Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Annu Rev Nutr ; 44(1): 205-229, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38759081

ABSTRACT

Linear growth during three distinct stages of life determines attained stature in adulthood: namely, in utero, early postnatal life, and puberty and the adolescent period. Individual host factors, genetics, and the environment, including nutrition, influence attained human stature. Each period of physical growth has its specific biological and environmental considerations. Recent epidemiologic investigations reveal a strong influence of prenatal factors on linear size at birth that in turn influence the postnatal growth trajectory. Although average population height changes have been documented in high-income regions, stature as a complex human trait is not well understood or easily modified. This review summarizes the biology of linear growth and its major drivers, including nutrition from a life-course perspective, the genetics of programmed growth patterns or height, and gene-environment interactions that determine human stature in toto over the life span. Implications for public health interventions and knowledge gaps are discussed.


Subject(s)
Body Height , Gene-Environment Interaction , Humans , Nutritional Status , Female , Environment , Pregnancy , Adolescent
2.
Exp Physiol ; 109(5): 662-671, 2024 May.
Article in English | MEDLINE | ID: mdl-38156734

ABSTRACT

Childhood stunting and wasting, or decreased linear and ponderal growth associated with undernutrition, continue to be a major global public health challenge. Although many of the current therapeutic and dietary interventions have significantly reduced childhood mortality caused by undernutrition, there remain great inefficacies in improving childhood stunting. Longitudinal bone growth in children is governed by different genetic, nutritional and other environmental factors acting systemically on the endocrine system and locally at the growth plate. Recent studies have shown that this intricate interplay between nutritional and hormonal regulation of the growth plate could involve the gut microbiota, highlighting the importance of a holistic approach in tackling childhood undernutrition. In this review, I focus on the mechanistic insights provided by these recent advances in gut microbiota research and discuss ongoing development of microbiota-based therapeutics in humans, which could be the missing link in solving undernutrition and childhood stunting.


Subject(s)
Bone Development , Gastrointestinal Microbiome , Growth Disorders , Humans , Gastrointestinal Microbiome/physiology , Bone Development/physiology , Child , Growth Disorders/microbiology , Growth Disorders/physiopathology , Animals , Malnutrition/microbiology , Malnutrition/physiopathology , Child Development/physiology
3.
PLoS Biol ; 16(7): e2005263, 2018 07.
Article in English | MEDLINE | ID: mdl-30036371

ABSTRACT

Bones at different anatomical locations vary dramatically in size. For example, human femurs are 20-fold longer than the phalanges in the fingers and toes. The mechanisms responsible for these size differences are poorly understood. Bone elongation occurs at the growth plates and advances rapidly in early life but then progressively slows due to a developmental program termed "growth plate senescence." This developmental program includes declines in cell proliferation and hypertrophy, depletion of cells in all growth plate zones, and extensive underlying changes in the expression of growth-regulating genes. Here, we show evidence that these functional, structural, and molecular senescent changes occur earlier in the growth plates of smaller bones (metacarpals, phalanges) than in the growth plates of larger bones (femurs, tibias) and that this differential aging contributes to the disparities in bone length. We also show evidence that the molecular mechanisms that underlie the differential aging between different bones involve modulation of critical paracrine regulatory pathways, including insulin-like growth factor (Igf), bone morphogenetic protein (Bmp), and Wingless and Int-1 (Wnt) signaling. Taken together, the findings reveal that the striking disparities in the lengths of different bones, which characterize normal mammalian skeletal proportions, is achieved in part by modulating the progression of growth plate senescence.


Subject(s)
Aging/physiology , Bone and Bones/anatomy & histology , Cartilage/growth & development , Growth Plate/growth & development , Animals , Bone Development , Cell Proliferation , Chondrocytes/pathology , Extremities/growth & development , Gene Expression Regulation, Developmental , Hypertrophy , Mice, Inbred C57BL , Paracrine Communication , Rats, Sprague-Dawley , Tibia/growth & development
4.
Genet Med ; 22(8): 1329-1337, 2020 08.
Article in English | MEDLINE | ID: mdl-32341572

ABSTRACT

PURPOSE: Impaired function of gonadotropin-releasing hormone (GnRH) neurons can cause a phenotypic spectrum ranging from delayed puberty to isolated hypogonadotropic hypogonadism (IHH). We sought to identify a new genetic etiology for these conditions. METHODS: Exome sequencing was performed in an extended family with autosomal dominant, markedly delayed puberty. The effects of the variant were studied in a GnRH neuronal cell line. Variants in the same gene were sought in a large cohort of individuals with IHH. RESULTS: We identified a rare missense variant (F900V) in DLG2 (which encodes PSD-93) that cosegregated with the delayed puberty. The variant decreased GnRH expression in vitro. PSD-93 is an anchoring protein of NMDA receptors, a type of glutamate receptor that has been implicated in the control of puberty in laboratory animals. The F900V variant impaired the interaction between PSD-93 and a known binding partner, Fyn, which phosphorylates NMDA receptors. Variants in DLG2 that also decreased GnRH expression were identified in three unrelated families with IHH. CONCLUSION: The findings indicate that variants in DLG2/PSD-93 cause autosomal dominant delayed puberty and may also contribute to IHH. The findings also suggest that the pathogenesis involves impaired NMDA receptor signaling and consequently decreased GnRH secretion.


Subject(s)
Gonadotropin-Releasing Hormone , Hypogonadism , Gonadotropin-Releasing Hormone/genetics , Guanylate Kinases , Humans , Hypogonadism/genetics , Proteins , Signal Transduction , Tumor Suppressor Proteins , Exome Sequencing
5.
Mol Ther ; 27(3): 673-680, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30765323

ABSTRACT

Recombinant human growth hormone (GH) is commonly used to treat short stature in children. However, GH treatment has limited efficacy, particularly in severe, non-GH-deficient conditions such as chondrodysplasias, and potential off-target effects. Because short stature results from decreased growth plate chondrogenesis, we developed a cartilage-targeting single-chain human antibody fragment (CaAb) aiming to deliver therapeutic molecules to the growth plate, thereby increasing treatment efficacy while minimizing adverse effects on other tissues. To this end, we created fusion proteins of these CaAbs conjugated with insulin-like growth factor 1 (IGF-1), an endocrine and/or paracrine factor that positively regulates chondrogenesis. These CaAb-IGF-1 fusion proteins retained both cartilage binding and IGF-1 biological activity, and they were able to stimulate bone growth in an organ culture system. Using a GH-deficient (lit) mouse model, we found that subcutaneous injections of these CaAb-IGF-1 fusion proteins increased overall growth plate height without increasing proliferation in kidney cortical cells, suggesting on-target efficacy at the growth plate and less off-target effect on the kidney than IGF-1 alone. Alternate-day injections of these fusion proteins, unlike IGF-1 alone, were sufficient to produce a therapeutic effect. Our findings provide proof of principle that targeting therapeutics to growth plate cartilage can potentially improve treatment for childhood growth disorders.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Animals , Cartilage/drug effects , Cartilage/metabolism , Chondrogenesis/drug effects , Growth Plate/drug effects , Growth Plate/metabolism , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mutation/genetics
6.
Clin Genet ; 95(1): 160-164, 2019 01.
Article in English | MEDLINE | ID: mdl-30281152

ABSTRACT

In many children with short stature, the etiology of the decreased linear growth remains unknown. We sought to identify the underlying genetic etiology in a patient with short stature, irregular growth plates of the proximal phalanges, developmental delay, and mildly dysmorphic facial features. Exome sequencing identified a de novo, heterozygous, nonsense mutation (c.1606C>T:p.R536X) in QRICH1. In vitro studies confirmed that the mutation impaired expression of the QRICH1 protein. SiRNA-mediated knockdown of Qrich1 in primary mouse epiphyseal chondrocytes caused downregulation of gene expression associated with hypertrophic differentiation. We then identified an unrelated individual with another heterozygous de novo nonsense mutation in QRICH1 who had a similar phenotype. A recently published study identified QRICH1 mutations in three patients with developmental delay, one of whom had short stature. Our findings indicate that QRICH1 mutations cause not only developmental delay but also a chondrodysplasia characterized by diminished linear growth and abnormal growth plate morphology due to impaired growth plate chondrocyte hypertrophic differentiation.


Subject(s)
Chondrogenesis/genetics , DNA-Binding Proteins/genetics , Developmental Disabilities/genetics , Microtubule Proteins/genetics , Osteochondrodysplasias/genetics , Transcription Factors/genetics , Animals , Child , Child, Preschool , Chondrocytes/metabolism , Chondrocytes/pathology , Codon, Nonsense/genetics , Developmental Disabilities/pathology , Exome/genetics , Female , Growth Plate/growth & development , Growth Plate/pathology , Heterozygote , Humans , Infant , Male , Mice , Mutation/genetics , Osteochondrodysplasias/pathology
7.
Proc Natl Acad Sci U S A ; 110(15): 6181-6, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23530192

ABSTRACT

Insulin-like growth factor 2 (IGF2) is an important fetal growth factor. Its expression is dramatically down-regulated in multiple organs after birth but is frequently up-regulated in cancers. The mechanisms that drive down-regulation of IGF2 in postnatal tissues or the up-regulation in malignancy are unclear. We found evidence that E2F transcription factor 3 (E2F3) drives these changes in expression. E2f3 mRNA expression, protein expression, and binding to the Igf2 promoter all decreased with age postnatally in multiple mouse organs. In late juvenile hepatocytes, restoration of high E2f3 expression restored high Igf2 expression, indicating a causal relationship, but this induction did not occur in fetal hepatocytes, which already have high E2f3 and Igf2 expression. Transient expression of E2f3 in both HEK293 cells and in late juvenile hepatocytes were able to activate reporter constructs containing the mouse Igf2 promoter P2, which includes consensus E2F-binding sites. In humans, microarray data revealed declines in E2F3 and IGF2 expression with age similar to the mouse. In addition, E2F3-overexpressing human prostate and bladder cancers showed increased IGF2 expression, and levels of E2F3 and IGF2 mRNA in these cancers were positively correlated. Taken together, the findings suggest that down-regulation of E2f3 with age helps drive the dramatic decline in Igf2 expression in postnatal organs, and E2F3 overexpression in human cancers induces IGF2 overexpression.


Subject(s)
E2F3 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor II/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Computational Biology , Down-Regulation , E2F3 Transcription Factor/genetics , Hepatocytes/cytology , Humans , Insulin-Like Growth Factor II/genetics , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Time Factors
8.
Pediatr Res ; 77(3): 406-15, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25521919

ABSTRACT

BACKGROUND: In juvenile mammals, the epiphyses of long bones grow by chondrogenesis within the articular cartilage. A better understanding of the molecular mechanisms that regulate the growth of articular cartilage may give insight into the antecedents of joint disease, such as osteoarthritis. METHODS: We used laser capture microdissection to isolate chondrocytes from the superficial, middle, and deep zones of growing tibial articular cartilage in the 1-wk-old mouse and then investigated expression patterns by microarray. To identify molecular markers for each zone of the growing articular cartilage, we found genes showing zone-specific expression and confirmed by real-time PCR and in situ hybridization. RESULTS: Bioinformatic analyses implicated ephrin receptor signaling, Wnt signaling, and bone morphogenetic protein signaling in the spatial regulation of chondrocyte differentiation during growth. Molecular markers were identified for superficial (e.g., Cilp, Prg4), middle (Cxcl14, Tnn), and deep zones (Sfrp5, Frzb). Comparison between juvenile articular and growth plate cartilage revealed that the superficial-to-deep zone transition showed similarity with the hypertrophic-to-resting zone transition. CONCLUSION: Laser capture microdissection combined with microarray analysis identified novel signaling pathways that are spatially regulated in growing mouse articular cartilage and revealed similarities between the molecular architecture of the growing articular cartilage and that of growth plate cartilage.


Subject(s)
Cartilage, Articular/growth & development , Chondrogenesis/physiology , Gene Expression Regulation, Developmental/physiology , Signal Transduction/physiology , Animals , Bone Morphogenetic Proteins/metabolism , Cartilage, Articular/metabolism , Cell Differentiation/physiology , Computational Biology , Gene Expression Profiling , In Situ Hybridization , Laser Capture Microdissection , Mice , Microarray Analysis , Real-Time Polymerase Chain Reaction , Receptors, Eph Family/metabolism , Wnt Proteins/metabolism
9.
Curr Opin Pediatr ; 27(4): 502-10, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26087427

