Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
2.
JCI Insight ; 5(4)2020 02 27.
Article in English | MEDLINE | ID: mdl-31990680

ABSTRACT

BACKGROUNDLiver disease in urea cycle disorders (UCDs) ranges from hepatomegaly and chronic hepatocellular injury to cirrhosis and end-stage liver disease. However, the prevalence and underlying mechanisms are unclear.METHODSWe estimated the prevalence of chronic hepatocellular injury in UCDs using data from a multicenter, longitudinal, natural history study. We also used ultrasound with shear wave elastography and FibroTest to evaluate liver stiffness and markers of fibrosis in individuals with argininosuccinate lyase deficiency (ASLD), a disorder with high prevalence of elevated serum alanine aminotransferase (ALT). To understand the human observations, we evaluated the hepatic phenotype of the AslNeo/Neo mouse model of ASLD.RESULTSWe demonstrate a high prevalence of elevated ALT in ASLD (37%). Hyperammonemia and use of nitrogen-scavenging agents, 2 markers of disease severity, were significantly (P < 0.001 and P = 0.001, respectively) associated with elevated ALT in ASLD. In addition, ultrasound with shear wave elastography and FibroTest revealed increased echogenicity and liver stiffness, even in individuals with ASLD and normal aminotransferases. The AslNeo/Neo mice mimic the human disorder with hepatomegaly, elevated aminotransferases, and excessive hepatic glycogen noted before death (3-5 weeks of age). This excessive hepatic glycogen is associated with impaired hepatic glycogenolysis and decreased glycogen phosphorylase and is rescued with helper-dependent adenovirus expressing Asl using a liver-specific (ApoE) promoter.CONCLUSIONOur results link urea cycle dysfunction and impaired hepatic glucose metabolism and identify a mouse model of liver disease in the setting of urea cycle dysfunction.TRIAL REGISTRATIONThis study has been registered at ClinicalTrials.gov (NCT03721367, NCT00237315).FUNDINGFunding was provided by NIH, Burroughs Wellcome Fund, NUCDF, Genzyme/ACMG Foundation, and CPRIT.


Subject(s)
Argininosuccinate Lyase/metabolism , Liver Diseases/metabolism , Liver Glycogen/metabolism , Alanine Transaminase/blood , Animals , Chronic Disease , Disease Models, Animal , Humans , Liver Diseases/complications , Liver Diseases/enzymology , Longitudinal Studies , Mice , Urea Cycle Disorders, Inborn/complications
3.
JBMR Plus ; 2(4): 235-239, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30283904

ABSTRACT

The heritable disorder osteogenesis imperfecta (OI) is characterized by bone fragility and low bone mass. OI type VI is an autosomal recessive form of the disorder with moderate to severe bone fragility. OI type VI is caused by mutations in the serpin peptidase inhibitor, clade F, member 1 (SERPINF1), the gene coding for pigment epithelium-derived factor (PEDF). Here, we report a patient with OI type VI caused by a novel homozygous intronic variant in SERPINF1 identified by whole-exome sequencing (WES). The mutation was not identified using a low bone mass gene panel based on next-generation sequencing. This variant creates a novel consensus splice donor site (AGGC to AGGT) in intron 4. Analysis of cDNA generated from fibroblasts revealed retention of a 32-bp intronic fragment between exons 4 and 5 in the cDNA, a result of alternative splicing from the novel splice-donor site. As a result, the aberrant insertion of this intronic fragment generated a frameshift pathogenic variant and induced nonsense-mediated decay. Furthermore, gene expression by quantitative PCR showed SERPINF1 expression was dramatically reduced in patient fibroblasts, and PEDF level was also significantly reduced in the patient's plasma. In conclusion, we report a novel homozygous variant that generates an alternative splice-donor in intron 4 of SERPINF1 which gives rise to severe bone fragility. The work also demonstrates clinical utility of WES analysis, and consideration of noncoding variants, in the diagnostic setting of rare bone diseases. © 2018 The Authors. JBMR Plus is published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.

4.
Am J Hum Genet ; 103(2): 276-287, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30075114

ABSTRACT

Primary hypertension is a major risk factor for ischemic heart disease, stroke, and chronic kidney disease. Insights obtained from the study of rare Mendelian forms of hypertension have been invaluable in elucidating the mechanisms causing primary hypertension and development of antihypertensive therapies. Endothelial cells play a key role in the regulation of blood pressure; however, a Mendelian form of hypertension that is primarily due to endothelial dysfunction has not yet been described. Here, we show that the urea cycle disorder, argininosuccinate lyase deficiency (ASLD), can manifest as a Mendelian form of endothelial-dependent hypertension. Using data from a human clinical study, a mouse model with endothelial-specific deletion of argininosuccinate lyase (Asl), and in vitro studies in human aortic endothelial cells and induced pluripotent stem cell-derived endothelial cells from individuals with ASLD, we show that loss of ASL in endothelial cells leads to endothelial-dependent vascular dysfunction with reduced nitric oxide (NO) production, increased oxidative stress, and impaired angiogenesis. Our findings show that ASLD is a unique model for studying NO-dependent endothelial dysfunction in human hypertension.


Subject(s)
Argininosuccinate Lyase/genetics , Argininosuccinic Aciduria/genetics , Endothelial Cells/pathology , Hypertension/genetics , Adolescent , Animals , Blood Pressure/genetics , Cells, Cultured , Child , Disease Models, Animal , Endothelium, Vascular/pathology , Female , Humans , Male , Mice , Mice, Transgenic , Neovascularization, Pathologic/genetics , Nitric Oxide/genetics , Oxidative Stress/genetics , Urea Cycle Disorders, Inborn/genetics
5.
Nat Commun ; 8: 15000, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28397831

ABSTRACT

Osteocytes are the terminally differentiated cell type of the osteoblastic lineage and have important functions in skeletal homeostasis. Although the transcriptional regulation of osteoblast differentiation has been well characterized, the factors that regulate differentiation of osteocytes from mature osteoblasts are poorly understood. Here we show that miR-23a∼27a∼24-2 (miR-23a cluster) promotes osteocyte differentiation. Osteoblast-specific miR-23a cluster gain-of-function mice have low bone mass associated with decreased osteoblast but increased osteocyte numbers. By contrast, loss-of-function transgenic mice overexpressing microRNA decoys for either miR-23a or miR-27a, but not miR24-2, show decreased osteocyte numbers. Moreover, RNA-sequencing analysis shows altered transforming growth factor-ß (TGF-ß) signalling. Prdm16, a negative regulator of the TGF-ß pathway, is directly repressed by miR-27a with concomitant alteration of sclerostin expression, and pharmacological inhibition of TGF-ß rescues the phenotypes observed in the gain-of-function transgenic mice. Taken together, the miR-23a cluster regulates osteocyte differentiation by modulating the TGF-ß signalling pathway through targeting of Prdm16.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation , MicroRNAs/genetics , Osteocytes/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Animals , Cell Line , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Female , HEK293 Cells , Humans , Mice, Transgenic , Models, Genetic , Multigene Family , Osteocytes/cytology , Osteogenesis/genetics , Transforming Growth Factor beta/metabolism
6.
J Clin Invest ; 127(4): 1475-1484, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28263186

ABSTRACT

Shohat-type spondyloepimetaphyseal dysplasia (SEMD) is a skeletal dysplasia that affects cartilage development. Similar skeletal disorders, such as spondyloepiphyseal dysplasias, are linked to mutations in type II collagen (COL2A1), but the causative gene in SEMD is not known. Here, we have performed whole-exome sequencing to identify a recurrent homozygous c.408+1G>A donor splice site loss-of-function mutation in DDRGK domain containing 1 (DDRGK1) in 4 families affected by SEMD. In zebrafish, ddrgk1 deficiency disrupted craniofacial cartilage development and led to decreased levels of the chondrogenic master transcription factor sox9 and its downstream target, col2a1. Overexpression of sox9 rescued the zebrafish chondrogenic and craniofacial phenotype generated by ddrgk1 knockdown, thus identifying DDRGK1 as a regulator of SOX9. Consistent with these results, Ddrgk1-/- mice displayed delayed limb bud chondrogenic condensation, decreased SOX9 protein expression and Col2a1 transcript levels, and increased apoptosis. Furthermore, we determined that DDRGK1 can directly bind to SOX9 to inhibit its ubiquitination and proteasomal degradation. Taken together, these data indicate that loss of DDRGK1 decreases SOX9 expression and causes a human skeletal dysplasia, identifying a mechanism that regulates chondrogenesis via modulation of SOX9 ubiquitination.


Subject(s)
Carrier Proteins/physiology , Osteochondrodysplasias/genetics , SOX9 Transcription Factor/metabolism , Ubiquitination , Adaptor Proteins, Signal Transducing , Animals , Cartilage/growth & development , Cells, Cultured , Chondrogenesis , Collagen Type II/genetics , Collagen Type II/metabolism , Gene Expression , Genetic Association Studies , Genetic Predisposition to Disease , Homozygote , Humans , Mice, Knockout , Mutation , Osteochondrodysplasias/metabolism , Pedigree , Protein Isoforms/physiology , Protein Stability , RNA Splice Sites , Zebrafish
7.
J Bone Miner Res ; 31(5): 1030-40, 2016 05.
Article in English | MEDLINE | ID: mdl-26716893

ABSTRACT

Osteogenesis imperfecta (OI) is characterized by low bone mass, poor bone quality, and fractures. Standard treatment for OI patients is limited to bisphosphonates, which only incompletely correct the bone phenotype, and seem to be less effective in adults. Sclerostin-neutralizing antibodies (Scl-Ab) have been shown to be beneficial in animal models of osteoporosis, and dominant OI resulting from mutations in the genes encoding type I collagen. However, Scl-Ab treatment has not been studied in models of recessive OI. Cartilage-associated protein (CRTAP) is involved in posttranslational type I collagen modification, and its loss of function results in recessive OI. In this study, we treated 1-week-old and 6-week-old Crtap(-/-) mice with Scl-Ab for 6 weeks (25 mg/kg, s.c., twice per week), to determine the effects on the bone phenotype in models of "pediatric" and "young adult" recessive OI. Vehicle-treated Crtap(-/-) and wild-type (WT) mice served as controls. Compared with control Crtap(-/-) mice, micro-computed tomography (µCT) analyses showed significant increases in bone volume and improved trabecular microarchitecture in Scl-Ab-treated Crtap(-/-) mice in both age cohorts, in both vertebrae and femurs. Additionally, Scl-Ab improved femoral cortical parameters in both age cohorts. Biomechanical testing showed that Scl-Ab improved parameters of whole-bone strength in Crtap(-/-) mice, with more robust effects in the week 6 to 12 cohort, but did not affect the increased bone brittleness. Additionally, Scl-Ab normalized the increased osteoclast numbers, stimulated bone formation rate (week 6 to 12 cohort only), but did not affect osteocyte density. Overall, our findings suggest that Scl-Ab treatment may be beneficial in the treatment of recessive OI caused by defects in collagen posttranslational modification. © 2015 American Society for Bone and Mineral Research.


Subject(s)
Antibodies/pharmacology , Genes, Recessive , Glycoproteins/antagonists & inhibitors , Osteoclasts/metabolism , Osteogenesis Imperfecta , Osteogenesis , Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , Extracellular Matrix Proteins , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Molecular Chaperones , Osteogenesis/drug effects , Osteogenesis/genetics , Osteogenesis Imperfecta/drug therapy , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/metabolism , Osteogenesis Imperfecta/pathology
8.
Mol Genet Metab ; 117(3): 378-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26693895

ABSTRACT

Osteogenesis imperfecta (OI) is a group of genetic disorders characterized by bone fragility and deformity. OI type VI is unique owing to the mineralization defects observed in patient biopsies. Furthermore, it has been reported to respond less well to standard therapy with bisphosphonates [1]. Others and we have previously identified SERPINF1 mutations in patients with OI type VI. SERPINF1 encodes pigment epithelium derived factor (PEDF), a secreted collagen-binding glycoprotein that is absent in the sera of patients with OI type VI. Serpinf1 null mice show increased osteoid and decreased bone mass, and thus recapitulate the OI type VI phenotype. We tested whether restoration of circulating PEDF in the blood could correct the phenotype of OI type VI in the context of protein replacement. To do so, we utilized a helper-dependent adenoviral vector (HDAd) to express human SERPINF1 in the mouse liver and assessed whether PEDF secreted from the liver was able to rescue the bone phenotype observed in Serpinf1(-/-) mice. We confirmed that expression of SERPINF1 in the liver restored the serum level of PEDF. We also demonstrated that PEDF secreted from the liver was biologically active by showing the expected metabolic effects of increased adiposity and impaired glucose tolerance in Serpinf1(-/-) mice. Interestingly, overexpression of PEDF in vitro increased mineralization with a concomitant increase in the expression of bone gamma-carboxyglutamate protein, alkaline phosphatase and collagen, type I, alpha I, but the increased serum PEDF level did not improve the bone phenotype of Serpinf1(-/-) mice. These results suggest that PEDF may function in a context-dependent and paracrine fashion in bone homeostasis.


Subject(s)
Bone and Bones/physiology , Eye Proteins/blood , Eye Proteins/genetics , Liver/metabolism , Nerve Growth Factors/blood , Nerve Growth Factors/genetics , Osteogenesis Imperfecta/physiopathology , Osteogenesis Imperfecta/therapy , Serpins/blood , Serpins/genetics , 1-Carboxyglutamic Acid/genetics , Adenoviridae/genetics , Alkaline Phosphatase/genetics , Animals , Bone Density , Collagen Type I/genetics , Gene Transfer Techniques , Glucose Intolerance , HEK293 Cells , Homeostasis , Humans , Mice , Mice, Knockout , Mutation , Nerve Growth Factors/deficiency , Phenotype , Serpins/deficiency
9.
Mol Genet Metab ; 115(1): 53-60, 2015 May.
Article in English | MEDLINE | ID: mdl-25779879

ABSTRACT

Angiotensin receptor blockers (ARBs) are a group of anti-hypertensive drugs that are widely used to treat pediatric hypertension. Recent application of ARBs to treat diseases such as Marfan syndrome or Alport syndrome has shown positive outcomes in animal and human studies, suggesting a broader therapeutic potential for this class of drugs. Multiple studies have reported a benefit of ARBs on adult bone homeostasis; however, its effect on the growing skeleton in children is unknown. We investigated the effect of Losartan, an ARB, in regulating bone mass and cartilage during development in mice. Wild type mice were treated with Losartan from birth until 6 weeks of age, after which bones were collected for microCT and histomorphometric analyses. Losartan increased trabecular bone volume vs. tissue volume (a 98% increase) and cortical thickness (a 9% increase) in 6-weeks old wild type mice. The bone changes were attributed to decreased osteoclastogenesis as demonstrated by reduced osteoclast number per bone surface in vivo and suppressed osteoclast differentiation in vitro. At the molecular level, Angiotensin II-induced ERK1/2 phosphorylation in RAW cells was attenuated by Losartan. Similarly, RANKL-induced ERK1/2 phosphorylation was suppressed by Losartan, suggesting a convergence of RANKL and angiotensin signaling at the level of ERK1/2 regulation. To assess the effect of Losartan on cartilage development, we examined the cartilage phenotype of wild type mice treated with Losartan in utero from conception to 1 day of age. Growth plates of these mice showed an elongated hypertrophic chondrocyte zone and increased Col10a1 expression level, with minimal changes in chondrocyte proliferation. Altogether, inhibition of the angiotensin pathway by Losartan increases bone mass and accelerates chondrocyte hypertrophy in growth plate during skeletal development.


Subject(s)
Bone Density/drug effects , Bone Development/drug effects , Bone and Bones/drug effects , Chondrocytes/drug effects , Losartan/pharmacology , Angiotensins/drug effects , Angiotensins/metabolism , Animals , Bone Density/physiology , Bone and Bones/diagnostic imaging , Bone and Bones/ultrastructure , Cartilage/drug effects , Cell Differentiation , Chondrocytes/physiology , Female , Growth Plate/drug effects , Hypertrophy/etiology , Losartan/administration & dosage , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Osteoclasts/drug effects , Osteoclasts/physiology , Phosphorylation , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , RAW 264.7 Cells , Radiography
10.
J Bone Miner Res ; 30(3): 489-98, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25251575

ABSTRACT

Osteogenesis imperfecta (OI) type V is characterized by increased bone fragility, long bone deformities, hyperplastic callus formation, and calcification of interosseous membranes. It is caused by a recurrent mutation in the 5' UTR of the IFITM5 gene (c.-14C > T). This mutation introduces an alternative start codon, adding 5 amino acid residues to the N-terminus of the protein. The mechanism whereby this novel IFITM5 protein causes OI type V is yet to be defined. To address this, we created transgenic mice expressing either the wild-type or the OI type V mutant IFITM5 under the control of an osteoblast-specific Col1a1 2.3-kb promoter. These mutant IFITM5 transgenic mice exhibited perinatal lethality, whereas wild-type IFITM5 transgenic mice showed normal growth and development. Skeletal preparations and radiographs performed on E15.5 and E18.5 OI type V transgenic embryos revealed delayed/abnormal mineralization and skeletal defects, including abnormal rib cage formation, long bone deformities, and fractures. Primary osteoblast cultures, derived from mutant mice calvaria at E18.5, showed decreased mineralization by Alizarin red staining, and RNA isolated from calvaria showed reduced expression of osteoblast differentiation markers such as Osteocalcin, compared with nontransgenic littermates and wild-type mice calvaria, consistent with the in vivo phenotype. Importantly, overexpression of wild-type Ifitm5 did not manifest a significant bone phenotype. Collectively, our results suggest that expression of mutant IFITM5 causes abnormal skeletal development, low bone mass, and abnormal osteoblast differentiation. Given that neither overexpression of the wild-type Ifitm5, as shown in our model, nor knock-out of Ifitm5, as previously published, showed significant bone abnormalities, we conclude that the IFITM5 mutation in OI type V acts in a neomorphic fashion.


Subject(s)
Membrane Proteins/genetics , Mutation , Animals , Mice , Mice, Transgenic
11.
Cancer Cell ; 26(3): 390-401, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25203324

ABSTRACT

Osteogenic sarcoma (OS) is a deadly skeletal malignancy whose cause is unknown. We report here a mouse model of OS based on conditional expression of the intracellular domain of Notch1 (NICD). Expression of the NICD in immature osteoblasts was sufficient to drive the formation of bone tumors, including OS, with complete penetrance. These tumors display features of human OS; namely, histopathology, cytogenetic complexity, and metastatic potential. We show that Notch activation combined with loss of p53 synergistically accelerates OS development in mice, although p53-driven OS is not Rbpj dependent, which demonstrates a dual dominance of the Notch oncogene and p53 mutation in the development of OS. Using this model, we also reveal the osteoblasts as the potential sources of OS.


Subject(s)
Bone Neoplasms/metabolism , Osteosarcoma/metabolism , Receptor, Notch1/genetics , Animals , Bone Neoplasms/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Neoplasm Transplantation , Osteoblasts/metabolism , Osteosarcoma/pathology , Protein Structure, Tertiary , Receptor, Notch1/metabolism , Transcriptome
12.
Nat Med ; 20(6): 670-5, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24793237

ABSTRACT

Osteogenesis imperfecta (OI) is a heritable disorder, in both a dominant and recessive manner, of connective tissue characterized by brittle bones, fractures and extraskeletal manifestations. How structural mutations of type I collagen (dominant OI) or of its post-translational modification machinery (recessive OI) can cause abnormal quality and quantity of bone is poorly understood. Notably, the clinical overlap between dominant and recessive forms of OI suggests common molecular pathomechanisms. Here, we show that excessive transforming growth factor-ß (TGF-ß) signaling is a mechanism of OI in both recessive (Crtap(-/-)) and dominant (Col1a2(tm1.1Mcbr)) OI mouse models. In the skeleton, we find higher expression of TGF-ß target genes, higher ratio of phosphorylated Smad2 to total Smad2 protein and higher in vivo Smad2 reporter activity. Moreover, the type I collagen of Crtap(-/-) mice shows reduced binding to the small leucine-rich proteoglycan decorin, a known regulator of TGF-ß activity. Anti-TGF-ß treatment using the neutralizing antibody 1D11 corrects the bone phenotype in both forms of OI and improves the lung abnormalities in Crtap(-/-) mice. Hence, altered TGF-ß matrix-cell signaling is a primary mechanism in the pathogenesis of OI and could be a promising target for the treatment of OI.


Subject(s)
Osteogenesis Imperfecta/physiopathology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Analysis of Variance , Animals , Collagen Type I/genetics , Electrophoresis, Polyacrylamide Gel , Extracellular Matrix Proteins , Female , Immunoblotting , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones , Osteogenesis Imperfecta/metabolism , Proteins/genetics , Real-Time Polymerase Chain Reaction , Surface Plasmon Resonance , X-Ray Microtomography
13.
Hum Mol Genet ; 23(18): 4822-31, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24777781

ABSTRACT

Osteogenesis imperfecta (OI) is an inherited brittle bone disorder characterized by bone fragility and low bone mass. Loss of function mutations in FK506-binding protein 10 (FKBP10), encoding the FKBP65 protein, result in recessive OI and Bruck syndrome, of which the latter is additionally characterized by joint contractures. FKBP65 is thought to act as a collagen chaperone, but it is unknown how loss of FKBP65 affects collagen synthesis and extracellular matrix formation. We evaluated the developmental and postnatal expression of Fkbp10 and analyzed the consequences of its generalized loss of function. Fkbp10 is expressed at low levels in E13.5 mouse embryos, particularly in skeletal tissues, and steadily increases through E17.5 with expression in not only skeletal tissues, but also in visceral tissues. Postnatally, expression is limited to developing bone and ligaments. In contrast to humans, with complete loss of function mutations, Fkbp10(-/-) mice do not survive birth, and embryos present with growth delay and tissue fragility. Type I calvarial collagen isolated from these mice showed reduced stable crosslink formation at telopeptide lysines. Furthermore, Fkbp10(-/-) mouse embryonic fibroblasts show retention of procollagen in the cell layer and associated dilated endoplasmic reticulum. These data suggest a requirement for FKBP65 function during embryonic connective tissue development in mice, but the restricted expression postnatally in bone, ligaments and tendons correlates with the bone fragility and contracture phenotype in humans.


Subject(s)
Connective Tissue/physiology , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Animals , Animals, Newborn , Bone and Bones/metabolism , Connective Tissue/embryology , Disease Models, Animal , Embryo, Mammalian , Genes, Lethal , Humans , Ligaments/metabolism , Mice , Mice, Inbred C57BL , Tendons/metabolism
14.
Hum Mol Genet ; 23(15): 4035-42, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24634143

ABSTRACT

Osteogenesis imperfecta (OI) is a heritable disorder of connective tissue characterized by bone fragility and low bone mass. Recently, our group and others reported that WNT1 recessive mutations cause OI, whereas WNT1 heterozygous mutations cause early onset osteoporosis. These findings support the hypothesis that WNT1 is an important WNT ligand regulating bone formation and bone homeostasis. While these studies provided strong human genetic and in vitro functional data, an in vivo animal model to study the mechanism of WNT1 function in bone is lacking. Here, we show that Swaying (Wnt1(sw/sw)) mice previously reported to carry a spontaneous mutation in Wnt1 share major features of OI including propensity to fractures and severe osteopenia. In addition, biomechanical and biochemical analyses showed that Wnt1(sw/sw) mice exhibit reduced bone strength with altered levels of mineral and collagen in the bone matrix that is also distinct from the type I collagen-related form of OI. Further histomorphometric analyses and gene expression studies demonstrate that the bone phenotype is associated with defects in osteoblast activity and function. Our study thus provides in vivo evidence that WNT1 mutations contribute to bone fragility in OI patients and demonstrates that the Wnt1(sw/sw) mouse is a murine model of OI caused by WNT1 mutations.


Subject(s)
Bone and Bones/metabolism , Fractures, Bone/genetics , Mutation , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis Imperfecta/genetics , Wnt1 Protein/genetics , Animals , Bone Density/genetics , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Bone and Bones/pathology , Disease Models, Animal , Female , Fractures, Bone/metabolism , Fractures, Bone/pathology , Gene Expression , Heterozygote , Homozygote , Humans , Male , Mice , Osteoblasts/pathology , Osteoclasts/pathology , Osteogenesis Imperfecta/metabolism , Osteogenesis Imperfecta/pathology , Phenotype , Wnt1 Protein/metabolism
15.
PLoS Genet ; 10(1): e1004121, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24465224

ABSTRACT

Mutations in the genes encoding cartilage associated protein (CRTAP) and prolyl 3-hydroxylase 1 (P3H1 encoded by LEPRE1) were the first identified causes of recessive Osteogenesis Imperfecta (OI). These proteins, together with cyclophilin B (encoded by PPIB), form a complex that 3-hydroxylates a single proline residue on the α1(I) chain (Pro986) and has cis/trans isomerase (PPIase) activity essential for proper collagen folding. Recent data suggest that prolyl 3-hydroxylation of Pro986 is not required for the structural stability of collagen; however, the absence of this post-translational modification may disrupt protein-protein interactions integral for proper collagen folding and lead to collagen over-modification. P3H1 and CRTAP stabilize each other and absence of one results in degradation of the other. Hence, hypomorphic or loss of function mutations of either gene cause loss of the whole complex and its associated functions. The relative contribution of losing this complex's 3-hydroxylation versus PPIase and collagen chaperone activities to the phenotype of recessive OI is unknown. To distinguish between these functions, we generated knock-in mice carrying a single amino acid substitution in the catalytic site of P3h1 (Lepre1(H662A) ). This substitution abolished P3h1 activity but retained ability to form a complex with Crtap and thus the collagen chaperone function. Knock-in mice showed absence of prolyl 3-hydroxylation at Pro986 of the α1(I) and α1(II) collagen chains but no significant over-modification at other collagen residues. They were normal in appearance, had no growth defects and normal cartilage growth plate histology but showed decreased trabecular bone mass. This new mouse model recapitulates elements of the bone phenotype of OI but not the cartilage and growth phenotypes caused by loss of the prolyl 3-hydroxylation complex. Our observations suggest differential tissue consequences due to selective inactivation of P3H1 hydroxylase activity versus complete ablation of the prolyl 3-hydroxylation complex.


Subject(s)
Collagen/genetics , Hydroxylation/genetics , Membrane Glycoproteins/genetics , Osteogenesis Imperfecta/genetics , Osteogenesis/genetics , Proteins/genetics , Proteoglycans/genetics , Animals , Collagen/chemistry , Cyclophilins/genetics , Extracellular Matrix Proteins , Gene Knock-In Techniques , Membrane Glycoproteins/metabolism , Mice , Molecular Chaperones , Osteogenesis Imperfecta/pathology , Protein Folding , Protein Interaction Maps , Protein Processing, Post-Translational , Proteins/metabolism , Proteoglycans/metabolism , Skeleton
16.
Mol Ther Oncolytics ; 1: 14008, 2014.
Article in English | MEDLINE | ID: mdl-27119096

ABSTRACT

Oncolytic adenoviruses (Onc.Ads) produce significant antitumor effects but as single agents they rarely eliminate tumors. Investigators have therefore incorporated sequences into these vectors that encode immunomodulatory molecules to enhance antitumor immunity. Successful implementation of this strategy requires multiple tumor immune inhibitory mechanisms to be overcome, and insertion of the corresponding multiple functional genes reduces the titer and replication of Onc.Ads, compromising their direct ant-tumor effects. By contrast, helper-dependent (HD) Ads are devoid of viral coding sequences, allowing inclusion of multiple transgenes. HDAds, however, lack replicative capacity. Since HDAds encode the adenoviral packaging signal, we hypothesized that the coadministration of Onc.Ad with HDAd would allow to be amplified and packaged during replication of Onc.Ad in transduced cancer cells. This combination could provide immunostimulation without losing oncolytic activity. We now show that coinfection of Onc.Ad with HDAd subsequently replicates HDAd vector DNA in trans in human cancer cell lines in vitro and in vivo, amplifying the transgenes the HDAd encode. This combinatorial treatment significantly suppresses the tumor growth compared to treatment with a single agent in an immunocompetent mouse model. Hence, combinatorial treatment of Onc.Ad with HDAd should overcome the inherent limitations of each agent and provide a highly immunogenic oncolytic therapy.

17.
Proc Natl Acad Sci U S A ; 110(18): 7336-41, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23589896

ABSTRACT

TGF-ß is abundantly produced in the skeletal system and plays a crucial role in skeletal homeostasis. E-selectin ligand-1 (ESL-1), a Golgi apparatus-localized protein, acts as a negative regulator of TGF-ß bioavailability by attenuating maturation of pro-TGF-ß during cartilage homeostasis. However, whether regulation of intracellular TGF-ß maturation by ESL-1 is also crucial during bone homeostasis has not been well defined. Here, we show that Esl-1(-/-) mice exhibit a severe osteopenia with elevated bone resorption and decreased bone mineralization. In primary culture, Esl-1(-/-) osteoclast progenitors show no difference in osteoclastogenesis. However, Esl-1(-/-) osteoblasts show delayed differentiation and mineralization and stimulate osteoclastogenesis more potently in the osteoblast-osteoclast coculture, suggesting that ESL-1 primarily acts in osteoblasts to regulate bone homeostasis. In addition, Esl-1(-/-) calvaria exhibit an elevated mature TGF-ß/pro-TGF-ß ratio, with increased expression of TGF-ß downstream targets (plasminogen activator inhibitor-1, parathyroid hormone-related peptide, connective tissue growth factor, and matrix metallopeptidase 13, etc.) and a key regulator of osteoclastogenesis (receptor activator of nuclear factor κB ligand). Moreover, in vivo treatment with 1D11, a pan-TGF-ß antibody, significantly improved the low bone mass of Esl-1(-/-) mice, suggesting that elevated TGF-ß signaling is the major cause of osteopenia in Esl-1(-/-) mice. In summary, our study identifies ESL-1 as an important regulator of bone remodeling and demonstrates that the modulation of TGF-ß maturation is pivotal in the maintenance of a homeostatic bone microenvironment and for proper osteoblast-osteoclast coupling.


Subject(s)
Bone Remodeling , Receptors, Fibroblast Growth Factor/metabolism , Sialoglycoproteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antibodies/pharmacology , Bone Diseases, Metabolic/complications , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Bone Diseases, Metabolic/physiopathology , Bone Remodeling/drug effects , Bone Remodeling/genetics , Bone Resorption/complications , Bone Resorption/genetics , Bone Resorption/pathology , Bone Resorption/physiopathology , Calcification, Physiologic/drug effects , Calcification, Physiologic/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Cell Lineage/genetics , Cells, Cultured , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Femur/physiopathology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Mice , Organ Size/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/drug effects , Osteogenesis/genetics , Phenotype , Radiography , Receptors, Fibroblast Growth Factor/deficiency , Sialoglycoproteins/deficiency , Signal Transduction/genetics
18.
Sci Transl Med ; 5(176): 176ra34, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23486780

ABSTRACT

Osteoarthritis (OA) is a common degenerative condition that afflicts more than 70% of the population between 55 and 77 years of age. Although its prevalence is rising globally with aging of the population, current therapy is limited to symptomatic relief and, in severe cases, joint replacement surgery. We report that intra-articular expression of proteoglycan 4 (Prg4) in mice protects against development of OA. Long-term Prg4 expression under the type II collagen promoter (Col2a1) does not adversely affect skeletal development but protects from developing signs of age-related OA. The protective effect is also shown in a model of posttraumatic OA created by cruciate ligament transection. Moreover, intra-articular injection of helper-dependent adenoviral vector expressing Prg4 protected against the development of posttraumatic OA when administered either before or after injury. Gene expression profiling of mouse articular cartilage and in vitro cell studies show that Prg4 expression inhibits the transcriptional programs that promote cartilage catabolism and hypertrophy through the up-regulation of hypoxia-inducible factor 3α. Analyses of available human OA data sets are consistent with the predictions of this model. Hence, our data provide insight into the mechanisms for OA development and offer a potential chondroprotective approach to its treatment.


Subject(s)
Osteoarthritis/metabolism , Osteoarthritis/prevention & control , Proteoglycans/metabolism , Animals , Cell Line , Collagen Type II/genetics , Humans , Laser Capture Microdissection , Male , Mice , Mice, Transgenic , Osteoarthritis/genetics , Promoter Regions, Genetic/genetics , Proteoglycans/genetics
19.
Mol Ther ; 21(4): 767-74, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23358188

ABSTRACT

Helper-dependent adenoviral (HDAd) vectors can mediate long-term, high-level transgene expression from transduced hepatocytes with no chronic toxicity. However, a toxic acute response with potentially lethal consequences has hindered their clinical applications. Liver sinusoidal endothelial cells (LSECs) and Kupffer cells are major barriers to efficient hepatocyte transduction. Understanding the mechanisms of adenoviral vector uptake by non-parenchymal cells may allow the development of strategies aimed at overcoming these important barriers and to achieve preferential hepatocyte gene transfer with reduced toxicity. Scavenger receptors on Kupffer cells bind adenoviral particles and remove them from the circulation, thus preventing hepatocyte transduction. In the present study, we show that HDAd particles interact in vitro and in vivo with scavenger receptor-A (SR-A) and with scavenger receptor expressed on endothelial cells-I (SREC-I) and we exploited this knowledge to increase the efficiency of hepatocyte transduction by HDAd vectors in vivo through blocking of SR-A and SREC-I with specific fragments antigen-binding (Fabs).


Subject(s)
Adenoviridae/genetics , Asialoglycoprotein Receptor/genetics , Genetic Vectors/genetics , Receptors, Immunologic/genetics , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class F/genetics , Animals , Cell Line , Fluorescent Antibody Technique , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
20.
Mol Ther ; 21(4): 796-805, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23319058

ABSTRACT

We previously dissected the components of the innate immune response to Helper-dependent adenoviral vectors (HDAds) using genetic models, and demonstrated that multiple pattern recognition receptor signaling pathways contribute to this host response to HDAds in vivo. Based on analysis of cytokine expression profiles, type I interferon (IFN) mRNA is induced in host mouse livers at 1 hour post-injection. This type I IFN signaling amplifies cytokine expression in liver independent of the nature of vector DNA sequences after 3 hours post-injection. This type I IFN signaling in response to HDAds administration contributes to transcriptional silencing of both HDAd prokaryotic and eukaryotic DNA in liver. This silencing occurs early and is mediated by epigenetic modification as shown by in vivo chromatin immunoprecipitation (ChIP) with anti-histone deacetylase (HDAC) and promyelocytic leukemia protein (PML). In contrast, self-complementary adeno-associated viral vectors (scAAVs) showed significantly lower induction of type I IFN mRNA in liver compared to HDAds at both early and late time points. These results show that the type I IFN signaling dependent transgene silencing differs between AAV and HDAd vectors after liver-directed gene transfer.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Interferon Type I/genetics , Animals , Chromatin Immunoprecipitation , Helper Viruses/genetics , Histone Deacetylases/metabolism , Liver/metabolism , Mice , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Transcription Factors/metabolism , Transgenes/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...