Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 333
Filter
1.
Int J Mol Sci ; 23(19)2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36232696

ABSTRACT

The NMDA receptor is a Ca2+-permeant glutamate receptor which plays key roles in health and disease. Canonical NMDARs contain two GluN2 subunits, of which 2A and 2B are predominant in the forebrain. Moreover, the relative contribution of 2A vs. 2B is controlled both developmentally and in an activity-dependent manner. The GluN2 subtype influences the biophysical properties of the receptor through difference in their N-terminal extracellular domain and transmembrane regions, but they also have large cytoplasmic Carboxyl (C)-terminal domains (CTDs) which have diverged substantially during evolution. While the CTD identity does not influence NMDAR subunit specific channel properties, it determines the nature of CTD-associated signalling molecules and has been implicated in mediating the control of subunit composition (2A vs. 2B) at the synapse. Historically, much of the research into the differential function of GluN2 CTDs has been conducted in vitro by over-expressing mutant subunits, but more recently, the generation of knock-in (KI) mouse models have allowed CTD function to be probed in vivo and in ex vivo systems without heterologous expression of GluN2 mutants. In some instances, findings involving KI mice have been in disagreement with models that were proposed based on earlier approaches. This review will examine the current research with the aim of addressing these controversies and how methodology may contribute to differences between studies. We will also discuss the outstanding questions regarding the role of GluN2 CTD sequences in regulating NMDAR subunit composition, as well as their relevance to neurodegenerative disease and neurodevelopmental disorders.


Subject(s)
Neurodegenerative Diseases , Neurodevelopmental Disorders , Receptors, N-Methyl-D-Aspartate , Animals , Disease Models, Animal , Growth and Development/genetics , Growth and Development/physiology , Mice , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/physiopathology , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Synapses/genetics , Synapses/metabolism , Synapses/physiology
2.
Mol Cell Probes ; 65: 101851, 2022 10.
Article in English | MEDLINE | ID: mdl-36007750

ABSTRACT

Environmental factors, genetic factors, and epigenetics are involved in animal growth and development. Among them, methylation is one of the abundant modifications of epigenetics. N6-methyladenosine(m6A) is extensive in cellular RNA, of which mRNA is the most common internal modification. m6A modification regulates life activities dynamically and reversibly, including expressed genes, RNA metabolism, and protein translation. The m6A modifications are closely related to human diseases involving heart failure, tumors, and cancer. It is relatively in-depth in the medical field. However, there are few studies on its biochemical function in animals. We summarized the latest paper related to the chemical structure and role of the writers, the erasers, and the readers to study exerting dynamic regulation of m6A modification of animal growth and development. Furthermore, the key roles of m6A modification were reported in the process of RNA metabolism. Finally, the dynamic regulation of m6A modification in animal growth and development was reviewed, including brain development, fertility, fat deposition, and muscle production. It reveals the key roles of m6A modification and the regulation of gene expression, aiming to provide new ideas for m6A methylation in animal growth and development.


Subject(s)
Adenosine , Neoplasms , Adenosine/genetics , Adenosine/metabolism , Animals , Growth and Development/genetics , Humans , Methylation , Neoplasms/genetics , RNA/metabolism , RNA, Messenger/genetics
3.
Proc Natl Acad Sci U S A ; 119(30): e2122148119, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35858396

ABSTRACT

In Darwin's and Mendel's times, researchers investigated a wealth of organisms, chosen to solve particular problems for which they seemed especially well suited. Later, a focus on a few organisms, which are accessible to systematic genetic investigations, resulted in larger repertoires of methods and applications in these few species. Genetic animal model organisms with large research communities are the nematode Caenorhabditis elegans, the fly Drosophila melanogaster, the zebrafish Danio rerio, and the mouse Mus musculus. Due to their specific strengths, these model organisms have their strongest impacts in rather different areas of biology. C. elegans is unbeatable in the analysis of cell-to-cell contacts by saturation mutagenesis, as worms can be grown very fast in very high numbers. In Drosophila, a rich pattern is generated in the embryo as well as in adults that is used to unravel the underlying mechanisms of morphogenesis. The transparent larvae of zebrafish are uniquely suited to study organ development in a vertebrate, and the superb versatility of reverse genetics in the mouse made it the model organism to study human physiology and diseases. The combination of these models allows the in-depth genetic analysis of many fundamental biological processes using a plethora of different methods, finally providing many specific approaches to combat human diseases. The plant model Arabidopsis thaliana provides an understanding of many aspects of plant biology that might ultimately be useful for breeding crops.


Subject(s)
Arabidopsis , Growth and Development , Models, Animal , Animals , Arabidopsis/genetics , Arabidopsis/growth & development , Caenorhabditis elegans/genetics , Drosophila melanogaster/genetics , Genetic Research , Growth and Development/genetics , Humans , Mice , Plant Breeding , Zebrafish/genetics
4.
Nature ; 607(7919): 604-609, 2022 07.
Article in English | MEDLINE | ID: mdl-35831506

ABSTRACT

Chromosome segregation errors during cell divisions generate aneuploidies and micronuclei, which can undergo extensive chromosomal rearrangements such as chromothripsis1-5. Selective pressures then shape distinct aneuploidy and rearrangement patterns-for example, in cancer6,7-but it is unknown whether initial biases in segregation errors and micronucleation exist for particular chromosomes. Using single-cell DNA sequencing8 after an error-prone mitosis in untransformed, diploid cell lines and organoids, we show that chromosomes have different segregation error frequencies that result in non-random aneuploidy landscapes. Isolation and sequencing of single micronuclei from these cells showed that mis-segregating chromosomes frequently also preferentially become entrapped in micronuclei. A similar bias was found in naturally occurring micronuclei of two cancer cell lines. We find that segregation error frequencies of individual chromosomes correlate with their location in the interphase nucleus, and show that this is highest for peripheral chromosomes behind spindle poles. Randomization of chromosome positions, Cas9-mediated live tracking and forced repositioning of individual chromosomes showed that a greater distance from the nuclear centre directly increases the propensity to mis-segregate. Accordingly, chromothripsis in cancer genomes9 and aneuploidies in early development10 occur more frequently for larger chromosomes, which are preferentially located near the nuclear periphery. Our findings reveal a direct link between nuclear chromosome positions, segregation error frequencies and micronucleus content, with implications for our understanding of tumour genome evolution and the origins of specific aneuploidies during development.


Subject(s)
Aneuploidy , Chromosome Positioning , Chromosome Segregation , Chromosomes , CRISPR-Associated Protein 9 , Cell Line , Cell Line, Tumor , Chromosome Segregation/genetics , Chromosomes/genetics , Chromosomes/metabolism , Chromothripsis , Growth and Development/genetics , Humans , Interphase , Micronuclei, Chromosome-Defective , Mitosis , Neoplasms/genetics , Neoplasms/pathology , Organoids/cytology , Organoids/metabolism , Sequence Analysis, DNA , Single-Cell Analysis
5.
Nat Rev Genet ; 23(3): 137-153, 2022 03.
Article in English | MEDLINE | ID: mdl-34608297

ABSTRACT

DNA methylation (DNAme) and histone post-translational modifications (PTMs) have important roles in transcriptional regulation. Although many reports have characterized the functions of such chromatin marks in isolation, recent genome-wide studies reveal surprisingly complex interactions between them. Here, we focus on the interplay between DNAme and methylation of specific lysine residues on the histone H3 tail. We describe the impact of genetic perturbation of the relevant methyltransferases in the mouse on the landscape of chromatin marks as well as the transcriptome. In addition, we discuss the specific neurodevelopmental growth syndromes and cancers resulting from pathogenic mutations in the human orthologues of these genes. Integrating these observations underscores the fundamental importance of crosstalk between DNA and histone H3 methylation in development and disease.


Subject(s)
Chromatin/metabolism , DNA Methylation/genetics , Disease/genetics , Growth and Development/genetics , Animals , Chromatin Assembly and Disassembly/physiology , Histones/metabolism , Humans , Mice , Protein Processing, Post-Translational
6.
Clin Epigenetics ; 13(1): 213, 2021 12 04.
Article in English | MEDLINE | ID: mdl-34863285

ABSTRACT

BACKGROUND: Cortisol-producing adrenocortical adenoma (CPA) during pregnancy rarely occurs in clinic. Growing evidence suggests that DNA methylation plays a key role in adrenocortical adenomas. The present study aims to examine the genome-wide DNA methylation profiles and identify the differences in DNA methylation signatures of non-pregnant and pregnant patients with CPA. RESULTS: Four pregnant and twelve non-pregnant patients with CPA were enrolled. The pregnant patients with CPA had higher serum cortisol, Estradiol, Progesterone, and human chorionic gonadotropin concentration, while having lower serum FSH (follicle-stimulating hormone) and luteinizing hormone concentrations (P < 0.01). Compared with the non-pregnant patients, the duration is shorter, and the growth rate of the tumor is faster in pregnant patients with CPA (P < 0.05). Morphology and cell proliferation assay showed that the percentage of Ki-67 positive cells in CPA were higher in pregnant group than non-pregnant group (8.0% vs 5.5%, P < 0.05). The DNA methylation analysis showed that Genome-wide DNA methylation signature difference between pregnant and non-pregnant with CPA, that the pregnant group had more hypermethylated DMPs (67.94% vs 22.16%) and less hypomethylated DMPs (32.93% vs 77.84%). The proportion of hypermethylated DMPs was relatively high on chromosomes 1 (9.68% vs 8.67%) and X (4.99% vs 3.35%) but lower on chromosome 2(7.98% vs 12.92%). In pregnant patients with CPA, 576 hypomethylated DMPs and 1109 hypermethylated DMPs were identified in the DNA promoter region. Bioinformatics analysis indicated that the Wnt/ß-Catenin pathway, Ras/MAPK Pathway and PI3K-AKT Pathway were associated with the development of CPA during pregnancy. CONCLUSIONS: Genome-wide DNA methylation profiling of CPA in non-pregnant and pregnant patients was identified in the present study. Alterations of DNA methylation were associated with the pathogenesis and exacerbation of CPA during pregnancy.


Subject(s)
Adrenocortical Adenoma/pathology , DNA Methylation/genetics , Adrenocortical Adenoma/physiopathology , Adult , DNA Methylation/immunology , Female , Growth and Development/genetics , Growth and Development/physiology , Humans , Pregnancy
7.
Plant Sci ; 313: 111073, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34763865

ABSTRACT

MicroRNAs (miRNAs), a class of single-stranded non-coding RNA of 20-24 nucleotides, regulate gene expression by target gene transcript cleavage or translation inhibition. The phytohormone auxin is a crucial regulator of almost every process involved in plant growth and development. Several studies have demonstrated the involvement of miRNA(s) in the regulation of the auxin signaling pathway and plant development. However, very few studies have identified the auxin-mediated regulation of miRNA(s). In this study, we reveal the detailed mechanism of auxin-mediated regulation of the cell wall-related miR775- Galactosyl transferase (GalT) module, which plays an important role in root growth in Arabidopsis thaliana. We also showed two interdependent mechanisms by which miR775 regulates root growth: miR775-GalT and light-mediated sucrose-dependent pathways. Treatment of GUS reporter lines with Indole Acetic Acid (IAA), sucrose, and light apparently enhanced the abundance of miR775 in root tissue. miR775 overexpressing (miR775OX) lines showed changes in root architecture, including increased primary root growth and root hair, by targeting GalT. miR775OX lines also showed tolerance toward low Pi. These results provide new insights into the auxin regulation of cell wall-related miR775 and suggest its significant role in plant root growth and development by modifying the cell wall.


Subject(s)
Arabidopsis/growth & development , Arabidopsis/genetics , Indoleacetic Acids/metabolism , MicroRNAs/metabolism , Plant Roots/growth & development , Plant Roots/genetics , Sucrose/metabolism , Adaptation, Ocular/drug effects , Adaptation, Ocular/genetics , Gene Expression Regulation, Plant , Genetic Variation , Genotype , Growth and Development/genetics , Metabolic Networks and Pathways/genetics
8.
Int J Mol Sci ; 22(21)2021 Nov 07.
Article in English | MEDLINE | ID: mdl-34769468

ABSTRACT

Mutations in the insulin gene (INS) are frequently associated with human permanent neonatal diabetes mellitus. However, the mechanisms underlying the onset of this genetic disease is not sufficiently decoded. We induced expression of two types of human mutant INSs in Drosophila using its ectopic expression system and investigated the resultant responses in development. Expression of the wild-type preproinsulin in the insulin-producing cells (IPCs) throughout the larval stage led to a stimulation of the overall and wing growth. However, ectopic expression of human mutant preproinsulins, hINSC96Y and hINSLB15YB16delinsH, neither of which secreted from the ß-cells, could not stimulate the Drosophila growth. Furthermore, neither of the mutant polypeptides induced caspase activation leading to apoptosis. Instead, they induced expression of several markers indicating the activation of unfolded protein response, such as ER stress-dependent Xbp1 mRNA splicing and ER chaperone induction. We newly found that the mutant polypeptides induced the expression of Growth arrest and DNA-damage-inducible 45 (Gadd45) in imaginal disc cells. ER stress induced by hINSC96Y also activated the JAK-STAT signaling, involved in inflammatory responses. Collectively, we speculate that the diabetes-like growth defects appeared as a consequence of the human mutant preproinsulin expression was involved in dysfunction of the IPCs, rather than apoptosis.


Subject(s)
Growth and Development/genetics , Insulin/genetics , Protein Precursors/genetics , Unfolded Protein Response , Animals , Animals, Genetically Modified , Down-Regulation/genetics , Drosophila/genetics , Drosophila/growth & development , Drosophila/metabolism , Embryo, Nonmammalian , Humans , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Janus Kinases/genetics , Janus Kinases/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Precursors/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction/genetics , Unfolded Protein Response/genetics , Up-Regulation/genetics , GADD45 Proteins
9.
J Neuroendocrinol ; 33(12): e13058, 2021 12.
Article in English | MEDLINE | ID: mdl-34748236

ABSTRACT

Prenatal exposure to excess androgens is associated with the development of polycystic ovary syndrome (PCOS). In prenatally androgenised (PNA) mice, a model of PCOS, progesterone receptor (PR) protein expression is reduced in arcuate nucleus (ARC) GABA neurons. This suggests a mechanism for PCOS-related impaired steroid hormone feedback and implicates androgen excess with respect to inducing transcriptional repression of the PR-encoding gene Pgr in the ARC. However, the androgen sensitivity of ARC neurons and the relative gene expression of PRs over development and following prenatal androgen exposure remain unknown. Here, we used a quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) of microdissected ARC to determine the relative androgen receptor (Ar) and progesterone receptor (Pgr) gene expression in PNA and control mice at five developmental timepoints. In a two-way analysis of variance, none of the genes examined showed expression changes with a statistically significant interaction between treatment and age, although PgrA showed a borderline interaction. For all genes, there was a statistically significant main effect of age on expression levels, reflecting a general increase in expression with increasing age, regardless of treatment. For PgrB and Ar, there was a statistically significant main effect of treatment, indicating a change in expression following PNA (increased for PgrB and decreased for Ar), regardless of age. For PgrA, there was a borderline main effect of treatment, suggesting a possible change in expression following PNA, regardless of age. PgrAB gene expression changes showed no significant main effect of treatment. We additionally examined androgen and progesterone responsiveness specifically in P60 ARC GABA neurons using RNAScope® (Advanced Cell Diagnostics, Inc.) in situ hybridization. This analysis revealed that Pgr and Ar were expressed in the majority of ARC GABA neurons in normal adult females. However, our RNAScope® analysis did not show significant changes in Pgr or Ar expression within ARC GABA neurons following PNA. Lastly, because GABA drive to gonadotropin-releasing hormone neurons is increased in PNA, we hypothesised that PNA mice would show increased expression of glutamic acid decarboxylase (GAD), the rate-limiting enzyme in GABA production. However, the RT-qPCR showed that the expression of GAD encoding genes (Gad1 and Gad2) was unchanged in adult PNA mice compared to controls. Our findings indicate that PNA treatment can impact Pgr and Ar mRNA expression in adulthood. This may reflect altered circulating steroid hormones in PNA mice or PNA-induced epigenetic changes in the regulation of Pgr and Ar gene expression in ARC neurons.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Prenatal Exposure Delayed Effects/genetics , Receptors, Androgen/genetics , Receptors, Progesterone/genetics , Virilism , Animals , Animals, Newborn , Arcuate Nucleus of Hypothalamus/growth & development , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Growth and Development/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Receptors, Progesterone/metabolism , Virilism/embryology , Virilism/genetics , Virilism/metabolism
10.
Nurs Res ; 70(6): 462-468, 2021.
Article in English | MEDLINE | ID: mdl-34380978

ABSTRACT

BACKGROUND: The gut microbiome is an important determinant of health and disease in preterm infants. OBJECTIVES: The objective of this article was to share our current protocol for other neonatal intensive care units to potentially expand their existing protocols, aiming to characterize the relationship between the intestinal microbiome and health outcomes in preterm infants. METHODS: This prospective, longitudinal study planned to recruit 160 preterm infants born <32 weeks gestational age or weighing <1,500 g and admitted to one of two Level III/IV neonatal intensive care units. During the neonatal intensive care unit period, the primary measures included events of early life pain/stress, gut microbiome, host genetic variations, and neurobehavioral assessment. During follow-up visits, gut microbiome; pain sensitivity; and medical, growth, and developmental outcomes at 4, 8-12, and 18-24 months corrected age were measured. DISCUSSION: As of February 14, 2020, 214 preterm infants have been recruited. We hypothesize that infants who experience greater levels of pain/stress will have altered gut microbiome, including potential adverse outcomes such as necrotizing enterocolitis and host genetic variations, feeding intolerance, and/or neurodevelopmental impairments. These will differ from the intestinal microbiome of preterm infants who do not develop these adverse outcomes. To test this hypothesis, we will determine how alterations in the intestinal microbiome affect the risk of developing necrotizing enterocolitis, feeding intolerance, and neurodevelopmental impairments in preterm infants. In addition, we will examine the interaction between the intestinal microbiome and host genetics in the regulation of intestinal health and neurodevelopmental outcomes.


Subject(s)
Gastrointestinal Microbiome , Growth and Development/genetics , Growth and Development/physiology , Health Status , Infant, Newborn/growth & development , Infant, Premature/growth & development , Neurodevelopmental Disorders/diagnosis , Age Factors , Child, Preschool , Connecticut , Female , Follow-Up Studies , Humans , Infant , Longitudinal Studies , Male , Prospective Studies
11.
Nurs Res ; 70(6): 481-486, 2021.
Article in English | MEDLINE | ID: mdl-34173371

ABSTRACT

BACKGROUND: Annually, approximately 15 million babies are born preterm (<37 weeks gestational age) globally. In the neonatal intensive care unit (NICU) environment, infants are exposed to repeated stressful or painful procedures as part of routine lifesaving care. These procedures have been associated with epigenetic alterations that may lead to an increased risk of neurodevelopmental disorders. Telomere length has been negatively associated with adverse life experiences in studies of adults. OBJECTIVES: This pilot study aimed to describe telomere length in a sample of preterm infants at NICU discharge and examine any associations with pain, feeding method, and neurodevelopment. METHODS: This descriptive pilot study sample includes baseline absolute telomere length (aTL) of 36 preterm infants immediately prior to discharge. Quantitative polymerase chain reaction was used to determine aTL. Infant demographics, pain/stress, type of feeding, antibiotic use, neurodevelopment, and buccal swab data were collected. Descriptive data analysis was used to describe the telomere length using graphs. RESULTS: Among our preterm infant samples, the mean aTL was far greater than the average adult telomere length. Although no significant associations were found between aTL and pain, feeding method, and neurodevelopment, a trend between sex was noted where male telomere lengths were shorter than females as they aged. DISCUSSION: This is one of few studies to evaluate preterm infant telomere length. Although other researchers have used relative telomere length, we used the more accurate aTL. We found nonsignificant shorter telomere lengths among males. Additional large-scale, longitudinal studies are needed to better identify the predictors of telomere length at the time of discharge from NICU.


Subject(s)
Feeding Behavior , Growth and Development/genetics , Infant, Premature/growth & development , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics , Pain/genetics , Telomere/genetics , Female , Humans , Infant, Newborn , Male , Pilot Projects
13.
Hist Philos Life Sci ; 43(1): 30, 2021 Feb 23.
Article in English | MEDLINE | ID: mdl-33624250

ABSTRACT

The discovery of RNA interference in 1998 has made a lasting impact on biological research. Identifying the regulatory role of small RNAs changed the modes of molecular biological inquiry as well as biologists' understanding of genetic regulation. This article examines the early years of small RNA biology's success story. I query which factors had to come together so that small RNA research came into life in the blink of an eye. I primarily look at scientific repertoires as facilitators of rapid scientific change. I show that for a short period of time, between the years 1998 and 2002, different model organism communities, investigative strategies, technological innovations, and research interests could be successfully aligned to take small RNA research off the ground. I discuss how the keystone discoveries were situated in specific experimental traditions and what strategies were employed to establish these discoveries as more general phenomena. Providing thus a practice-based approach of rapid scientific change, I ask how to relate the change in propositional bits of scientific knowledge with changes in scientific practice.


Subject(s)
Biochemistry/history , Caenorhabditis elegans/genetics , Crops, Agricultural/genetics , Genetic Techniques/history , RNA Interference , Animals , Biochemistry/methods , Caenorhabditis elegans/growth & development , Crops, Agricultural/growth & development , Disease/genetics , Growth and Development/genetics , History, 20th Century , History, 21st Century , RNA/genetics
14.
Reprod Biol Endocrinol ; 19(1): 22, 2021 Feb 13.
Article in English | MEDLINE | ID: mdl-33581723

ABSTRACT

BACKGROUND: Irisin, which is cleaved from fibronectin type III domain-containing protein 5 (Fndc5), plays an important role in energy homeostasis. The link between energy metabolism and reproduction is well known. However, the biological actions of irisin in reproduction remain largely unexplored. METHODS: In this study, we generated Fndc5 gene mutation to create irisin deficient mice. Female wild-type (WT) and Fndc5 mutant mice were fed with standard chow for 48 weeks. Firstly, the survival rate, body weight and fertility were described in mice. Secondly, the levels of steroid hormones in serum were measured by ELISA, and the estrus cycle and the appearance of follicles were determined by vaginal smears and ovarian continuous sections. Thirdly, mRNA-sequencing analysis was used to compare gene expression between the ovaries of Fndc5 mutant mice and those of WT mice. Finally, the effects of exogenous irisin on steroid hormone production was investigated in KGN cells. RESULTS: The mice lacking irisin presented increased mortality, reduced body weight and poor fertility. Analysis of sex hormones showed decreased levels of estradiol, follicle-stimulating hormone and luteinizing hormone, and elevated progesterone levels in Fndc5 mutant mice. Irisin deficiency in mice was associated with irregular estrus, reduced ratio of antral follicles. The expressions of Akr1c18, Mamld1, and Cyp19a1, which are involved in the synthesis of steroid hormones, were reduced in the ovaries of mutant mice. Exogenous irisin could promote the expression of Akr1c18, Mamld1, and Cyp19a1 in KGN cells, stimulating estradiol production and inhibiting progesterone secretion. CONCLUSIONS: Irisin deficiency was related to disordered endocrinology metabolism in mice. The irisin deficient mice showed poor growth and development, and decreased fertility. Irisin likely have effects on the expressions of Akr1c18, Mamld1 and Cyp19a1 in ovary, regulating the steroid hormone production. This study provides novel insights into the potential role of irisin in mammalian growth and reproduction.


Subject(s)
Fertility/genetics , Fibronectins/genetics , Growth and Development/genetics , Animals , Cells, Cultured , Female , Gene Deletion , Granulosa Cells/physiology , Humans , Infertility, Female/genetics , Infertility, Female/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovarian Follicle/metabolism , Ovarian Follicle/physiology
15.
J Cell Mol Med ; 25(3): 1350-1358, 2021 02.
Article in English | MEDLINE | ID: mdl-33393222

ABSTRACT

The hair follicle (HF) is an important mini-organ of the skin, composed of many types of cells. Dermal papilla cells are important signalling components that guide the proliferation, upward migration and differentiation of HF stem cell progenitor cells to form other types of HF cells. Thymosin ß4 (Tß4), a major actin-sequestering protein, is involved in various cellular responses and has recently been shown to play key roles in HF growth and development. Endogenous Tß4 can activate the mouse HF cycle transition and affect HF growth and development by promoting the migration and differentiation of HF stem cells and their progeny. In addition, exogenous Tß4 increases the rate of hair growth in mice and promotes cashmere production by increasing the number of secondary HFs (hair follicles) in cashmere goats. However, the molecular mechanisms through which Tß4 promotes HF growth and development have rarely been reported. Herein, we review the functions and mechanisms of Tß4 in HF growth and development and describe the endogenous and exogenous actions of Tß4 in HFs to provide insights into the roles of Tß4 in HF growth and development.


Subject(s)
Hair Follicle/cytology , Hair Follicle/physiology , Organogenesis , Thymosin/genetics , Thymosin/metabolism , Animals , Gene Expression Regulation/drug effects , Growth and Development/drug effects , Growth and Development/genetics , Hair Follicle/drug effects , Humans , Organogenesis/drug effects , Signal Transduction , Structure-Activity Relationship , Thymosin/chemistry , Thymosin/pharmacology
16.
Int J Mol Sci ; 22(2)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33478078

ABSTRACT

Long non-coding RNAs (lncRNAs) have structural and functional roles in development and disease. We have previously shown that the LINC00961/SPAAR (small regulatory polypeptide of amino acid response) locus regulates endothelial cell function, and that both the lncRNA and micropeptide counter-regulate angiogenesis. To assess human cardiac cell SPAAR expression, we mined a publicly available scRNSeq dataset and confirmed LINC00961 locus expression and hypoxic response in a murine endothelial cell line. We investigated post-natal growth and development, basal cardiac function, the cardiac functional response, and tissue-specific response to myocardial infarction. To investigate the influence of the LINC00961/SPAAR locus on longitudinal growth, cardiac function, and response to myocardial infarction, we used a novel CRISPR/Cas9 locus knockout mouse line. Data mining suggested that SPAAR is predominantly expressed in human cardiac endothelial cells and fibroblasts, while murine LINC00961 expression is hypoxia-responsive in mouse endothelial cells. LINC00961-/- mice displayed a sex-specific delay in longitudinal growth and development, smaller left ventricular systolic and diastolic areas and volumes, and greater risk area following myocardial infarction compared with wildtype littermates. These data suggest the LINC00961/SPAAR locus contributes to cardiac endothelial cell and fibroblast function and hypoxic response, growth and development, and basal cardiovascular function in adulthood.


Subject(s)
Growth and Development/genetics , Heart/physiology , Myocardial Infarction/physiopathology , Peptides/physiology , Animals , Endothelial Cells/physiology , Female , Genetic Loci/physiology , Heart/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardium/metabolism , Neovascularization, Physiologic/genetics , Peptides/genetics
17.
BMC Plant Biol ; 21(1): 13, 2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33407131

ABSTRACT

BACKGROUND: The sequencing and annotations of cotton genomes provide powerful theoretical support to unravel more physiological and functional information. Plant homeodomain (PHD) protein family has been reported to be involved in regulating various biological processes in plants. However, their functional studies have not yet been carried out in cotton. RESULTS: In this study, 108, 55, and 52 PHD genes were identified in G. hirsutum, G. raimondii, and G. arboreum, respectively. A total of 297 PHD genes from three cotton species, Arabidopsis, and rice were divided into five groups. We performed chromosomal location, phylogenetic relationship, gene structure, and conserved domain analysis for GhPHD genes. GhPHD genes were unevenly distributed on each chromosome. However, more GhPHD genes were distributed on At_05, Dt_05, and At_07 chromosomes. GhPHD proteins depicted conserved domains, and GhPHD genes exhibiting similar gene structure were clustered together. Further, whole genome duplication (WGD) analysis indicated that purification selection greatly contributed to the functional maintenance of GhPHD gene family. Expression pattern analysis based on RNA-seq data showed that most GhPHD genes showed clear tissue-specific spatiotemporal expression patterns elucidating the multiple functions of GhPHDs in plant growth and development. Moreover, analysis of cis-acting elements revealed that GhPHDs may respond to a variety of abiotic and phytohormonal stresses. In this regard, some GhPHD genes showed good response against abiotic and phytohormonal stresses. Additionally, co-expression network analysis indicated that GhPHDs are essential for plant growth and development, while GhPHD genes response against abiotic and phytohormonal stresses may help to improve plant tolerance in adverse environmental conditions. CONCLUSION: This study will provide useful information to facilitate further research related to the vital roles of GhPHD gene family in plant growth and development.


Subject(s)
Arabidopsis/genetics , Gossypium/growth & development , Gossypium/genetics , Homeodomain Proteins/genetics , Oryza/genetics , Phytochrome/genetics , Plant Growth Regulators/genetics , Stress, Physiological/genetics , Crops, Agricultural/genetics , Crops, Agricultural/growth & development , Gene Expression Regulation, Developmental , Gene Expression Regulation, Plant , Genes, Plant , Genome, Plant , Growth and Development/genetics , Homeodomain Proteins/metabolism , Multigene Family , Phylogeny , Phytochrome/metabolism , Plant Growth Regulators/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Sequence Analysis , Stress, Physiological/physiology
18.
J Dev Orig Health Dis ; 12(6): 876-882, 2021 12.
Article in English | MEDLINE | ID: mdl-33407969

ABSTRACT

Rapid infant growth increases the risk for adult obesity. The gut microbiome is associated with early weight status; however, no study has examined how interactions between microbial and host ribonucleic acid (RNA) expression influence infant growth. We hypothesized that dynamics in infant stool micro-ribonucleic acids (miRNAs) would be associated with both microbial activity and infant growth via putative metabolic targets. Stool was collected twice from 30 full-term infants, at 1 month and again between 6 and 12 months. Stool RNA were measured with high-throughput sequencing and aligned to human and microbial databases. Infant growth was measured by weight-for-length z-score at birth and 12 months. Increased RNA transcriptional activity of Clostridia (R = 0.55; Adj p = 3.7E-2) and Burkholderia (R = -0.820, Adj p = 2.62E-3) were associated with infant growth. Of the 25 human RNAs associated with growth, 16 were miRNAs. The miRNAs demonstrated significant target enrichment (Adj p < 0.05) for four metabolic pathways. There were four associations between growth-related miRNAs and growth-related phyla. We have shown that longitudinal trends in gut microbiota activity and human miRNA levels are associated with infant growth and the metabolic targets of miRNAs suggest these molecules may regulate the biosynthetic landscape of the gut and influence microbial activity.


Subject(s)
Feces/microbiology , Gastrointestinal Microbiome/genetics , Growth and Development/physiology , Female , Follow-Up Studies , Gastrointestinal Microbiome/physiology , Gene Expression Profiling/methods , Gene Expression Profiling/statistics & numerical data , Growth and Development/genetics , Humans , Infant , Male , Pennsylvania
19.
Mol Cell Endocrinol ; 519: 111044, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33053393

ABSTRACT

Human size is achieved by the coordinated expression of many genes. From conception to adulthood, a given genomic endowment is modified by highly variable environmental circumstances. During each stage of a person's life, distinct nutritional and hormonal influences continuously shape growing physical features until mature characteristics are attained. Underlying processes depend on precise provision of substrates and energy extracted by insulin action from nutrients, which allows cell proliferation, differentiation, and survival, under the concerted actions of growth hormone and insulin-like growth factor-I (IGF-I). It should be noted that growth and metabolic signaling pathways are interdependent and superimposed at multiple levels. Attainment of a fully developed human phenotype should be considered as a harmonious increment in body size rather than a simple increase in height. From this perspective we herein analyze adult features of individuals with an inactive growth hormone receptor, who consequently have severely diminished concentrations of serum insulin and endocrine IGF-I.


Subject(s)
Growth Disorders/genetics , Hearing Loss, Sensorineural/genetics , Human Growth Hormone/genetics , Insulin Resistance/genetics , Insulin-Like Growth Factor I/deficiency , Mutation/genetics , Growth and Development/genetics , Humans , Insulin-Like Growth Factor I/genetics , Signal Transduction
20.
Nat Rev Genet ; 22(1): 38-58, 2021 01.
Article in English | MEDLINE | ID: mdl-32958894

ABSTRACT

The Trithorax group (TrxG) of proteins is a large family of epigenetic regulators that form multiprotein complexes to counteract repressive developmental gene expression programmes established by the Polycomb group of proteins and to promote and maintain an active state of gene expression. Recent studies are providing new insights into how two crucial families of the TrxG - the COMPASS family of histone H3 lysine 4 methyltransferases and the SWI/SNF family of chromatin remodelling complexes - regulate gene expression and developmental programmes, and how misregulation of their activities through genetic abnormalities leads to pathologies such as developmental disorders and malignancies.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/metabolism , Drosophila/genetics , Growth and Development/genetics , Histone-Lysine N-Methyltransferase/metabolism , Neoplasms/genetics , Polycomb-Group Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Animals , Chromatin/genetics , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice , Models, Molecular , Multiprotein Complexes , Polycomb-Group Proteins/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...