Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 405
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 176(5): 949-951, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30794779

RESUMO

Single-cell transcriptomics coupled with dynamic two-color fluorescence are used by Gehart et al. (2019) to elucidate adult mammalian cell trajectories in real time. The authors' close examination of intestinal enteroendocrine differentiation reveals new lineage features and shifting cell identities, and experiments in organoids uncover specific roles for transcriptional regulators identified by this approach.


Assuntos
Organoides , Células-Tronco , Animais , Diferenciação Celular , Linhagem da Célula , Intestinos
2.
Cell ; 165(6): 1389-1400, 2016 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-27212235

RESUMO

Bivalent promoters in embryonic stem cells (ESCs) carry methylation marks on two lysine residues, K4 and K27, in histone3 (H3). K4me2/3 is generally considered to promote transcription, and Polycomb Repressive Complex 2 (PRC2) places K27me3, which is erased at lineage-restricted genes when ESCs differentiate in culture. Molecular defects in various PRC2 null adult tissues lack a unifying explanation. We found that epigenomes in adult mouse intestine and other self-renewing tissues show fewer and distinct bivalent promoters compared to ESCs. Groups of tissue-specific genes that carry bivalent marks are repressed, despite the presence of promoter H3K4me2/3. These are the predominant genes de-repressed in PRC2-deficient adult cells, where aberrant expression is proportional to the H3K4me2/3 levels observed at their promoters in wild-type cells. Thus, in adult animals, PRC2 specifically represses genes with acquired, tissue-restricted promoter bivalency. These findings provide new insights into specificity in chromatin-based gene regulation.


Assuntos
Células-Tronco Embrionárias/metabolismo , Complexo Repressor Polycomb 2/genética , Regiões Promotoras Genéticas , Animais , Diferenciação Celular/genética , Metilação de DNA , Regulação da Expressão Gênica , Histonas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/citologia , Lisina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Complexo Repressor Polycomb 2/metabolismo
3.
Genes Dev ; 36(11-12): 684-698, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35738677

RESUMO

The progeny of intestinal stem cells (ISCs) dedifferentiate in response to ISC attrition. The precise cell sources, transitional states, and chromatin remodeling behind this activity remain unclear. In the skin, stem cell recovery after injury preserves an epigenetic memory of the damage response; whether similar memories arise and persist in regenerated ISCs is not known. We addressed these questions by examining gene activity and open chromatin at the resolution of single Neurog3-labeled mouse intestinal crypt cells, hence deconstructing forward and reverse differentiation of the intestinal secretory (Sec) lineage. We show that goblet, Paneth, and enteroendocrine cells arise by multilineage priming in common precursors, followed by selective access at thousands of cell-restricted cis-elements. Selective ablation of the ISC compartment elicits speedy reversal of chromatin and transcriptional features in large fractions of precursor and mature crypt Sec cells without obligate cell cycle re-entry. ISC programs decay and reappear along a cellular continuum lacking discernible discrete interim states. In the absence of gross tissue damage, Sec cells simply reverse their forward trajectories, without invoking developmental or other extrinsic programs, and starting chromatin identities are effectively erased. These findings identify strikingly plastic molecular frameworks in assembly and regeneration of a self-renewing tissue.


Assuntos
Cromatina , Células-Tronco , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/genética , Cromatina/metabolismo , Mucosa Intestinal/metabolismo , Intestinos , Camundongos , Proteínas do Tecido Nervoso/metabolismo
4.
Genes Dev ; 36(1-2): 38-52, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34969824

RESUMO

Barrett's esophagus (BE) and gastric intestinal metaplasia are related premalignant conditions in which areas of human stomach epithelium express mixed gastric and intestinal features. Intestinal transcription factors (TFs) are expressed in both conditions, with unclear causal roles and cis-regulatory mechanisms. Ectopic CDX2 reprogrammed isogenic mouse stomach organoid lines to a hybrid stomach-intestinal state transcriptionally similar to clinical metaplasia; squamous esophageal organoids resisted this CDX2-mediated effect. Reprogramming was associated with induced activity at thousands of previously inaccessible intestine-restricted enhancers, where CDX2 occupied DNA directly. HNF4A, a TF recently implicated in BE pathogenesis, induced weaker intestinalization by binding a novel shadow Cdx2 enhancer and hence activating Cdx2 expression. CRISPR/Cas9-mediated germline deletion of that cis-element demonstrated its requirement in Cdx2 induction and in the resulting activation of intestinal genes in stomach cells. dCas9-conjugated KRAB repression mapped this activity to the shadow enhancer's HNF4A binding site. Altogether, we show extensive but selective recruitment of intestinal enhancers by CDX2 in gastric cells and that HNF4A-mediated ectopic CDX2 expression in the stomach occurs through a conserved shadow cis-element. These findings identify mechanisms for TF-driven intestinal metaplasia and a likely pathogenic TF hierarchy.


Assuntos
Esôfago de Barrett , Fatores de Transcrição , Animais , Esôfago de Barrett/genética , Esôfago de Barrett/metabolismo , Esôfago de Barrett/patologia , Fator de Transcrição CDX2/genética , Proteínas de Homeodomínio/genética , Metaplasia/genética , Camundongos , Fatores de Transcrição/genética
5.
Cell ; 158(4): 889-902, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25126792

RESUMO

Engineering clinically relevant cells in vitro holds promise for regenerative medicine, but most protocols fail to faithfully recapitulate target cell properties. To address this, we developed CellNet, a network biology platform that determines whether engineered cells are equivalent to their target tissues, diagnoses aberrant gene regulatory networks, and prioritizes candidate transcriptional regulators to enhance engineered conversions. Using CellNet, we improved B cell to macrophage conversion, transcriptionally and functionally, by knocking down predicted B cell regulators. Analyzing conversion of fibroblasts to induced hepatocytes (iHeps), CellNet revealed an unexpected intestinal program regulated by the master regulator Cdx2. We observed long-term functional engraftment of mouse colon by iHeps, thereby establishing their broader potential as endoderm progenitors and demonstrating direct conversion of fibroblasts into intestinal epithelium. Our studies illustrate how CellNet can be employed to improve direct conversion and to uncover unappreciated properties of engineered cells.


Assuntos
Engenharia Celular/métodos , Biologia de Sistemas/métodos , Animais , Linfócitos B/citologia , Linfócitos B/metabolismo , Engenharia Celular/normas , Redes Reguladoras de Genes , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos
6.
Mol Cell ; 78(1): 141-151.e5, 2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32027840

RESUMO

Polycomb repressive complex 2 (PRC2) places H3K27me3 at developmental genes and is causally implicated in keeping bivalent genes silent. It is unclear if that silence requires minimum H3K27me3 levels and how the mark transmits faithfully across mammalian somatic cell generations. Mouse intestinal cells lacking EZH2 methyltransferase reduce H3K27me3 proportionately at all PRC2 target sites, but ∼40% uniform residual levels keep target genes inactive. These genes, derepressed in PRC2-null villus cells, remain silent in intestinal stem cells (ISCs). Quantitative chromatin immunoprecipitation and computational modeling indicate that because unmodified histones dilute H3K27me3 by 50% each time DNA replicates, PRC2-deficient ISCs initially retain sufficient H3K27me3 to avoid gene derepression. EZH2 mutant human lymphoma cells also require multiple divisions before H3K27me3 dilution relieves gene silencing. In both cell types, promoters with high basal H3K4me2/3 activate in spite of some residual H3K27me3, compared to less-poised promoters. These findings have implications for PRC2 inhibition in cancer therapy.


Assuntos
Replicação do DNA , Proteína Potenciadora do Homólogo 2 de Zeste/fisiologia , Inativação Gênica , Código das Histonas , Regiões Promotoras Genéticas , Animais , Linhagem Celular Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Histonas/metabolismo , Humanos , Intestinos/citologia , Camundongos , Complexo Repressor Polycomb 2/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/metabolismo , Ativação Transcricional
7.
Mol Cell ; 74(3): 542-554.e5, 2019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-30905509

RESUMO

Developing and adult tissues use different cis-regulatory elements. Although DNA at some decommissioned embryonic enhancers is hypomethylated in adult cells, it is unknown whether this putative epigenetic memory is complete and recoverable. We find that, in adult mouse cells, hypomethylated CpG dinucleotides preserve a nearly complete archive of tissue-specific developmental enhancers. Sites that carry the active histone mark H3K4me1, and are therefore considered "primed," are mainly cis elements that act late in organogenesis. In contrast, sites decommissioned early in development retain hypomethylated DNA as a singular property. In adult intestinal and blood cells, sustained absence of polycomb repressive complex 2 indirectly reactivates most-and only-hypomethylated developmental enhancers. Embryonic and fetal transcriptional programs re-emerge as a result, in reverse chronology to cis element inactivation during development. Thus, hypomethylated DNA in adult cells preserves a "fossil record" of tissue-specific developmental enhancers, stably marking decommissioned sites and enabling recovery of this epigenetic memory.


Assuntos
Metilação de DNA/genética , Elementos Facilitadores Genéticos/genética , Epigenômica , Histonas/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos
8.
Genes Dev ; 32(21-22): 1430-1442, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30366903

RESUMO

After acquiring competence for selected cell fates, embryonic primordia may remain plastic for variable periods before tissue identity is irrevocably determined (commitment). We investigated the chromatin basis for these developmental milestones in mouse endoderm, a tissue with recognizable rostro-caudal patterning and transcription factor (TF)-dependent interim plasticity. Foregut-specific enhancers are as accessible and active in early midgut as in foregut endoderm, and intestinal enhancers and identity are established only after ectopic cis-regulatory elements are decommissioned. Depletion of the intestinal TF CDX2 before this cis element transition stabilizes foregut enhancers, reinforces ectopic transcriptional programs, and hence imposes foregut identities on the midgut. Later in development, as the window of chromatin plasticity elapses, CDX2 depletion weakens intestinal, without strengthening foregut, enhancers. Thus, midgut endoderm is primed for heterologous cell fates, and TFs act on a background of shifting chromatin access to determine intestinal at the expense of foregut identity. Similar principles likely govern other fate commitments.


Assuntos
Endoderma/metabolismo , Elementos Facilitadores Genéticos , Mucosa Intestinal/metabolismo , Intestinos/embriologia , Transcrição Gênica , Animais , Fator de Transcrição CDX2/genética , Fator de Transcrição CDX2/metabolismo , Cromatina/metabolismo , Endoderma/embriologia , Intestinos/anatomia & histologia , Camundongos
9.
Annu Rev Physiol ; 83: 405-427, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33234018

RESUMO

The cardinal properties of adult tissue stem cells are self-renewal and the ability to generate diverse resident cell types. The daily losses of terminally differentiated intestinal, skin, and blood cells require "professional" stem cells to produce replacements. This occurs by continuous expansion of stem cells and their immediate progeny, followed by coordinated activation of divergent transcriptional programs to generate stable cells with diverse functions. Other tissues turn over slowly, if at all, and vary widely in strategies for facultative stem cell activity or interconversion among mature resident cell types (transdifferentiation). Cell fate potential is programmed in tissue-specific configurations of chromatin, which restrict the complement of available genes and cis-regulatory elements, hence allowing specific cell types to arise. Using as a model the transcriptional and chromatin basis of cell differentiation and dedifferentiation in intestinal crypts, we discuss here how self-renewing and other tissues execute homeostatic and injury-responsive stem cell activity.


Assuntos
Plasticidade Celular/genética , Plasticidade Celular/fisiologia , Epigênese Genética/genética , Epigênese Genética/fisiologia , Intestinos/fisiologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem da Célula/genética , Linhagem da Célula/fisiologia , Humanos
10.
Genes Dev ; 31(23-24): 2391-2404, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29321178

RESUMO

Compacted chromatin and nucleosomes are known barriers to gene expression; the nature and relative importance of other transcriptional constraints remain unclear, especially at distant enhancers. Polycomb repressor complex 2 (PRC2) places the histone mark H3K27me3 predominantly at promoters, where its silencing activity is well documented. In adult tissues, enhancers lack H3K27me3, and it is unknown whether intergenic H3K27me3 deposits affect nearby genes. In primary intestinal villus cells, we identified hundreds of tissue-restricted enhancers that require the transcription factor (TF) CDX2 to prevent the incursion of H3K27me3 from adjoining areas of elevated basal marking into large well-demarcated genome domains. Similarly, GATA1-dependent enhancers exclude H3K27me3 from extended regions in erythroid blood cells. Excess intergenic H3K27me3 in both TF-deficient tissues is associated with extreme mRNA deficits, which are significantly rescued in intestinal cells lacking PRC2. Explaining these observations, enhancers show TF-dependent binding of the H3K27 demethylase KDM6A. Thus, in diverse cell types, certain genome regions far from promoters accumulate H3K27me3, and optimal gene expression depends on enhancers clearing this repressive mark. These findings reveal new "anti-repressive" function for hundreds of tissue-specific enhancers.


Assuntos
Elementos Facilitadores Genéticos/fisiologia , Regulação da Expressão Gênica , Genoma/genética , Histonas/metabolismo , Intestino Delgado/metabolismo , Fatores de Transcrição/metabolismo , Animais , Fator de Transcrição CDX2/genética , Células Eritroides/metabolismo , Feminino , Histona Desmetilases/metabolismo , Intestino Delgado/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Ligação Proteica
11.
J Org Chem ; 89(13): 9389-9404, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38913823

RESUMO

An efficient one-pot metal-free, base-catalyzed method has been developed for the regioselective [3 + 2]/[4 + 2] annulation reactions of electrophilic methyleneindolinones with haloalcohols to furnish spirooxindole derivatives under mild reaction conditions. This reaction afforded the corresponding products with two contiguous stereocenters including a quaternary center in good to excellent yield (up to 95%) with moderate to good diastereoselectivities (up to 12.5:1 dr) with complete regioselectivity.

12.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33653947

RESUMO

Pancreatic ductal adenocarcinoma (PDA) is a lethal, therapy-resistant cancer that thrives in a highly desmoplastic, nutrient-deprived microenvironment. Several studies investigated the effects of depriving PDA of either glucose or glutamine alone. However, the consequences on PDA growth and metabolism of limiting both preferred nutrients have remained largely unknown. Here, we report the selection for clonal human PDA cells that survive and adapt to limiting levels of both glucose and glutamine. We find that adapted clones exhibit increased growth in vitro and enhanced tumor-forming capacity in vivo. Mechanistically, adapted clones share common transcriptional and metabolic programs, including amino acid use for de novo glutamine and nucleotide synthesis. They also display enhanced mTORC1 activity that prevents the proteasomal degradation of glutamine synthetase (GS), the rate-limiting enzyme for glutamine synthesis. This phenotype is notably reversible, with PDA cells acquiring alterations in open chromatin upon adaptation. Silencing of GS suppresses the enhanced growth of adapted cells and mitigates tumor growth. These findings identify nongenetic adaptations to nutrient deprivation in PDA and highlight GS as a dependency that could be targeted therapeutically in pancreatic cancer patients.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Glutamato-Amônia Ligase/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/genética , Linhagem Celular Tumoral , Estabilidade Enzimática , Glutamato-Amônia Ligase/genética , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética
13.
J Org Chem ; 88(14): 9987-10001, 2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37406306

RESUMO

A facile one-pot metal-free, base-mediated formal [3 + 2] and [4 + 2] dearomative ipso-cycloaddition of para-quinone methides (p-QMs) with halo alcohols has been designed for the efficient construction of 2-oxa-spirocyclohexadienones in excellent yield under mild reaction conditions. The commercial availability of the bases, reagents, and convenient reaction procedure makes it an attractive method for ipso-cyclization.

14.
Biol Lett ; 19(4): 20220602, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37016813

RESUMO

Habitat fragmentation can have negative impacts on migratory organisms that rely on the functional connectivity between growing and breeding grounds. Quantifying the population-level phenotypic consequences of such fragmentation requires fine-scaled tracking of individual behaviour and movements across relevant scales. Here we make use of a natural experiment where some populations of 'migrant' three-spined sticklebacks (Gasterosteus aculeatus) became 'residents', following habitat fragmentation five decades ago. To test whether residents have a lower movement tendency than migrants, we developed a novel experimental platform that allows the automated tracking of individual movements via RFID technology in a semi-natural mesocosm where spatio-temporal scales and environmental conditions can be manipulated. We found that residents moved significantly less than migrants at large but not at small spatial scale. This pattern was consistent across time and contexts (water flow and group size). Our study substantiates prior literature on rapid phenotypic divergence in sticklebacks in response to human-induced isolation and highlights the importance of observing behaviour in ecologically relevant set-ups that bridge the gap between laboratory and field studies.


Assuntos
Smegmamorpha , Animais , Humanos , Smegmamorpha/fisiologia , Ecossistema
15.
Gastroenterology ; 161(3): 924-939.e11, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34090884

RESUMO

BACKGROUND & AIMS: Tissue metaplasia is uncommon in adults because established cis-element programs resist rewiring. In Barrett's esophagus, the distal esophageal mucosa acquires a predominantly intestinal character, with notable gastric features, and is predisposed to developing invasive cancers. We sought to understand the chromatin underpinnings of Barrett's metaplasia and why it commonly displays simultaneous gastric and intestinal properties. METHODS: We profiled cis-regulatory elements with active histone modifications in primary human biopsy materials using chromatin immunoprecipitation followed by DNA sequencing. Mutations in Barrett's esophagus were examined in relation to tissue-specific enhancer landscapes using a random forest machine-learning algorithm. We also profiled open chromatin at single-cell resolution in primary Barrett's biopsy specimens using the assay for transposase-accessible chromatin. We used 1- and 2-color immunohistochemistry to examine protein expression of tissue-restricted genes. RESULTS: Barrett's esophagus bears epigenome fingerprints of human stomach and intestinal columnar, but not esophageal squamous, epithelia. Mutational patterns were best explained as arising on the epigenome background of active gastric cis-elements, supporting the view that adjoining stomach epithelium is a likely tissue source. Individual cells in Barrett's metaplasia coexpress gastric and intestinal genes, reflecting concomitant chromatin access at enhancers ordinarily restricted to one or the other epithelium. Protein expression of stomach-specific mucins; CLDN18; and a novel gastric marker, ANXA10, showed extensive tissue and subclonal heterogeneity of dual stomach-intestinal cell states. CONCLUSIONS: These findings reveal mixed and dynamic tissue-restricted chromatin states and phenotypic heterogeneity in Barrett's esophagus. Pervasive intragland variation argues against stem-cell governance of this phenotype.


Assuntos
Esôfago de Barrett/genética , Esôfago de Barrett/patologia , Plasticidade Celular , Montagem e Desmontagem da Cromatina , Epigenoma , Mucosa Esofágica/patologia , Células-Tronco/patologia , Linhagem da Célula , Sequenciamento de Cromatina por Imunoprecipitação , Análise Mutacional de DNA , Elementos Facilitadores Genéticos , Epigenômica , Predisposição Genética para Doença , Humanos , Imuno-Histoquímica , Metaplasia , Mutação , Fenótipo , Análise de Célula Única
16.
Development ; 146(5)2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30745430

RESUMO

Lineage-restricted transcription factors, such as the intestine-specifying factor CDX2, often have dual requirements across developmental time. Embryonic loss of CDX2 triggers homeotic transformation of intestinal fate, whereas adult-onset loss compromises crucial physiological functions but preserves intestinal identity. It is unclear how such diverse requirements are executed across the developmental continuum. Using primary and engineered human tissues, mouse genetics, and a multi-omics approach, we demonstrate that divergent CDX2 loss-of-function phenotypes in embryonic versus adult intestines correspond to divergent CDX2 chromatin-binding profiles in embryonic versus adult stages. CDX2 binds and activates distinct target genes in developing versus adult mouse and human intestinal cells. We find that temporal shifts in chromatin accessibility correspond to these context-specific CDX2 activities. Thus, CDX2 is not sufficient to activate a mature intestinal program; rather, CDX2 responds to its environment, targeting stage-specific genes to contribute to either intestinal patterning or mature intestinal function. This study provides insights into the mechanisms through which lineage-specific regulatory factors achieve divergent functions over developmental time.


Assuntos
Fator de Transcrição CDX2/metabolismo , Cromatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Intestinos/embriologia , Animais , Fator de Transcrição CDX2/genética , Sistemas CRISPR-Cas , Diferenciação Celular , Linhagem da Célula , Feminino , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Knockout , Mutação , Células-Tronco Pluripotentes/citologia , Ligação Proteica , Domínios Proteicos , Transativadores/metabolismo
17.
Mol Biol Rep ; 49(12): 11829-11846, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36214948

RESUMO

BACKGROUND: Porcine circovirus 2 is globally noted swine pathogen with multiple genotypes associated with vast clinical and subclinical outcomes. This study aimed to isolate and characterize PCV2 genotypes circulating in southern states of India. METHODS AND RESULTS: A total of 434 field samples comprising of serum (n = 273), tissues (n = 109) and swabs (n = 52) collected from swine during 2019 to 2021 from southern states of India were screened for PCV2 by specific polymerase chain reaction (PCR) assay. Molecular prevalence of PCV2 in southern India was found to be 12.21% (n = 53). All the 53 PCV2 positive samples were further subjected to the PCR assay with designed primers targeting full length amplification of ORF2 gene of PCV2 for molecular characterization. Randomly 32 positive samples by full length PCV2-ORF2 gene PCR were sequenced for genotyping. Signature motif and phylogenetic analysis of 32 PCV2 sequences revealed 62.5% (n = 20) prevalence of PCV2d genotype followed by 21.8% (n = 7) of PCV2h or PCV2-IM1 and 15.6% (n = 5) of PCV2b genotypes. Twenty five PCR positive field samples were subjected for virus isolation in PK15 cells and characterized. Out of 25 samples processed 5 (20%) PCV2 isolates obtained in this study were confirmed by PCR and immune fluorescence assay. Molecular characterization of PK15 adapted five PCV2 isolates confirmed circulation of PCV2d, PCV2h and PCV2b genotypes in pigs under field conditions in southern India. CONCLUSIONS: Isolation and molecular epidemiological study of PCV2 in southern states of India evidences high circulation of PCV2d genotypes in field conditions in comparison to other genotypes.


Assuntos
Infecções por Circoviridae , Circovirus , Doenças dos Suínos , Suínos , Animais , Circovirus/genética , Infecções por Circoviridae/epidemiologia , Infecções por Circoviridae/veterinária , Filogenia , Doenças dos Suínos/epidemiologia , Genótipo
18.
Mol Divers ; 26(5): 2535-2548, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34822095

RESUMO

Herein, we identified a potent lead compound RRA2, within a series of 54 derivatives of 1,2,4-triazolethiols (exhibit good potency as an anti-mycobacterial agents) against intracellular Mycobacterium tuberculosis (Mtb). Compound RRA2 showed significant mycobactericidal activity against active stage Mycobacterium bovis BCG and Mtb with minimum inhibitory concentration (MIC) values of 2.3 and 2.0 µg/mL, respectively. At MIC value, RRA2 compound yielded 0.82 log reduction of colony-forming unit (cfu) against non-replicating Mtb. Furthermore, RRA2 compound was selected for further target identification due to the presence of alkyne group, showing higher selectivity index (> 66.66 ± 0.22, in non-replicating stage). Using "click" chemistry, we synthesized the biotin linker-RRA2 conjugate, purified with HPLC method and confirmed the conjugation of biotin linker-RRA2 complex by HR-MS analysis. Furthermore, we successfully pulled down and identified a specific target protein GroEl2, from Mtb whole-cell extract. Furthermore, computational molecular modeling indicated RRA2 could interact with GroEl2, which explains the structure-activity relationship observed in this study. GroEL-2 identified a potent and specific target protein for RRA 2 compound in whole cell extract of Mtb H37Ra.


