Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Development ; 151(7)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38456551

ABSTRACT

Adhesion between stem cells and their niche provides stable anchorage and signaling cues to sustain properties such as quiescence. Skeletal muscle stem cells (MuSCs) adhere to an adjacent myofiber via cadherin-catenin complexes. Previous studies on N- and M-cadherin in MuSCs revealed that although N-cadherin is required for quiescence, they are collectively dispensable for MuSC niche localization and regenerative activity. Although additional cadherins are expressed at low levels, these findings raise the possibility that cadherins are unnecessary for MuSC anchorage to the niche. To address this question, we conditionally removed from MuSCs ß- and γ-catenin, and, separately, αE- and αT-catenin, factors that are essential for cadherin-dependent adhesion. Catenin-deficient MuSCs break quiescence similarly to N-/M-cadherin-deficient MuSCs, but exit the niche and are depleted. Combined in vivo, ex vivo and single cell RNA-sequencing approaches reveal that MuSC attrition occurs via precocious differentiation, re-entry to the niche and fusion to myofibers. These findings indicate that cadherin-catenin-dependent adhesion is required for anchorage of MuSCs to their niche and for preservation of the stem cell compartment. Furthermore, separable cadherin-regulated functions govern niche localization, quiescence and MuSC maintenance.


Subject(s)
Cadherins , Stem Cell Niche , Stem Cell Niche/genetics , Cadherins/genetics , Cadherins/metabolism , Muscle Fibers, Skeletal/metabolism , Signal Transduction , Catenins/genetics , Catenins/metabolism , Muscle, Skeletal/metabolism , Cell Adhesion/genetics
2.
J Cell Sci ; 136(24)2023 12 15.
Article in English | MEDLINE | ID: mdl-38149870

ABSTRACT

Skeletal muscle stem cells (MuSCs, also called satellite cells) are the source of the robust regenerative capability of this tissue. The hallmark property of MuSCs at homeostasis is quiescence, a reversible state of cell cycle arrest required for long-term preservation of the stem cell population. MuSCs reside between an individual myofiber and an enwrapping basal lamina, defining the immediate MuSC niche. Additional cell types outside the basal lamina, in the interstitial space, also contribute to niche function. Quiescence is actively maintained by multiple niche-derived signals, including adhesion molecules presented from the myofiber surface and basal lamina, as well as soluble signaling factors produced by myofibers and interstitial cell types. In this Cell Science at a Glance article and accompanying poster, we present the most recent information on how niche signals promote MuSC quiescence and provide perspectives for further research.


Subject(s)
Muscle, Skeletal , Satellite Cells, Skeletal Muscle , Stem Cell Niche , Muscle Fibers, Skeletal , Cell Division , Stem Cells/metabolism
3.
Nat Commun ; 13(1): 1626, 2022 03 25.
Article in English | MEDLINE | ID: mdl-35338152

ABSTRACT

The combination of EGF, CHIR99021, A83-01, SB431542, VPA, and Y27632 (EGF/CASVY) facilitates the derivation of trophoblast stem (TS) cells from human blastocysts and first-trimester, but not term, cytotrophoblasts. The mechanism underlying this chemical induction of TS cells remains elusive. Here we demonstrate that the induction efficiency of cytotrophoblast is determined by functional antagonism of the placental transcription factor GCM1 and the stemness regulator ΔNp63α. ΔNp63α reduces GCM1 transcriptional activity, whereas GCM1 inhibits ΔNp63α oligomerization and autoregulation. EGF/CASVY cocktail activates ΔNp63α, thereby partially inhibiting GCM1 activity and reverting term cytotrophoblasts into stem cells. By applying hypoxia condition, we can further reduce GCM1 activity and successfully induce term cytotrophoblasts into TS cells. Consequently, we identify mitochondrial creatine kinase 1 (CKMT1) as a key GCM1 target crucial for syncytiotrophoblast differentiation and reveal decreased CKMT1 expression in preeclampsia. Our study delineates the molecular underpinnings of trophoblast stemness and differentiation and an efficient method to establish TS cells from term placentas.


Subject(s)
Epidermal Growth Factor , Trophoblasts , Cell Differentiation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epidermal Growth Factor/metabolism , Female , Humans , Nuclear Proteins/metabolism , Placenta/metabolism , Pregnancy , Transcription Factors/genetics , Transcription Factors/metabolism , Trophoblasts/metabolism , Tumor Suppressor Proteins
4.
Development ; 148(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34610637

ABSTRACT

Many developmental disorders are thought to arise from an interaction between genetic and environmental risk factors. The Hedgehog (HH) signaling pathway regulates myriad developmental processes, and pathway inhibition is associated with birth defects, including holoprosencephaly (HPE). Cannabinoids are HH pathway inhibitors, but little is known of their effects on HH-dependent processes in mammalian embryos, and their mechanism of action is unclear. We report that the psychoactive cannabinoid Δ9-tetrahydrocannabinol (THC) induces two hallmark HH loss-of-function phenotypes (HPE and ventral neural tube patterning defects) in Cdon mutant mice, which have a subthreshold deficit in HH signaling. THC therefore acts as a 'conditional teratogen', dependent on a complementary but insufficient genetic insult. In vitro findings indicate that THC is a direct inhibitor of the essential HH signal transducer smoothened. The canonical THC receptor, cannabinoid receptor-type 1, is not required for THC to inhibit HH signaling. Cannabis consumption during pregnancy may contribute to a combination of risk factors underlying specific developmental disorders. These findings therefore have significant public health relevance.


Subject(s)
Body Patterning/drug effects , Cannabinoid Receptor Agonists/toxicity , Dronabinol/toxicity , Holoprosencephaly/chemically induced , Smoothened Receptor/metabolism , Teratogens/toxicity , Animals , Cannabinoid Receptor Agonists/pharmacology , Cell Adhesion Molecules/genetics , Cells, Cultured , Dronabinol/pharmacology , Female , Mice , Mice, Inbred C57BL , Neural Tube/drug effects , Neural Tube/embryology , Neural Tube/metabolism , Signal Transduction/drug effects , Teratogens/pharmacology
5.
Front Cell Dev Biol ; 9: 795194, 2021.
Article in English | MEDLINE | ID: mdl-35004690

ABSTRACT

Many common developmental disorders are thought to arise from a complex set of genetic and environmental risk factors. These factors interact with each other to affect the strength and duration of key developmental signaling pathways, thereby increasing the possibility that they fail to achieve the thresholds required for normal embryonic patterning. One such disorder, holoprosencephaly (HPE), serves as a useful model system in understanding various forms of multifactorial etiology. Genomic analysis of HPE cases, epidemiology, and mechanistic studies of animal models have illuminated multiple potential ways that risk factors interact to produce adverse developmental outcomes. Among these are: 1) interactions between driver and modifier genes; 2) oligogenic inheritance, wherein each parent provides predisposing variants in one or multiple distinct loci; 3) interactions between genetic susceptibilities and environmental risk factors that may be insufficient on their own; and 4) interactions of multiple genetic variants with multiple non-genetic risk factors. These studies combine to provide concepts that illuminate HPE and are also applicable to additional disorders with complex etiology, including neural tube defects, congenital heart defects, and oro-facial clefting.

6.
Sci Rep ; 9(1): 10098, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31417113

ABSTRACT

Iron granules containing superparamagnetic magnetite act as magnetoreceptor for magnetoreception in honey bees. Biomineralization of iron granules occurs in the iron deposition vesicles of trophocytes and requires the participation of actin, myosin, ferritin2, and ATP synthase. The mechanism of magnetoreception in honey bees can be explored by suppressing the formation of iron granules. Toward this goal, we injected double-stranded RNA of ferritin2 and ferritin1 into newly emerged worker honey bees to knock down these genes via RNA interference. We confirmed that mRNA and protein production of the ferritins was inhibited, leading to immature iron granules. Downregulating ferritin2 and ferritin1, moreover, leads to different deposition morphology of 7.5-nm diameter iron particles, indicating that the two genes play different roles in the formation of iron granules in worker honey bees.


Subject(s)
Adipocytes/metabolism , Bees/physiology , Behavior, Animal/physiology , Ferritins/genetics , Ferritins/metabolism , Iron/metabolism , RNA Interference , Animals , Ferrosoferric Oxide/metabolism , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , RNA, Double-Stranded/administration & dosage , RNA, Double-Stranded/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
7.
FASEB J ; 33(1): 314-326, 2019 01.
Article in English | MEDLINE | ID: mdl-29979633

ABSTRACT

Migration of placental extravillous trophoblast (EVT) cells into uterine decidua facilitates the establishment of blood circulation between mother and fetus and is modulated by EVT-decidual cell interaction. Poor or excessive EVT migration is associated with pregnancy complications such as preeclampsia or placenta accreta. Glial cells missing 1 (GCM1) transcription factor is essential for placental development, and decreased GCM1 activity is detected in preeclampsia. To study whether GCM1 regulates trophoblast cell migration, here we showed that GCM1 promotes BeWo and JAR trophoblast cell migration through a novel target gene, WNT10B. Moreover, WNT10B signaling stimulated cytoskeletal remodeling via Rac1 and frizzled 7 (FZD7) was identified as the cognate receptor for WNT10B to up-regulate cell migration. We further showed that secreted frizzled-related protein 3 (SFRP3) is expressed in uterine decidual cells by immunohistochemistry and that SFRP3 expression in telomerase-transformed human endometrial stromal cells (T-HESCs) is elevated under decidualization stimuli and further enhanced by bone morphogenetic protein 2 via SMAD1. SFRP3 blocked the interaction between FZD7 and WNT10B to decrease BeWo cell migration, which corroborated the elevated BeWo cell migration when cocultured with decidualized and SFRP3-knockdown T-HESC monolayer. Our results suggest that GCM1 up-regulates EVT cell migration through WNT10B and FZD7, which is negatively modulated by decidual SFRP3.-Wang, L.-J., Lo, H.-F., Lin, C.-F., Ng, P.-S., Wu, Y.-H., Lee, Y.-S., Cheong, M.-L., Chen, H. SFRP3 negatively regulates placental extravillous trophoblast cell migration mediated by the GCM1-WNT10B-FZD7 axis.


Subject(s)
Cell Movement , Frizzled Receptors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Placenta/physiology , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Trophoblasts/physiology , Wnt Proteins/metabolism , Cells, Cultured , DNA-Binding Proteins , Decidua/cytology , Decidua/physiology , Endometrium/cytology , Endometrium/physiology , Female , Frizzled Receptors/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Neuroglia/cytology , Neuroglia/physiology , Nuclear Proteins/genetics , Placenta/cytology , Pregnancy , Proto-Oncogene Proteins/genetics , Stromal Cells/cytology , Stromal Cells/physiology , Transcription Factors/genetics , Trophoblasts/cytology , Wnt Proteins/genetics
8.
J Pathol ; 241(3): 324-336, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27917469

ABSTRACT

The transcription factor glial cells missing 1 (GCM1) regulates trophoblast differentiation and function during placentation. Decreased GCM1 expression is associated with pre-eclampsia, suggesting that abnormal expression of GCM1 target genes may contribute to the pathogenesis of pregnancy complications. Here we identified a novel GCM1 target gene, synapse defective 1 (SYDE1), which encodes a RhoGAP that is highly expressed in human placenta, and demonstrated that SYDE1 promotes cytoskeletal remodelling and cell migration and invasion. Importantly, genetic ablation of murine Syde1 results in small fetuses and placentas with aberrant phenotypes in the placental-yolk sac barrier, maternal-trophoblast interface, and placental vascularization. Microarray analysis revealed altered expression of renin-1, angiotensin I converting enzyme 2, angiotensin II type 1a receptor, and membrane metalloendopeptidase of the renin-angiotensin system in Syde1-knockout placenta, which may compensate for the vascular defects to maintain normal blood pressure. As pregnancy proceeds, growth restriction of the Syde1-/- fetuses and placentas continues, with elevated expression of the Syde1 homologue Syde2 in placenta. Syde2 may compensate for the loss of Syde1 function because SYDE2, but not the GAP-dead SYDE2 mutant, reverses migration and invasion activities of SYDE1-knockdown JAR trophoblast cells. Clinically, we further detected decreased SYDE1 expression in preterm and term IUGR placentas compared with gestational age-matched controls. Our study suggests a novel mechanism for GCM1 and SYDE1 in regulation of trophoblast cell migration and invasion during placental development and that decreased SYDE1 expression is associated with IUGR. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cell Differentiation/genetics , Cell Movement/genetics , GTPase-Activating Proteins/genetics , Membrane Proteins/genetics , Placenta/metabolism , Placentation/genetics , Animals , DNA-Binding Proteins , Female , Humans , Mice , Nuclear Proteins/genetics , Pregnancy , Renin-Angiotensin System , Transcription Factors/genetics , Transcription Factors/metabolism , Trophoblasts/cytology
9.
Mol Cell Biol ; 36(1): 197-209, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26503785

ABSTRACT

Human chorionic gonadotropin (hCG) is composed of a common α subunit and a placenta-specific ß subunit. Importantly, hCG is highly expressed in the differentiated and multinucleated syncytiotrophoblast, which is formed via trophoblast cell fusion and stimulated by cyclic AMP (cAMP). Although the ubiquitous activating protein 2 (AP2) transcription factors TFAP2A and TFAP2C may regulate hCGß expression, it remains unclear how cAMP stimulates placenta-specific hCGß gene expression and trophoblastic differentiation. Here we demonstrated that the placental transcription factor glial cells missing 1 (GCM1) binds to a highly conserved promoter region in all six hCGß paralogues by chromatin immunoprecipitation-on-chip (ChIP-chip) analyses. We further showed that cAMP stimulates GCM1 and the CBP coactivator to activate the hCGß promoter through a GCM1-binding site (GBS1), which also constitutes a previously identified AP2 site. Given that TFAP2C may compete with GCM1 for GBS1, cAMP enhances the association between the hCGß promoter and GCM1 but not TFAP2C. Indeed, the hCG-cAMP-protein kinase A (PKA) signaling pathway also stimulates Ser269 and Ser275 phosphorylation of GCM1, which recruits CBP to mediate GCM1 acetylation and stabilization. Consequently, hCG stimulates the expression of GCM1 target genes, including the fusogenic protein syncytin-1, to promote placental cell fusion. Our study reveals a positive feedback loop between GCM1 and hCG regulating placental hCGß expression and cell differentiation.


Subject(s)
Cell Differentiation/physiology , Chorionic Gonadotropin, beta Subunit, Human/metabolism , Chorionic Gonadotropin/metabolism , Neuropeptides/metabolism , Nuclear Proteins/metabolism , Placenta/metabolism , Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Line , Chorionic Gonadotropin/genetics , Chorionic Gonadotropin, beta Subunit, Human/genetics , DNA-Binding Proteins , Female , Humans , Neuroglia/metabolism , Neuropeptides/genetics , Nuclear Proteins/genetics , Pregnancy , Promoter Regions, Genetic/genetics , Transcription Factors/genetics
10.
J Biol Chem ; 289(33): 22958-22968, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25002585

ABSTRACT

Human trophoblast invasion of decidualized endometrium is essential for placentation and is tightly regulated and involves trophoblast-decidual cell interaction. High temperature requirement A4 (HtrA4) is a secreted serine protease highly expressed in the invasive extravillous trophoblasts that invade decidua. In contrast, both HtrA1 and HtrA3 have been shown to inhibit trophoblast invasion. Here we provide evidence that decidua-secreted HtrA1 and HtrA3 antagonize HtrA4-mediated trophoblast invasion. We demonstrated that HtrA1 and HtrA3 interact with and degrade HtrA4 and thereby inhibit trophoblast-like JAR cell invasion. Specifically, HtrA1 and HtrA3 expression is up-regulated under decidualization conditions in endometrial stromal and epithelial cells, T-HESCs and Ishikawa cells, respectively. Conditioned media from these two cell lines after decidualization treatment suppress HtrA4-expressing JAR cell invasion in an HtrA1- or HtrA3-dependent manner. Co-culture of the HtrA4-expressing JAR cells with decidualization stimuli-treated T-HESC or Ishikawa monolayer also impairs JAR cell invasion, which can be reversed by HtrA1 or HtrA3 knockdown, supporting that HtrA1 and HtrA3 are crucial for trophoblast-decidual cell interaction in the control of trophoblast invasion. Our study reveals a novel regulatory mechanism of trophoblast invasion through physical and functional interaction between HtrA family members.


Subject(s)
Serine Endopeptidases/metabolism , Serine Proteases/metabolism , Trophoblasts/enzymology , Female , HEK293 Cells , High-Temperature Requirement A Serine Peptidase 1 , Humans , Trophoblasts/cytology
11.
Biochem J ; 453(2): 201-8, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23651062

ABSTRACT

GCM1 (glial cell missing 1) is a short-lived transcription factor essential for placental development. The F-box protein, FBW2 (F-box and WD-repeat domain-containing 2), which contains five WD (tryptophan-aspartate) repeats, recognizes GCM1 and mediates its ubiquitination via the SCFFBW2 E3 ligase complex. Although the interaction between GCM1 and FBW2 is facilitated by GCM1 phosphorylation, it is possible that this interaction might be regulated by additional cellular factors. In the present study, we perform tandem-affinity purification coupled with MS analysis identifying RACK1 (receptor for activated C-kinase 1) as an FBW2-interacting protein. RACK1 is a multifaceted scaffold protein containing seven WD repeats. We demonstrate that the WD repeats in both RACK1 and FBW2 are required for the interaction of RACK1 and FBW2. Furthermore, RACK1 competes with GCM1 for FBW2 and thereby prevents GCM1 ubiquitination, which is also supported by the observation that GCM1 is destabilized in RACK1-knockdown BeWo placental cells. Importantly, RACK1 knockdown leads to decreased expression of the GCM1 target gene HTRA4 (high-temperature requirement protein A4), which encodes a serine protease crucial for cell migration and invasion. As a result, migration and invasion activities are down-regulated in RACK1-knockdown BeWo cells. The present study reveals a novel function for RACK1 to regulate GCM1 activity and placental cell migration and invasion.


Subject(s)
F-Box Proteins/metabolism , GTP-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Placenta/metabolism , Receptors, Cell Surface/metabolism , Transcription Factors/metabolism , Up-Regulation , Base Sequence , Cell Line , Cell Movement , DNA Primers , DNA-Binding Proteins , Female , Humans , Placenta/cytology , Pregnancy , Real-Time Polymerase Chain Reaction , Receptors for Activated C Kinase , Ubiquitination
12.
FASEB J ; 27(7): 2818-28, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23580611

ABSTRACT

Glial cell missing 1 (GCM1) transcription factor regulates placental cell fusion into the syncytiotrophoblast. Caspase-14 is proteolytically activated to mediate filaggrin processing during keratinocyte differentiation. Interestingly, altered expression of nonactivated caspase-14 proenzyme is associated with tumorigenesis and diabetic retinopathy, suggesting that caspase-14 may perform physiological functions independently of its protease activity. Here, we performed tandem affinity purification coupled with mass spectrometry analysis to identify caspase-14 proenzyme as a GCM1-interacting protein that suppresses GCM1 activity and syncytiotrophoblast differentiation. Immunohistochemistry revealed that caspase-14 and GCM1 colocalize to placental cytotrophoblast cells at 8 wk of gestation and syncytiotrophoblast layer at term. Further, we demonstrated that caspase-14 mRNA level is decreased by 40% in placental BeWo cells treated with forskolin (FSK). To the contrary, stimulation of GCM1-regulated placental cell fusion and human chorionic gonadotropin ß (hCGß) expression by FSK is enhanced by caspase-14 knockdown. Indeed, GCM1 protein level is increased by 40% in the caspase-14-knockdown BeWo cells. Because GCM1 is stabilized by acetylation, we subsequently showed that caspase-14 impedes the interaction between GCM1 and cAMP response element-binding protein (CREB)-binding protein (CBP) to suppress CBP-mediated acetylation and transcriptional coactivation of GCM1. Therefore, caspase-14 can suppress placental cell differentiation through down-regulation of GCM1 activity.


Subject(s)
Caspase 14/genetics , Cell Differentiation/genetics , Nuclear Proteins/genetics , Placenta/metabolism , Transcription Factors/genetics , Acetylation , CREB-Binding Protein/metabolism , Caspase 14/metabolism , Cell Line, Tumor , Cells, Cultured , Colforsin/pharmacology , DNA-Binding Proteins , Female , Filaggrin Proteins , Gene Expression Regulation, Developmental/drug effects , HEK293 Cells , Humans , Immunoblotting , Immunohistochemistry , Microscopy, Fluorescence , Nuclear Proteins/metabolism , Placenta/cytology , Placentation , Pregnancy , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism
13.
Cardiovasc Res ; 75(3): 575-83, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17499231

ABSTRACT

OBJECTIVE: Caveolin-1 and ATP-binding cassette transporter A1 (ABCA1) are proteins that are involved in cellular cholesterol efflux. In this study, we analyzed the relationships between caveolin-1 and ABCA1 on high-density lipoprotein (HDL)-mediated cholesterol efflux in rat aortic endothelial cells. METHODS AND RESULTS: Overexpression of caveolin-1 by transfection with caveolin-1 cDNA in aortic endothelial cells up-regulated ABCA1 expression and enhanced cholesterol efflux. Suppression of caveolin-1 by siRNA decreased ABCA1 expression and reduced cholesterol efflux. The number of caveolae increased after transfection with caveolin-1 into cells. Immunoprecipitation assays revealed a molecular interaction between caveolin-1 and ABCA1 in the plasma membrane and in the cytoplasm after HDL incubation. Immunoelectron microscopy demonstrated that caveolin-1 colocalized with ABCA1 in the caveolae and in the cytoplasmic vesicles; it was also found that caveolin-1 and ABCA1 colocalized with cellular cholesterol by immunofluorescence microscopy. Blocking of intracellular lipid transport by inhibitors disrupted the interaction between caveolin-1 and ABCA1 and reduced cholesterol to methyl-beta-cyclodextrin and HDL. CONCLUSIONS: The molecular interaction between caveolin-1 and ABCA1 is associated with the HDL-mediated cholesterol efflux pathway in aortic endothelial cells.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Caveolin 1/metabolism , Cholesterol/metabolism , Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/analysis , Animals , Aorta , Biological Transport , Caveolin 1/analysis , Cell Membrane/chemistry , Cell Membrane/metabolism , Cells, Cultured , Cholesterol/pharmacology , Cytoplasm/chemistry , Cytoplasm/metabolism , Endothelial Cells/chemistry , Gene Expression Regulation , Immunoblotting/methods , Lipoproteins, HDL/pharmacology , Microscopy, Fluorescence , Microscopy, Immunoelectron , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transduction, Genetic/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...