Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 77
1.
Cancer Res Commun ; 4(4): 1100-1110, 2024 Apr 18.
Article En | MEDLINE | ID: mdl-38551394

PURPOSE: TPST-1120 is a first-in-class oral inhibitor of peroxisome proliferator-activated receptor α (PPARα), a fatty acid ligand-activated transcription factor that regulates genes involved in fatty acid oxidation, angiogenesis, and inflammation, and is a novel target for cancer therapy. TPST-1120 displayed antitumor activity in xenograft models and synergistic tumor reduction in syngeneic tumor models when combined with anti-PD-1 agents. EXPERIMENTAL DESIGN: This phase I, open-label, dose-escalation study (NCT03829436) evaluated TPST-1120 as monotherapy in patients with advanced solid tumors and in combination with nivolumab in patients with renal cell carcinoma (RCC), cholangiocarcinoma (CCA), or hepatocellular carcinoma. Objectives included evaluation of safety, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity (RECIST v1.1). RESULTS: A total of 39 patients enrolled with 38 treated (20 monotherapy, 18 combination; median 3 prior lines of therapy). The most common treatment-related adverse events (TRAE) were grade 1-2 nausea, fatigue, and diarrhea. No grade 4-5 TRAEs or dose-limiting toxicities were reported. In the monotherapy group, 53% (10/19) of evaluable patients had a best objective response of stable disease. In the combination group, 3 patients had partial responses, for an objective response rate of 20% (3/15) across all doses and 30% (3/10) at TPST-1120 ≥400 mg twice daily. Responses occurred in 2 patients with RCC, both of whom had previously progressed on anti-PD-1 therapy, and 1 patient with late-line CCA. CONCLUSIONS: TPST-1120 was well tolerated as monotherapy and in combination with nivolumab and the combination showed preliminary evidence of clinical activity in PD-1 inhibitor refractory and immune compromised cancers. SIGNIFICANCE: TPST-1120 is a first-in-class oral inhibitor of PPARα, whose roles in metabolic and immune regulation are implicated in tumor proliferation/survival and inhibition of anticancer immunity. This first-in-human study of TPST-1120 alone and in combination with nivolumab supports proof-of-concept of PPARα inhibition as a target of therapeutic intervention in solid tumors.


Carcinoma, Renal Cell , Kidney Neoplasms , Liver Neoplasms , PPAR alpha , Humans , Carcinoma, Renal Cell/drug therapy , Fatty Acids , Kidney Neoplasms/drug therapy , Liver Neoplasms/drug therapy , Nivolumab/therapeutic use , PPAR alpha/antagonists & inhibitors
2.
PLoS One ; 19(2): e0293072, 2024.
Article En | MEDLINE | ID: mdl-38349913

We performed a forward genetic screen to discover peptides that specifically target breast cancer cells using a Penetratin tagged, random 15mer peptide library. We identified a group of novel peptides that specifically inhibited the proliferation and survival of breast cancer cells without affecting normal primary mammary epithelial cells or fibroblasts. The intrinsic apoptotic pathway is activated by these peptides in the face of abnormal expression of numerous cell cycle regulatory genes. Associated alterations in histone marks, nuclear structure, and levels of critical RNA binding proteins vary in a peptide specific manner. This study demonstrates a novel method for the discovery of new potential therapeutic peptides.


Breast Neoplasms , Peptide Library , Humans , Female , Cell Proliferation , Peptides/chemistry , Breast/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism
3.
Invest Ophthalmol Vis Sci ; 64(5): 1, 2023 05 01.
Article En | MEDLINE | ID: mdl-37126314

Purpose: Familial exudative vitreoretinopathy (FEVR) and Norrie disease are examples of genetic disorders in which the retinal vasculature fails to fully form (hypovascular), leading to congenital blindness. While studying the role of a factor expressed during retinal development, T-box factor Tbx3, we discovered that optic cup loss of Tbx3 caused the retina to become hypovascular. The purpose of this study was to characterize how loss of Tbx3 affects retinal vasculature formation. Methods: Conditional removal of Tbx3 from both retinal progenitors and astrocytes was done using the optic cup-Cre recombinase driver BAC-Dkk3-Cre and was analyzed using standard immunohistochemical techniques. Results: With Tbx3 loss, the retinas were hypovascular, as seen in patients with retinopathy of prematurity (ROP) and FEVR. Retinal vasculature failed to form the stereotypic tri-layered plexus in the dorsal-temporal region. Astrocyte precursors were reduced in number and failed to form a lattice at the dorsal-temporal edge. We next examined retinal ganglion cells, as they have been shown to play a critical role in retinal angiogenesis. We found that melanopsin expression and Islet1/2-positive retinal ganglion cells were reduced in the dorsal half of the retina. In previous studies, the loss of melanopsin has been linked to hyaloid vessel persistence, which we also observed in the Tbx3 conditional knockout (cKO) retinas, as well as in infants with ROP or FEVR. Conclusions: To the best of our knowledge, these studies are the first demonstration that Tbx3 is required for normal mammalian eye formation. Together, the results provide a potential genetic model for retinal hypovascular diseases.


Retinal Degeneration , Retinopathy of Prematurity , Mice , Animals , Infant, Newborn , Humans , Retina , Retinal Ganglion Cells , Retinal Vessels , Familial Exudative Vitreoretinopathies , Mammals , T-Box Domain Proteins
4.
Dev Biol ; 499: 10-21, 2023 07.
Article En | MEDLINE | ID: mdl-37060937

Development of the outflow tract of the heart requires specification, proliferation and deployment of a progenitor cell population from the second heart field to generate the myocardium at the arterial pole of the heart. Disruption of these processes leads to lethal defects in rotation and septation of the outflow tract. We previously showed that Fibroblast Growth Factor 8 (FGF8) directs a signaling cascade in the second heart field that regulates critical aspects of OFT morphogenesis. Here we show that in addition to the survival and proliferation cues previously described, FGF8 provides instructive and patterning information to OFT myocardial cells and their progenitors that prevents their aberrant differentiation along a working myocardial program.


Heart , Myocardium , Cell Differentiation/physiology , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Mesoderm/metabolism , Myocardium/metabolism , Myocytes, Cardiac , Animals , Mice
5.
BMC Biol ; 21(1): 55, 2023 03 20.
Article En | MEDLINE | ID: mdl-36941669

BACKGROUND: The reactivation of genetic programs from early development is a common mechanism for injury-induced organ regeneration. T-box 3 (TBX3) is a member of the T-box family of transcription factors previously shown to regulate pluripotency and subsequent lineage commitment in a number of tissues, including limb and lung. TBX3 is also involved in lung and heart organogenesis. Here, we provide a comprehensive and thorough characterization of TBX3 and its role during pancreatic organogenesis and regeneration. RESULTS: We interrogated the level and cell specificity of TBX3 in the developing and adult pancreas at mRNA and protein levels at multiple developmental stages in mouse and human pancreas. We employed conditional mutagenesis to determine its role in murine pancreatic development and in regeneration after the induction of acute pancreatitis. We found that Tbx3 is dynamically expressed in the pancreatic mesenchyme and epithelium. While Tbx3 is expressed in the developing pancreas, its absence is likely compensated by other factors after ablation from either the mesenchymal or epithelial compartments. In an adult model of acute pancreatitis, we found that a lack of Tbx3 resulted in increased proliferation and fibrosis as well as an enhanced inflammatory gene programs, indicating that Tbx3 has a role in tissue homeostasis and regeneration. CONCLUSIONS: TBX3 demonstrates dynamic expression patterns in the pancreas. Although TBX3 is dispensable for proper pancreatic development, its absence leads to altered organ regeneration after induction of acute pancreatitis.


Pancreatitis , Adult , Humans , Animals , Mice , Acute Disease , Pancreatitis/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Pancreas/metabolism , Organogenesis/genetics
6.
Dev Dyn ; 251(10): 1711-1727, 2022 10.
Article En | MEDLINE | ID: mdl-35618654

BACKGROUND: Asymmetries in craniofacial anomalies are commonly observed. In the facial skeleton, the left side is more commonly and/or severely affected than the right. Such asymmetries complicate treatment options. Mechanisms underlying variation in disease severity between individuals as well as within individuals (asymmetries) are still relatively unknown. RESULTS: Developmental reductions in fibroblast growth factor 8 (Fgf8) have a dosage dependent effect on jaw size, shape, and symmetry. Further, Fgf8 mutants have directionally asymmetric jaws with the left side being more affected than the right. Defects in lower jaw development begin with disruption to Meckel's cartilage, which is discontinuous. All skeletal elements associated with the proximal condensation are dysmorphic, exemplified by a malformed and misoriented malleus. At later stages, Fgf8 mutants exhibit syngnathia, which falls into two broad categories: bony fusion of the maxillary and mandibular alveolar ridges and zygomatico-mandibular fusion. All of these morphological defects exhibit both inter- and intra-specimen variation. CONCLUSIONS: We hypothesize that these asymmetries are linked to heart development resulting in higher levels of Fgf8 on the right side of the face, which may buffer the right side to developmental perturbations. This mouse model may facilitate future investigations of mechanisms underlying human syngnathia and facial asymmetry.


Branchial Region , Heart , Animals , Fibroblast Growth Factor 8/genetics , Humans , Jaw Abnormalities , Maxilla , Mice , Mouth Abnormalities
7.
Dev Dyn ; 251(9): 1613-1627, 2022 09.
Article En | MEDLINE | ID: mdl-35506352

The limb phenotypes of Tbx2 and Tbx3 mutants are distinct: loss of Tbx2 results in isolated duplication of digit 4 in the hindlimb while loss of Tbx3 results in anterior polydactyly and posterior oligodactly in the forelimb. In the face of such disparate phenotypes, we sought to determine whether Tbx2 and Tbx3 have functional redundancy during development of the mouse limb. We found that sequential loss of alleles generates defects that are not simply additive of those observed in single mutants and that multiple structures in both the forelimb and hindlimb display compound sensitivity to decreased gene dosage.


Extremities , T-Box Domain Proteins/metabolism , Animals , Hindlimb , Mice , Phenotype , T-Box Domain Proteins/genetics
8.
Mol Ther Oncolytics ; 23: 342-354, 2021 Dec 17.
Article En | MEDLINE | ID: mdl-34820504

hnRNPK is a multifunctional protein that plays an important role in cancer cell proliferation and metastasis via its RNA- and DNA-binding properties. Previously we showed that cell-penetrating peptides derived from the RGG RNA-binding domain of SAFA (hnRNPU) disrupt cancer cell proliferation and survival. Here we explore the efficacy of a peptide derived from the RGG domain of hnRNPK. This peptide acts in a dominant-negative manner on several hnRNPK functions to induce death of multiple types of cancer cells. The peptide phenocopies the effect of hnRNPK knockdown on its mRNA-stability targets such as KLF4 and EGR1 and alters the levels and locations of long non-coding RNAs (lncRNAs) and proteins required for nuclear and paraspeckle formation and function. The RGG-derived peptide also decreases euchromatin as evidenced by loss of active marks and polymerase II occupancy. Our findings reveal the potential therapeutic utility of the hnRNPK RGG-derived peptide in a range of cancers.

9.
Cell Rep ; 35(9): 109156, 2021 06 01.
Article En | MEDLINE | ID: mdl-34077726

RBM39 is a known splicing factor and coactivator. Here, we report that RBM39 functions as a master transcriptional regulator that interacts with the MLL1 complex to facilitate chromatin binding and H3K4 trimethylation in breast cancer cells. We identify RBM39 functional domains required for DNA and complex binding and show that the loss of RBM39 has widespread effects on H3K4me3 and gene expression, including key oncogenic pathways. RBM39's RNA recognition motif 3 (RRM3) functions as a dominant-negative domain; namely, it disrupts the complex and H3K4me trimethylation and expression of RBM/MLL1 target genes. RRM3-derived cell-penetrating peptides phenocopy the effects of the loss of RBM39 to decrease growth and survival of all major subtypes of breast cancer and yet are nontoxic to normal cells. These findings establish RBM39/MLL1 as a major contributor to the abnormal epigenetic landscape in breast cancer and lay the foundation for peptide-mediated cancer-specific therapy based on disruption of RBM39 epigenomic functions.


Breast Neoplasms/genetics , Breast Neoplasms/pathology , Epigenomics , Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Peptides/metabolism , RNA-Binding Proteins/genetics , Transcription, Genetic , Animals , Carcinogenesis/pathology , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/genetics , Cell Survival , Cell-Penetrating Peptides/metabolism , Female , HEK293 Cells , Histone Code , Histone-Lysine N-Methyltransferase/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Inbred NOD , Mice, SCID , Myeloid-Lymphoid Leukemia Protein/metabolism , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding , Protein Domains , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism
10.
Development ; 148(8)2021 04 15.
Article En | MEDLINE | ID: mdl-33795231

All epithelial components of the inner ear, including sensory hair cells and innervating afferent neurons, arise by patterning and differentiation of epithelial progenitors residing in a simple sphere, the otocyst. Here, we identify the transcriptional repressors TBX2 and TBX3 as novel regulators of these processes in the mouse. Ablation of Tbx2 from the otocyst led to cochlear hypoplasia, whereas loss of Tbx3 was associated with vestibular malformations. The loss of function of both genes (Tbx2/3cDKO) prevented inner ear morphogenesis at midgestation, resulting in indiscernible cochlear and vestibular structures at birth. Morphogenetic impairment occurred concomitantly with increased apoptosis in ventral and lateral regions of Tbx2/3cDKO otocysts around E10.5. Expression analyses revealed partly disturbed regionalisation, and a posterior-ventral expansion of the neurogenic domain in Tbx2/3cDKO otocysts at this stage. We provide evidence that repression of FGF signalling by TBX2 is important to restrict neurogenesis to the anterior-ventral otocyst and implicate another T-box factor, TBX1, as a crucial mediator in this regulatory network.


Apoptosis , Ear, Inner/embryology , Gene Expression Regulation, Developmental , Organogenesis , Signal Transduction , T-Box Domain Proteins/biosynthesis , Animals , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Mice , Mice, Knockout , T-Box Domain Proteins/genetics
11.
Front Oncol ; 11: 621825, 2021.
Article En | MEDLINE | ID: mdl-33859938

Scaffold-attachment-factor A (SAFA) has important roles in many normal and pathologic cellular processes but the scope of its function in cancer cells is unknown. Here, we report dominant-negative activity of novel peptides derived from the SAP and RGG-domains of SAFA and their effects on proliferation, survival and the epigenetic landscape in a range of cancer cell types. The RGG-derived peptide dysregulates SAFA binding and regulation of alternatively spliced targets and decreases levels of key spliceosome proteins in a cell-type specific manner. In contrast, the SAP-derived peptide reduces active histone marks, promotes chromatin compaction, and activates the DNA damage response and cell death in a subset of cancer cell types. Our findings reveal an unprecedented function of SAFA-derived peptides in regulating diverse SAFA molecular functions as a tumor suppressive mechanism and demonstrate the potential therapeutic utility of SAFA-peptides in a wide range of cancer cells.

12.
Development ; 147(22)2020 11 19.
Article En | MEDLINE | ID: mdl-33046506

FGF8 signaling plays diverse roles in inner ear development, acting at multiple stages from otic placode induction to cellular differentiation in the organ of Corti. As a secreted morphogen with diverse functions, Fgf8 expression is likely to be spatially restricted and temporally dynamic throughout inner ear development. We evaluated these characteristics using genetic labeling mediated by Fgf8mcm gene-targeted mice and determined that Fgf8 expression is a specific and early marker of Type-I vestibular hair cell identity. Fgf8mcm expression initiates at E11.5 in the future striolar region of the utricle, labeling hair cells following EdU birthdating, and demonstrates that sub-type identity is determined shortly after terminal mitosis. This early fate specification is not apparent using markers or morphological criteria that are not present before birth in the mouse. Although analyses of Fgf8 conditional knockout mice did not reveal developmental phenotypes, the restricted pattern of Fgf8 expression suggests that functionally redundant FGF ligands may contribute to vestibular hair cell differentiation and supports a developmental model in which Type-I and Type-II hair cells develop in parallel rather than from an intermediate precursor.


Fibroblast Growth Factor 8/metabolism , Hair Cells, Vestibular/metabolism , Saccule and Utricle/embryology , Animals , Fibroblast Growth Factor 8/genetics , Hair Cells, Vestibular/cytology , Mice , Mice, Knockout , Saccule and Utricle/cytology
13.
Development ; 147(17)2020 09 11.
Article En | MEDLINE | ID: mdl-32778568

The role played by the Notch pathway in cardiac progenitor cell biology remains to be elucidated. Delta-like ligand 4 (Dll4), the arterial-specific Notch ligand, is expressed by second heart field (SHF) progenitors at time-points that are crucial in SHF biology. Dll4-mediated Notch signaling is required for maintaining an adequate pool of SHF progenitors, such that Dll4 knockout results in a reduction in proliferation and an increase in apoptosis. A reduced SHF progenitor pool leads to an underdeveloped right ventricle (RV) and outflow tract (OFT). In its most severe form, there is severe RV hypoplasia and poorly developed OFT resulting in early embryonic lethality. In its milder form, the OFT is foreshortened and misaligned, resulting in a double outlet right ventricle. Dll4-mediated Notch signaling maintains Fgf8 expression by transcriptional regulation at the promoter level. Combined heterozygous knockout of Dll4 and Fgf8 demonstrates genetic synergy in OFT alignment. Exogenous supplemental Fgf8 rescues proliferation in Dll4 mutants in ex-vivo culture. Our results establish a novel role for Dll4-mediated Notch signaling in SHF biology. More broadly, our model provides a platform for understanding oligogenic inheritance that results in clinically relevant OFT malformations.


Adaptor Proteins, Signal Transducing/metabolism , Calcium-Binding Proteins/metabolism , Cell Proliferation , Fibroblast Growth Factor 8/biosynthesis , Gene Expression Regulation, Developmental , Heart Ventricles/embryology , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Calcium-Binding Proteins/genetics , Fibroblast Growth Factor 8/genetics , Mice , Mice, Knockout , Receptors, Notch/genetics
14.
Proc Natl Acad Sci U S A ; 117(27): 15712-15723, 2020 07 07.
Article En | MEDLINE | ID: mdl-32561646

The mechanisms used by embryos to pattern tissues across their axes has fascinated developmental biologists since the founding of embryology. Here, using single-cell technology, we interrogate complex patterning defects and define a Hedgehog (Hh)-fibroblast growth factor (FGF) signaling axis required for anterior mesoderm lineage development during gastrulation. Single-cell transcriptome analysis of Hh-deficient mesoderm revealed selective deficits in anterior mesoderm populations, culminating in defects to anterior embryonic structures, including the pharyngeal arches, heart, and anterior somites. Transcriptional profiling of Hh-deficient mesoderm during gastrulation revealed disruptions to both transcriptional patterning of the mesoderm and FGF signaling for mesoderm migration. Mesoderm-specific Fgf4/Fgf8 double-mutants recapitulated anterior mesoderm defects and Hh-dependent GLI transcription factors modulated enhancers at FGF gene loci. Cellular migration defects during gastrulation induced by Hh pathway antagonism were mitigated by the addition of FGF4 protein. These findings implicate a multicomponent signaling hierarchy activated by Hh ligands from the embryonic node and executed by FGF signals in nascent mesoderm to control anterior mesoderm patterning.


Fibroblast Growth Factor 4/genetics , Fibroblast Growth Factor 8/genetics , Gastrulation/genetics , Zinc Finger Protein GLI1/genetics , Animals , Body Patterning/genetics , Cell Lineage/genetics , Chick Embryo , Fibroblast Growth Factors/genetics , Gastrula/growth & development , Gastrula/metabolism , Gene Expression Regulation, Developmental/genetics , Hedgehog Proteins/genetics , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Signal Transduction/genetics , Single-Cell Analysis , Transcriptome/genetics
15.
Pediatr Cardiol ; 40(7): 1388-1400, 2019 Oct.
Article En | MEDLINE | ID: mdl-31372681

In this article, we provide a brief summary of work by us and others to discover the molecular underpinnings of early conduction system development and function. We focus on how the multifunctional protein Tbx3 contributes to acquisition and homeostasis of the tissue-specific properties of the sinoatrial and atrioventricular nodes. We also provide unpublished, preliminary findings supporting the role of Tbx3-regulated alternative RNA processing in the developing conduction system.


Gene Expression Regulation, Developmental , Heart Conduction System/metabolism , Animals , Atrioventricular Node/physiopathology , Gene Expression , Heart Conduction System/embryology , Humans , RNA/metabolism , T-Box Domain Proteins
16.
Development ; 146(4)2019 02 20.
Article En | MEDLINE | ID: mdl-30787001

Congenital heart disease (CHD) is the most common type of birth defect. In recent years, research has focussed on identifying the genetic causes of CHD. However, only a minority of CHD cases can be attributed to single gene mutations. In addition, studies have identified different environmental stressors that promote CHD, but the additive effect of genetic susceptibility and environmental factors is poorly understood. In this context, we have investigated the effects of short-term gestational hypoxia on mouse embryos genetically predisposed to heart defects. Exposure of mouse embryos heterozygous for Tbx1 or Fgfr1/Fgfr2 to hypoxia in utero increased the incidence and severity of heart defects while Nkx2-5+/- embryos died within 2 days of hypoxic exposure. We identified the molecular consequences of the interaction between Nkx2-5 and short-term gestational hypoxia, which suggest that reduced Nkx2-5 expression and a prolonged hypoxia-inducible factor 1α response together precipitate embryo death. Our study provides insight into the causes of embryo loss and variable penetrance of monogenic CHD, and raises the possibility that cases of foetal death and CHD in humans could be caused by similar gene-environment interactions.


Gene-Environment Interaction , Heart Defects, Congenital/genetics , Heart/embryology , Homeobox Protein Nkx-2.5/genetics , Homeodomain Proteins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Animals , Apoptosis , Cell Proliferation , Embryo, Mammalian/metabolism , Female , Genetic Predisposition to Disease , Heart/diagnostic imaging , Heterozygote , Homeobox Protein Nkx-2.5/physiology , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxygen/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , T-Box Domain Proteins/genetics , Time Factors
17.
Nat Metab ; 1(2): 222-235, 2019 02.
Article En | MEDLINE | ID: mdl-32694784

Heterogeneous populations of hypothalamic neurons orchestrate energy balance via the release of specific signatures of neuropeptides. However, how specific intracellular machinery controls peptidergic identities and function of individual hypothalamic neurons remains largely unknown. The transcription factor T-box 3 (Tbx3) is expressed in hypothalamic neurons sensing and governing energy status, whereas human TBX3 haploinsufficiency has been linked with obesity. Here, we demonstrate that loss of Tbx3 function in hypothalamic neurons causes weight gain and other metabolic disturbances by disrupting both the peptidergic identity and plasticity of Pomc/Cart and Agrp/Npy neurons. These alterations are observed after loss of Tbx3 in both immature hypothalamic neurons and terminally differentiated mouse neurons. We further establish the importance of Tbx3 for body weight regulation in Drosophila melanogaster and show that TBX3 is implicated in the differentiation of human embryonic stem cells into hypothalamic Pomc neurons. Our data indicate that Tbx3 directs the terminal specification of neurons as functional components of the melanocortin system and is required for maintaining their peptidergic identity. In summary, we report the discovery of a key mechanistic process underlying the functional heterogeneity of hypothalamic neurons governing body weight and systemic metabolism.


Hypothalamus/metabolism , Melanocortins/metabolism , Neurons/metabolism , T-Box Domain Proteins/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Body Weight , Energy Metabolism , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Hypothalamus/cytology , Mice , Mice, Inbred C57BL , Pro-Opiomelanocortin/genetics , RNA, Messenger/genetics , T-Box Domain Proteins/genetics
18.
Development ; 145(23)2018 11 26.
Article En | MEDLINE | ID: mdl-30478225

The organized array of smooth muscle cells (SMCs) and fibroblasts in the walls of visceral tubular organs arises by patterning and differentiation of mesenchymal progenitors surrounding the epithelial lumen. Here, we show that the TBX2 and TBX3 transcription factors have novel and required roles in regulating these processes in the murine ureter. Co-expression of TBX2 and TBX3 in the inner mesenchymal region of the developing ureter requires canonical WNT signaling. Loss of TBX2/TBX3 in this region disrupts activity of two crucial drivers of the SMC program, Foxf1 and BMP4 signaling, resulting in decreased SMC differentiation and increased extracellular matrix. Transcriptional profiling and chromatin immunoprecipitation experiments revealed that TBX2/TBX3 directly repress expression of the WNT antagonists Dkk2 and Shisa2, the BMP antagonist Bmper and the chemokine Cxcl12 These findings suggest that TBX2/TBX3 are effectors of canonical WNT signaling in the ureteric mesenchyme that promote SMC differentiation by maintaining BMP4 and WNT signaling in the inner region, while restricting CXCL12 signaling to the outer layer of fibroblast-fated mesenchyme.


Body Patterning , Cell Differentiation , Mesoderm/embryology , T-Box Domain Proteins/metabolism , Ureter/embryology , Wnt Signaling Pathway , Animals , Bone Morphogenetic Protein 4/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Mice , Models, Biological , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Peristalsis , T-Box Domain Proteins/genetics , Transcriptome/genetics , Ureter/metabolism , Ureter/pathology
19.
Dev Biol ; 444 Suppl 1: S337-S351, 2018 12 01.
Article En | MEDLINE | ID: mdl-30292786

Transcription factors that coordinate migration, differentiation or proliferation of enteric nervous system (ENS) precursors are not well defined. To identify novel transcriptional regulators of ENS development, we performed microarray analysis at embryonic day (E) 17.5 and identified many genes that were enriched in the ENS compared to other bowel cells. We decided to investigate the T-box transcription factor Tbx3, which is prominently expressed in developing and mature ENS. Haploinsufficiency for TBX3 causes ulnar-mammary syndrome (UMS) in humans, a multi-organ system disorder. TBX3 also regulates several genes known to be important for ENS development. To test the hypothesis that Tbx3 is important for ENS development or function, we inactivated Tbx3 in all neural crest derivatives, including ENS progenitors using Wnt1-Cre and a floxed Tbx3 allele. Tbx3 fl/fl; Wnt1-Cre conditional mutant mice die shortly after birth with cleft palate and difficulty feeding. The ENS of mutants was well-organized with a normal density of enteric neurons and nerve fiber bundles, but small bowel glial cell density was reduced. Despite this, bowel motility appeared normal. Furthermore, although Tbx3 is expressed in cardiac neural crest, Tbx3 fl/fl; Wnt1-Cre mice had structurally normal hearts. Thus, loss of Tbx3 within neural crest has selective effects on Tbx3-expressing neural crest derivatives.


Enteric Nervous System/embryology , Neural Crest/embryology , T-Box Domain Proteins/physiology , Animals , Cell Differentiation , Cell Movement , Cleft Palate/embryology , Cleft Palate/genetics , Heart/embryology , Intestines/embryology , Mice , Mice, Inbred C57BL , Neural Crest/metabolism , Neural Crest/physiology , Neurogenesis , Neuroglia/physiology , Neurons , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Wnt1 Protein
20.
PLoS One ; 12(9): e0184678, 2017.
Article En | MEDLINE | ID: mdl-28961240

BACKGROUND: Genetic and epigenetic programs regulate dramatic structural changes during cardiac morphogenesis. Concurrent biomechanical forces within the heart created by blood flow and pressure in turn drive downstream cellular, molecular and genetic responses. Thus, a genetic-morphogenetic-biomechanical feedback loop is continually operating to regulate heart development. During the evolution of a congenital heart defect, concomitant abnormalities in blood flow, hemodynamics, and patterns of mechanical loading would be predicted to change the output of this feedback loop, impacting not only the ultimate morphology of the defect, but potentially altering tissue-level biomechanical properties of structures that appear structurally normal. AIM: The goal of this study was to determine if abnormal hemodynamics present during outflow tract formation and remodeling in a genetically engineered mouse model of persistent truncus arteriosus (PTA) causes tissue-level biomechanical abnormalities. METHODS: The passive stiffness of surface locations on the left ventricle (LV), right ventricle (RV), and outflow tract (OFT) was measured with a pipette aspiration technique in Fgf8;Isl1Cre conditional mutant embryonic mouse hearts and controls. Control and mutant experimental results were compared by a strain energy metric based on the measured relationship between pressure and aspirated height, and also used as target behavior for finite element models of the ventricles. Model geometry was determined from 3D reconstructions of whole-mount, confocal-imaged hearts. The stress-strain relationship of the model was adjusted to achieve an optimal match between model and experimental behavior. RESULTS AND CONCLUSION: Although the OFT is the most severely affected structure in Fgf8;Isl1Cre hearts, its passive stiffness was the same as in control hearts. In contrast, both the LV and RV showed markedly increased passive stiffness, doubling in LVs and quadrupling in RVs of mutant hearts. These differences are not attributable to differences in ventricular volume, wall thickness, or trabecular density. Excellent agreement was obtained between the model and experimental results. Overall our findings show that hearts developing PTA have early changes in ventricular tissue biomechanics relevant to cardiac function and ongoing development.


Disease Models, Animal , Myocardium/pathology , Truncus Arteriosus, Persistent/pathology , Animals , Female , Male , Mice , Mice, Transgenic , Microscopy, Confocal
...