Assuntos
Proteínas de Bactérias/análise , Mycobacterium tuberculosis , Alcinos , Antibacterianos , Antituberculosos/química , Antituberculosos/farmacologia , Vacina BCG , Biotina , Extratos Celulares , Testes de Sensibilidade Microbiana , Proteínas , Compostos de Sulfidrila , Triazóis
19.
J Assist Reprod Genet ; 39(9): 2103-2114, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35849255

RESUMO

PURPOSE: To identify clinically significant genomic copy number (CNV) and single nucleotide variants (SNV) in males with unexplained spermatogenic failure (SPGF). MATERIALS AND METHODS: Peripheral blood DNA from 97/102 study participants diagnosed with oligozoospermia, severe oligozoospermia, or non-obstructive azoospermia (NOA) was analyzed for CNVs via array comparative genomic hybridization (aCGH) and SNVs using whole-exome sequencing (WES). RESULTS: Of the 2544 CNVs identified in individuals with SPGF, > 90% were small, ranging from 0.6 to 75 kb. Thirty, clinically relevant genomic aberrations, were detected in 28 patients (~ 29%). These included likely diagnostic CNVs in 3/41 NOA patients (~ 7%): 1 hemizygous, intragenic TEX11 deletion, 1 hemizygous DDX53 full gene deletion, and 1 homozygous, intragenic STK11 deletion. High-level mosaicism for X chromosome disomy (~ 10% 46,XY and ~ 90% 47,XXY) was also identified in 3 of 41 NOA patients who previously tested normal with conventional karyotyping. The remaining 24 CNVs detected were heterozygous, autosomal recessive carrier variants. Follow-up WES analysis confirmed 8 of 27 (30%) CNVs (X chromosome disomy excluded). WES analysis additionally identified 13 significant SNVs and/or indels in 9 patients (~ 9%) including X-linked AR, KAL1, and NR0B1 variants. CONCLUSION: Using a combined genome-wide aCGH/WES approach, we identified pathogenic and likely pathogenic SNVs and CNVs in 15 patients (15%) with unexplained SPGF. This value equals the detection rate of conventional testing for aneuploidies and is considerably higher than the prevalence of Y chromosome microdeletions. Our results underscore the importance of comprehensive genomic analysis in emerging diagnostic testing of complex conditions like male infertility.


Assuntos
Variações do Número de Cópias de DNA , Oligospermia , Azoospermia , Hibridização Genômica Comparativa , Variações do Número de Cópias de DNA/genética , Testes Genéticos/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Nucleotídeos , Oligospermia/diagnóstico , Oligospermia/genética
20.
Genes Dev ; 28(16): 1827-39, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25128499

RESUMO

Tissues may adopt diverse strategies to establish specific transcriptional programs in daughter lineages. In intestinal crypts, enhancers for genes expressed in both major cell types appear broadly permissive in stem and specified progenitor cells. In blood, another self-renewing tissue, it is unclear when chromatin becomes permissive for transcription of genes expressed in distinct terminal lineages. Using chromatin immunoprecipitation (ChIP) combined with deep sequencing (ChIP-seq) to profile activating histone marks, we studied enhancer dynamics in primary mouse blood stem, progenitor, and specified cells. Stem and multipotent progenitor cells show scant H3K4me2 marking at enhancers bound by specific transcription factors in their committed progeny. Rather, enhancers are modulated dynamically and serially, with substantial loss and gain of H3K4me2, at each cellular transition. Quantitative analysis of these dynamics accurately modeled hematopoiesis according to Waddington's notion of epigenotypes. Delineation of enhancers in terminal blood lineages coincides with cell specification, and enhancers active in single lineages show well-positioned H3K4me2- and H3K27ac-marked nucleosomes and DNaseI hypersensitivity in other cell types, revealing limited lineage fidelity. These findings demonstrate that enhancer chronology in blood cells differs markedly from that in intestinal crypts. Chromatin dynamics in hematopoiesis provide a useful foundation to consider classical observations such as cellular reprogramming and multilineage locus priming.


Assuntos
Células Sanguíneas/citologia , Células Sanguíneas/metabolismo , Elementos Facilitadores Genéticos/genética , Hematopoese/genética , Animais , Diferenciação Celular , Linhagem da Célula , Epigênese Genética , Histonas/metabolismo , Camundongos , Células-Tronco/citologia , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA