 g (P = 0.0014). These results indicated that catecholamine neurotransmission was involved in the development of PONV in the NTS.

16.
J Cell Biol ; 220(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34037658

ABSTRACT

Oncogene-induced senescence (OIS) is a stable cell cycle arrest that occurs in normal cells upon oncogene activation. Cells undergoing OIS express a wide variety of secreted factors that affect the senescent microenvironment termed the senescence-associated secretory phenotype (SASP), which is beneficial or detrimental in a context-dependent manner. OIS cells are also characterized by marked epigenetic changes. We globally assessed histone modifications of OIS cells and discovered an increase in the active histone marks H3K79me2/3. The H3K79 methyltransferase disruptor of telomeric silencing 1-like (DOT1L) was necessary and sufficient for increased H3K79me2/3 occupancy at the IL1A gene locus, but not other SASP genes, and was downstream of STING. Modulating DOT1L expression did not affect the cell cycle arrest. Together, our studies establish DOT1L as an epigenetic regulator of the SASP, whose expression is uncoupled from the senescence-associated cell cycle arrest, providing a potential strategy to inhibit the negative side effects of senescence while maintaining the beneficial inhibition of proliferation.


Subject(s)
Cellular Senescence , DNA Methylation , Epigenesis, Genetic , Fibroblasts/enzymology , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Interleukin-1alpha/metabolism , 9,10-Dimethyl-1,2-benzanthracene , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Cycle Checkpoints , Cell Proliferation , Female , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Interleukin-1alpha/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Microscopy, Fluorescence , Papilloma/chemically induced , Papilloma/genetics , Papilloma/metabolism , Papilloma/pathology , Phenotype , Secretory Pathway , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate
17.
J Perinatol ; 41(3): 551-561, 2021 03.
Article in English | MEDLINE | ID: mdl-33177681

ABSTRACT

OBJECTIVE: Extreme preterm infants are a growing population in neonatal intensive care units who carry a high mortality and morbidity. Multiple factors play a role in preterm birth, resulting in major impact on organogenesis leading to complications including bronchopulmonary dysplasia (BPD). The goal of this study was to identify biomarker signatures associated with prematurity and BPD. STUDY DESIGN: We analyzed miRNA and mRNA profiles in tracheal aspirates (TAs) from 55 infants receiving invasive mechanical ventilation. Twenty-eight infants were extremely preterm and diagnosed with BPD, and 27 were term babies receiving invasive mechanical ventilation for elective procedures. RESULT: We found 22 miRNAs and 33 genes differentially expressed (FDR < 0.05) in TAs of extreme preterm infants with BPD vs. term babies without BPD. Pathway analysis showed associations with inflammatory response, cellular growth/proliferation, and tissue development. CONCLUSIONS: Specific mRNA-miRNA signatures in TAs may serve as biomarkers for BPD pathogenesis, a consequence of extreme prematurity.


Subject(s)
Bronchopulmonary Dysplasia , MicroRNAs , Premature Birth , Bronchopulmonary Dysplasia/genetics , Female , Humans , Infant , Infant, Newborn , Infant, Premature , MicroRNAs/genetics , Pregnancy , Transcriptome
18.
Elife ; 92020 12 23.
Article in English | MEDLINE | ID: mdl-33355091

ABSTRACT

Synapse formation and regulation require signaling interactions between pre- and postsynaptic proteins, notably cell adhesion molecules (CAMs). It has been proposed that the functions of neuroligins (Nlgns), postsynaptic CAMs, rely on the formation of trans-synaptic complexes with neurexins (Nrxns), presynaptic CAMs. Nlgn3 is a unique Nlgn isoform that localizes at both excitatory and inhibitory synapses. However, Nlgn3 function mediated via Nrxn interactions is unknown. Here we demonstrate that Nlgn3 localizes at postsynaptic sites apposing vesicular glutamate transporter 3-expressing (VGT3+) inhibitory terminals and regulates VGT3+ inhibitory interneuron-mediated synaptic transmission in mouse organotypic slice cultures. Gene expression analysis of interneurons revealed that the αNrxn1+AS4 splice isoform is highly expressed in VGT3+ interneurons as compared with other interneurons. Most importantly, postsynaptic Nlgn3 requires presynaptic αNrxn1+AS4 expressed in VGT3+ interneurons to regulate inhibitory synaptic transmission. Our results indicate that specific Nlgn-Nrxn signaling generates distinct functional properties at synapses.


Subject(s)
Calcium-Binding Proteins/physiology , Cell Adhesion Molecules, Neuronal/physiology , GABAergic Neurons/physiology , Hippocampus/physiology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Neural Cell Adhesion Molecules/physiology , Animals , CA1 Region, Hippocampal/physiology , Female , Gene Knockdown Techniques , Male , Mice , Mice, Inbred C57BL , Synapses/physiology
19.
Nat Commun ; 11(1): 6118, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33257658

ABSTRACT

Inhibitors of poly-ADP-ribose polymerase 1 (PARPi) are highly effective in killing cells deficient in homologous recombination (HR); thus, PARPi have been clinically utilized to successfully treat BRCA2-mutant tumors. However, positive response to PARPi is not universal, even among patients with HR-deficiency. Here, we present the results of genome-wide CRISPR knockout and activation screens which reveal genetic determinants of PARPi response in wildtype or BRCA2-knockout cells. Strikingly, we report that depletion of the ubiquitin ligase HUWE1, or the histone acetyltransferase KAT5, top hits from our screens, robustly reverses the PARPi sensitivity caused by BRCA2-deficiency. We identify distinct mechanisms of resistance, in which HUWE1 loss increases RAD51 levels to partially restore HR, whereas KAT5 depletion rewires double strand break repair by promoting 53BP1 binding to double-strand breaks. Our work provides a comprehensive set of putative biomarkers that advance understanding of PARPi response, and identifies novel pathways of PARPi resistance in BRCA2-deficient cells.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Poly(ADP-ribose) Polymerase Inhibitors/isolation & purification , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/drug effects , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Biomarkers , DNA Damage , DNA Repair , Gene Knockout Techniques , HeLa Cells , Homologous Recombination/drug effects , Humans , Lysine Acetyltransferase 5/metabolism , Mad2 Proteins/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1 , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
20.
Neuron ; 107(6): 1197-1211.e9, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32707082

ABSTRACT

Neural stem cells directly or indirectly generate all neurons and macroglial cells and guide migrating neurons by using a palisade-like scaffold made of their radial fibers. Here, we describe an unexpected role for the radial fiber scaffold in directing corticospinal and other axons at the junction between the striatum and globus pallidus. The maintenance of this scaffold, and consequently axon pathfinding, is dependent on the expression of an atypical RHO-GTPase, RND3/RHOE, together with its binding partner ARHGAP35/P190A, a RHO GTPase-activating protein, in the radial glia-like neural stem cells within the ventricular zone of the medial ganglionic eminence. This role is independent of RND3 and ARHGAP35 expression in corticospinal neurons, where they regulate dendritic spine formation, axon elongation, and pontine midline crossing in a FEZF2-dependent manner. The prevalence of neural stem cell scaffolds and their expression of RND3 and ARHGAP35 suggests that these observations might be broadly relevant for axon guidance and neural circuit formation.


Subject(s)
Axon Guidance , Neural Stem Cells/cytology , Neuroglia/cytology , Animals , Axons/metabolism , Corpus Striatum/cytology , Corpus Striatum/growth & development , Dendritic Spines/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Globus Pallidus/cytology , Globus Pallidus/growth & development , Humans , Mice , Neural Stem Cells/metabolism , Neuroglia/metabolism , Pyramidal Tracts/cytology , Pyramidal Tracts/growth & development , Repressor Proteins/genetics , Repressor Proteins/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL