Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
STAR Protoc ; 4(3): 102535, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37682716

ABSTRACT

Exploring early embryonic gene expression is challenging due to the rate of development and the limited material available. Here, we present a protocol for ordering Drosophila embryos along a developmental pseudo-time trajectory and determining the sex of the embryos using RNA-seq data. We describe steps for sample collection, RNA isolation, RNA-seq, and RNA-seq data processing. We then detail the establishment of a continuous transcriptome dataset for assessing gene expression throughout early development and in a sex-specific manner. For complete details on the use and execution of this protocol, please refer to Pérez-Mojica et al.1.


Subject(s)
Drosophila , Gene Expression Profiling , Female , Male , Animals , Drosophila/genetics , Sequence Analysis, RNA , RNA-Seq , Transcriptome/genetics
2.
Chemosphere ; 330: 138738, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37084897

ABSTRACT

Maternal exposure to environmental contaminants during pregnancy poses a significant threat to a developing fetus, as these substances can easily cross the placenta and disrupt the neurodevelopment of offspring. Specifically, the hypothalamus is essential in the regulation of metabolism, notably during critical windows of development. An abnormal hormonal and inflammatory milieu during development can trigger persistent changes in the function of hypothalamic circuits, leading to long-lasting effects on the body's energy homeostasis and metabolism. We recently demonstrated that gestational exposure to clinically relevant levels of benzene induces severe metabolic dysregulation in the offspring. Given the central role of the hypothalamus in metabolic control, we hypothesized that prenatal exposure to benzene impacts hypothalamic development, contributing to the adverse metabolic effects in the offspring. C57BL/6JB dams were exposed to benzene at 50 ppm in the inhalation chambers exclusively during pregnancy (from E0.5 to E19). Transcriptomic analysis of the exposed offspring at postnatal day 21 (P21) revealed hypothalamic changes in genes related to metabolic regulation, inflammation, and neurodevelopment exclusively in males. Moreover, the hypothalamus of prenatally benzene-exposed male offspring displayed alterations in orexigenic and anorexigenic projections, impairments in leptin signaling, and increased microgliosis. Additional exposure to benzene during lactation did not promote further microgliosis or astrogliosis in the offspring, while the high-fat diet (HFD) challenge in adulthood exacerbated glucose metabolism and hypothalamic inflammation in benzene-exposed offspring of both sexes. These findings reveal the persistent adverse effects of prenatal benzene exposure on hypothalamic circuits and neuroinflammation, predisposing the offspring to long-lasting metabolic health conditions.


Subject(s)
Metabolic Diseases , Prenatal Exposure Delayed Effects , Pregnancy , Humans , Female , Mice , Male , Animals , Benzene/toxicity , Benzene/metabolism , Prenatal Exposure Delayed Effects/metabolism , Mice, Inbred C57BL , Hypothalamus/metabolism , Diet, High-Fat/adverse effects , Inflammation/metabolism , Metabolic Diseases/metabolism
3.
Cell Genom ; 3(3): 100265, 2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36950383

ABSTRACT

The transformative events during early organismal development lay the foundation for body formation and long-term phenotype. The rapid progression of events and the limited material available present major barriers to studying these earliest stages of development. Herein, we report an operationally simple RNA sequencing approach for high-resolution, time-sensitive transcriptome analysis in early (≤3 h) Drosophila embryos. This method does not require embryo staging but relies on single-embryo RNA sequencing and transcriptome ordering along a developmental trajectory (pseudo-time). The resulting high-resolution, time-sensitive mRNA expression profiles reveal the exact onset of transcription and degradation for thousands of transcripts. Further, using sex-specific transcription signatures, embryos can be sexed directly, eliminating the need for Y chromosome genotyping and revealing patterns of sex-biased transcription from the beginning of zygotic transcription. Our data provide an unparalleled resolution of gene expression during early development and enhance the current understanding of early transcriptional processes.

4.
Cell Metab ; 35(5): 821-836.e7, 2023 05 02.
Article in English | MEDLINE | ID: mdl-36948185

ABSTRACT

The mechanisms that specify and stabilize cell subtypes remain poorly understood. Here, we identify two major subtypes of pancreatic ß cells based on histone mark heterogeneity (ßHI and ßLO). ßHI cells exhibit ∼4-fold higher levels of H3K27me3, distinct chromatin organization and compaction, and a specific transcriptional pattern. ßHI and ßLO cells also differ in size, morphology, cytosolic and nuclear ultrastructure, epigenomes, cell surface marker expression, and function, and can be FACS separated into CD24+ and CD24- fractions. Functionally, ßHI cells have increased mitochondrial mass, activity, and insulin secretion in vivo and ex vivo. Partial loss of function indicates that H3K27me3 dosage regulates ßHI/ßLO ratio in vivo, suggesting that control of ß cell subtype identity and ratio is at least partially uncoupled. Both subtypes are conserved in humans, with ßHI cells enriched in humans with type 2 diabetes. Thus, epigenetic dosage is a novel regulator of cell subtype specification and identifies two functionally distinct ß cell subtypes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Humans , Insulin-Secreting Cells/metabolism , Histones/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Epigenesis, Genetic , Insulin Secretion
5.
bioRxiv ; 2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36711607

ABSTRACT

The hypothalamus is essential in the regulation of metabolism, notably during critical windows of development. An abnormal hormonal and inflammatory milieu during development can trigger persistent changes in the function of hypothalamic circuits, leading to long-lasting effects on the body’s energy homeostasis and metabolism. We recently demonstrated that gestational exposure to benzene at smoking levels induces severe metabolic dysregulation in the offspring. Given the central role of the hypothalamus in metabolic control, we hypothesized that prenatal exposure to benzene impacts hypothalamic development, contributing to the adverse metabolic effects in the offspring. C57BL/6JB dams were exposed to benzene in the inhalation chambers exclusively during pregnancy (from E0.5 to E19). The transcriptome analysis of the offspring hypothalamus at postnatal day 21 (P21) revealed changes in genes related to metabolic regulation, inflammation, and neurodevelopment exclusively in benzene-exposed male offspring. Moreover, the hypothalamus of prenatally benzene-exposed male offspring displayed alterations in orexigenic and anorexigenic projections, impairments in leptin signaling, and increased microgliosis. Additional exposure to benzene during lactation did not promote further microgliosis or astrogliosis in the offspring, while the high-fat diet (HFD) challenge in adulthood exacerbated glucose metabolism and hypothalamic inflammation in benzene-exposed offspring of both sexes. These findings reveal the persistent impact of prenatal benzene exposure on hypothalamic circuits and neuroinflammation, predisposing the offspring to long-lasting metabolic health conditions.

6.
J Vis Exp ; (184)2022 06 16.
Article in English | MEDLINE | ID: mdl-35786676

ABSTRACT

Obesity is a complex disease influenced by genetics, epigenetics, the environment, and their interactions. Mature adipocytes represent the major cell type in white adipose tissue. Understanding how adipocytes function and respond to (epi)genetic and environmental signals is essential for identifying the cause(s) of obesity. RNA and chromatin have previously been isolated from adipocytes using enzymatic digestion. In addition, protocols have been developed for nuclear isolation, where purification is achieved by fluorescence-activated cell sorting (FACS) of adipocyte-specific transgenic reporters. One of the greatest challenges to achieving high yield and quality during such protocols is the substantial amount of lipid contained in adipose tissue. The present protocol describes an optimized procedure for isolating mature adipocytes that leverages heptane to separate lipids from the targets of interest (RNA/chromatin). The resulting RNA has high integrity and generates high-quality RNA-seq results. Likewise, the procedure improves nuclei yield rate and generates reproducible ChIP-seq results across samples. Therefore, the current study provides a reliable and universal murine adipocyte isolation protocol suitable for whole-genome transcriptome and epigenome studies.


Subject(s)
Adipocytes, White , Transcriptome , Animals , Chromatin/metabolism , Epigenome , Mice , Obesity/metabolism , RNA/metabolism
7.
Ann N Y Acad Sci ; 1506(1): 118-141, 2021 12.
Article in English | MEDLINE | ID: mdl-34791665

ABSTRACT

The human transcriptome contains many types of noncoding RNAs, which rival the number of protein-coding species. From long noncoding RNAs (lncRNAs) that are over 200 nucleotides long to piwi-interacting RNAs (piRNAs) of only 20 nucleotides, noncoding RNAs play important roles in regulating transcription, epigenetic modifications, translation, and cell signaling. Roles for noncoding RNAs in disease mechanisms are also being uncovered, and several species have been identified as potential drug targets. On May 11-14, 2021, the Keystone eSymposium "Noncoding RNAs: Biology and Applications" brought together researchers working in RNA biology, structure, and technologies to accelerate both the understanding of RNA basic biology and the translation of those findings into clinical applications.


Subject(s)
Congresses as Topic/trends , Epigenesis, Genetic/genetics , Gene Targeting/trends , RNA, Untranslated/administration & dosage , RNA, Untranslated/genetics , Research Report , Animals , Drug Delivery Systems/methods , Drug Delivery Systems/trends , Gene Targeting/methods , Humans , MicroRNAs/administration & dosage , MicroRNAs/genetics , RNA, Long Noncoding/administration & dosage , RNA, Long Noncoding/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Untranslated/administration & dosage , RNA, Small Untranslated/genetics , Signal Transduction/genetics
8.
Semin Cell Dev Biol ; 97: 116-122, 2020 01.
Article in English | MEDLINE | ID: mdl-31404658

ABSTRACT

It is well established that lifestyle and other environmental factors have the potential to shape our own health and future. Research from the last two decades, however, provides mounting evidence that parental exposures or experiences such as dietary challenges, toxin exposure, or stress can impact the health and future of our offspring. There are indications that both the paternal and maternal germline are able to store information of the parental environment and pass certain information on to their progeny. These intergenerational effects are mediated by epigenetic mechanisms. This review summarizes and discusses insights into germline epigenetic plasticity caused by environmental stimuli and how such alterations are transmitted to induce a stable phenotype in the offspring.


Subject(s)
Epigenesis, Genetic/genetics , Germ Cells/metabolism , Inheritance Patterns , Humans
10.
Nat Genet ; 51(3): 452-469, 2019 03.
Article in English | MEDLINE | ID: mdl-30778226

ABSTRACT

Body-fat distribution is a risk factor for adverse cardiovascular health consequences. We analyzed the association of body-fat distribution, assessed by waist-to-hip ratio adjusted for body mass index, with 228,985 predicted coding and splice site variants available on exome arrays in up to 344,369 individuals from five major ancestries (discovery) and 132,177 European-ancestry individuals (validation). We identified 15 common (minor allele frequency, MAF ≥5%) and nine low-frequency or rare (MAF <5%) coding novel variants. Pathway/gene set enrichment analyses identified lipid particle, adiponectin, abnormal white adipose tissue physiology and bone development and morphology as important contributors to fat distribution, while cross-trait associations highlight cardiometabolic traits. In functional follow-up analyses, specifically in Drosophila RNAi-knockdowns, we observed a significant increase in the total body triglyceride levels for two genes (DNAH10 and PLXND1). We implicate novel genes in fat distribution, stressing the importance of interrogating low-frequency and protein-coding variants.


Subject(s)
Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Homeostasis/genetics , Lipids/genetics , Proteins/genetics , Animals , Body Fat Distribution/methods , Body Mass Index , Case-Control Studies , Drosophila/genetics , Exome/genetics , Female , Gene Frequency/genetics , Genome-Wide Association Study/methods , Humans , Male , Risk Factors , Waist-Hip Ratio/methods
11.
Cell Metab ; 27(6): 1294-1308.e7, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29754954

ABSTRACT

To date, it remains largely unclear to what extent chromatin machinery contributes to the susceptibility and progression of complex diseases. Here, we combine deep epigenome mapping with single-cell transcriptomics to mine for evidence of chromatin dysregulation in type 2 diabetes. We find two chromatin-state signatures that track ß cell dysfunction in mice and humans: ectopic activation of bivalent Polycomb-silenced domains and loss of expression at an epigenomically unique class of lineage-defining genes. ß cell-specific Polycomb (Eed/PRC2) loss of function in mice triggers diabetes-mimicking transcriptional signatures and highly penetrant, hyperglycemia-independent dedifferentiation, indicating that PRC2 dysregulation contributes to disease. The work provides novel resources for exploring ß cell transcriptional regulation and identifies PRC2 as necessary for long-term maintenance of ß cell identity. Importantly, the data suggest a two-hit (chromatin and hyperglycemia) model for loss of ß cell identity in diabetes.


Subject(s)
Chromatin/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Gene Silencing , Insulin-Secreting Cells/metabolism , Polycomb Repressive Complex 2/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Chromosome Mapping , Diabetes Mellitus, Type 2/genetics , Epigenomics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Hyperglycemia/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Polycomb Repressive Complex 2/genetics , Single-Cell Analysis
13.
Nat Genet ; 50(5): 766-767, 2018 05.
Article in English | MEDLINE | ID: mdl-29549330

ABSTRACT

In the version of this article originally published, one of the two authors with the name Wei Zhao was omitted from the author list and the affiliations for both authors were assigned to the single Wei Zhao in the author list. In addition, the ORCID for Wei Zhao (Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA) was incorrectly assigned to author Wei Zhou. The errors have been corrected in the HTML and PDF versions of the article.

14.
Nat Genet ; 50(1): 26-41, 2018 01.
Article in English | MEDLINE | ID: mdl-29273807

ABSTRACT

Genome-wide association studies (GWAS) have identified >250 loci for body mass index (BMI), implicating pathways related to neuronal biology. Most GWAS loci represent clusters of common, noncoding variants from which pinpointing causal genes remains challenging. Here we combined data from 718,734 individuals to discover rare and low-frequency (minor allele frequency (MAF) < 5%) coding variants associated with BMI. We identified 14 coding variants in 13 genes, of which 8 variants were in genes (ZBTB7B, ACHE, RAPGEF3, RAB21, ZFHX3, ENTPD6, ZFR2 and ZNF169) newly implicated in human obesity, 2 variants were in genes (MC4R and KSR2) previously observed to be mutated in extreme obesity and 2 variants were in GIPR. The effect sizes of rare variants are ~10 times larger than those of common variants, with the largest effect observed in carriers of an MC4R mutation introducing a stop codon (p.Tyr35Ter, MAF = 0.01%), who weighed ~7 kg more than non-carriers. Pathway analyses based on the variants associated with BMI confirm enrichment of neuronal genes and provide new evidence for adipocyte and energy expenditure biology, widening the potential of genetically supported therapeutic targets in obesity.


Subject(s)
Body Mass Index , Energy Intake/genetics , Energy Metabolism/genetics , Genetic Variation , Obesity/genetics , Adult , Animals , Drosophila/genetics , Female , Gene Frequency , Humans , Male , Proteins/genetics , Syndrome
15.
Cell Metab ; 26(1): 171-184.e6, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28683285

ABSTRACT

Natural killer (NK) cells contribute to the development of obesity-associated insulin resistance. We demonstrate that in mice obesity promotes expansion of a distinct, interleukin-6 receptor (IL6R)a-expressing NK subpopulation, which also expresses a number of other myeloid lineage genes such as the colony-stimulating factor 1 receptor (Csf1r). Selective ablation of this Csf1r-expressing NK cell population prevents obesity and insulin resistance. Moreover, conditional inactivation of IL6Ra or Stat3 in NK cells limits obesity-associated formation of these myeloid signature NK cells, protecting from obesity, insulin resistance, and obesity-associated inflammation. Also in humans IL6Ra+ NK cells increase in obesity and correlate with markers of systemic low-grade inflammation, and their gene expression profile overlaps with characteristic gene sets of NK cells in obese mice. Collectively, we demonstrate that obesity-associated inflammation and metabolic disturbances depend on interleukin-6/Stat3-dependent formation of a distinct NK population, which may provide a target for the treatment of obesity, metaflammation-associated pathologies, and diabetes.


Subject(s)
Energy Metabolism , Glucose/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Killer Cells, Natural/pathology , Obesity/metabolism , STAT3 Transcription Factor/metabolism , Adult , Animals , Homeostasis , Humans , Inflammation/complications , Inflammation/pathology , Insulin Resistance , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Obesity/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Interleukin-6/metabolism , Signal Transduction , Young Adult
16.
Cell ; 164(3): 353-64, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26824653

ABSTRACT

More than one-half billion people are obese, and despite progress in genetic research, much of the heritability of obesity remains enigmatic. Here, we identify a Trim28-dependent network capable of triggering obesity in a non-Mendelian, "on/off" manner. Trim28(+/D9) mutant mice exhibit a bi-modal body-weight distribution, with isogenic animals randomly emerging as either normal or obese and few intermediates. We find that the obese-"on" state is characterized by reduced expression of an imprinted gene network including Nnat, Peg3, Cdkn1c, and Plagl1 and that independent targeting of these alleles recapitulates the stochastic bi-stable disease phenotype. Adipose tissue transcriptome analyses in children indicate that humans too cluster into distinct sub-populations, stratifying according to Trim28 expression, transcriptome organization, and obesity-associated imprinted gene dysregulation. These data provide evidence of discrete polyphenism in mouse and man and thus carry important implications for complex trait genetics, evolution, and medicine.


Subject(s)
Epigenesis, Genetic , Haploinsufficiency , Nuclear Proteins/genetics , Obesity/genetics , Repressor Proteins/genetics , Thinness/genetics , Adolescent , Animals , Body Mass Index , Child , Child, Preschool , Humans , Mice , Nutrition Surveys , Polymorphism, Genetic , Tripartite Motif-Containing Protein 28
17.
Nat Rev Genet ; 16(11): 665-81, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26460345

ABSTRACT

Obesity and its associated diseases are expected to affect more than 1 billion people by the year 2030. These figures have sparked intensive research into the molecular control of food intake, nutrient distribution, storage and metabolism--processes that are collectively termed energy homeostasis. Recent decades have also seen dramatic developments in our understanding of gene regulation at the signalling, chromatin and post-transcriptional levels. The seemingly exponential growth in this complexity now poses a major challenge for translational researchers in need of simplified but accurate paradigms for clinical use. In this Review, we consider the current understanding of transcriptional control of energy homeostasis, including both transcriptional and epigenetic regulators, and crosstalk between pathways. We also provide insights into emerging developments and challenges in this field.


Subject(s)
Energy Metabolism/genetics , Gene Expression Regulation , Homeostasis/genetics , Signal Transduction/genetics , Epigenesis, Genetic , Humans , Models, Genetic , Obesity/genetics , Transcription, Genetic , Translational Research, Biomedical/methods , Translational Research, Biomedical/trends
18.
Cell ; 159(6): 1352-64, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25480298

ABSTRACT

The global rise in obesity has revitalized a search for genetic and epigenetic factors underlying the disease. We present a Drosophila model of paternal-diet-induced intergenerational metabolic reprogramming (IGMR) and identify genes required for its encoding in offspring. Intriguingly, we find that as little as 2 days of dietary intervention in fathers elicits obesity in offspring. Paternal sugar acts as a physiological suppressor of variegation, desilencing chromatin-state-defined domains in both mature sperm and in offspring embryos. We identify requirements for H3K9/K27me3-dependent reprogramming of metabolic genes in two distinct germline and zygotic windows. Critically, we find evidence that a similar system may regulate obesity susceptibility and phenotype variation in mice and humans. The findings provide insight into the mechanisms underlying intergenerational metabolic reprogramming and carry profound implications for our understanding of phenotypic variation and evolution.


Subject(s)
Disease Models, Animal , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Epigenesis, Genetic , Obesity/genetics , Animals , Carbohydrate Metabolism , Diet , Embryo, Nonmammalian/metabolism , Eye Color , Female , Genetic Predisposition to Disease , Heterochromatin/metabolism , Humans , Male , Mice , Obesity/metabolism , Spermatozoa/metabolism
19.
Nat Genet ; 46(9): 973-981, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108384

ABSTRACT

Polycomb/Trithorax response elements (PRE/TREs) can switch their function reversibly between silencing and activation by mechanisms that are poorly understood. Here we show that a switch in forward and reverse noncoding transcription from the Drosophila melanogaster vestigial (vg) PRE/TRE switches the status of the element between silencing (induced by the forward strand) and activation (induced by the reverse strand). In vitro, both noncoding RNAs inhibit PRC2 histone methyltransferase activity, but, in vivo, only the reverse strand binds PRC2. Overexpression of the reverse strand evicts PRC2 from chromatin and inhibits its enzymatic activity. We propose that the interaction of RNAs with PRC2 is differentially regulated in vivo, allowing regulated inhibition of local PRC2 activity. Genome-wide analysis shows that strand switching of noncoding RNAs occurs at several hundred Polycomb-binding sites in fly and vertebrate genomes. This work identifies a previously unreported and potentially widespread class of PRE/TREs that switch function by switching the direction of noncoding RNA transcription.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Drosophila Proteins/genetics , Genes, Switch , Polycomb-Group Proteins/genetics , RNA, Untranslated , Response Elements , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Chromatin/genetics , DNA-Binding Proteins/genetics , Drosophila melanogaster , Genome, Insect , Histone-Lysine N-Methyltransferase/genetics , Molecular Sequence Data , Transcription Factors/genetics
20.
Cell Mol Life Sci ; 70(9): 1609-21, 2013 May.
Article in English | MEDLINE | ID: mdl-23463237

ABSTRACT

The DNA sequence largely defines gene expression and phenotype. However, it is becoming increasingly clear that an additional chromatin-based regulatory network imparts both stability and plasticity to genome output, modifying phenotype independently of the genetic blueprint. Indeed, alterations in this "epigenetic" control layer underlie, at least in part, the reason for monozygotic twins being discordant for disease. Functionally, this regulatory layer comprises post-translational modifications of DNA and histones, as well as small and large noncoding RNAs. Together these regulate gene expression by changing chromatin organization and DNA accessibility. Successive technological advances over the past decade have enabled researchers to map the chromatin state with increasing accuracy and comprehensiveness, catapulting genetic research into a genome-wide era. Here, aiming particularly at the genomics/epigenomics newcomer, we review the epigenetic basis that has helped drive the technological shift and how this progress is shaping our understanding of complex disease.


Subject(s)
Epigenomics/methods , Animals , Chromatin/genetics , Chromatin/metabolism , DNA/genetics , DNA/metabolism , Epigenesis, Genetic , Genetic Predisposition to Disease , Histones/genetics , Histones/metabolism , Humans , Protein Processing, Post-Translational , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...