Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Am J Respir Crit Care Med ; 208(6): 709-725, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37463497

ABSTRACT

Rationale: Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a lethal developmental disorder of lung morphogenesis caused by insufficiency of FOXF1 (forkhead box F1) transcription factor function. The cellular and transcriptional mechanisms by which FOXF1 deficiency disrupts human lung formation are unknown. Objectives: To identify cell types, gene networks, and cell-cell interactions underlying the pathogenesis of ACDMPV. Methods: We used single-nucleus RNA and assay for transposase-accessible chromatin sequencing, immunofluorescence confocal microscopy, and RNA in situ hybridization to identify cell types and molecular networks influenced by FOXF1 in ACDMPV lungs. Measurements and Main Results: Pathogenic single-nucleotide variants and copy-number variant deletions involving the FOXF1 gene locus in all subjects with ACDMPV (n = 6) were accompanied by marked changes in lung structure, including deficient alveolar development and a paucity of pulmonary microvasculature. Single-nucleus RNA and assay for transposase-accessible chromatin sequencing identified alterations in cell number and gene expression in endothelial cells (ECs), pericytes, fibroblasts, and epithelial cells in ACDMPV lungs. Distinct cell-autonomous roles for FOXF1 in capillary ECs and pericytes were identified. Pathogenic variants involving the FOXF1 gene locus disrupt gene expression in EC progenitors, inhibiting the differentiation or survival of capillary 2 ECs and cell-cell interactions necessary for both pulmonary vasculogenesis and alveolar type 1 cell differentiation. Loss of the pulmonary microvasculature was associated with increased VEGFA (vascular endothelial growth factor A) signaling and marked expansion of systemic bronchial ECs expressing COL15A1 (collagen type XV α 1 chain). Conclusions: Distinct FOXF1 gene regulatory networks were identified in subsets of pulmonary endothelial and fibroblast progenitors, providing both cellular and molecular targets for the development of therapies for ACDMPV and other diffuse lung diseases of infancy.


Subject(s)
Persistent Fetal Circulation Syndrome , Infant, Newborn , Humans , Persistent Fetal Circulation Syndrome/genetics , Persistent Fetal Circulation Syndrome/pathology , Gene Regulatory Networks/genetics , Vascular Endothelial Growth Factor A/genetics , Endothelial Cells/pathology , Multiomics , Lung/pathology , RNA , Forkhead Transcription Factors/genetics
2.
Circulation ; 142(4): 365-379, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32393053

ABSTRACT

BACKGROUND: Metabolic disorders such as obesity and diabetes mellitus can cause dysfunction of endothelial cells (ECs) and vascular rarefaction in adipose tissues. However, the modulatory role of ECs in adipose tissue function is not fully understood. Other than vascular endothelial growth factor-vascular endothelial growth factor receptor-mediated angiogenic signaling, little is known about the EC-derived signals in adipose tissue regulation. We previously identified Argonaute 1 (AGO1; a key component of microRNA-induced silencing complex) as a crucial regulator in hypoxia-induced angiogenesis. In this study, we intend to determine the AGO1-mediated EC transcriptome, the functional importance of AGO1-regulated endothelial function in vivo, and the relevance to adipose tissue function and obesity. METHODS: We generated and subjected mice with EC-AGO1 deletion (EC-AGO1-knockout [KO]) and their wild-type littermates to a fast food-mimicking, high-fat high-sucrose diet and profiled the metabolic phenotypes. We used crosslinking immunoprecipitation- and RNA-sequencing to identify the AGO1-mediated mechanisms underlying the observed metabolic phenotype of EC-AGO1-KO. We further leveraged cell cultures and mouse models to validate the functional importance of the identified molecular pathway, for which the translational relevance was explored using human endothelium isolated from healthy donors and donors with obesity/type 2 diabetes mellitus. RESULTS: We identified an antiobesity phenotype of EC-AGO1-KO, evident by lower body weight and body fat, improved insulin sensitivity, and enhanced energy expenditure. At the organ level, we observed the most significant phenotype in the subcutaneous and brown adipose tissues of KO mice, with greater vascularity and enhanced browning and thermogenesis. Mechanistically, EC-AGO1 suppression results in inhibition of thrombospondin-1 (THBS1/TSP1), an antiangiogenic and proinflammatory cytokine that promotes insulin resistance. In EC-AGO1-KO mice, overexpression of TSP1 substantially attenuated the beneficial phenotype. In human endothelium isolated from donors with obesity or type 2 diabetes mellitus, AGO1 and THBS1 are expressed at higher levels than the healthy controls, supporting a pathological role of this pathway. CONCLUSIONS: Our study suggests a novel mechanism by which ECs, through the AGO1-TSP1 pathway, control vascularization and function of adipose tissues, insulin sensitivity, and whole-body metabolic state.


Subject(s)
Adipose Tissue, Brown/metabolism , Argonaute Proteins/metabolism , Disease Susceptibility , Endothelium/metabolism , Eukaryotic Initiation Factors/metabolism , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Adult , Animals , Argonaute Proteins/genetics , Diet, High-Fat , Disease Models, Animal , Energy Metabolism , Eukaryotic Initiation Factors/genetics , Female , Gene Expression Profiling , Gene Targeting , Genetic Loci , Humans , Insulin Resistance , Male , Metabolic Diseases/diagnosis , Mice , Mice, Knockout , Middle Aged , Models, Biological , Obesity , Phenotype
4.
Proc Natl Acad Sci U S A ; 113(42): 11883-11888, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27688768

ABSTRACT

The aryl hydrocarbon receptor (AhR) is now recognized as an important physiological regulator in the immune and reproductive systems, and in the development of the liver and vascular system. AhR regulates cell cycle, cell proliferation, and differentiation through interacting with other signaling pathways, like estrogen receptor α (ERα), androgen receptor (AR), and Notch signaling. In the present study, we investigated Notch and estrogen signaling in AhR-/- mice. We found low fertility with degenerative changes in the testes, germ cell apoptosis, and a reduced number of early spermatids. There was no change in aromatase, AR, ERα, or ERß expression in the testis and no detectable change in serum estrogen levels. However, expression of Notch receptors (Notch1 and Notch3) and their target Hairy and Enhancer of Split homolog 1 (HES1) was reduced. In addition, the testosterone level was slightly reduced in the serum. In the mammary fat pad, AhR appeared to regulate estrogen signaling because, in AhR-/- males, there was significant growth of the mammary ducts with high expression of ERα in the ductal epithelium. The enhanced mammary ductal growth appears to be related to overexpression of ERα accompanied by a high proliferation index, whereas the reduced fertility appears to be related defects in Notch signaling that leads to reduced expression of HES1 and, consequently, early maturation of spermatocytes and a depletion of primary spermatids. Previous reports have indicated that AhR pathway is associated with infertility in men. Our results provide a mechanistic explanation for this defect.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/deficiency , Estrogen Receptor alpha/metabolism , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Notch/metabolism , Signal Transduction , Animals , Apoptosis/genetics , Aromatase/metabolism , Biomarkers , Cell Proliferation , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Fertility/genetics , Gene Deletion , Gene Expression , Germ Cells/metabolism , Immunohistochemistry , Male , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Spermatocytes/metabolism , Testis/metabolism
5.
Proc Natl Acad Sci U S A ; 113(27): 7614-9, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27335465

ABSTRACT

The etiology of peripheral squamous cell lung cancer (PSCCa) remains unknown. Here, we show that this condition spontaneously develops in mice in which the genes for two oxysterol receptors, Liver X Receptor (LXR) α (Nr1h3) and ß (Nr1h2), are inactivated. By 1 y of age, most of these mice have to be euthanized because of severe dyspnea. Starting at 3 mo, the lungs of LXRα,ß(Dko) mice, but not of LXRα or LXRß single knockout mice, progressively accumulate foam cells, so that by 1 y, the lungs are covered by a "golden coat." There is infiltration of inflammatory cells and progressive accumulation of lipid in the alveolar wall, type 2 pneumocytes, and macrophages. By 14 mo, there are three histological lesions: one resembling adenomatous hyperplasia, one squamous metaplasia, and one squamous cell carcinoma characterized by expression of transformation-related protein (p63), sex determining region Y-box 2 (Sox2), cytokeratin 14 (CK14), and cytokeratin 13 (CK13) and absence of thyroid transcription factor 1 (TTF1), and prosurfactant protein C (pro-SPC). RNA sequencing analysis at 12 mo confirmed a massive increase in markers of M1 macrophages and lymphocytes. The data suggest a previously unidentified etiology of PSCCa: cholesterol dysregulation and M1 macrophage-predominant lung inflammation combined with damage to, and aberrant repair of, lung tissue, particularly the peripheral parenchyma. The results raise the possibility that components of the LXR signaling may be useful targets in the treatment of PSCCa.


Subject(s)
Lipid Metabolism , Liver X Receptors/physiology , Lung Neoplasms/etiology , Lung/metabolism , Neoplasms, Squamous Cell/etiology , Alveolar Epithelial Cells/metabolism , Animals , Fibroblasts/metabolism , Homeostasis , Lung/pathology , Macrophages/metabolism , Mice, Inbred C57BL , Pneumonia/etiology , Sequence Analysis, RNA
6.
Proc Natl Acad Sci U S A ; 112(45): 14006-11, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26504234

ABSTRACT

The recent discovery of browning of white adipose tissue (WAT) has raised great research interest because of its significant potential in counteracting obesity and type 2 diabetes. Browning is the result of the induction in WAT of a newly discovered type of adipocyte, the beige cell. When mice are exposed to cold or several kinds of hormones or treatments with chemicals, specific depots of WAT undergo a browning process, characterized by highly activated mitochondria and increased heat production and energy expenditure. However, the mechanisms underlying browning are still poorly understood. Liver X receptors (LXRs) are one class of nuclear receptors, which play a vital role in regulating cholesterol, triglyceride, and glucose metabolism. Following our previous finding that LXRs serve as repressors of uncoupling protein-1 (UCP1) in classic brown adipose tissue in female mice, we found that LXRs, especially LXRß, also repress the browning process of subcutaneous adipose tissue (SAT) in male rodents fed a normal diet. Depletion of LXRs activated thyroid-stimulating hormone (TSH)-releasing hormone (TRH)-positive neurons in the paraventricular nucleus area of the hypothalamus and thus stimulated secretion of TSH from the pituitary. Consequently, production of thyroid hormones in the thyroid gland and circulating thyroid hormone level were increased. Moreover, the activity of thyroid signaling in SAT was markedly increased. Together, our findings have uncovered the basis of increased energy expenditure in male LXR knockout mice and provided support for targeting LXRs in treatment of obesity.


Subject(s)
Adipose Tissue, White/metabolism , Brain/metabolism , Energy Metabolism/physiology , Mitochondria/metabolism , Orphan Nuclear Receptors/metabolism , Thyroid Hormones/metabolism , Analysis of Variance , Animals , Body Composition/physiology , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Immunohistochemistry , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors/genetics , Thyrotropin-Releasing Hormone/metabolism
7.
Proc Natl Acad Sci U S A ; 112(27): 8397-402, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26100911

ABSTRACT

The antidiuretic hormone arginine vasopressin is a systemic effector in urinary concentration. However, increasing evidence suggests that other locally produced factors may also play an important role in the regulation of water reabsorption in renal collecting ducts. Recently, prostaglandin E2 (PGE2) receptor EP4 has emerged as a potential therapeutic target for the treatment of nephrogenic diabetes insipidus, but the underlying mechanism is unknown. To evaluate the role of EP4 in regulating water homeostasis, mice with renal tubule-specific knockout of EP4 (Ksp-EP4(-/-)) and collecting duct-specific knockout of EP4 (AQP2-EP4(-/-)) were generated using the Cre-loxP recombination system. Urine concentrating defect was observed in both Ksp-EP4(-/-) and AQP2-EP4(-/-) mice. Decreased aquaporin 2 (AQP2) abundance and apical membrane targeting in renal collecting ducts were evident in Ksp-EP4(-/-) mice. In vitro studies demonstrated that AQP2 mRNA and protein levels were significantly up-regulated in mouse primary inner medullary collecting duct (IMCD) cells after pharmacological activation or adenovirus-mediated overexpression of EP4 in a cAMP/cAMP-response element binding protein-dependent manner. In addition, EP4 activation or overexpression also increased AQP2 membrane accumulation in a mouse IMCD cell line (IMCD3) stably transfected with the AQP2 gene, mainly through the cAMP/protein kinase A and extracellular signal-regulated kinase pathways. In summary, the EP4 receptor in renal collecting ducts plays an important role in regulating urinary concentration under physiological conditions. The ability of EP4 to promote AQP2 membrane targeting and increase AQP2 abundance makes it a potential therapeutic target for the treatment of clinical disorders including acquired and congenital diabetes insipidus.


Subject(s)
Aquaporin 2/genetics , Kidney Concentrating Ability/genetics , Kidney Tubules, Collecting/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Animals , Aquaporin 2/metabolism , Blotting, Western , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dinoprostone/analogs & derivatives , Dinoprostone/biosynthesis , Dinoprostone/pharmacology , Kidney Tubules, Collecting/cytology , MAP Kinase Signaling System , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Primary Cell Culture , Pyrrolidinones/pharmacology , RNA Interference , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Water/metabolism
8.
J Am Soc Nephrol ; 28(11): 3251-3261, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28696247

ABSTRACT

CKD is an independent risk factor for cardiovascular disease (CVD). The accumulation of uremic toxins in CKD induces oxidative stress and endothelial dysfunction. MicroRNA-92a (miR-92a) is induced by oxidative stress in endothelial cells (ECs) and involved in angiogenesis and atherosclerosis. We investigated a role for oxidative stress-responsive miR-92a in CKD. Our study of patients at three clinical sites showed increased serum miR-92a level with decreased kidney function. In cultured ECs, human CKD serum or uremic toxins (such as indoxyl sulfate), compared with non-CKD serum, induced the levels of miR-92a and suppressed the expression of miR-92a targets, including key endothelial-protective molecules. The antioxidant N-acetylcysteine inhibited these vasculopathic properties. In rats, adenine-induced CKD associated with increased levels of miR-92a in aortas, serum, and CD144+ endothelial microparticles. Furthermore, CD144+ microparticles from human uremic serum contained more miR-92a than those from control serum. Additional analysis showed a positive correlation between serum levels of miR-92a and indoxyl sulfate in a cohort of patients with ESRD undergoing hemodialysis. Collectively, our findings suggest that the uremic toxins accumulated in CKD can upregulate miR-92a in ECs, which impairs EC function and predisposes patients to CVD.


Subject(s)
Endothelial Cells/physiology , MicroRNAs/physiology , Renal Insufficiency, Chronic/physiopathology , Animals , Female , Humans , Male , MicroRNAs/blood , Middle Aged , Rats , Rats, Wistar , Renal Insufficiency, Chronic/blood
9.
Proc Natl Acad Sci U S A ; 111(6): 2277-82, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24464484

ABSTRACT

The farnesoid X receptor (FXR) is a ligand-activated transcription factor belonging to the nuclear receptor superfamily. FXR is mainly expressed in liver and small intestine, where it plays an important role in bile acid, lipid, and glucose metabolism. The kidney also has a high FXR expression level, with its physiological function unknown. Here we demonstrate that FXR is ubiquitously distributed in renal tubules. FXR agonist treatment significantly lowered urine volume and increased urine osmolality, whereas FXR knockout mice exhibited an impaired urine concentrating ability, which led to a polyuria phenotype. We further found that treatment of C57BL/6 mice with chenodeoxycholic acid, an FXR endogenous ligand, significantly up-regulated renal aquaporin 2 (AQP2) expression, whereas FXR gene deficiency markedly reduced AQP2 expression levels in the kidney. In vitro studies showed that the AQP2 gene promoter contained a putative FXR response element site, which can be bound and activated by FXR, resulting in a significant increase of AQP2 transcription in cultured primary inner medullary collecting duct cells. In conclusion, the present study demonstrates that FXR plays a critical role in the regulation of urine volume, and its activation increases urinary concentrating capacity mainly via up-regulating its target gene AQP2 expression in the collecting ducts.


Subject(s)
Kidney Concentrating Ability/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Aquaporin 2/genetics , Aquaporin 2/metabolism , Base Sequence , DNA Primers , Kidney/metabolism , Male , Mice , Mice, Knockout , Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL