Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Curr Top Dev Biol ; 153: 145-180, 2023.
Article in English | MEDLINE | ID: mdl-36967193

ABSTRACT

Understanding how the body plan is established during embryogenesis remains a fundamental biological question. The Wnt/ß-catenin signaling pathway plays a crucial and highly conserved role in body plan formation, functioning to polarize the primary anterior-posterior (AP) or head-to-tail body axis in most metazoans. In this chapter, we focus on the roles that the mammalian Wnt/ß-catenin pathway plays to prepare the pluripotent epiblast for gastrulation, and to elicit the emergence of multipotent axial progenitors from the caudal epiblast. Interactions between Wnt and retinoic acid (RA), another powerful family of developmental signaling molecules, in axial progenitors will also be discussed. Gastrulation movements and somitogenesis result in the anterior displacement of the RA source (the rostral somites and lateral plate mesoderm (LPM)), from the posterior Wnt source (the primitive streak (PS)), leading to the establishment of antiparallel gradients of RA and Wnt that control the self-renewal and successive differentiation of neck, trunk and tail progenitors.


Subject(s)
Body Patterning , Wnt Signaling Pathway , Animals , Germ Layers , Mesoderm/metabolism , Somites , Tretinoin , Cell Differentiation , Mammals
2.
Curr Top Dev Biol ; 153: xv-xvi, 2023.
Article in English | MEDLINE | ID: mdl-36967204
3.
Mol Psychiatry ; 28(2): 767-779, 2023 02.
Article in English | MEDLINE | ID: mdl-36203006

ABSTRACT

Opioids are the frontline analgesics for managing various types of pain. Paradoxically, repeated use of opioid analgesics may cause an exacerbated pain state known as opioid-induced hyperalgesia (OIH), which significantly contributes to dose escalation and consequently opioid overdose. Neuronal malplasticity in pain circuits has been the predominant proposed mechanism of OIH expression. Although glial cells are known to become reactive in OIH animal models, their biological contribution to OIH remains to be defined and their activation mechanism remains to be elucidated. Here, we show that reactive astrocytes (a.k.a. astrogliosis) are critical for OIH development in both male and female mice. Genetic reduction of astrogliosis inhibited the expression of OIH and morphine-induced neural circuit polarization (NCP) in the spinal dorsal horn (SDH). We found that Wnt5a is a neuron-to-astrocyte signal that is required for morphine-induced astrogliosis. Conditional knock-out of Wnt5a in neurons or its co-receptor ROR2 in astrocytes blocked not only morphine-induced astrogliosis but also OIH and NCP. Furthermore, we showed that the Wnt5a-ROR2 signaling-dependent astrogliosis contributes to OIH via inflammasome-regulated IL-1ß. Our results reveal an important role of morphine-induced astrogliosis in OIH pathogenesis and elucidate a neuron-to-astrocyte intercellular Wnt signaling pathway that controls the astrogliosis.


Subject(s)
Analgesics, Opioid , Hyperalgesia , Animals , Female , Male , Mice , Astrocytes/metabolism , Gliosis , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Hyperalgesia/metabolism , Morphine , Pain , Wnt Signaling Pathway
4.
Blood ; 139(5): 690-703, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34657154

ABSTRACT

The cellular mechanisms required to ensure homeostasis of the hematopoietic niche and the ability of this niche to support hematopoiesis upon stress remain elusive. We here identify Wnt5a in Osterix+ mesenchymal progenitor and stem cells (MSPCs) as a critical factor for niche-dependent hematopoiesis. Mice lacking Wnt5a in MSPCs suffer from stress-related bone marrow (BM) failure and increased mortality. Niche cells devoid of Wnt5a show defective actin stress fiber orientation due to an elevated activity of the small GTPase CDC42. This results in incorrect positioning of autophagosomes and lysosomes, thus reducing autophagy and increasing oxidative stress. In MSPCs from patients from BM failure states which share features of peripheral cytopenia and hypocellular BM, we find similar defects in actin stress fiber orientation, reduced and incorrect colocalization of autophagosomes and lysosomes, and CDC42 activation. Strikingly, a short pharmacological intervention to attenuate elevated CDC42 activation in vivo in mice prevents defective actin-anchored autophagy in MSPCs, salvages hematopoiesis and protects against lethal cytopenia upon stress. In summary, our study identifies Wnt5a as a restriction factor for niche homeostasis by affecting CDC42-regulated actin stress-fiber orientation and autophagy upon stress. Our data further imply a critical role for autophagy in MSPCs for adequate support of hematopoiesis by the niche upon stress and in human diseases characterized by peripheral cytopenias and hypocellular BM.


Subject(s)
Autophagy , Bone Marrow Failure Disorders/metabolism , Hematopoiesis , Mesenchymal Stem Cells/cytology , Animals , Cells, Cultured , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mice , Oxidative Stress , Wnt-5a Protein/metabolism
5.
PLoS One ; 15(4): e0232025, 2020.
Article in English | MEDLINE | ID: mdl-32353019

ABSTRACT

The actin cytoskeleton plays a central role in establishing cell polarity and shape during embryonic morphogenesis. Daam1, a member of the Formin family of actin cytoskeleton regulators, is a Dvl2-binding protein that functions in the Wnt/Planar Cell Polarity (PCP) pathway. To examine the role of the Daam proteins in mammalian development, we generated Daam-deficient mice by gene targeting and found that Daam1, but not Daam2, is necessary for fetal survival. Embryonic development of Daam1 mutants was delayed most likely due to functional defects in the labyrinthine layer of the placenta. Examination of Daam2 and Daam1/2 double mutants revealed that Daam1 and Daam2 are functionally redundant during placental development. Of note, neural tube closure defects (NTD), which are observed in several mammalian PCP mutants, are not observed in Wnt5a or Daam1 single mutants, but arise in Daam1;Wnt5a double mutants. These findings demonstrate a unique function for Daam genes in placental development and are consistent with a role for Daam1 in the Wnt/PCP pathway in mammals.


Subject(s)
Microfilament Proteins/genetics , Placentation/genetics , rho GTP-Binding Proteins/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carrier Proteins/metabolism , Cell Polarity , Cytoskeleton/metabolism , Embryonic Development , Female , Formins/genetics , Formins/metabolism , Gene Expression Regulation, Developmental/genetics , Male , Mice/embryology , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Placenta/embryology , Pregnancy , Wnt Signaling Pathway , rho GTP-Binding Proteins/metabolism
6.
Development ; 147(8)2020 04 12.
Article in English | MEDLINE | ID: mdl-32156757

ABSTRACT

Despite the importance of Wnt signaling for adult intestinal stem cell homeostasis and colorectal cancer, relatively little is known about its role in colon formation during embryogenesis. The development of the colon starts with the formation and extension of the hindgut. We show that Wnt3a is expressed in the caudal embryo in a dorsal-ventral (DV) gradient across all three germ layers, including the hindgut. Using genetic and lineage-tracing approaches, we describe novel dorsal and ventral hindgut domains, and show that ventrolateral hindgut cells populate the majority of the colonic epithelium. A Wnt3a-ß-catenin-Sp5/8 pathway, which is active in the dorsal hindgut endoderm, is required for hindgut extension and colon formation. Interestingly, the absence of Wnt activity in the ventral hindgut is crucial for proper hindgut morphogenesis, as ectopic stabilization of ß-catenin in the ventral hindgut via gain- or loss-of-function mutations in Ctnnb1 or Apc, respectively, leads to severe colonic hyperplasia. Thus, the DV Wnt gradient is required to coordinate growth between dorsal and ventral hindgut domains to regulate the extension of the hindgut that leads to colon formation.


Subject(s)
Body Patterning , Colon/embryology , Colon/metabolism , Wnt Signaling Pathway , Wnt3A Protein/metabolism , beta Catenin/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Proliferation , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Mice, Transgenic , Morphogenesis
7.
EMBO Rep ; 21(1): e48469, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31789450

ABSTRACT

Amino acids are essential for cellular metabolism, and it is important to understand how nutrient supply is coordinated with changing energy requirements during embryogenesis. Here, we show that the amino acid transporter Slc7a5/Lat1 is highly expressed in tissues undergoing morphogenesis and that Slc7a5-null mouse embryos have profound neural and limb bud outgrowth defects. Slc7a5-null neural tissue exhibited aberrant mTORC1 activity and cell proliferation; transcriptomics, protein phosphorylation and apoptosis analyses further indicated induction of the integrated stress response as a potential cause of observed defects. The pattern of stress response gene expression induced in Slc7a5-null embryos was also detected at low level in wild-type embryos and identified stress vulnerability specifically in tissues undergoing morphogenesis. The Slc7a5-null phenotype is reminiscent of Wnt pathway mutants, and we show that Wnt/ß-catenin loss inhibits Slc7a5 expression and induces this stress response. Wnt signalling therefore normally supports the metabolic demands of morphogenesis and constrains cellular stress. Moreover, operation in the embryo of the integrated stress response, which is triggered by pathogen-mediated as well as metabolic stress, may provide a mechanistic explanation for a range of developmental defects.


Subject(s)
Large Neutral Amino Acid-Transporter 1 , Wnt Signaling Pathway , Animals , Cell Proliferation/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Morphogenesis
8.
Development ; 145(8)2018 04 13.
Article in English | MEDLINE | ID: mdl-29615464

ABSTRACT

Embryonic morphogenesis of a complex organism requires proper regulation of patterning and directional growth. Planar cell polarity (PCP) signaling is emerging as a crucial evolutionarily conserved mechanism whereby directional information is conveyed. PCP is thought to be established by global cues, and recent studies have revealed an instructive role of a Wnt signaling gradient in epithelial tissues of both invertebrates and vertebrates. However, it remains unclear whether Wnt/PCP signaling is regulated in a coordinated manner with embryonic patterning during morphogenesis. Here, in mouse developing limbs, we find that apical ectoderm ridge-derived Fgfs required for limb patterning regulate PCP along the proximal-distal axis in a Wnt5a-dependent manner. We demonstrate with genetic evidence that the Wnt5a gradient acts as a global cue that is instructive in establishing PCP in the limb mesenchyme, and that Wnt5a also plays a permissive role to allow Fgf signaling to orient PCP. Our results indicate that limb morphogenesis is regulated by coordination of directional growth and patterning through integration of Wnt5a and Fgf signaling.


Subject(s)
Body Patterning/physiology , Cell Polarity/physiology , Fibroblast Growth Factor 4/physiology , Fibroblast Growth Factor 8/physiology , Wnt-5a Protein/physiology , Animals , Body Patterning/genetics , Embryonic Development/genetics , Embryonic Development/physiology , Extremities/embryology , Fibroblast Growth Factor 4/deficiency , Fibroblast Growth Factor 4/genetics , Fibroblast Growth Factor 8/deficiency , Fibroblast Growth Factor 8/genetics , Mesoderm/embryology , Mice , Mice, Knockout , Mice, Transgenic , Signal Transduction , Wnt-5a Protein/deficiency , Wnt-5a Protein/genetics
9.
PLoS One ; 12(8): e0181549, 2017.
Article in English | MEDLINE | ID: mdl-28846685

ABSTRACT

Resident tissue myeloid cells play a role in many aspects of physiology including development of the vascular systems. In the blood vasculature, myeloid cells use VEGFC to promote angiogenesis and can use Wnt ligands to control vascular branching and to promote vascular regression. Here we show that myeloid cells also regulate development of the dermal lymphatic vasculature using Wnt ligands. Using myeloid-specific deletion of the WNT transporter Wntless we show that myeloid Wnt ligands are active at two distinct stages of development of the dermal lymphatics. As lymphatic progenitors are emigrating from the cardinal vein and intersomitic vessels, myeloid Wnt ligands regulate both their numbers and migration distance. Later in lymphatic development, myeloid Wnt ligands regulate proliferation of lymphatic endothelial cells (LEC) and thus control lymphatic vessel caliber. Myeloid-specific deletion of WNT co-receptor Lrp5 or Wnt5a gain-of-function also produce elevated caliber in dermal lymphatic capillaries. These data thus suggest that myeloid cells produce Wnt ligands to regulate lymphatic development and use Wnt pathway co-receptors to regulate the balance of Wnt ligand activity during the macrophage-LEC interaction.


Subject(s)
Dermis/metabolism , Endothelial Cells/metabolism , Lymphangiogenesis/physiology , Lymphatic Vessels/metabolism , Myeloid Cells/metabolism , Wnt Proteins/metabolism , Animals , Cell Proliferation/physiology , Mice , Wnt Signaling Pathway
10.
Dev Cell ; 40(5): 439-452.e4, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28292423

ABSTRACT

Polarization of node cells along the anterior-posterior axis of mouse embryos is responsible for left-right symmetry breaking. How node cells become polarized has remained unknown, however. Wnt5a and Wnt5b are expressed posteriorly relative to the node, whereas genes for Sfrp inhibitors of Wnt signaling are expressed anteriorly. Here we show that polarization of node cells is impaired in Wnt5a-/-Wnt5b-/- and Sfrp mutant embryos, and also in the presence of a uniform distribution of Wnt5a or Sfrp1, suggesting that Wnt5 and Sfrp proteins act as instructive signals in this process. The absence of planar cell polarity (PCP) core proteins Prickle1 and Prickle2 in individual cells or local forced expression of Wnt5a perturbed polarization of neighboring wild-type cells. Our results suggest that opposing gradients of Wnt5a and Wnt5b and of their Sfrp inhibitors, together with intercellular signaling via PCP proteins, polarize node cells along the anterior-posterior axis for breaking of left-right symmetry.


Subject(s)
Body Patterning , Cell Polarity , Signal Transduction , Wnt Proteins/metabolism , Wnt-5a Protein/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Communication , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Models, Biological , Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 113(50): E8079-E8088, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911818

ABSTRACT

Blastocyst implantation is a complex process requiring coordination of a dynamic sequence of embryo-uterine interactions. Blood vessels enter the uterus from the mesometrium, demarcating the uterus into mesometrial (M) and antimesometrial (AM) domains. Implantation occurs along the uterine longitudinal axis within specialized implantation chambers (crypts) that originate within the evaginations directed from the primary lumen toward the AM domain. The morphological orientation of crypts in rodent uteri was recognized more than a century ago, but the mechanism remained unknown. Here we provide evidence that planar cell polarity (PCP) signaling orchestrates directed epithelial evaginations to form crypts for implantation in mice. Uterine deletion of Vang-like protein 2, but not Vang-like protein 1, conferred aberrant PCP signaling, misdirected epithelial evaginations, defective crypt formation, and blastocyst attachment, leading to severely compromised pregnancy outcomes. The study reveals a previously unrecognized role for PCP in executing spatial cues for crypt formation and implantation. Because PCP is an evolutionarily conserved phenomenon, our study is likely to inspire implantation studies of this signaling pathway in humans and other species.


Subject(s)
Cell Polarity/physiology , Embryo Implantation/physiology , Uterus/physiology , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/physiology , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Communication/physiology , Dishevelled Proteins/physiology , Epithelium/anatomy & histology , Epithelium/physiology , Female , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Pregnancy , Pregnancy Outcome , Receptor Tyrosine Kinase-like Orphan Receptors/deficiency , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/physiology , Signal Transduction/physiology , Uterus/anatomy & histology , Wnt-5a Protein/deficiency , Wnt-5a Protein/genetics , Wnt-5a Protein/physiology
12.
Genesis ; 54(9): 497-502, 2016 09.
Article in English | MEDLINE | ID: mdl-27411055

ABSTRACT

Wnt/ß-catenin signals are important regulators of embryonic and adult stem cell self-renewal and differentiation and play causative roles in tumorigenesis. Purified recombinant Wnt3a protein, or Wnt3a-conditioned culture medium, has been widely used to study canonical Wnt signaling in vitro or ex vivo. To study the role of Wnt3a in embryogenesis and cancer models, we developed a Cre recombinase activatable Rosa26(Wnt3a) allele, in which a Wnt3a cDNA was inserted into the Rosa26 locus to allow for conditional, spatiotemporally defined expression of Wnt3a ligand for gain-of-function (GOF) studies in mice. To validate this reagent, we ectopically overexpressed Wnt3a in early embryonic progenitors using the T-Cre transgene. This resulted in up-regulated expression of a ß-catenin/Tcf-Lef reporter and of the universal Wnt/ß-catenin pathway target genes, Axin2 and Sp5. Importantly, T-Cre; Rosa26(Wnt3a) mutants have expanded presomitic mesoderm (PSM) and compromised somitogenesis and closely resemble previously studied T-Cre; Ctnnb1(ex3) (ß-catenin(GOF) ) mutants. These data indicate that the exogenously expressed Wnt3a stimulates the Wnt/ß-catenin signaling pathway, as expected. The Rosa26(Wnt3a) mouse line should prove to be an invaluable tool to study the function of Wnt3a in vivo.


Subject(s)
Gene Targeting/methods , Transgenes , Wnt3A Protein/genetics , Animals , Genes, Reporter , Genetic Vectors/genetics , Integrases/genetics , Mice , Mice, Inbred C57BL , Mutation , Up-Regulation , Wnt3A Protein/metabolism
13.
Mol Cell Biol ; 36(12): 1793-802, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27090637

ABSTRACT

The Wnt/ß-catenin signaling pathway controls embryonic development and adult stem cell maintenance through the regulation of transcription. Failure to downregulate Wnt signaling can result in embryonic malformations and cancer, highlighting the important role of negative regulators of the pathway. The Wnt pathway activates several negative feedback targets, including axin2 and Dkk1, that function at different levels of the signaling cascade; however, none have been identified that directly target active ß-catenin/Tcf1 transcriptional complexes. We show that Zfp703 is a Wnt target gene that inhibits Wnt/ß-catenin activity in Wnt reporter assays and in Wnt-dependent mesoderm differentiation in embryonic stem cells. Zfp703 binds directly to Tcf1 to inhibit ß-catenin/Tcf1 complex formation and does so independently of the Groucho/Tle transcriptional corepressor. We propose that Zfp703 is a novel feedback suppressor of Wnt/ß-catenin signaling that functions by inhibiting the association of ß-catenin with Tcf1 on Wnt response elements in target gene enhancers.


Subject(s)
Embryonic Stem Cells/cytology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Repressor Proteins/metabolism , beta Catenin/metabolism , Animals , Cell Differentiation , Embryonic Development , Embryonic Stem Cells/metabolism , Humans , Mesoderm/cytology , Mice , Wnt Proteins/metabolism , Wnt Signaling Pathway
14.
Proc Natl Acad Sci U S A ; 113(13): 3545-50, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26969725

ABSTRACT

The ancient, highly conserved, Wnt signaling pathway regulates cell fate in all metazoans. We have previously shown that combined null mutations of the specificity protein (Sp) 1/Klf-like zinc-finger transcription factors Sp5 and Sp8 (i.e., Sp5/8) result in an embryonic phenotype identical to that observed when core components of the Wnt/ß-catenin pathway are mutated; however, their role in Wnt signal transduction is unknown. Here, we show in mouse embryos and differentiating embryonic stem cells that Sp5/8 are gene-specific transcriptional coactivators in the Wnt/ß-catenin pathway. Sp5/8 bind directly to GC boxes in Wnt target gene enhancers and to adjacent, or distally positioned, chromatin-bound T-cell factor (Tcf) 1/lymphoid enhancer factor (Lef) 1 to facilitate recruitment of ß-catenin to target gene enhancers. Because Sp5 is itself directly activated by Wnt signals, we propose that Sp5 is a Wnt/ß-catenin pathway-specific transcript on factor that functions in a feed-forward loop to robustly activate select Wnt target genes.


Subject(s)
DNA-Binding Proteins/metabolism , Hepatocyte Nuclear Factor 1-alpha/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/metabolism , Animals , DNA-Binding Proteins/genetics , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Female , Hepatocyte Nuclear Factor 1-alpha/genetics , Lymphoid Enhancer-Binding Factor 1/genetics , Mice , Mice, Transgenic , Pregnancy , Transcription Factors/genetics , Transcriptional Activation , beta Catenin/genetics
15.
Dev Biol ; 412(1): 18-31, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26916252

ABSTRACT

Wnt5a, a non-canonical Wnt ligand critical for outflow tract (OFT) morphogenesis, is expressed specifically in second heart field (SHF) progenitors in the caudal splanchnic mesoderm (SpM) near the inflow tract (IFT). Using a conditional Wnt5a gain of function (GOF) allele and Islet1-Cre, we broadly over-expressed Wnt5a throughout the SHF lineage, including the entire SpM between the IFT and OFT. Wnt5a over-expression in Wnt5a null mutants can rescue the cell polarity and actin polymerization defects as well as severe SpM shortening, but fails to rescue OFT shortening. Moreover, Wnt5a over-expression in wild-type background is able to cause OFT shortening. We find that Wnt5a over-expression does not perturb SHF cell proliferation, apoptosis or differentiation, but affects the deployment of SHF cells by causing them to accumulate into a large bulge at the rostral SpM and fail to enter the OFT. Our immunostaining analyses suggest an inverse correlation between cell cohesion and Wnt5a level in the wild-type SpM. Ectopic Wnt5a expression in the rostral SpM of Wn5a-GOF mutants diminishes the upregulation of adherens junction; whereas loss of Wnt5a in Wnt5a null mutants causes premature increase in adherens junction level in the caudal SpM. Over-expression of mouse Wnt5a in Xenopus animal cap cells also reduces C-cadherin distribution on the plasma membrane without affecting its overall protein level, suggesting that Wnt5a may play an evolutionarily conserved role in controlling the cell surface level of cadherin to modulate cell cohesion during tissue morphogenesis. Collectively, our data indicate that restricted expression of Wnt5a in the caudal SpM is essential for normal OFT morphogenesis, and uncover a novel function of spatially regulated cell cohesion by Wnt5a in driving the deployment of SHF cells from the SpM into the OFT.


Subject(s)
Myocardium/cytology , Stem Cells/cytology , Wnt Proteins/physiology , Animals , Mice , Signal Transduction , Wnt-5a Protein
16.
Elife ; 5: e07727, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26845523

ABSTRACT

Endothelial cells respond to molecular and physical forces in development and vascular homeostasis. Deregulation of endothelial responses to flow-induced shear is believed to contribute to many aspects of cardiovascular diseases including atherosclerosis. However, how molecular signals and shear-mediated physical forces integrate to regulate vascular patterning is poorly understood. Here we show that endothelial non-canonical Wnt signalling regulates endothelial sensitivity to shear forces. Loss of Wnt5a/Wnt11 renders endothelial cells more sensitive to shear, resulting in axial polarization and migration against flow at lower shear levels. Integration of flow modelling and polarity analysis in entire vascular networks demonstrates that polarization against flow is achieved differentially in artery, vein, capillaries and the primitive sprouting front. Collectively our data suggest that non-canonical Wnt signalling stabilizes forming vascular networks by reducing endothelial shear sensitivity, thus keeping vessels open under low flow conditions that prevail in the primitive plexus.


Subject(s)
Endothelial Cells/physiology , Stress, Mechanical , Vascular Remodeling , Wnt Signaling Pathway , Animals , Cell Line , Cell Movement , Cell Polarity , Gene Expression Regulation , Mice
18.
Dev Biol ; 408(1): 126-39, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26526197

ABSTRACT

Wnt ligands regulate heart morphogenesis but the underlying mechanisms remain unclear. Two Formin-related proteins, DAAM1 and 2, were previously found to bind the Wnt effector Disheveled. Here, since DAAM1 and 2 nucleate actin and mediate Wnt-induced cytoskeletal changes, a floxed-allele of Daam1 was used to disrupt its function specifically in the myocardium and investigate Wnt-associated pathways. Homozygous Daam1 conditional knockout (CKO) mice were viable but had misshapen hearts and poor cardiac function. The defects in Daam1 CKO mice were observed by mid-gestation and were associated with a loss of protrusions from cardiomyocytes invading the outflow tract. Further, these mice exhibited noncompaction cardiomyopathy (NCM) and deranged cardiomyocyte polarity. Interestingly, Daam1 CKO mice that were also homozygous for an insertion disrupting Daam2 (DKO) had stronger NCM, severely reduced cardiac function, disrupted sarcomere structure, and increased myocardial proliferation, suggesting that DAAM1 and DAAM2 have redundant functions. While RhoA was unaffected in the hearts of Daam1/2 DKO mice, AKT activity was lower than in controls, raising the issue of whether DAAM1/2 are only mediating Wnt signaling. Daam1-floxed mice were thus bred to Wnt5a null mice to identify genetic interactions. The hearts of Daam1 CKO mice that were also heterozygous for the null allele of Wnt5a had stronger NCM and more severe loss of cardiac function than Daam1 CKO mice, consistent with DAAM1 and Wnt5a acting in a common pathway. However, deleting Daam1 further disrupted Wnt5a homozygous-null hearts, suggesting that DAAM1 also has Wnt5a-independent roles in cardiac development.


Subject(s)
Microfilament Proteins/metabolism , Myocardium/metabolism , Sarcomeres/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Adhesion , Cell Proliferation , Cytoskeleton/metabolism , Embryo, Mammalian/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3/metabolism , Heart Function Tests , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Heterozygote , Mice, Knockout , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Morphogenesis , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Organ Specificity , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Wnt Proteins , Wnt-5a Protein , rho GTP-Binding Proteins/deficiency , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein/metabolism
19.
Am J Pathol ; 185(8): 2194-205, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26100214

ABSTRACT

Activation of Wnt/ß-catenin signaling during liver regeneration (LR) after partial hepatectomy (PH) is observed in several species. However, how this pathway is turned off when hepatocyte proliferation is no longer required is unknown. We assessed LR in liver-specific knockouts of Wntless (Wls-LKO), a protein required for Wnt secretion from a cell. When subjected to PH, Wls-LKO showed prolongation of hepatocyte proliferation for up to 4 days compared with littermate controls. This coincided with increased ß-catenin-T-cell factor 4 interaction and cyclin-D1 expression. Wls-LKO showed decreased expression and secretion of inhibitory Wnt5a during LR. Wnt5a expression increased between 24 and 48 hours, and Frizzled-2 between 24 and 72 hours, after PH in normal mice. Treatment of primary mouse hepatocytes and liver tumor cells with Wnt5a led to a notable decrease in ß-catenin-T-cell factor activity, cyclin-D1 expression, and cell proliferation. Intriguingly, Wnt5a-LKO did not display any prolongation of LR because of compensation by other cells. In addition, Wnt5a-LKO hepatocytes failed to respond to exogenous Wnt5a treatment in culture because of a compensatory decrease in Frizzled-2 expression. In conclusion, we demonstrate Wnt5a to be, by default, a negative regulator of ß-catenin signaling and hepatocyte proliferation, both in vitro and in vivo. We also provide evidence that the Wnt5a/Frizzled-2 axis suppresses ß-catenin signaling in hepatocytes in an autocrine manner, thereby contributing to timely conclusion of the LR process.


Subject(s)
Cell Proliferation/physiology , Hepatocytes/metabolism , Liver Regeneration/physiology , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Liver/drug effects , Liver/metabolism , Liver Regeneration/drug effects , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Wnt Proteins/genetics , Wnt Proteins/pharmacology , Wnt Signaling Pathway/drug effects , Wnt-5a Protein
20.
Development ; 142(9): 1628-38, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25922526

ABSTRACT

In the development of the vertebrate body plan, Wnt3a is thought to promote the formation of paraxial mesodermal progenitors (PMPs) of the trunk region while suppressing neural specification. Recent lineage-tracing experiments have demonstrated that these trunk neural progenitors and PMPs derive from a common multipotent progenitor called the neuromesodermal progenitor (NMP). NMPs are known to reside in the anterior primitive streak (PS) region; however, the extent to which NMPs populate the PS and contribute to the vertebrate body plan, and the precise role that Wnt3a plays in regulating NMP self-renewal and differentiation are unclear. To address this, we used cell-specific markers (Sox2 and T) and tamoxifen-induced Cre recombinase-based lineage tracing to locate putative NMPs in vivo. We provide functional evidence for NMP location primarily in the epithelial PS, and to a lesser degree in the ingressed PS. Lineage-tracing studies in Wnt3a/ß-catenin signaling pathway mutants provide genetic evidence that trunk progenitors normally fated to enter the mesodermal germ layer can be redirected towards the neural lineage. These data, combined with previous PS lineage-tracing studies, support a model that epithelial anterior PS cells are Sox2(+)T(+) multipotent NMPs and form the bulk of neural progenitors and PMPs of the posterior trunk region. Finally, we find that Wnt3a/ß-catenin signaling directs trunk progenitors towards PMP fates; however, our data also suggest that Wnt3a positively supports a progenitor state for both mesodermal and neural progenitors.


Subject(s)
Body Patterning/physiology , Cell Differentiation/physiology , Cell Lineage/physiology , Mesoderm/embryology , Neural Stem Cells/physiology , Primitive Streak/cytology , Signal Transduction/physiology , Animals , Histological Techniques , Immunohistochemistry , In Situ Hybridization , Mesoderm/cytology , Mice , Mice, Knockout , Models, Biological , Wnt3A Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...