ABSTRACT

PURPOSE OF REVIEW: Recent basic studies have yielded important new insights into the molecular mechanisms that regulate growth locally. Simultaneously, clinical studies have identified new molecular defects that cause growth failure and overgrowth, and genome-wide association studies have elucidated the genetic basis for normal human height variation. RECENT FINDINGS: The Hippo pathway has emerged as one of the major mechanisms controlling organ size. In addition, an extensive genetic program has been described that allows rapid body growth in the fetus and infant but then causes growth to slow with age in multiple tissues. In human genome-wide association studies, hundreds of loci associated with adult stature have been identified; many appear to involve genes that function locally in the growth plate. Clinical genetic studies have identified a new genetic abnormality, microduplication of Xq26.3, that is responsible for growth hormone excess, and a gene, DNMT3A, in which mutations cause an overgrowth syndrome through epigenetic mechanisms. SUMMARY: These recent advances in our understanding of somatic growth not only provide insight into childhood growth disorders but also have broader medical applications because disruption of these regulatory systems contributes to oncogenesis.


Subject(s)
Body Height/genetics , Genome-Wide Association Study , Growth Disorders/genetics , Growth Plate/metabolism , Hippocampus/physiology , Body Height/physiology , Child , Child Nutritional Physiological Phenomena , Child, Preschool , Gene Expression Profiling , Growth Disorders/physiopathology , Growth Plate/physiology , Humans , Infant , Infant, Newborn , Mutation , Phenotype , Quantitative Trait Loci/genetics , Signal Transduction
10.
Hum Mol Genet ; 21(23): 5193-201, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22914739

ABSTRACT

Previous meta-analysis of genome-wide association (GWA) studies has identified 180 loci that influence adult height. However, each GWA locus typically comprises a set of contiguous genes, only one of which presumably modulates height. We reasoned that many of the causative genes within these loci influence height because they are expressed in and function in the growth plate, a cartilaginous structure that causes bone elongation and thus determines stature. Therefore, we used expression microarray studies of mouse and rat growth plate, human disease databases and a mouse knockout phenotype database to identify genes within the GWAS loci that are likely required for normal growth plate function. Each of these approaches identified significantly more genes within the GWA height loci than at random genomic locations (P < 0.0001 each), supporting the validity of the approach. The combined analysis strongly implicates 78 genes in growth plate function, including multiple genes that participate in PTHrP-IHH, BMP and CNP signaling, and many genes that have not previously been implicated in the growth plate. Thus, this analysis reveals a large number of novel genes that regulate human growth plate chondrogenesis and thereby contribute to the normal variations in human adult height. The analytic approach developed for this study may be applied to GWA studies for other common polygenic traits and diseases, thus providing a new general strategy to identify causative genes within GWA loci and to translate genetic associations into mechanistic biological insights.


Subject(s)
Body Height/genetics , Gene Expression Profiling , Genome-Wide Association Study , Growth Plate/metabolism , Animals , Computational Biology/methods , Genomics , Growth Plate/growth & development , Humans , Male , Mice , Phenotype , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Reproducibility of Results
11.
Front Endocrinol (Lausanne) ; 15: 1327378, 2024.
Article in English | MEDLINE | ID: mdl-38370361

ABSTRACT

Epigenetic modifications play an important role in regulation of transcription and gene expression. The molecular machinery governing epigenetic modifications, also known as epigenetic regulators, include non-coding RNA, chromatin remodelers, and enzymes or proteins responsible for binding, reading, writing and erasing DNA and histone modifications. Recent advancement in human genetics and high throughput sequencing technology have allowed the identification of causative variants, many of which are epigenetic regulators, for a wide variety of childhood growth disorders that include skeletal dysplasias, idiopathic short stature, and generalized overgrowth syndromes. In this review, we highlight the connection between epigenetic modifications, genetic variants in epigenetic regulators and childhood growth disorders being established over the past decade, discuss their insights into skeletal biology, and the potential of epidrugs as a new type of therapeutic intervention.


Subject(s)
Chromatin , Epigenesis, Genetic , Humans , DNA Methylation , DNA , Growth Disorders/genetics
12.
J Clin Endocrinol Metab ; 109(2): 312-320, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-37450557

ABSTRACT

Human overgrowth disorders are characterized by excessive prenatal and/or postnatal growth of various tissues. These disorders often present with tall stature, macrocephaly, and/or abdominal organomegaly and are sometimes associated with additional phenotypic abnormalities such as intellectual disability and increased cancer risk. As the genetic etiology of these disorders have been elucidated, a surprising pattern has emerged. Multiple monogenic overgrowth syndromes result from variants in epigenetic regulators: variants in histone methyltransferases NSD1 and EZH2 cause Sotos syndrome and Weaver syndrome, respectively, variants in DNA methyltransferase DNMT3A cause Tatton-Brown-Rahman syndrome, and variants in chromatin remodeler CHD8 cause an autism spectrum disorder with overgrowth. In addition, very recently, a variant in histone reader protein SPIN4 was identified in a new X-linked overgrowth disorder. In this review, we discuss the genetics of these overgrowth disorders and explore possible common underlying mechanisms by which epigenetic pathways regulate human body size.


Subject(s)
Abnormalities, Multiple , Autism Spectrum Disorder , Intellectual Disability , Humans , Abnormalities, Multiple/genetics , Syndrome , Histone Methyltransferases/genetics , Intellectual Disability/genetics , Epigenesis, Genetic
13.
J Endocr Soc ; 8(8): bvae117, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38957653

ABSTRACT

Gut microbiota plays an important role in the regulation of bone homeostasis and bone health. Recent studies showed that these effects could be mediated through microbial metabolites released by the microbiota like short-chain fatty acids, metabolism of endogenous molecules such as bile acids, or a complex interplay between microbiota, the endocrine system, and the immune system. Importantly, some studies showed a reciprocal relationship between the endocrine system and gut microbiota. For instance, postmenopausal estrogen deficiency could lead to dysbiosis of the gut microbiota, which could in turn affect various immune response and bone remodeling. In addition, evidence showed that shift in the indigenous gut microbiota caused by antibiotics treatment may also impact normal skeletal growth and maturation. In this mini-review, we describe recent findings on the role of microbiome in bone homeostasis, with a particular focus on molecular mechanisms and their interactions with the endocrine and immune system. We will also discuss the recent findings on estrogen deficiency and microbiota dysbiosis, and the clinical implications for the development of new therapeutic strategies for osteoporosis and other bone disorders.

14.
JCI Insight ; 8(9)2023 05 08.
Article in English | MEDLINE | ID: mdl-36927955

ABSTRACT

Overgrowth syndromes can be caused by pathogenic genetic variants in epigenetic writers, such as DNA and histone methyltransferases. However, no overgrowth disorder has previously been ascribed to variants in a gene that acts primarily as an epigenetic reader. Here, we studied a male individual with generalized overgrowth of prenatal onset. Exome sequencing identified a hemizygous frameshift variant in Spindlin 4 (SPIN4), with X-linked inheritance. We found evidence that SPIN4 binds specific histone modifications, promotes canonical WNT signaling, and inhibits cell proliferation in vitro and that the identified frameshift variant had lost all of these functions. Ablation of Spin4 in mice recapitulated the human phenotype with generalized overgrowth, including increased longitudinal bone growth. Growth plate analysis revealed increased cell proliferation in the proliferative zone and an increased number of progenitor chondrocytes in the resting zone. We also found evidence of decreased canonical Wnt signaling in growth plate chondrocytes, providing a potential explanation for the increased number of resting zone chondrocytes. Taken together, our findings provide strong evidence that SPIN4 is an epigenetic reader that negatively regulates mammalian body growth and that loss of SPIN4 causes an overgrowth syndrome in humans, expanding our knowledge of the epigenetic regulation of human growth.


Subject(s)
Epigenesis, Genetic , Genes, X-Linked , Male , Humans , Mice , Animals , Syndrome , Cell Cycle Proteins , Mammals
15.
Front Endocrinol (Lausanne) ; 14: 1258313, 2023.
Article in English | MEDLINE | ID: mdl-38152138

ABSTRACT

Very tall people attract much attention and represent a clinically and genetically heterogenous group of individuals. Identifying the genetic etiology can provide important insights into the molecular mechanisms regulating linear growth. We studied a three-generation pedigree with five isolated (non-syndromic) tall members and one individual with normal stature by whole exome sequencing; the tallest man had a height of 211 cm. Six heterozygous gene variants predicted as damaging were shared among the four genetically related tall individuals and not present in a family member with normal height. To gain insight into the putative role of these candidate genes in bone growth, we assessed the transcriptome of murine growth plate by microarray and RNA Seq. Two (Ift140, Nav2) of the six genes were well-expressed in the growth plate. Nav2 (p-value 1.91E-62) as well as Ift140 (p-value of 2.98E-06) showed significant downregulation of gene expression between the proliferative and hypertrophic zone, suggesting that these genes may be involved in the regulation of chondrocyte proliferation and/or hypertrophic differentiation. IFT140, NAV2 and SCAF11 have also significantly associated with height in GWAS studies. Pathway and network analysis indicated functional connections between IFT140, NAV2 and SCAF11 and previously associated (tall) stature genes. Knockout of the all-trans retinoic acid responsive gene, neuron navigator 2 NAV2, in Xenopus supports its functional role as a growth promotor. Collectively, our data expand the spectrum of genes with a putative role in tall stature phenotypes and, among other genes, highlight NAV2 as an interesting gene to this phenotype.


Subject(s)
Body Height , DNA Helicases , Animals , Humans , Male , Mice , Bone Development , Growth Plate , Tretinoin , Body Height/genetics , DNA Helicases/genetics
16.
Pediatr Res ; 71(1): 32-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22289848

ABSTRACT

INTRODUCTION: In many normal tissues, proliferation rates decline postnatally, causing somatic growth to slow. Previous evidence suggests that this decline is due, in part, to decline in the expression of growth-promoting imprinted genes including Mest, Plagl1, Peg3, Dlk1, and Igf2. Embryonal cancers are composed of cells that maintain embryonic characteristics and proliferate rapidly in childhood. We hypothesized that the abnormal persistent rapid proliferation in embryonal cancers occurs in part because of abnormal persistent high expression of growth-promoting imprinted genes. RESULTS: Analysis of microarray data showed elevated expression of MEST, PLAGL1, PEG3, DLK1, and IGF2 in various embryonal cancers, especially rhabdomyosarcoma, as compared to nonembryonal cancers and normal tissues. Similarly, mRNA expression, assessed by real-time PCR, of MEST, PEG3, and IGF2 in rhabdomyosarcoma cell lines was increased as compared to nonembryonal cancer cell lines. Furthermore, siRNA-mediated knockdown of MEST, PLAGL1, PEG3, and IGF2 expression inhibited proliferation in Rh30 rhabdomyosarcoma cells. DISCUSSION: These findings suggest that the normal postnatal downregulation of growth-promoting imprinted genes fails to occur in some embryonal cancers, particularly rhabdomyosarcoma, and contributes to the persistent rapid proliferation of rhabdomyosarcoma cells and, more generally, that failure of the mechanisms responsible for normal somatic growth deceleration can promote tumorigenesis.


Subject(s)
Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Genomic Imprinting , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Child , Humans , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Microarray Analysis , Proteins/genetics , Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
Nat Commun ; 13(1): 700, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35121733

ABSTRACT

SP7/Osterix is a transcription factor critical for osteoblast maturation and bone formation. Homozygous loss-of-function mutations in SP7 cause osteogenesis imperfecta type XII, but neomorphic (gain-of-new-function) mutations of SP7 have not been reported in humans. Here we describe a de novo dominant neomorphic missense variant (c.926 C > G:p.S309W) in SP7 in a patient with craniosynostosis, cranial hyperostosis, and long bone fragility. Histomorphometry shows increased osteoblasts but decreased bone mineralization. Mice with the corresponding variant also show a complex skeletal phenotype distinct from that of Sp7-null mice. The mutation alters the binding specificity of SP7 from AT-rich motifs to a GC-consensus sequence (typical of other SP family members) and produces an aberrant gene expression profile, including increased expression of Col1a1 and endogenous Sp7, but decreased expression of genes involved in matrix mineralization. Our study identifies a pathogenic mechanism in which a mutation in a transcription factor shifts DNA binding specificity and provides important in vivo evidence that the affinity of SP7 for AT-rich motifs, unique among SP proteins, is critical for normal osteoblast differentiation.


Subject(s)
Bone Diseases/genetics , Bone and Bones/metabolism , Gene Expression Regulation , Mutation , Sp7 Transcription Factor/genetics , Animals , Bone Diseases/metabolism , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Child , HEK293 Cells , Humans , In Situ Hybridization , Male , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/metabolism , Sp7 Transcription Factor/metabolism , X-Ray Microtomography
18.
FASEB J ; 24(8): 3083-92, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20371622

ABSTRACT

Children grow, but adults do not. The cessation of growth in multiple organs is the end result of a progressive decline in cell proliferation beginning in early life. The mechanisms responsible for this growth deceleration are largely unknown. Using expression microarray and real-time PCR, we identified a common program of gene expression in lung, kidney, and liver during growth deceleration in juvenile rats. Gene ontology analyses and siRNA-mediated knockdown in vitro indicated that many of the down-regulated genes are growth promoting. Down-regulated genes in the program showed declining histone H3K4 trimethylation with age, implicating underlying epigenetic mechanisms. To investigate the physiological processes driving the genetic program, a tryptophan-deficient diet was used to temporarily inhibit juvenile growth in newborn rats for 4 wk. Afterward, microarray analysis showed that the genetic program had been delayed, implying that it is driven by body growth itself rather than age. Taken together, the findings suggest that growth in early life induces progressive down-regulation of a large set of proliferation-stimulating genes, causing organ growth to slow and eventually cease.


Subject(s)
Down-Regulation/genetics , Epigenesis, Genetic , Gene Regulatory Networks , Growth/genetics , Organ Size/genetics , Animals , Cell Proliferation , Gene Expression Profiling , Histones/metabolism , Kidney , Liver , Lung , Methylation , Rats
19.
Methods Mol Biol ; 2245: 105-119, 2021.
Article in English | MEDLINE | ID: mdl-33315198

ABSTRACT

The ability to identify, isolate, and study pure populations of cells is critical for understanding normal physiology in organs and tissues, which involves spatial regulation of signaling pathways and interactions between cells with different functions, expression profiles, and lineages. Here, we focus on assessing the growth plate cartilage, composed of multiple functionally and histologically distinct zones, to investigate temporally and spatially dependent gene expression differences. In this chapter, we describe the method of laser capture microdissection to isolate chondrocytes from different zones of differentiation in the mouse growth plate cartilage for RNA isolation, and subsequent downstream applications, such as RNA-sequencing and quantitative real-time PCR. We also provide an assessment of different factors contributing to the integrity of the isolated RNA, such as staining methods and procedures in RNA isolation.


Subject(s)
Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Growth Plate/cytology , Growth Plate/metabolism , Laser Capture Microdissection/methods , Animals , Immunohistochemistry , Mice
20.
Front Endocrinol (Lausanne) ; 12: 660731, 2021.
Article in English | MEDLINE | ID: mdl-34194391

ABSTRACT

Human growth is a complex trait. A considerable number of gene defects have been shown to cause short stature, but there are only few examples of genetic causes of non-syndromic tall stature. Besides rare variants with large effects and common risk alleles with small effect size, oligogenic effects may contribute to this phenotype. Exome sequencing was carried out in a tall male (height 3.5 SDS) and his parents. Filtered damaging variants with high CADD scores were validated by Sanger sequencing in the trio and three other affected and one unaffected family members. Network analysis was carried out to assess links between the candidate genes, and the transcriptome of murine growth plate was analyzed by microarray as well as RNA Seq. Heterozygous gene variants in CEP104, CROCC, NEK1, TOM1L2, and TSTD2 predicted as damaging were found to be shared between the four tall family members. Three of the five genes (CEP104, CROCC, and NEK1) belong to the ciliary gene family. All genes are expressed in mouse growth plate. Pathway and network analyses indicated close functional connections. Together, these data expand the spectrum of genes with a role in linear growth and tall stature phenotypes.


Subject(s)
Body Height/genetics , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , Growth Disorders/genetics , NIMA-Related Kinase 1/genetics , Thiosulfate Sulfurtransferase/genetics , Adolescent , Animals , Child , Child, Preschool , Exome , Female , Gene Expression , Growth Plate/metabolism , Humans , Infant , Infant, Newborn , Male , Mice , Netherlands , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL