Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 9(1): 3544, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30837642

ABSTRACT

The cellular and molecular mechanisms underlying senile osteoporosis remain poorly understood. In this study, transgenic mCol1α1-Pitx1 mice overexpressing paired-like homeodomain 1 (PITX1), a homeobox transcription factor, rapidly develop a severe type-II osteoporotic phenotype with significant reduction in bone mass and biomechanical strength similar to that seen in humans and reminiscent of the phenotype previously observed in Sca-1 (Ly6a)-null mice. PITX1 plays a critical role in hind limb formation during fetal development, while loss of expression is associated with primary knee/hip osteoarthritis in aging humans. Through in vivo and in vitro analyses, we demonstrate that Pitx1 directly regulates the self-renewal of mesenchymal progenitors and indirectly regulates osteoclast differentiation through the upregulation of Wnt signaling inhibitors DKK1, SOST, and GSK3-ß. This is confirmed by elevated levels of plasma DKK1 and the accumulation of phospho-ß-catenin in transgenic mice osteoblasts. Furthermore, overexpressed Pitx1 in mice osteoblasts results in severe repression of Sca-1 (Ly6a) that was previously associated with senile osteoporosis. Our study is the first to demonstrate the novel roles of PITX1 in senile osteoporosis where PITX1 regulates the self-renewal of mesenchymal stem cells or progenitor cells through Sca-1 (Ly6a) repression and, in addition, inhibits the Wnt signaling pathway.


Subject(s)
Bone and Bones/metabolism , Cell Self Renewal , Gene Expression Regulation , Mesenchymal Stem Cells/pathology , Osteoporosis/pathology , Paired Box Transcription Factors/genetics , Wnt Signaling Pathway/genetics , Animals , Bone Density , Bone and Bones/physiopathology , Mice , Organ Specificity , Osteoclasts/pathology , Osteogenesis , Osteoporosis/genetics , Osteoporosis/physiopathology , Phenotype
2.
Am J Pathol ; 180(4): 1441-53, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22342522

ABSTRACT

The Hedgehog (Hh) signaling pathway regulates tissue patterning during development, including patterning and growth of limbs and face, but whether Hh signaling plays a role in adult kidney remains undefined. In this study, using a panel of hedgehog-reporter mice, we show that the two Hh ligands (Indian hedgehog and sonic hedgehog ligands) are expressed in tubular epithelial cells. We report that the Hh effectors (Gli1 and Gli2) are expressed exclusively in adjacent platelet-derived growth factor receptor-ß-positive interstitial pericytes and perivascular fibroblasts, suggesting a paracrine signaling loop. In two models of renal fibrosis, Indian Hh ligand was upregulated with a dramatic activation of downstream Gli effector expression. Hh-responsive Gli1-positive interstitial cells underwent 11-fold proliferative expansion during fibrosis, and both Gli1- and Gli2-positive cells differentiated into α-smooth muscle actin-positive myofibroblasts. In the pericyte-like cell line 10T1/2, hedgehog ligand triggered cell proliferation, suggesting a possible role for this pathway in the regulation of cell cycle progression of myofibroblast progenitors during the development of renal fibrosis. The hedgehog antagonist IPI-926 abolished Gli1 induction in vivo but did not decrease kidney fibrosis. However, the transcriptional induction of Gli2 was unaffected by IPI-926, suggesting the existence of smoothened-independent Gli activation in this model. This study is the first detailed description of paracrine hedgehog signaling in adult kidney, which indicates a possible role for hedgehog-Gli signaling in fibrotic chronic kidney disease.


Subject(s)
Hedgehog Proteins/metabolism , Kidney/pathology , Animals , Cell Line , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Epithelial Cells/metabolism , Fibroblasts/metabolism , Fibrosis , Kidney/metabolism , Kidney Tubules/metabolism , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/metabolism , Ligands , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Paracrine Communication/physiology , Patched Receptors , Pericytes/metabolism , Pericytes/pathology , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Up-Regulation/physiology , Veratrum Alkaloids/pharmacology , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2
3.
Dev Dyn ; 240(2): 404-12, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21246657

ABSTRACT

Mice possessing no Has2 expression in chondrocytes died near birth and displayed abnormalities throughout their skeleton. By embryonic day 18.5, the long bones were short and wide, and possessed excessive mineralization within their diaphysis, with little evidence of diaphyseal bone modeling. However, this does not appear to be associated with an absence of blood vessel invasion or the reduced presence of osteoclasts. There was no evidence for the formation of an organized growth plate between the epiphysis and diaphysis, and while hypertrophic chondrocytes were present in this region they were abnormal in both appearance and organization. There was also increased cellularity in the epiphyseal cartilage and a corresponding decrease in the abundance of extracellular matrix, but aggrecan was still present. Thus, hyaluronan production by chondrocytes is not only essential for formation of an organized growth plate and subsequent long bone growth but also for normal modeling of the diaphyseal bone.


Subject(s)
Bone Development/physiology , Chondrocytes/physiology , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Hyaluronic Acid/metabolism , Animals , Bone and Bones/anatomy & histology , Bone and Bones/pathology , Bone and Bones/physiology , Cartilage/cytology , Cartilage/metabolism , Cartilage/pathology , Chondrocytes/cytology , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Hyaluronan Synthases , Mice , Mice, Knockout , Phenotype
4.
Dev Biol ; 320(1): 242-55, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18582859

ABSTRACT

The development of extraocular orbital structures, in particular the choroid and sclera, is regulated by a complex series of interactions between neuroectoderm, neural crest and mesoderm derivatives, although in many instances the signals that mediate these interactions are not known. In this study we have investigated the function of Indian hedgehog (Ihh) in the developing mammalian eye. We show that Ihh is expressed in a population of non-pigmented cells located in the developing choroid adjacent to the RPE. The analysis of Hh mutant mice demonstrates that the RPE and developing scleral mesenchyme are direct targets of Ihh signaling and that Ihh is required for the normal pigmentation pattern of the RPE and the condensation of mesenchymal cells to form the sclera. Our findings also indicate that Ihh signals indirectly to promote proliferation and photoreceptor specification in the neural retina. This study identifies Ihh as a novel choroid-derived signal that regulates RPE, sclera and neural retina development.


Subject(s)
Endothelial Cells/metabolism , Hedgehog Proteins/metabolism , Pigment Epithelium of Eye/embryology , Sclera/embryology , Signal Transduction , Animals , Biomarkers/metabolism , Choroid/embryology , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Homeodomain Proteins/metabolism , Hypopigmentation/pathology , Kruppel-Like Transcription Factors , Mesoderm/embryology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Orbit/metabolism , Pigment Epithelium of Eye/abnormalities , Pigment Epithelium of Eye/ultrastructure , Retina/embryology , Retina/pathology , Sclera/abnormalities , Sclera/ultrastructure , Trans-Activators/metabolism , Zinc Finger Protein GLI1
5.
Dev Biol ; 299(1): 272-82, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16935278

ABSTRACT

The synchondroses consist of mirror-image growth plates and are critical for cranial base elongation, but relatively little is known about their formation and regulation. Here we show that synchondrosis development is abnormal in Indian hedgehog-null mice. The Ihh(-/-) cranial bases displayed reduced growth and chondrocyte proliferation, but chondrocyte hypertrophy was widespread. Rather than forming a typical narrow zone, Ihh(-/-) hypertrophic chondrocytes occupied an elongated central portion of each growth plate and were flanked by immature collagen II-expressing chondrocytes facing perichondrial tissues. Endochondral ossification was delayed in much of the Ihh(-/-) cranial bases but, surprisingly, was unaffected most posteriorly. Searching for an explanation, we found that notochord remnants near incipient spheno-occipital synchondroses at E13.5 expressed Sonic hedgehog and local chondrocytes expressed Patched, suggesting that Shh had sustained chondrocyte maturation and occipital ossification. Equally unexpected, Ihh(-/-) growth plates stained poorly with Alcian blue and contained low aggrecan transcript levels. A comparable difference was seen in cultured wild-type versus Ihh(-/-) synchondrosis chondrocytes. Treatment with exogenous Ihh did not fully restore normal proteoglycan levels in mutant cultures, but a combination of Ihh and BMP-2 did. In summary, Ihh is required for multiple processes during synchondrosis and cranial base development, including growth plate zone organization, chondrocyte orientation, and proteoglycan production. The cranial base appears to be a skeletal structure in which growth and ossification patterns along its antero-posterior axis are orchestrated by both Ihh and Shh.


Subject(s)
Chondrogenesis/physiology , Growth Plate/embryology , Growth Plate/metabolism , Hedgehog Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Skull Base/embryology , Aggrecans/genetics , Animals , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/pathology , Gene Expression Regulation, Developmental , Genetic Markers , Growth Plate/cytology , Hypertrophy , Intracellular Signaling Peptides and Proteins/deficiency , Mice , Occipital Lobe/cytology , Osteogenesis/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skull Base/cytology , Skull Base/pathology
6.
Development ; 132(5): 1057-67, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15689378

ABSTRACT

A null mutation in the morphogen Indian hedgehog (IHH) results in an embryonic lethal phenotype characterized by the conspicuous absence of bony tissue in the extremities. We show that this ossification defect is not attributable to a permanent arrest in cartilage differentiation, since Ihh(-/-) chondrocytes undergo hypertrophy and terminal differentiation, express angiogenic markers such as Vegf, and are invaded, albeit aberrantly, by blood vessels. Subsequent steps, including vessel expansion and persistence, are impaired, and the net result is degraded cartilage matrix that is devoid of blood vessels. The absence of blood vessels is not because the Ihh(-/-) skeleton is anti-angiogenic; in fact, in an ex vivo environment, both wild-type and Ihh mutant vessels invade the Ihh(-/-) cartilage, though only wild-type vessels expand to create the marrow cavity. In the ex vivo setting, Ihh(-/-) cells differentiate into osteoblasts and deposit a bony matrix, without benefit of exogenous hedgehog in the new environment. Even more surprising is our finding that the earliest IHH-dependent skeletal defect is obvious by the time the limb mesenchyme segregates into chondrogenic and perichondrogenic condensations. Although Ihh(-/-) cells organize into chondrogenic condensations similar in size and shape to wild-type condensations, perichondrial cells surrounding the mutant condensations are clearly faulty. They fail to aggregate, elongate and flatten into a definitive, endothelial cell-rich perichondrium like their wild-type counterparts. Normally, these cells surrounding the chondrogenic condensation are exposed to IHH, as evidenced by their expression of the hedgehog target genes, patched (Ptch) and Gli1. In the mutant environment, the milieu surrounding the cartilage -- comprising osteoblast precursors and endothelial cells -- as well as the cartilage itself, develop in the absence of this important morphogen. In conclusion, the skeletal phenotype of Ihh(-/-) embryos represents the sum of disturbances in three separate cell populations, the chondrocytes, the osteoblasts and the vasculature, each of which is a direct target of hedgehog signaling.


Subject(s)
Cartilage/embryology , Neovascularization, Physiologic , Pericytes/metabolism , Trans-Activators/physiology , Alleles , Animals , Bone and Bones/embryology , Cartilage/metabolism , Cell Differentiation , Cell Lineage , Endothelium, Vascular/cytology , Hedgehog Proteins , Hypertrophy , Immunohistochemistry , In Situ Hybridization , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Osteoblasts/cytology , Phenotype , Signal Transduction , Time Factors , Trans-Activators/genetics
7.
Development ; 129(2): 361-72, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11807029

ABSTRACT

Blood islands, the precursors of yolk sac blood vessels, contain primitive erythrocytes surrounded by a layer of endothelial cells. These structures differentiate from extra-embryonic mesodermal cells that underlie the visceral endoderm. Our previous studies have shown that Indian hedgehog (Ihh) is expressed in the visceral endoderm both in the visceral yolk sac in vivo and in embryonic stem (ES) cell-derived embryoid bodies. Differentiating embryoid bodies form blood islands, providing an in vitro model for studying vasculogenesis and hematopoiesis. A role for Ihh in yolk sac function is suggested by the observation that roughly 50% of Ihh(-/-) mice die at mid-gestation, potentially owing to vascular defects in the yolk sac. To address the nature of the possible vascular defects, we have examined the ability of ES cells deficient for Ihh or smoothened (Smo), which encodes a receptor component essential for all hedgehog signaling, to form blood islands in vitro. Embryoid bodies derived from these cell lines are unable to form blood islands, and express reduced levels of both PECAM1, an endothelial cell marker, and alpha-SMA, a vascular smooth muscle marker. RT-PCR analysis in the Ihh(-/-) lines shows a substantial decrease in the expression of Flk1 and Tal1, markers for the hemangioblast, the precursor of both blood and endothelial cells, as well as Flt1, an angiogenesis marker. To extend these observations, we have examined the phenotypes of embryo yolk sacs deficient for Ihh or SMO: Whereas Ihh(-/-) yolk sacs can form blood vessels, the vessels are fewer in number and smaller, perhaps owing to their inability to undergo vascular remodeling. Smo(-/-) yolk sacs arrest at an earlier stage: the endothelial tubes are packed with hematopoietic cells, and fail to undergo even the limited vascular remodeling observed in the Ihh(-/-) yolk sacs. Our study supports a role for hedgehog signaling in yolk sac angiogenesis.


Subject(s)
Embryo, Mammalian/physiology , Neovascularization, Physiologic , Receptors, G-Protein-Coupled , Trans-Activators/metabolism , Yolk Sac/blood supply , Animals , Biomarkers , Cell Differentiation/physiology , Embryo, Mammalian/cytology , Genes, Reporter , Hedgehog Proteins , Hematopoiesis , Immunohistochemistry , In Situ Hybridization , Mice , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Smoothened Receptor , Stem Cells/physiology , Trans-Activators/genetics , Yolk Sac/growth & development , Yolk Sac/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...