Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Sci Transl Med ; 14(665): eabh2369, 2022 10 05.
Article in English | MEDLINE | ID: mdl-36197968

ABSTRACT

The nitric oxide (NO) signaling pathway in hypothalamic neurons plays a key role in the regulation of the secretion of gonadotropin-releasing hormone (GnRH), which is crucial for reproduction. We hypothesized that a disruption of neuronal NO synthase (NOS1) activity underlies some forms of hypogonadotropic hypogonadism. Whole-exome sequencing was performed on a cohort of 341 probands with congenital hypogonadotropic hypogonadism to identify ultrarare variants in NOS1. The activity of the identified NOS1 mutant proteins was assessed by their ability to promote nitrite and cGMP production in vitro. In addition, physiological and pharmacological characterization was carried out in a Nos1-deficient mouse model. We identified five heterozygous NOS1 loss-of-function mutations in six probands with congenital hypogonadotropic hypogonadism (2%), who displayed additional phenotypes including anosmia, hearing loss, and intellectual disability. NOS1 was found to be transiently expressed by GnRH neurons in the nose of both humans and mice, and Nos1 deficiency in mice resulted in dose-dependent defects in sexual maturation as well as in olfaction, hearing, and cognition. The pharmacological inhibition of NO production in postnatal mice revealed a critical time window during which Nos1 activity shaped minipuberty and sexual maturation. Inhaled NO treatment at minipuberty rescued both reproductive and behavioral phenotypes in Nos1-deficient mice. In summary, lack of NOS1 activity led to GnRH deficiency associated with sensory and intellectual comorbidities in humans and mice. NO treatment during minipuberty reversed deficits in sexual maturation, olfaction, and cognition in Nos1 mutant mice, suggesting a potential therapy for humans with NO deficiency.


Subject(s)
Hypogonadism , Nitric Oxide , Animals , Cognition , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Humans , Hypogonadism/complications , Hypogonadism/congenital , Hypogonadism/genetics , Mice , Mutant Proteins , Mutation/genetics , Nitric Oxide Synthase Type I/genetics , Nitrites
2.
Article in English | MEDLINE | ID: mdl-34152287

ABSTRACT

SUMMARY: Complete androgen-insensitivity syndrome (CAIS), a disorder of sex development (46,XY DSD), is caused primarily by mutations in the androgen receptor (AR). Gonadectomy is recommended due to the increased risk of gonadoblastoma, however, surgical intervention is often followed by loss of libido. We present a 26-year-old patient with CAIS who underwent gonadectomy followed by a significant decrease in libido, which was improved with testosterone treatment but not with estradiol. Genetic testing was performed and followed by molecular characterization. We found that this patient carried a previously unidentified start loss mutation in the androgen receptor. This variant resulted in an N-terminal truncated protein with an intact DNA binding domain and was confirmed to be loss-of-function in vitro. This unique CAIS case and detailed functional studies raise intriguing questions regarding the relative roles of testosterone and estrogen in libido, and in particular, the potential non-genomic actions of androgens. LEARNING POINTS: N-terminal truncation of androgen receptor can cause androgen-insensitivity syndrome. Surgical removal of testosterone-producing gonads can result in loss of libido. Libido may be improved with testosterone treatment but not with estradiol in some forms of CAIS. A previously unreported AR mutation - p.Glu2_Met190del (c.2T>C) - is found in a CAIS patient and results in blunted AR transcriptional activity under testosterone treatment.

3.
Genet Med ; 22(11): 1759-1767, 2020 11.
Article in English | MEDLINE | ID: mdl-32724172

ABSTRACT

PURPOSE: Congenital hypogonadotropic hypogonadism (CHH) is a rare disorder resulting in absent puberty and infertility. The genetic architecture is complex with multiple loci involved, variable expressivity, and incomplete penetrance. The majority of cases are sporadic, consistent with a disease affecting fertility. The current study aims to investigate mosaicism as a genetic mechanism for CHH, focusing on de novo rare variants in CHH genes. METHODS: We evaluated 60 trios for de novo rare sequencing variants (RSV) in known CHH genes using exome sequencing. Potential mosaicism was suspected among RSVs with altered allelic ratios and confirmed using customized ultradeep sequencing (UDS) in multiple tissues. RESULTS: Among the 60 trios, 10 probands harbored de novo pathogenic variants in CHH genes. Custom UDS demonstrated that three of these de novo variants were in fact postzygotic mosaicism-two in FGFR1 (p.Leu630Pro and p.Gly348Arg), and one in CHD7 (p.Arg2428*). Statistically significant variation across multiple tissues (DNA from blood, buccal, hair follicle, urine) confirmed their mosaic nature. CONCLUSIONS: We identified a significant number of de novo pathogenic variants in CHH of which a notable number (3/10) exhibited mosaicism. This report of postzygotic mosaicism in CHH patients provides valuable information for accurate genetic counseling.


Subject(s)
Hypogonadism , Infertility , Genetic Counseling , Humans , Hypogonadism/genetics , Mosaicism , Exome Sequencing
4.
Am J Hum Genet ; 106(1): 58-70, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31883645

ABSTRACT

Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic disorder characterized by infertility and the absence of puberty. Defects in GnRH neuron migration or altered GnRH secretion and/or action lead to a severe gonadotropin-releasing hormone (GnRH) deficiency. Given the close developmental association of GnRH neurons with the olfactory primary axons, CHH is often associated with anosmia or hyposmia, in which case it is defined as Kallmann syndrome (KS). The genetics of CHH are heterogeneous, and >40 genes are involved either alone or in combination. Several CHH-related genes controlling GnRH ontogeny encode proteins containing fibronectin-3 (FN3) domains, which are important for brain and neural development. Therefore, we hypothesized that defects in other FN3-superfamily genes would underlie CHH. Next-generation sequencing was performed for 240 CHH unrelated probands and filtered for rare, protein-truncating variants (PTVs) in FN3-superfamily genes. Compared to gnomAD controls the CHH cohort was statistically enriched for PTVs in neuron-derived neurotrophic factor (NDNF) (p = 1.40 × 10-6). Three heterozygous PTVs (p.Lys62∗, p.Tyr128Thrfs∗55, and p.Trp469∗, all absent from the gnomAD database) and an additional heterozygous missense mutation (p.Thr201Ser) were found in four KS probands. Notably, NDNF is expressed along the GnRH neuron migratory route in both mouse embryos and human fetuses and enhances GnRH neuron migration. Further, knock down of the zebrafish ortholog of NDNF resulted in altered GnRH migration. Finally, mice lacking Ndnf showed delayed GnRH neuron migration and altered olfactory axonal projections to the olfactory bulb; both results are consistent with a role of NDNF in GnRH neuron development. Altogether, our results highlight NDNF as a gene involved in the GnRH neuron migration implicated in KS.


Subject(s)
Cell Movement , Hypogonadism/congenital , Hypogonadism/genetics , Mutation , Nerve Growth Factors/genetics , Neurons/pathology , Adolescent , Animals , Cohort Studies , Female , Heterozygote , Humans , Hypogonadism/pathology , Male , Mice , Mice, Knockout , Nerve Growth Factors/physiology , Neurons/metabolism , Pedigree , Zebrafish
5.
Am J Physiol Endocrinol Metab ; 315(5): E833-E847, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29944388

ABSTRACT

ß-Klotho (encoded by Klb) is an obligate coreceptor, mediating both fibroblast growth factor (FGF)15 and FGF21 signaling. Klb-/- mice are refractory to metabolic FGF15 and FGF21 action and exhibit derepressed (increased) bile acid (BA) synthesis. Here, we deeply phenotyped male Klb-/- mice on a pure C57BL/6J genetic background, fed a chow diet focusing on metabolic aspects. This aims to better understand the physiological consequences of concomitant FGF15 and FGF21 signaling deficiency, in particular on the gut-liver axis. Klb-/- mice present permanent growth restriction independent of adiposity and energy balance. Klb-/- mice also exhibit few changes in carbohydrate metabolism, combining normal gluco-tolerance, insulin sensitivity, and fasting response with increased gluconeogenic capacity and decreased glycogen mobilization. Livers of Klb-/- mice reveal pathologic features, including a proinflammatory status and initiation of fibrosis. These defects are associated to a massive shift in BA composition in the enterohepatic system and blood circulation featured by a large excess of microbiota-derived deoxycholic acid, classically known for its genotoxicity in the gastrointestinal tract. In conclusion, ß-Klotho is a gatekeeper of hepatic integrity through direct action (mediating FGF21 anti-inflammatory signaling) and indirect mechanisms (mediating FGF15 signaling that maintains BA level and composition).


Subject(s)
Bile Acids and Salts/metabolism , Body Weight/physiology , Gastrointestinal Tract/metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , Membrane Proteins/metabolism , Adiposity/genetics , Animals , Energy Metabolism/genetics , Fibroblast Growth Factors/metabolism , Gluconeogenesis/physiology , Ketone Bodies/blood , Klotho Proteins , Membrane Proteins/genetics , Mice , Mice, Knockout , Signal Transduction/physiology
6.
Hum Mol Genet ; 27(2): 359-372, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29202173

ABSTRACT

Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic disease characterized by absent puberty and infertility due to GnRH deficiency, and is often associated with anosmia [Kallmann syndrome (KS)]. The genetic etiology of CHH is heterogeneous, and more than 30 genes have been implicated in approximately 50% of patients with CHH. We hypothesized that genes encoding axon-guidance proteins containing fibronectin type-III (FN3) domains (similar to ANOS1, the first gene associated with KS), are mutated in CHH. We performed whole-exome sequencing in a cohort of 133 CHH probands to test this hypothesis, and identified rare sequence variants (RSVs) in genes encoding for the FN3-domain encoding protein deleted in colorectal cancer (DCC) and its ligand Netrin-1 (NTN1). In vitro studies of these RSVs revealed altered intracellular signaling associated with defects in cell morphology, and confirmed five heterozygous DCC mutations in 6 probands-5 of which presented as KS. Two KS probands carry heterozygous mutations in both DCC and NTN1 consistent with oligogenic inheritance. Further, we show that Netrin-1 promotes migration in immortalized GnRH neurons (GN11 cells). This study implicates DCC and NTN1 mutations in the pathophysiology of CHH consistent with the role of these two genes in the ontogeny of GnRH neurons in mice.


Subject(s)
DCC Receptor/genetics , Hypogonadism/genetics , Netrin-1/genetics , Adult , Cohort Studies , DCC Receptor/metabolism , Female , Fibronectin Type III Domain , Gonadotropin-Releasing Hormone/deficiency , Humans , Hypogonadism/metabolism , Hypogonadism/pathology , Male , Mutation , Netrin-1/metabolism , Neurons/metabolism , Neurons/pathology , Pedigree , Exome Sequencing
7.
Genet Med ; 20(8): 872-881, 2018 08.
Article in English | MEDLINE | ID: mdl-29144511

ABSTRACT

PURPOSE: Congenital hypogonadotropic hypogonadism (CHH), a rare genetic disease caused by gonadotropin-releasing hormone deficiency, can also be part of complex syndromes (e.g., CHARGE syndrome). CHD7 mutations were reported in 60% of patients with CHARGE syndrome, and in 6% of CHH patients. However, the definition of CHD7 mutations was variable, and the associated CHARGE signs in CHH were not systematically examined. METHODS: Rare sequencing variants (RSVs) in CHD7 were identified through exome sequencing in 116 CHH probands, and were interpreted according to American College of Medical Genetics and Genomics guidelines. Detailed phenotyping was performed in CHH probands who were positive for CHD7 RSVs, and genotype-phenotype correlations were evaluated. RESULTS: Of the CHH probands, 16% (18/116) were found to harbor heterozygous CHD7 RSVs, and detailed phenotyping was performed in 17 of them. Of CHH patients with pathogenic or likely pathogenic CHD7 variants, 80% (4/5) were found to exhibit multiple CHARGE features, and 3 of these patients were reclassified as having CHARGE syndrome. In contrast, only 8% (1/12) of CHH patients with nonpathogenic CHD7 variants exhibited multiple CHARGE features (P = 0.01). CONCLUSION: Pathogenic or likely pathogenic CHD7 variants rarely cause isolated CHH. Therefore a detailed clinical investigation is indicated to clarify the diagnosis (CHH versus CHARGE) and to optimize clinical management.


Subject(s)
CHARGE Syndrome/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Hypogonadism/genetics , CHARGE Syndrome/diagnosis , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Family , Female , Genetic Association Studies , Genetic Variation/genetics , Heterozygote , Humans , Male , Mutation , Pedigree , Phenotype , Sequence Analysis, DNA
8.
EMBO Mol Med ; 9(10): 1379-1397, 2017 10.
Article in English | MEDLINE | ID: mdl-28754744

ABSTRACT

Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic form of isolated gonadotropin-releasing hormone (GnRH) deficiency caused by mutations in > 30 genes. Fibroblast growth factor receptor 1 (FGFR1) is the most frequently mutated gene in CHH and is implicated in GnRH neuron development and maintenance. We note that a CHH FGFR1 mutation (p.L342S) decreases signaling of the metabolic regulator FGF21 by impairing the association of FGFR1 with ß-Klotho (KLB), the obligate co-receptor for FGF21. We thus hypothesized that the metabolic FGF21/KLB/FGFR1 pathway is involved in CHH Genetic screening of 334 CHH patients identified seven heterozygous loss-of-function KLB mutations in 13 patients (4%). Most patients with KLB mutations (9/13) exhibited metabolic defects. In mice, lack of Klb led to delayed puberty, altered estrous cyclicity, and subfertility due to a hypothalamic defect associated with inability of GnRH neurons to release GnRH in response to FGF21. Peripheral FGF21 administration could indeed reach GnRH neurons through circumventricular organs in the hypothalamus. We conclude that FGF21/KLB/FGFR1 signaling plays an essential role in GnRH biology, potentially linking metabolism with reproduction.


Subject(s)
Fibroblast Growth Factors/metabolism , Gonadotropin-Releasing Hormone/metabolism , Kallmann Syndrome/genetics , Membrane Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , COS Cells , Caenorhabditis elegans/genetics , Chlorocebus aethiops , Cohort Studies , Female , Fibroblast Growth Factors/genetics , Gonadotropin-Releasing Hormone/genetics , HEK293 Cells , Humans , Hypothalamus/metabolism , Klotho Proteins , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Neurons/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics
9.
J Mol Cell Cardiol ; 92: 63-74, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26826378

ABSTRACT

Calreticulin deficiency causes myocardial developmental defects that culminate in an embryonic lethal phenotype. Recent studies have linked loss of this calcium binding chaperone to failure in myofibrillogenesis through an as yet undefined mechanism. The purpose of the present study was to identify cellular processes corrupted by calreticulin deficiency that precipitate dysregulation of cardiac myofibrillogenesis related to acquisition of cardiac phenotype. In an embryonic stem cell knockout model, calreticulin deficit (crt(-/-)) compromised nucleocytoplasmic transport of nuclear localization signal-dependent and independent pathways, disrupting nuclear import of the cardiac transcription factor MEF2C. The expression of nucleoporins and associated nuclear transport proteins in derived crt(-/-) cardiomyocytes revealed an abnormal nuclear pore complex (NPC) configuration. Altered protein content in crt(-/-) cells resulted in remodeled NPC architecture that caused decreased pore diameter and diminished probability of central channel occupancy versus wild type counterparts. Ionophore treatment of impaired calcium handling in crt(-/-) cells corrected nuclear pore microarchitecture and rescued nuclear import resulting in normalized myofibrillogenesis. Thus, calreticulin deficiency alters nuclear pore function and structure, impeding myofibrillogenesis in nascent cardiomyocytes through a calcium dependent mechanism. This essential role of calreticulin in nucleocytoplasmic communication competency ties its regulatory action with proficiency of cardiac myofibrillogenesis essential for proper cardiac development.


Subject(s)
Calreticulin/genetics , Cardiomyopathies/genetics , Muscle Development/genetics , Nuclear Pore/genetics , Active Transport, Cell Nucleus/genetics , Animals , Calcium/metabolism , Calcium Signaling/genetics , Calreticulin/deficiency , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Differentiation/genetics , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Gene Knockout Techniques , Humans , MEF2 Transcription Factors/genetics , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , Nuclear Pore/metabolism , Nuclear Pore/ultrastructure
10.
J Mol Recognit ; 26(12): 679-88, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24277613

ABSTRACT

Acetyl-CoA carboxylase 2 (ACC2) is an isoform of ACC functioning as a negative regulator of fatty acid ß-oxidation. Spot14, a thyroid hormone responsive protein, and Mig12, a Spot14 paralog, have recently been identified as regulators of fatty acid synthesis targeting ACC1, a distinctive subtype of ACC. Here, we examined whether Spot14/Mig12 modulates ACC2. Nanoscale protein topography mapped putative protein-protein interactions between purified human Spot14/Mig12 and ACC2, validated by functional assays. Human ACC2 displayed consistent enzymatic activity, and homogeneous particle distribution was probed by atomic force microscopy. Citrate-induced polymerization and enzymatic activity of ACC2 were restrained by the addition of the recombinant Spot14/Mig12 heterocomplex but only partially by the oligo-heterocomplex, demonstrating that the heterocomplex is a designated metabolic inhibitor of human ACC2. Moreover, Spot14/Mig12 demonstrated a sequestering role preventing an initial ACC2 nucleation step during filamentous polymer formation. Thus, the Spot14/Mig12 heterocomplex controls human ACC2 polymerization and catalytic function, emerging as a previously unrecognized molecular regulator in catalytic lipid metabolism.


Subject(s)
Acetyl-CoA Carboxylase/chemistry , Acetyl-CoA Carboxylase/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Catalysis , Fatty Acids/metabolism , Humans , Microscopy, Atomic Force , Oxidation-Reduction , Protein Binding
11.
Stem Cells ; 28(7): 1281-91, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20506533

ABSTRACT

Genomic perturbations that challenge normal signaling at the pluripotent stage may trigger unforeseen ontogenic aberrancies. Anticipatory systems biology identification of transcriptome landscapes that underlie latent phenotypes would offer molecular diagnosis before the onset of symptoms. The purpose of this study was to assess the impact of calreticulin-deficient embryonic stem cell transcriptomes on molecular functions and physiological systems. Bioinformatic surveillance of calreticulin-null stem cells, a monogenic insult model, diagnosed a disruption in transcriptome dynamics, which re-prioritized essential cellular functions. Calreticulin-calibrated signaling axes were uncovered, and network-wide cartography of undifferentiated stem cell transcripts suggested cardiac manifestations. Calreticulin-deficient stem cell-derived cardiac cells verified disorganized sarcomerogenesis, mitochondrial paucity, and cytoarchitectural aberrations to validate calreticulin-dependent network forecasts. Furthermore, magnetic resonance imaging and histopathology detected a ventricular septal defect, revealing organogenic manifestation of calreticulin deletion. Thus, bioinformatic deciphering of a primordial calreticulin-deficient transcriptome decoded at the pluripotent stem cell stage a reconfigured multifunctional molecular registry to anticipate predifferentiation susceptibility toward abnormal cardiophenotype.


Subject(s)
Calreticulin/metabolism , Embryonic Stem Cells/metabolism , Myocardium/metabolism , Animals , Calreticulin/deficiency , Cell Death , Cell Differentiation , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Gene Expression Profiling , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Myocardium/cytology , Phenotype
12.
Nat Cell Biol ; 10(7): 825-36, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18516091

ABSTRACT

Expression of p16(Ink4a) and p19(Arf) increases with age in both rodent and human tissues. However, whether these tumour suppressors are effectors of ageing remains unclear, mainly because knockout mice lacking p16(Ink4a) or p19(Arf) die early of tumours. Here, we show that skeletal muscle and fat, two tissues that develop early ageing-associated phenotypes in response to BubR1 insufficiency, have high levels of p16(Ink4a) and p19(Arf). Inactivation of p16(Ink4a) in BubR1-insufficient mice attenuates both cellular senescence and premature ageing in these tissues. Conversely, p19(Arf) inactivation exacerbates senescence and ageing in BubR1 mutant mice. Thus, we identify BubR1 insufficiency as a trigger for activation of the Cdkn2a locus in certain mouse tissues, and demonstrate that p16(Ink4a) is an effector and p19(Arf) an attenuator of senescence and ageing in these tissues.


Subject(s)
Aging/physiology , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Protein Serine-Threonine Kinases/metabolism , Adipose Tissue/anatomy & histology , Adipose Tissue/metabolism , Animals , Biomarkers/metabolism , Cell Cycle Proteins , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Eye/anatomy & histology , Eye/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Genotype , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neoplasms/metabolism , Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Survival Rate , Tissue Distribution
13.
Stem Cells ; 26(2): 387-400, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17991915

ABSTRACT

In the developing embryo, instructive guidance from the ventral endoderm secures cardiac program induction within the anterolateral mesoderm. Endoderm-guided cardiogenesis, however, has yet to be resolved at the proteome level. Here, through cardiopoietic priming of the endoderm with the reprogramming cytokine tumor necrosis factor alpha (TNFalpha), candidate effectors of embryonic stem cell cardiac differentiation were delineated by comparative proteomics. Differential two-dimensional gel electrophoretic mapping revealed that more than 75% of protein species increased >1.5-fold in the TNFalpha-primed versus unprimed endodermal secretome. Protein spot identification by linear ion trap quadrupole (LTQ) tandem mass spectrometry (MS/MS) and validation by shotgun LTQ-Fourier transform MS/MS following multidimensional chromatography mapped 99 unique proteins from 153 spot assignments. A definitive set of 48 secretome proteins was deduced by iterative bioinformatic screening using algorithms for detection of canonical and noncanonical indices of secretion. Protein-protein interaction analysis, in conjunction with respective expression level changes, revealed a nonstochastic TNFalpha-centric secretome network with a scale-free hierarchical architecture. Cardiovascular development was the primary developmental function of the resolved TNFalpha-anchored network. Functional cooperativity of the derived cardioinductive network was validated through direct application of the TNFalpha-primed secretome on embryonic stem cells, potentiating cardiac commitment and sarcomerogenesis. Conversely, inhibition of primary network hubs negated the procardiogenic effects of TNFalpha priming. Thus, proteomic cartography establishes a systems biology framework for the endodermal secretome network guiding stem cell cardiopoiesis.


Subject(s)
Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fetal Heart/cytology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line , Embryonic Stem Cells/cytology , Endoderm/cytology , Endoderm/drug effects , Endoderm/metabolism , Fetal Heart/drug effects , Fetal Heart/embryology , Fetal Heart/metabolism , Mice , Protein Interaction Mapping , Proteome/isolation & purification , Proteomics , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
14.
J Exp Med ; 204(2): 405-20, 2007 Feb 19.
Article in English | MEDLINE | ID: mdl-17283208

ABSTRACT

Embryonic stem cells have the distinct potential for tissue regeneration, including cardiac repair. Their propensity for multilineage differentiation carries, however, the liability of neoplastic growth, impeding therapeutic application. Here, the tumorigenic threat associated with embryonic stem cell transplantation was suppressed by cardiac-restricted transgenic expression of the reprogramming cytokine TNF-alpha, enhancing the cardiogenic competence of recipient heart. The in vivo aptitude of TNF-alpha to promote cardiac differentiation was recapitulated in embryoid bodies in vitro. The procardiogenic action required an intact endoderm and was mediated by secreted cardio-inductive signals. Resolved TNF-alpha-induced endoderm-derived factors, combined in a cocktail, secured guided differentiation of embryonic stem cells in monolayers produce cardiac progenitors termed cardiopoietic cells. Characterized by a down-regulation of oncogenic markers, up-regulation, and nuclear translocation of cardiac transcription factors, this predetermined population yielded functional cardiomyocyte progeny. Recruited cardiopoietic cells delivered in infarcted hearts generated cardiomyocytes that proliferated into scar tissue, integrating with host myocardium for tumor-free repair. Thus, cardiopoietic programming establishes a strategy to hone stem cell pluripotency, offering a tumor-resistant approach for regeneration.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Heart/physiology , Myocytes, Cardiac/cytology , Regeneration/physiology , Stem Cell Transplantation/methods , Tumor Necrosis Factor-alpha/metabolism , Animals , DNA Primers , Embryonic Stem Cells/transplantation , Gene Expression Regulation , Mice , Mice, Transgenic , Microarray Analysis , Neoplasms/prevention & control , Transcription Factors/metabolism
15.
J Biol Chem ; 282(14): 10465-71, 2007 Apr 06.
Article in English | MEDLINE | ID: mdl-17289667

ABSTRACT

Green fluorescent proteins (GFP) are widely used in biology for tracking purposes. Although expression of GFP is considered to be innocuous for the cells, deleterious effects have been reported. We recently demonstrated that expression of eGFP in muscle impairs its contractile properties (Agbulut, O., Coirault, C., Niederlander, N., Huet, A., Vicart, P., Hagege, A., Puceat, M., and Menasche, P. (2006) Nat. Meth. 3, 331). This prompted us to identify the molecular mechanisms linking eGFP expression to contractile dysfunction and, particularly, to test the hypothesis that eGFP could inhibit actin-myosin interactions. Therefore, we assessed the cellular, mechanical, enzymatic, biochemical, and structural properties of myosin in the presence of eGFP and F-actin. In vitro motility assays, the maximum actin-activated ATPase rate (V(max)) and the associated constant of myosin for actin (K(m)) were determined at 1:0.5, 1:1, and 1:3 myosin:eGFP molar ratios. At a myosin:eGFP ratio of 1:0.5, there was a nearly 10-fold elevation of K(m). As eGFP concentration increased relative to myosin, the percentage of moving filaments, the myosin-based velocity, and V(max) significantly decreased compared with controls. Moreover, myosin co-precipitated with eGFP. Crystal structures of myosin, actin, and GFP indicated that GFP and actin exhibited similar electrostatic surface patterns and the ClusPro docking model showed that GFP bound preferentially to the myosin head and especially to the actin-binding site. In conclusion, our data demonstrate that expression of eGFP in muscle resulted in the binding of eGFP to myosin, thereby disturbing the actin-myosin interaction and in turn the contractile function of the transduced cells. This potential adverse effect of eGFP should be kept in mind when using this marker to track cells following transplantation.


Subject(s)
Actins/chemistry , Green Fluorescent Proteins/chemistry , Models, Molecular , Myosins/chemistry , Actins/antagonists & inhibitors , Actins/metabolism , Animals , Animals, Genetically Modified , Animals, Newborn , Binding Sites , Crystallography, X-Ray , Genetic Markers , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Kinetics , Muscle Contraction/genetics , Myosins/antagonists & inhibitors , Myosins/metabolism , Protein Binding/genetics , Protein Structure, Quaternary , Rats , Rats, Sprague-Dawley
16.
Nat Clin Pract Cardiovasc Med ; 4 Suppl 1: S68-76, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17230218

ABSTRACT

Nuclear transport of transcription factors is a critical step in stem cell commitment to a tissue-specific lineage. While it is recognized that nuclear pores are gatekeepers of nucleocytoplasmic exchange, it is unknown how the nuclear transport machinery becomes competent to support genetic reprogramming and cell differentiation. Here, we report the dynamics of nuclear transport factor expression and nuclear pore microanatomy during cardiac differentiation of embryonic stem cells. Cardiac progeny derived from pluripotent stem cells displayed a distinct proteomic profile characterized by the emergence of cardiac-specific proteins. This profile correlated with the nuclear translocation of cardiac transcription factors. The nuclear transport genes, including nucleoporins, importins, exportins, transportins, and Ran-related factors, were globally downregulated at the genomic level, streamlining the differentiation program underlying stem cell-derived cardiogenesis. Establishment of the cardiac molecular phenotype was associated with an increased density of nuclear pores spanning the nuclear envelope. At nanoscale resolution, individual nuclear pores exhibited conformational changes resulting in the expansion of the pore diameter and an augmented probability of conduit occupancy. Thus, embryonic stem cells undergo adaptive remodeling of the nuclear transport infrastructure associated with nuclear translocation of cardiac transcription factors and execution of the cardiogenic program, underscoring the plasticity of the nucleocytoplasmic trafficking machinery in accommodating differentiation requirements.


Subject(s)
Active Transport, Cell Nucleus/physiology , Embryonic Stem Cells/physiology , Myocytes, Cardiac/physiology , Animals , Cell Differentiation , Cells, Cultured , Mice , Nuclear Pore/physiology , Phenotype
17.
Cardiovasc Res ; 72(1): 175-83, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16934240

ABSTRACT

OBJECTIVES: To assess the functional effects of multipotent adult progenitor cells (MAPCs) transplanted in a rat model of chronic myocardial infarction. METHODS: Forty-four rats underwent coronary ligation and, 14 days later, were randomly allocated to receive in-scar injections (5 x 10(6) cells/150 microL) of green fluorescent protein (eGFP)-transduced allogeneic MAPCs (n = 25) or culture medium (controls, n = 19). Nine of the MAPC-treated hearts were employed for functional studies while the remaining 16 received cells co-labeled with Resovist and were only used for serial histological assessments. Left ventricular (LV) function was assessed echocardiographically before transplantation and 1 month thereafter in a blinded manner. Immunohistochemistry, electron microscopy and PCR were used to detect grafted cells. All data were compared by nonparametric tests. RESULTS: Baseline ejection fractions (EF, median;[interquartile range]) did not differ significantly among the groups: 30% [0.23;0.37] and 37% [0.32;0.38] in control and rMAPC-transplanted hearts, respectively. One month later, LV function of control hearts was found to have deteriorated, as reflected by a decline in EF to 24% [0.21;0.30], and although EF tended to remain more stable after cell transplantation (37% [0.27;0.41]), the difference between the two groups failed to achieve statistical significance (p = 0.06). While MAPCs could be identified early post-transplant, no evidence of engraftment was further observed at 1 month by immunohistochemistry, electron microscopy or PCR. CONCLUSIONS: In this model, MAPCs did not improve global pump function, and although some of these cells expressed endothelial markers during the early post-transplant period, we could not detect any evidence for differentiation into cardiomyocytes and no engraftment was further identified beyond 2 weeks after cell injections.


Subject(s)
Bone Marrow Cells/cytology , Multipotent Stem Cells/transplantation , Myocardial Infarction/surgery , Myocardium/pathology , Animals , Female , Graft Rejection , Models, Animal , Multipotent Stem Cells/ultrastructure , Myocardial Contraction , Myocardial Infarction/pathology , Myocardium/ultrastructure , Rats , Rats, Sprague-Dawley , Treatment Failure
19.
Eur J Biochem ; 271(22): 4572-81, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15560799

ABSTRACT

Titin is known to interact with actin thin filaments within the I-band region of striated muscle sarcomeres. In this study, we have used a titin fragment of 800 kDa (T800) purified from striated skeletal muscle to measure the effect of this interaction on the functional properties of the actin-myosin complex. MALDI-TOF MS revealed that T800 contains the entire titin PEVK (Pro, Glu, Val, Lys-rich) domain. In the presence of tropomyosin-troponin, T800 increased the sliding velocity (both average and maximum values) of actin filaments on heavy-meromyosin (HMM)-coated surfaces and dramatically decreased the number of stationary filaments. These results were correlated with a 30% reduction in actin-activated HMM ATPase activity and with an inhibition of HMM binding to actin N-terminal residues as shown by chemical cross-linking. At the same time, T800 did not affect the efficiency of the Ca(2+)-controlled on/off switch, nor did it alter the overall binding energetics of HMM to actin, as revealed by cosedimentation experiments. These data are consistent with a competitive effect of PEVK domain-containing T800 on the electrostatic contacts at the actin-HMM interface. They also suggest that titin may participate in the regulation of the active tension generated by the actin-myosin complex.


Subject(s)
Actins/metabolism , Muscle Proteins/metabolism , Myosin Subfragments/metabolism , Protein Kinases/metabolism , Actins/chemistry , Adenosine Triphosphatases/metabolism , Animals , Connectin , Cross-Linking Reagents/pharmacology , Electrophoresis, Polyacrylamide Gel , Ethyldimethylaminopropyl Carbodiimide/pharmacology , Humans , Muscle Proteins/chemistry , Muscle Proteins/pharmacology , Muscle, Skeletal/chemistry , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myosin Subfragments/chemistry , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Protein Binding , Protein Kinases/chemistry , Protein Kinases/pharmacology , Protein Structure, Tertiary , Rabbits , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Tropomyosin/chemistry , Tropomyosin/metabolism , Troponin/chemistry , Troponin/metabolism
20.
Biochem Biophys Res Commun ; 325(4): 1304-11, 2004 Dec 24.
Article in English | MEDLINE | ID: mdl-15555569

ABSTRACT

We have identified a fourth member of the enigma homolog (ENH) family within a pool of human transcripts specifically expressed in skeletal muscle tissue. This new ENH isoform of 215 amino acids is the shorter of the family, it lacks the C-terminal LIM domains present in ENH1 but contains the N-terminal PDZ domain. Northern blot analysis confirmed the muscle specificity of ENH4. Western blot studies of muscle tissues using a non-isoform-specific anti-ENH antibody revealed that ENH4 is present only in skeletal muscle and that there is a specific distribution of ENH members between skeletal and cardiac muscles, which is different in human and mouse. ENH4 was found to co-localize in the sarcomeric Z-band and to interact with alpha-actinin like the other members of the ENH family. Two additional new ENH4 partners of about 34 and 54kDa were also identified. These results bring new lights on the ENH protein family members.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Cells, Cultured , Cytoskeletal Proteins , Genetic Vectors , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins , Mice , Molecular Sequence Data , Molecular Weight , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...