Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Dev Biol ; 505: 42-57, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37827362

ABSTRACT

BAZ1B is one of 25-27 coding genes deleted in canonical Williams syndrome, a multi-system disorder causing slow growth, vascular stenosis, and gastrointestinal complaints, including constipation. BAZ1B is involved in (among other processes) chromatin organization, DNA damage repair, and mitosis, suggesting reduced BAZ1B may contribute to Williams syndrome symptoms. In mice, loss of Baz1b causes early neonatal death. 89.6% of Baz1b-/- mice die within 24 h of birth without vascular anomalies or congenital heart disease (except for patent ductus arteriosus). Some (<50%) Baz1b-/- were noted to have prolonged neonatal cyanosis, patent ductus arteriosus, or reduced lung aeration, and none developed a milk spot. Meanwhile, 35.5% of Baz1b+/- mice die over the first three weeks after birth. Surviving Baz1b heterozygotes grow slowly (with variable severity). 66.7% of Baz1b+/- mice develop bowel dilation, compared to 37.8% of wild-type mice, but small bowel and colon transit studies were normal. Additionally, enteric neuron density appeared normal in Baz1b-/- mice except in distal colon myenteric plexus, where neuron density was modestly elevated. Combined with several rare phenotypes (agnathia, microphthalmia, bowel dilation) recovered, our work confirms the importance of BAZ1B in survival and growth and suggests that reduced copy number of BAZ1B may contribute to the variability in Williams syndrome phenotypes.


Subject(s)
Ductus Arteriosus, Patent , Williams Syndrome , Animals , Mice , Colon , DNA Repair , Neurons , Williams Syndrome/genetics
2.
Int J Mol Sci ; 23(12)2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35743192

ABSTRACT

Lysyl oxidase (LOX) is a copper-binding enzyme that cross-links elastin and collagen. The dominant LOX variation contributes to familial thoracic aortic aneurysm. Previously reported murine Lox mutants had a mild phenotype and did not dilate without drug-induced provocation. Here, we present a new, more severe mutant, Loxb2b370.2Clo (c.G854T; p.Cys285Phe), whose mutation falls just N-terminal to the copper-binding domain. Unlike the other mutants, the C285F Lox protein was stably produced/secreted, and male C57Bl/6J Lox+/C285F mice exhibit increased systolic blood pressure (BP; p < 0.05) and reduced caliber aortas (p < 0.01 at 100mmHg) at 3 months that independently dilate by 6 months (p < 0.0001). Multimodal imaging reveals markedly irregular elastic sheets in the mutant (p = 2.8 × 10−8 for breaks by histology) that become increasingly disrupted with age (p < 0.05) and breeding into a high BP background (p = 6.8 × 10−4). Aortic dilation was amplified in males vs. females (p < 0.0001 at 100mmHg) and ameliorated by castration. The transcriptome of young Lox mutants showed alteration in dexamethasone (p = 9.83 × 10−30) and TGFß-responsive genes (p = 7.42 × 10−29), and aortas from older C57Bl/6J Lox+/C285F mice showed both enhanced susceptibility to elastase (p < 0.01 by ANOVA) and increased deposition of aggrecan (p < 0.05). These findings suggest that the secreted Lox+/C285F mutants produce dysfunctional elastic fibers that show increased susceptibility to proteolytic damage. Over time, the progressive weakening of the connective tissue, modified by sex and blood pressure, leads to worsening aortic disease.


Subject(s)
Elastic Tissue , Protein-Lysine 6-Oxidase , Animals , Aorta/metabolism , Blood Pressure , Copper , Dilatation, Pathologic/pathology , Elastic Tissue/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism
3.
Function (Oxf) ; 2(3): zqab015, 2021.
Article in English | MEDLINE | ID: mdl-34223172

ABSTRACT

Elastin (ELN) insufficiency leads to the cardiovascular hallmarks of the contiguous gene deletion disorder, Williams-Beuren syndrome, including hypertension and vascular stiffness. Previous studies showed that Williams-Beuren syndrome deletions, which extended to include the NCF1 gene, were associated with lower blood pressure (BP) and reduced vascular stiffness. NCF1 encodes for p47phox, the regulatory component of the NOX1 NADPH oxidase complex that generates reactive oxygen species (ROS) in the vascular wall. Dihydroethidium and 8-hydroxyguanosine staining of mouse aortas confirmed that Eln heterozygotes (Eln+/- ) had greater ROS levels than the wild-types (Eln+/+ ), a finding that was negated in vessels cultured without hemodynamic stressors. To analyze the Nox effect on ELN insufficiency, we used both genetic and chemical manipulations. Both Ncf1 haploinsufficiency (Ncf1+/- ) and Nox1 insufficiency (Nox1-/y ) decreased oxidative stress and systolic BP in Eln+/- without modifying vascular structure. Chronic treatment with apocynin, a p47phox inhibitor, lowered systolic BP in Eln+/- , but had no impact on Eln+/+ controls. In vivo dosing with phenylephrine (PE) produced an augmented BP response in Eln+/- relative to Eln+/+ , and genetic modifications or drug-based interventions that lower Nox1 expression reduced the hypercontractile response to PE in Eln+/- mice to Eln+/+ levels. These results indicate that the mechanical and structural differences caused by ELN insufficiency leading to oscillatory flow can perpetuate oxidative stress conditions, which are linked to hypertension, and that by lowering the Nox1-mediated capacity for vascular ROS production, BP differences can be normalized.


Subject(s)
Elastin , Hypertension , Williams Syndrome , Animals , Mice , Blood Pressure , Elastin/genetics , Hypertension/genetics , Phenylephrine/pharmacology , Reactive Oxygen Species/metabolism , Williams Syndrome/genetics
4.
Am J Physiol Heart Circ Physiol ; 315(1): H18-H32, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29498532

ABSTRACT

Increased vascular stiffness correlates with a higher risk of cardiovascular complications in aging adults. Elastin (ELN) insufficiency, as observed in patients with Williams-Beuren syndrome or with familial supravalvular aortic stenosis, also increases vascular stiffness and leads to arterial narrowing. We used Eln+/- mice to test the hypothesis that pathologically increased vascular stiffness with concomitant arterial narrowing leads to decreased blood flow to end organs such as the brain. We also hypothesized that drugs that remodel arteries and increase lumen diameter would improve flow. To test these hypotheses, we compared carotid blood flow using ultrasound and cerebral blood flow using MRI-based arterial spin labeling in wild-type (WT) and Eln+/- mice. We then studied how minoxidil, an ATP-sensitive K+ channel opener and vasodilator, affects vessel mechanics, blood flow, and gene expression. Both carotid and cerebral blood flows were lower in Eln+/- mice than in WT mice. Treatment of Eln+/- mice with minoxidil lowered blood pressure and reduced functional arterial stiffness to WT levels. Minoxidil also improved arterial diameter and restored carotid and cerebral blood flows in Eln+/- mice. The beneficial effects persisted for weeks after drug removal. RNA-Seq analysis revealed differential expression of 127 extracellular matrix-related genes among the treatment groups. These results indicate that ELN insufficiency impairs end-organ perfusion, which may contribute to the increased cardiovascular risk. Minoxidil, despite lowering blood pressure, improves end-organ perfusion. Changes in matrix gene expression and persistence of treatment effects after drug withdrawal suggest arterial remodeling. Such remodeling may benefit patients with genetic or age-dependent ELN insufficiency. NEW & NOTEWORTHY Our work with a model of chronic vascular stiffness, the elastin ( Eln)+/- mouse, shows reduced brain perfusion as measured by carotid ultrasound and MRI arterial spin labeling. Vessel caliber, functional stiffness, and blood flow improved with minoxidil. The ATP-sensitive K+ channel opener increased Eln gene expression and altered 126 other matrix-associated genes.


Subject(s)
Cerebrovascular Circulation/drug effects , Extracellular Matrix/metabolism , Minoxidil/pharmacology , Vascular Stiffness/drug effects , Vasodilator Agents/pharmacology , Animals , Cerebral Arteries/drug effects , Cerebral Arteries/metabolism , Cerebral Arteries/physiology , Elastin/genetics , Elastin/metabolism , Extracellular Matrix/drug effects , Mice , Mice, Inbred C57BL
5.
J Clin Invest ; 127(11): 4124-4135, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28990935

ABSTRACT

Acute lung injury is a leading cause of death in bacterial sepsis due to the wholesale destruction of the lung endothelial barrier, which results in protein-rich lung edema, influx of proinflammatory leukocytes, and intractable hypoxemia. Pyroptosis is a form of programmed lytic cell death that is triggered by inflammatory caspases, but little is known about its role in EC death and acute lung injury. Here, we show that systemic exposure to the bacterial endotoxin lipopolysaccharide (LPS) causes severe endothelial pyroptosis that is mediated by the inflammatory caspases, human caspases 4/5 in human ECs, or the murine homolog caspase-11 in mice in vivo. In caspase-11-deficient mice, BM transplantation with WT hematopoietic cells did not abrogate endotoxemia-induced acute lung injury, indicating a central role for nonhematopoietic caspase-11 in endotoxemia. Additionally, conditional deletion of caspase-11 in ECs reduced endotoxemia-induced lung edema, neutrophil accumulation, and death. These results establish the requisite role of endothelial pyroptosis in endotoxemic tissue injury and suggest that endothelial inflammatory caspases are an important therapeutic target for acute lung injury.


Subject(s)
Caspases/physiology , Endothelial Cells/enzymology , Endotoxemia/enzymology , Lung Injury/enzymology , Pyroptosis , Animals , Case-Control Studies , Caspases, Initiator , Cells, Cultured , Endothelium, Vascular/pathology , Endotoxemia/immunology , Female , Humans , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Lung/enzymology , Lung/immunology , Lung/pathology , Lung Injury/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptor 4/metabolism
6.
Stem Cell Reports ; 9(3): 796-806, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28781077

ABSTRACT

The generation of functional arterial endothelial cells (aECs) from embryonic stem cells (ESCs) holds great promise for vascular tissue engineering. However, the mechanisms underlying their generation and the potential of aECs in revascularizing ischemic tissue are not fully understood. Here, we observed that hypoxia exposure of mouse ESCs induced an initial phase of HIF1α-mediated upregulation of the transcription factor Etv2, which in turn induced the commitment to the EC fate. However, sustained activation of HIF1α in these EC progenitors thereafter induced NOTCH1 signaling that promoted the transition to aEC fate. We observed that transplantation of aECs mediated arteriogenesis in the mouse hindlimb ischemia model. Furthermore, transplantation of aECs in mice showed engraftment in ischemic myocardium and restored cardiac function in contrast to ECs derived under normoxia. Thus, HIF1α activation of Etv2 in ESCs followed by NOTCH1 signaling is required for the generation aECs that are capable of arteriogenesis and revascularization of ischemic tissue.


Subject(s)
Arteries/cytology , Cell Differentiation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mouse Embryonic Stem Cells/cytology , Receptors, Notch/metabolism , Transcription Factors/metabolism , Animals , Cell Hypoxia , Cell Lineage , Endothelial Cells/transplantation , Hindlimb/pathology , Ischemia/pathology , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Organogenesis , Signal Transduction , Transcription Factors/genetics , Up-Regulation/genetics
7.
Mol Ther ; 22(2): 451-463, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24002691

ABSTRACT

There is a constant shortage of red blood cells (RBCs) from sufficiently matched donors for patients who need chronic transfusion. Ex vivo expansion and maturation of human erythroid precursors (erythroblasts) from the patients or optimally matched donors could represent a potential solution. Proliferating erythroblasts can be expanded from umbilical cord blood mononuclear cells (CB MNCs) ex vivo for 10(6)-10(7)-fold (in ~50 days) before proliferation arrest and reaching sufficient number for broad application. Here, we report that ectopic expression of three genetic factors (Sox2, c-Myc, and an shRNA against TP53 gene) associated with iPSC derivation enables CB-derived erythroblasts to undergo extended expansion (~10(68)-fold in ~12 months) in a serum-free culture condition without change of cell identity or function. These expanding erythroblasts maintain immature erythroblast phenotypes and morphology, a normal diploid karyotype and dependence on a specific combination of growth factors for proliferation throughout expansion period. When being switched to a terminal differentiation condition, these immortalized erythroblasts gradually exit cell cycle, decrease cell size, accumulate hemoglobin, condense nuclei and eventually give rise to enucleated hemoglobin-containing erythrocytes that can bind and release oxygen. Our result may ultimately lead to an alternative approach to generate unlimited numbers of RBCs for personalized transfusion medicine.


Subject(s)
Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythropoiesis/physiology , Fetal Blood/cytology , Cell Differentiation/genetics , Cell Proliferation , Cluster Analysis , Cytokines/metabolism , Cytokines/pharmacology , Diploidy , Erythroblasts/cytology , Erythroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genes, myb , Genes, myc , Hemoglobins/genetics , Hemoglobins/metabolism , Hormones/metabolism , Hormones/pharmacology , Humans , Karyotype , SOXB1 Transcription Factors/genetics
8.
Hum Mol Genet ; 20(1): 165-75, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20940146

ABSTRACT

Patients with genetic defects of the cyclic (c) adenosine-monophosphate (AMP)-signaling pathway and those with neonatal-onset multisystem inflammatory disease (NOMID) develop tumor-like lesions of the long bones. The molecular basis of this similarity is unknown. NOMID is caused by inappropriate caspase-1 activity, which in turn activates the inflammasome. The present study demonstrates that NOMID bone lesions are derived from the same osteoblast progenitor cells that form fibroblastoid tumors in mice and humans with defects that lead to increased cAMP-dependent protein kinase A (PKA) signaling. NOMID tumor cells showed high PKA activity, and an increase in their cAMP signaling led to PKA-specific activation of caspase-1. Increased PKA led to inflammation-independent activation of caspase-1 via over-expression of the proto-oncogene (and early osteoblast factor) Ets-1. In NOMID tumor cells, as in cells with defective PKA regulation, increased prostaglandin E2 (PGE2) led to increased cAMP levels and activation of Wnt signaling, like in other states of inappropriate PKA activity. Caspase-1 and PGE2 inhibition led to a decrease in cell proliferation of both NOMID and cells with abnormal PKA. These data reveal a previously unsuspected link between abnormal cAMP signaling and defective regulation of the inflammasome and suggest that caspase-1 and PGE2 inhibition may be therapeutic targets in bone lesions associated with defects of these two pathways.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Osteoblasts/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Stem Cells/metabolism , Animals , Bone and Bones/pathology , Caspase 1/genetics , Cells, Cultured , Cryopyrin-Associated Periodic Syndromes/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Dinoprostone/genetics , Dinoprostone/metabolism , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Mice , Proto-Oncogene Mas , Proto-Oncogene Protein c-ets-1/genetics , RNA, Small Interfering/genetics , Signal Transduction/genetics , Stromal Cells/metabolism , Transcriptional Activation/genetics , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
9.
Proc Natl Acad Sci U S A ; 107(19): 8683-8, 2010 May 11.
Article in English | MEDLINE | ID: mdl-20421483

ABSTRACT

A population of stromal cells that retains osteogenic capacity in adult bone (adult bone stromal cells or aBSCs) exists and is under intense investigation. Mice heterozygous for a null allele of prkar1a (Prkar1a(+/-)), the primary receptor for cyclic adenosine monophosphate (cAMP) and regulator of protein kinase A (PKA) activity, developed bone lesions that were derived from cAMP-responsive osteogenic cells and resembled fibrous dysplasia (FD). Prkar1a(+/-) mice were crossed with mice that were heterozygous for catalytic subunit Calpha (Prkaca(+/-)), the main PKA activity-mediating molecule, to generate a mouse model with double heterozygosity for prkar1a and prkaca (Prkar1a(+/-)Prkaca(+/-)). Unexpectedly, Prkar1a(+/-)Prkaca(+/-) mice developed a greater number of osseous lesions starting at 3 months of age that varied from the rare chondromas in the long bones and the ubiquitous osteochondrodysplasia of vertebral bodies to the occasional sarcoma in older animals. Cells from these lesions originated from an area proximal to the growth plate, expressed osteogenic cell markers, and showed higher PKA activity that was mostly type II (PKA-II) mediated by an alternate pattern of catalytic subunit expression. Gene expression profiling confirmed a preosteoblastic nature for these cells but also showed a signature that was indicative of mesenchymal-to-epithelial transition and increased Wnt signaling. These studies show that a specific subpopulation of aBSCs can be stimulated in adult bone by alternate PKA and catalytic subunit activity; abnormal proliferation of these cells leads to skeletal lesions that have similarities to human FD and bone tumors.


Subject(s)
Aging/pathology , Bone and Bones/enzymology , Bone and Bones/pathology , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Animals , Bone and Bones/diagnostic imaging , Calcification, Physiologic , Catalytic Domain , Heterozygote , Mesoderm/metabolism , Mice , Phosphoric Diester Hydrolases/metabolism , Spectrum Analysis, Raman , Stromal Cells/enzymology , Stromal Cells/pathology , Tomography, X-Ray Computed
10.
Hum Mol Genet ; 19(8): 1387-98, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20080939

ABSTRACT

PRKAR1A inactivation leads to dysregulated cAMP signaling and Carney complex (CNC) in humans, a syndrome associated with skin, endocrine and other tumors. The CNC phenotype is not easily explained by the ubiquitous cAMP signaling defect; furthermore, Prkar1a(+/-) mice did not develop skin and other CNC tumors. To identify whether a Prkar1a defect is truly a generic but weak tumorigenic signal that depends on tissue-specific or other factors, we investigated Prkar1a(+/-) mice when bred within the Rb1(+/-) or Trp53(+/-) backgrounds, or treated with a two-step skin carcinogenesis protocol. Prkar1a(+/-) Trp53(+/-) mice developed more sarcomas than Trp53(+/-) mice (P < 0.05) and Prkar1a(+/-) Rb1(+/-) mice grew more (and larger) pituitary and thyroid tumors than Rb1(+/-) mice. All mice with double heterozygosity had significantly reduced life-spans compared with their single-heterozygous counterparts. Prkar1a(+/-) mice also developed more papillomas than wild-type animals. A whole-genome transcriptome profiling of tumors produced by all three models identified Wnt signaling as the main pathway activated by abnormal cAMP signaling, along with cell cycle abnormalities; all changes were confirmed by qRT-PCR array and immunohistochemistry. siRNA down-regulation of Ctnnb1, E2f1 or Cdk4 inhibited proliferation of human adrenal cells bearing a PRKAR1A-inactivating mutation and Prkar1a(+/-) mouse embryonic fibroblasts and arrested both cell lines at the G0/G1 phase of the cell cycle. In conclusion, Prkar1a haploinsufficiency is a relatively weak tumorigenic signal that can act synergistically with other tumor suppressor gene defects or chemicals to induce tumors, mostly through Wnt-signaling activation and cell cycle dysregulation, consistent with studies in human neoplasms carrying PRKAR1A defects.


Subject(s)
Cell Cycle , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Neoplasms/genetics , Neoplasms/pathology , Retinoblastoma Protein/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics , Wnt Proteins/metabolism , Animals , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Haploidy , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasms/metabolism , Neoplasms/physiopathology , Neoplastic Processes , Papilloma/chemically induced , Papilloma/genetics , Papilloma/metabolism , Papilloma/physiopathology , Retinoblastoma Protein/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/physiopathology , Tumor Suppressor Protein p53/metabolism , Wnt Proteins/genetics
11.
Cancer Res ; 69(13): 5301-6, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19549888

ABSTRACT

Inactivating germline mutations in phosphodiesterase 11A (PDE11A) have been implicated in adrenal tumor susceptibility. PDE11A is highly expressed in endocrine steroidogenic tissues, especially the testis, and mice with inactivated Pde11a exhibit male infertility, a known testicular germ cell tumor (TGCT) risk factor. We sequenced the PDE11A gene-coding region in 95 patients with TGCT from 64 unrelated kindreds. We identified 8 nonsynonymous substitutions in 20 patients from 15 families: four (R52T, F258Y, G291R, and V820M) were newly recognized, three (R804H, R867G, and M878V) were functional variants previously implicated in adrenal tumor predisposition, and one (Y727C) was a known polymorphism. We compared the frequency of these variants in our patients to unrelated controls that had been screened and found negative for any endocrine diseases: only the two previously reported variants, R804H and R867G, known to be frequent in general population, were detected in these controls. The frequency of all PDE11A-gene variants (combined) was significantly higher among patients with TGCT (P = 0.0002), present in 19% of the families of our cohort. Most variants were detected in the general population, but functional studies showed that all these mutations reduced PDE activity, and that PDE11A protein expression was decreased (or absent) in TGCT samples from carriers. This is the first demonstration of the involvement of a PDE gene in TGCT, although the cyclic AMP signaling pathway has been investigated extensively in reproductive organ function and their diseases. In conclusion, we report that PDE11A-inactivating sequence variants may modify the risk of familial and bilateral TGCT.


Subject(s)
Neoplasms, Germ Cell and Embryonal/genetics , Phosphoric Diester Hydrolases/genetics , Testicular Neoplasms/genetics , 3',5'-Cyclic-GMP Phosphodiesterases , Adolescent , Adult , Age of Onset , Cell Line , Gene Frequency , Genetic Variation , Humans , Kidney , Male , Middle Aged , Mutation , National Cancer Institute (U.S.) , Neoplasms, Germ Cell and Embryonal/epidemiology , Risk Factors , Testicular Neoplasms/epidemiology , Transfection , United States , White People/genetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...