Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.979
Filter
1.
Cell Mol Life Sci ; 81(1): 225, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38769116

ABSTRACT

Ischemic stroke induces neovascularization of the injured tissue as an attempt to promote structural repair and neurological recovery. Angiogenesis is regulated by pericytes that potently react to ischemic stroke stressors, ranging from death to dysfunction. Platelet-derived growth factor (PDGF) receptor (PDGFR)ß controls pericyte survival, migration, and interaction with brain endothelial cells. PDGF-D a specific ligand of PDGFRß is expressed in the brain, yet its regulation and role in ischemic stroke pathobiology remains unexplored. Using experimental ischemic stroke mouse model, we found that PDGF-D is transiently induced in brain endothelial cells at the injury site in the subacute phase. To investigate the biological significance of PDGF-D post-ischemic stroke regulation, its subacute expression was either downregulated using siRNA or upregulated using an active recombinant form. Attenuation of PDGF-D subacute induction exacerbates neuronal loss, impairs microvascular density, alters vascular permeability, and increases microvascular stalling. Increasing PDGF-D subacute bioavailability rescues neuronal survival and improves neurological recovery. PDGF-D subacute enhanced bioavailability promotes stable neovascularization of the injured tissue and improves brain perfusion. Notably, PDGF-D enhanced bioavailability improves pericyte association with brain endothelial cells. Cell-based assays using human brain pericyte and brain endothelial cells exposed to ischemia-like conditions were applied to investigate the underlying mechanisms. PDGF-D stimulation attenuates pericyte loss and fibrotic transition, while increasing the secretion of pro-angiogenic and vascular protective factors. Moreover, PDGF-D stimulates pericyte migration required for optimal endothelial coverage and promotes angiogenesis. Our study unravels new insights into PDGF-D contribution to neurovascular protection after ischemic stroke by rescuing the functions of pericytes.


Subject(s)
Endothelial Cells , Ischemic Stroke , Lymphokines , Pericytes , Platelet-Derived Growth Factor , Pericytes/metabolism , Pericytes/pathology , Animals , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Mice , Lymphokines/metabolism , Lymphokines/genetics , Platelet-Derived Growth Factor/metabolism , Humans , Endothelial Cells/metabolism , Male , Mice, Inbred C57BL , Brain/metabolism , Brain/pathology , Disease Models, Animal , Neovascularization, Physiologic , Cell Movement
2.
Cell Biol Int ; 47(12): 1942-1949, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37615370

ABSTRACT

Platelet-derived growth factor C (PDGF-C) is a member of PDGF/VEGF family, which is well-known for important functions in the vascular system. It is widely reported that PDGF-C is able to modulate cell proliferation. However, it is still not very clear about this cell modulating mechanism at the molecular level. In a screening of factors regulated by PDGF-C protein, we fished out a factor called block of proliferation 1 (BOP1), which is a pivotal regulator of ribosome biogenesis and cell proliferation. In this study, we investigated the regulation of BOP1 by PDGF-C and its role in modulating cell proliferation. We found that BOP1 was downregulated at both mRNA and protein levels in cells treated with PDGF-C-containing conditioned medium. On the other hand, BOP1 was upregulated in PDGF-C deficient mice. Furthermore, we confirmed that overexpression of BOP1 inhibited HEK293A cell proliferation, whereas knockdown of BOP1 promoted cell proliferation. The mitogenic effect of PDGF-C could be attenuated by downregulation of BOP1. Our results demonstrate a clear PDGF-C-BOP1 signaling that modulates cell proliferation.


Subject(s)
Lymphokines , Platelet-Derived Growth Factor , Animals , Mice , Platelet-Derived Growth Factor/metabolism , Cell Proliferation , Lymphokines/genetics , Lymphokines/metabolism , Lymphokines/pharmacology , Signal Transduction
3.
Cancer Lett ; 567: 216266, 2023 07 28.
Article in English | MEDLINE | ID: mdl-37321532

ABSTRACT

Drug resistance is a major problem in cancer treatment with traditional or targeted therapeutics. Gemcitabine is approved for several human cancers and the first line treatment for locally advanced or metastatic pancreatic ductal adenocarcinoma (PDAC). However, gemcitabine resistance frequently occurs and is a major problem in successful treatments of these cancers and the mechanism of gemcitabine resistance remains largely unknown. In this study, we identified 65 genes that had reversible methylation changes in their promoters in gemcitabine resistant PDAC cells using whole genome Reduced Representation Bisulfite Sequencing analyses. One of these genes, PDGFD, was further studied in detail for its reversible epigenetic regulation in expression and shown to contribute to gemcitabine resistance in vitro and in vivo via stimulating STAT3 signaling in both autocrine and paracrine manners to upregulate RRM1 expression. Analyses of TCGA datasets showed that PDGFD positively associates with poor outcome of PDAC patients. Together, we conclude that the reversible epigenetic upregulation plays an important role in gemcitabine resistance development and targeting PDGFD signaling alleviates gemcitabine resistance for PDAC treatment.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Gemcitabine , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Up-Regulation , Epigenesis, Genetic , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/pathology , Demethylation , Ribonucleoside Diphosphate Reductase/genetics , Lymphokines/genetics , Lymphokines/metabolism , Lymphokines/therapeutic use , Platelet-Derived Growth Factor/genetics , Pancreatic Neoplasms
4.
Aging (Albany NY) ; 15(10): 4071-4083, 2023 03 02.
Article in English | MEDLINE | ID: mdl-37253627

ABSTRACT

Hepatocellular carcinoma (HCC) ranks high in morbidity and mortality among notorious malignancies because of the lack of effective biomarkers and treatments. LncRNA PITPNA antisense RNA 1 (PITPNA-AS1) plays an oncogenic role in HCC, yet the mechanistic basis remains unprobed. Here using Bioinformatics and PCR analyses, we validated that PITPNA-AS1 expression was significantly increased in HCC. The levels of PITPNA-AS1 in tumors were reversely correlated with the prognosis in HCC patients. Downregulation of PITPNA-AS1 inhibited malignant activities of HCC cells. Next, we elucidated that PITPNA-AS1 acts as a competing endogenous RNA (ceRNA) to sponge miR-363-5p, thereby regulating the expression of platelet-derived growth factor-D (PDGFD). Moreover, the suppression of HCC cell activities by PITPNA-AS1 downregulation can be removed by PDGFD overexpression or miR-363-5p inhibition. Collectively, our work reveals the involvement of the PITPNA-AS1/miR-363-5p/PDGFD regulatory axis in HCC progression.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Disease Progression , Cell Proliferation/genetics , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Lymphokines/genetics , Lymphokines/metabolism
5.
Nat Cancer ; 4(4): 468-484, 2023 04.
Article in English | MEDLINE | ID: mdl-36914817

ABSTRACT

Patients with estrogen receptor (ER)-positive breast cancer are at risk of metastatic relapse for decades after primary tumor resection and treatment, a consequence of dormant disseminated tumor cells (DTCs) reawakening at secondary sites. Here we use syngeneic ER+ mouse models in which DTCs display a dormant phenotype in young mice but accelerated metastatic outgrowth in an aged or fibrotic microenvironment. In young mice, low-level Pdgfc expression by ER+ DTCs is required for their maintenance in secondary sites but is insufficient to support development of macrometastases. By contrast, the platelet-derived growth factor (PDGF)-Chi environment of aging or fibrotic lungs promotes DTC proliferation and upregulates tumor cell Pdgfc expression stimulating further stromal activation, events that can be blocked by pharmacological inhibition of PDGFRα or with a PDGF-C-blocking antibody. These results highlight the role of the changing microenvironment in regulating DTC outgrowth and the opportunity to target PDGF-C signaling to limit metastatic relapse in ER+ breast cancer.


Subject(s)
Breast Neoplasms , Platelet-Derived Growth Factor , Tumor Microenvironment , Animals , Mice , Lymphokines/genetics , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Recurrence , Tumor Microenvironment/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology
6.
J Cutan Pathol ; 49(3): 274-277, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34628665

ABSTRACT

Dermatofibrosarcoma protuberans (DFSP) is a mesenchymal neoplasm that is usually located in the dermis or subcutis and is locally aggressive. Rarely, these lesions may undergo fibrosarcomatous transformation, which is thought to increase their metastatic potential. DFSP is classically associated with a 17;22 translocation (or ring chromosome thereof) resulting in fusion of the COL1A1 and PDGFB genes. However, variant fusions involving PDGFD have been recently reported. Herein, we present two morphologically diverse cases of DFSP with PDGFD rearrangement. Case 1 is a 68-year-old female with a left dorsal foot lesion. Morphologically, the lesion is unusual as it is a well-circumscribed, hypercellular, subcutaneous nodule with uniform CD34-positive spindle cells arranged in a herringbone pattern without storiform arrangement or "honeycombing" fat entrapment. It was diagnosed as pure fibrosarcomatous DFSP. Case 2 is a 37-year-old male with a right supra-auricular lesion. Morphologically, the lesion displays classic DFSP features including bland CD34-positive spindle cells with storiform growth, fat entrapment, and infiltrative borders. Both lesions were negative for COL1A1-PDGFB fusion but positive for PDGFD rearrangement by fluorescence in situ hybridization (FISH) analysis. FISH testing for PDGFD rearrangement should be performed in cases where there is a high suspicion for DFSP but initial studies for COL1A1-PDGFB are negative.


Subject(s)
Dermatofibrosarcoma/genetics , Gene Rearrangement , Lymphokines/genetics , Platelet-Derived Growth Factor/genetics , Skin Neoplasms/genetics , Adult , Aged , Dermatofibrosarcoma/pathology , Female , Humans , In Situ Hybridization, Fluorescence , Male , Skin Neoplasms/pathology
7.
J Cereb Blood Flow Metab ; 42(2): 264-279, 2022 02.
Article in English | MEDLINE | ID: mdl-34689641

ABSTRACT

Platelet-derived growth factor B (PDGFB) released from endothelial cells is indispensable for pericyte recruitment during angiogenesis in embryonic and postnatal organ growth. Constitutive genetic loss-of-function of PDGFB leads to pericyte hypoplasia and the formation of a sparse, dilated and venous-shifted brain microvasculature with dysfunctional blood-brain barrier (BBB) in mice, as well as the formation of microvascular calcification in both mice and humans. Endothelial PDGFB is also expressed in the adult quiescent microvasculature, but here its importance is unknown. We show that deletion of Pdgfb in endothelial cells in 2-months-old mice causes a slowly progressing pericyte loss leading, at 12-18 months of age, to ≈50% decrease in endothelial:pericyte cell ratio, ≈60% decrease in pericyte longitudinal capillary coverage and >70% decrease in pericyte marker expression. Similar to constitutive loss of Pdgfb, this correlates with increased BBB permeability. However, in contrast to the constitutive loss of Pdgfb, adult-induced loss does not lead to vessel dilation, impaired arterio-venous zonation or the formation of microvascular calcifications. We conclude that PDFGB expression in quiescent adult microvascular brain endothelium is critical for the maintenance of pericyte coverage and normal BBB function, but that microvessel dilation, rarefaction, arterio-venous skewing and calcification reflect developmental roles of PDGFB.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability , Endothelium, Vascular/metabolism , Lymphokines/metabolism , Pericytes/metabolism , Platelet-Derived Growth Factor/metabolism , Vascular Calcification/metabolism , Animals , Blood-Brain Barrier/pathology , Endothelium, Vascular/pathology , Gene Expression Regulation , Lymphokines/genetics , Mice , Mice, Knockout , Pericytes/pathology , Platelet-Derived Growth Factor/genetics , Vascular Calcification/genetics , Vascular Calcification/pathology
8.
Front Immunol ; 12: 668391, 2021.
Article in English | MEDLINE | ID: mdl-34539622

ABSTRACT

The binding of platelet-derived growth factor D (PDGF-DD) to the NKp44 receptor activates a distinct transcriptional program in primary IL-2 expanded human natural killer (NK) cells. We were interested in knowing if the PDGF-DD-NKp44 pathway of NK cell activation might play a clinically relevant role in anti-tumor immunity. In order to address this question, we determined transcriptional signatures unique to resting, IL-2 expanded, and PDGF-DD activated, NK cells, in addition to different T cell subsets, and established the abundance of these immune cell phenotypes in The Cancer Genome Atlas (TCGA) low-grade glioma (LGG) dataset using CIBERSORT. Our results show that LGG patient tumors enriched for either the PDGF-DD activated NK cell or memory CD8+ T cell phenotypes are associated with a more favorable prognosis. Combined cell phenotype analyses revealed that patients with LGG tumors enriched for the PDGF-DD activated NK cell phenotype and the CD4+ T helper cell phenotype had a more favorable prognosis. High expression of transcripts encoding members of the killer cell lectin-like receptor (KLR) family, such as KLRK1 and KLRC2, KLRC3 and KLRC4 in LGG tumors were associated with more favorable prognosis, suggesting that these NK cell family receptors may play a prominent role in LGG anti-tumor immunity. Finally, many of the TCGA findings were reciprocated in LGG patients from the Chinese Glioma Genome Atlas (CGGA) dataset. Our results provide transcriptomic evidence that PDGF-DD activated NK cells and KLR family receptors may play an important clinical role in immune surveillance of LGG.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression Profiling , Glioma/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Transcriptome , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Databases, Genetic , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/immunology , Glioma/pathology , Humans , Interleukin-2/metabolism , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphokines/genetics , Natural Cytotoxicity Triggering Receptor 2/metabolism , Neoplasm Grading , Phenotype , Platelet-Derived Growth Factor/genetics , Predictive Value of Tests , Progression-Free Survival , Signal Transduction , Tumor Microenvironment
9.
J Exp Clin Cancer Res ; 40(1): 278, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34470658

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM), a lethal brain tumor, remains the most daunting challenge in cancer therapy. Overexpression and constitutive activation of PDGFs and PDGFRα are observed in most GBM; however, available inhibitors targeting isolated signaling pathways are minimally effective. Therefore, better understanding of crucial mechanisms underlying GBM is needed for developing more effective targeted therapies. METHODS: Target genes controlled by HIF1α in GBM were identified by analysis of TCGA database and by RNA-sequencing of GBM cells with HIF1α knockout by sgRNA-Cas9 method. Functional roles of HIF1α, PDGFs and PDGFRs were elucidated by loss- or gain-of-function assays or chemical inhibitors, and compared in response to oxygen tension. Pharmacological efficacy and gene expression in mice with intracranial xenografts of primary GBM were analyzed by bioluminescence imaging and immunofluorescence. RESULTS: HIF1α binds the PDGFD proximal promoter and PDGFRA intron enhancers in GBM cells under normoxia or mild-hypoxia to induce their expression and maintain constitutive activation of AKT signaling, which in turn increases HIF1α protein level and activity. Paradoxically, severe hypoxia abrogates PDGFRα expression despite enhancing HIF1α accumulation and corresponding PDGF-D expression. Knockout of HIF1A, PDGFD or PDGFRA in U251 cells inhibits cell growth and invasion in vitro and eradicates tumor growth in vivo. HIF1A knockdown in primary GBM extends survival of xenograft mice, whereas PDGFD overexpression in GL261 shortens survival. HIF1α inhibitor Echinomycin induces GBM cell apoptosis and effectively inhibits growth of GBM in vivo by simultaneously targeting HIF1α-PDGFD/PDGFRα-AKT feedforward pathway. CONCLUSIONS: HIF1α orchestrates expression of PDGF-D and PDGFRα for constitutive activation of AKT pathway and is crucial for GBM malignancy. Therefore, therapies targeting HIF1α should provide an effective treatment for GBM.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Brain Neoplasms/pathology , Echinomycin/therapeutic use , Glioblastoma/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Lymphokines/metabolism , Oxygen/metabolism , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Enzyme Activation , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Lymphokines/genetics , Mice , Mice, Inbred NOD , Neoplasm Invasiveness , Platelet-Derived Growth Factor/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics
10.
Science ; 373(6550)2021 07 02.
Article in English | MEDLINE | ID: mdl-34210853

ABSTRACT

The mechanisms by which macrophages regulate energy storage remain poorly understood. We identify in a genetic screen a platelet-derived growth factor (PDGF)/vascular endothelial growth factor (VEGF)-family ortholog, Pvf3, that is produced by macrophages and is required for lipid storage in fat-body cells of Drosophila larvae. Genetic and pharmacological experiments indicate that the mouse Pvf3 ortholog PDGFcc, produced by adipose tissue-resident macrophages, controls lipid storage in adipocytes in a leptin receptor- and C-C chemokine receptor type 2-independent manner. PDGFcc production is regulated by diet and acts in a paracrine manner to control lipid storage in adipose tissues of newborn and adult mice. At the organismal level upon PDGFcc blockade, excess lipids are redirected toward thermogenesis in brown fat. These data identify a macrophage-dependent mechanism, conducive to the design of pharmacological interventions, that controls energy storage in metazoans.


Subject(s)
Adipocytes/immunology , Diet, High-Fat , Drosophila Proteins/metabolism , Energy Metabolism , Lymphokines/metabolism , Macrophages/immunology , Obesity/immunology , Platelet-Derived Growth Factor/metabolism , Thermogenesis , Adipose Tissue, Brown/immunology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Hemocytes/immunology , Liver/immunology , Lymphokines/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet-Derived Growth Factor/genetics , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism
11.
J Exp Med ; 218(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34236404

ABSTRACT

Obesity-induced secretory disorder of adipose tissue-derived factors is important for cardiac damage. However, whether platelet-derived growth factor-D (PDGF-D), a newly identified adipokine, regulates cardiac remodeling in angiotensin II (AngII)-infused obese mice is unclear. Here, we found obesity induced PDGF-D expression in adipose tissue as well as more severe cardiac remodeling compared with control lean mice after AngII infusion. Adipocyte-specific PDGF-D knockout attenuated hypertensive cardiac remodeling in obese mice. Consistently, adipocyte-specific PDGF-D overexpression transgenic mice (PA-Tg) showed exacerbated cardiac remodeling after AngII infusion without high-fat diet treatment. Mechanistic studies indicated that AngII-stimulated macrophages produce urokinase plasminogen activator (uPA) that activates PDGF-D by splicing full-length PDGF-D into the active PDGF-DD. Moreover, bone marrow-specific uPA knockdown decreased active PDGF-DD levels in the heart and improved cardiac remodeling in HFD hypertensive mice. Together, our data provide for the first time a new interaction pattern between macrophage and adipocyte: that macrophage-derived uPA activates adipocyte-secreted PDGF-D, which finally accelerates AngII-induced cardiac remodeling in obese mice.


Subject(s)
Lymphokines/metabolism , Obesity/physiopathology , Platelet-Derived Growth Factor/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Ventricular Remodeling/physiology , Adipocytes/metabolism , Adipocytes/pathology , Angiotensin II/pharmacology , Animals , Heart/drug effects , Hypertension/genetics , Hypertension/physiopathology , Lymphokines/genetics , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mice, Transgenic , Myocardium/pathology , Obesity/metabolism , Platelet-Derived Growth Factor/genetics , Urokinase-Type Plasminogen Activator/genetics
12.
Genes Genomics ; 43(9): 1087-1094, 2021 09.
Article in English | MEDLINE | ID: mdl-34302633

ABSTRACT

BACKGROUND: In tooth bioengineering for replacement therapy of missing teeth, the utilized cells must possess an inductive signal-forming ability to initiate odontogenesis. This ability is called odontogenic potential. In mice, the odontogenic potential signal is known to be translocated from the epithelium to the mesenchyme at the early bud stage in the developing molar tooth germ. However, the identity of the molecular constituents of this process remains unclear. OBJECTIVE: The purpose of this study is to determine the molecular identity of odontogenic potential and to provide a new perspective in the field of tooth development research. METHODS: In this study, whole transcriptome profiles of the mouse molar tooth germ epithelium and mesenchyme were investigated using the RNA sequencing (RNA-seq) technique. The analyzed transcriptomes corresponded to two developmental stages, embryonic day 11.5 (E11.5) and 14.5 (E14.5), which represent the odontogenic potential shifts. RESULTS: We identified differentially expressed genes (DEGs), which were specifically overexpressed in both the E11.5 epithelium and E14.5 mesenchyme, but not expressed in their respective counterparts. Of the 55 DEGs identified, the top three most expressed transcription factor genes (transcription factor AP-2 beta isoform 3 [TFAP2B], developing brain homeobox protein 2 [DBX2], and insulin gene enhancer protein ISL-1 [ISL1]) and three tooth development-related genes (transcription factor HES-5 [HES5], platelet-derived growth factor D precursor [PDGFD], semaphrin-3 A precursor [SEMA3A]) were selected and validated by quantitative RT-PCR. Using immunofluorescence staining, the TFAP2B protein expression was found to be localized only at the E11.5 epithelium and E14.5 mesenchyme. CONCLUSIONS: Thus, our empirical findings in the present study may provide a new perspective into the characterization of the molecules responsible for the odontogenic potential and may have an implication in the cell-based whole tooth regeneration strategy.


Subject(s)
Molar/growth & development , Odontogenesis/genetics , Tooth Germ/growth & development , Transcriptome/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Epithelium/growth & development , Epithelium/metabolism , Gene Expression Regulation, Developmental/genetics , Genetic Association Studies , Homeodomain Proteins/genetics , Humans , LIM-Homeodomain Proteins/genetics , Lymphokines/genetics , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Molar/metabolism , Platelet-Derived Growth Factor/genetics , RNA-Seq , Repressor Proteins/genetics , Semaphorin-3A/genetics , Tooth Germ/metabolism , Transcription Factor AP-2/genetics , Transcription Factors/genetics
13.
Genome Med ; 13(1): 80, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33971972

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension (PAH) is a lethal vasculopathy characterized by pathogenic remodeling of pulmonary arterioles leading to increased pulmonary pressures, right ventricular hypertrophy, and heart failure. PAH can be associated with other diseases (APAH: connective tissue diseases, congenital heart disease, and others) but often the etiology is idiopathic (IPAH). Mutations in bone morphogenetic protein receptor 2 (BMPR2) are the cause of most heritable cases but the vast majority of other cases are genetically undefined. METHODS: To identify new risk genes, we utilized an international consortium of 4241 PAH cases with exome or genome sequencing data from the National Biological Sample and Data Repository for PAH, Columbia University Irving Medical Center, and the UK NIHR BioResource - Rare Diseases Study. The strength of this combined cohort is a doubling of the number of IPAH cases compared to either national cohort alone. We identified protein-coding variants and performed rare variant association analyses in unrelated participants of European ancestry, including 1647 IPAH cases and 18,819 controls. We also analyzed de novo variants in 124 pediatric trios enriched for IPAH and APAH-CHD. RESULTS: Seven genes with rare deleterious variants were associated with IPAH with false discovery rate smaller than 0.1: three known genes (BMPR2, GDF2, and TBX4), two recently identified candidate genes (SOX17, KDR), and two new candidate genes (fibulin 2, FBLN2; platelet-derived growth factor D, PDGFD). The new genes were identified based solely on rare deleterious missense variants, a variant type that could not be adequately assessed in either cohort alone. The candidate genes exhibit expression patterns in lung and heart similar to that of known PAH risk genes, and most variants occur in conserved protein domains. For pediatric PAH, predicted deleterious de novo variants exhibited a significant burden compared to the background mutation rate (2.45×, p = 2.5e-5). At least eight novel pediatric candidate genes carrying de novo variants have plausible roles in lung/heart development. CONCLUSIONS: Rare variant analysis of a large international consortium identified two new candidate genes-FBLN2 and PDGFD. The new genes have known functions in vasculogenesis and remodeling. Trio analysis predicted that ~ 15% of pediatric IPAH may be explained by de novo variants.


Subject(s)
Biomarkers , Calcium-Binding Proteins/genetics , Extracellular Matrix Proteins/genetics , Genetic Predisposition to Disease , Genetic Variation , Lymphokines/genetics , Platelet-Derived Growth Factor/genetics , Pulmonary Arterial Hypertension/epidemiology , Pulmonary Arterial Hypertension/etiology , Adolescent , Adult , Age of Onset , Aged , Alleles , Amino Acid Substitution , Calcium-Binding Proteins/chemistry , Child , Child, Preschool , Extracellular Matrix Proteins/chemistry , Female , Genotype , Humans , Lymphokines/chemistry , Male , Middle Aged , Mutation , Phenotype , Platelet-Derived Growth Factor/chemistry , Population Surveillance , United Kingdom/epidemiology , United States/epidemiology , Young Adult
14.
Oncogene ; 40(11): 1957-1973, 2021 03.
Article in English | MEDLINE | ID: mdl-33603171

ABSTRACT

Targeted therapies for gastrointestinal stromal tumor (GIST) are modestly effective, but GIST cannot be cured with single agent tyrosine kinase inhibitors. In this study, we sought to identify new therapeutic targets in GIST by investigating the tumor microenvironment. Here, we identified a paracrine signaling network by which cancer-associated fibroblasts (CAFs) drive GIST growth and metastasis. Specifically, CAFs isolated from human tumors were found to produce high levels of platelet-derived growth factor C (PDGFC), which activated PDGFC-PDGFRA signal transduction in GIST cells that regulated the expression of SLUG, an epithelial-mesenchymal transition (EMT) transcription factor and downstream target of PDGFRA signaling. Together, this paracrine induce signal transduction cascade promoted tumor growth and metastasis in vivo. Moreover, in metastatic GIST patients, SLUG expression positively correlated with tumor size and mitotic index. Given that CAF paracrine signaling modulated GIST biology, we directly targeted CAFs with a dual PI3K/mTOR inhibitor, which synergized with imatinib to increase tumor cell killing and in vivo disease response. Taken together, we identified a previously unappreciated cellular target for GIST therapy in order to improve disease control and cure rates.


Subject(s)
Gastrointestinal Stromal Tumors/genetics , Lymphokines/genetics , Platelet-Derived Growth Factor/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Snail Family Transcription Factors/genetics , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/pathology , Humans , Neoplasm Metastasis , Paracrine Communication/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Tumor Microenvironment/drug effects
15.
Eur J Pharmacol ; 895: 173868, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33460613

ABSTRACT

Complex of platelet-derived growth factor (PDGF) isoforms and PDGF receptors have important functions in the regulation of growth and survival of various cell types. Herein, it was found that aberrant PDGFC expression is closely associated with survival rates in triple-negative breast cancer (TNBC) patients. In addition, PDGFC expression was identified to be significantly increased in TNBC cells unlike other subtypes such as PDGFA, PDGFB, and PDGFD. Apparently, the effects of specific PDGF receptor (PDGFR) inhibitors such as sunitinib and ponatinib on HCC1806 and Hs578T TNBC cells were investigated. Both inhibitors decreased cell viability in a dose-dependent manner. In addition, the inhibitors completely inhibited cell growth in both the cell lines and decreased the expression of matrix metalloproteinase-1 (MMP-1), one of the metastasis-related genes. Cell migration was also decreased by the inhibitors. Finally, the combined effects of the inhibitors with doxorubicin (DOX) were investigated. The results showed that the combination of two PDGFR inhibitors with DOX inhibited the growth of cells and enhanced the apoptotic cell death more uniformly than DOX. Consequently, it is demonstrated that PDGFR inhibitors, sunitinib and ponatinib hold the potential for effective treatment of TNBC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Doxorubicin/pharmacology , Imidazoles/pharmacology , Lymphokines/antagonists & inhibitors , Platelet-Derived Growth Factor/antagonists & inhibitors , Pyridazines/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Sunitinib/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphokines/genetics , Lymphokines/metabolism , Matrix Metalloproteinase 1/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
16.
Circ Res ; 128(4): e46-e62, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33375813

ABSTRACT

RATIONALE: Pericytes are capillary mural cells playing a role in stabilizing newly formed blood vessels during development and tissue repair. Loss of pericytes has been described in several brain disorders, and genetically induced pericyte deficiency in the brain leads to increased macromolecular leakage across the blood-brain barrier (BBB). However, the molecular details of the endothelial response to pericyte deficiency remain elusive. OBJECTIVE: To map the transcriptional changes in brain endothelial cells resulting from lack of pericyte contact at single-cell level and to correlate them with regional heterogeneities in BBB function and vascular phenotype. METHODS AND RESULTS: We reveal transcriptional, morphological, and functional consequences of pericyte absence for brain endothelial cells using a combination of methodologies, including single-cell RNA sequencing, tracer analyses, and immunofluorescent detection of protein expression in pericyte-deficient adult Pdgfbret/ret mice. We find that endothelial cells without pericyte contact retain a general BBB-specific gene expression profile, however, they acquire a venous-shifted molecular pattern and become transformed regarding the expression of numerous growth factors and regulatory proteins. Adult Pdgfbret/ret brains display ongoing angiogenic sprouting without concomitant cell proliferation providing unique insights into the endothelial tip cell transcriptome. We also reveal heterogeneous modes of pericyte-deficient BBB impairment, where hotspot leakage sites display arteriolar-shifted identity and pinpoint putative BBB regulators. By testing the causal involvement of some of these using reverse genetics, we uncover a reinforcing role for angiopoietin 2 at the BBB. CONCLUSIONS: By elucidating the complexity of endothelial response to pericyte deficiency at cellular resolution, our study provides insight into the importance of brain pericytes for endothelial arterio-venous zonation, angiogenic quiescence, and a limited set of BBB functions. The BBB-reinforcing role of ANGPT2 (angiopoietin 2) is paradoxical given its wider role as TIE2 (TEK receptor tyrosine kinase) receptor antagonist and may suggest a unique and context-dependent function of ANGPT2 in the brain.


Subject(s)
Blood-Brain Barrier/metabolism , Pericytes/cytology , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/pathology , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/physiology , Lymphokines/deficiency , Lymphokines/genetics , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic , Pericytes/metabolism , Pericytes/pathology , Platelet-Derived Growth Factor/deficiency , Platelet-Derived Growth Factor/genetics , Single-Cell Analysis , Transcriptome
17.
J Hepatol ; 74(2): 380-393, 2021 02.
Article in English | MEDLINE | ID: mdl-32916216

ABSTRACT

BACKGROUND & AIMS: Angiocrine signaling by liver sinusoidal endothelial cells (LSECs) regulates hepatic functions such as growth, metabolic maturation, and regeneration. Recently, we identified GATA4 as the master regulator of LSEC specification during development. Herein, we studied the role of endothelial GATA4 in the adult liver and in hepatic pathogenesis. METHODS: We generated adult Clec4g-icretg/0xGata4fl/fl (Gata4LSEC-KO) mice with LSEC-specific depletion of Gata4. Livers were analyzed by histology, electron microscopy, immunohistochemistry/immunofluorescence, in situ hybridization, and LSECs were isolated for gene expression profiling, ChIP- and ATAC-sequencing. Partial hepatectomy was performed to assess regeneration. We used choline-deficient, l-amino acid-defined (CDAA) diet and chronic carbon tetrachloride exposure to model liver fibrosis. Human single cell RNA-seq data sets were analyzed for endothelial alterations in healthy and cirrhotic livers. RESULTS: Genetic Gata4 deficiency in LSECs of adult mice caused perisinusoidal liver fibrosis, hepatopathy and impaired liver regeneration. Sinusoidal capillarization and LSEC-to-continuous endothelial transdifferentiation were accompanied by a profibrotic angiocrine switch involving de novo endothelial expression of hepatic stellate cell-activating cytokine PDGFB. Increased chromatin accessibility and amplification by activated MYC mediated angiocrine Pdgfb expression. As observed in Gata4LSEC-KO livers, CDAA diet-induced perisinusoidal liver fibrosis was associated with GATA4 repression, MYC activation and a profibrotic angiocrine switch in LSECs. Comparison of CDAA-fed Gata4LSEC-KO and control mice demonstrated that endothelial GATA4 indeed protects against dietary-induced perisinusoidal liver fibrosis. In human cirrhotic livers, GATA4-positive LSECs and endothelial GATA4 target genes were reduced, while non-LSEC endothelial cells and MYC target genes including PDGFB were enriched. CONCLUSIONS: Endothelial GATA4 protects against perisinusoidal liver fibrosis by repressing MYC activation and profibrotic angiocrine signaling at the chromatin level. Therapies targeting the GATA4/MYC/PDGFB/PDGFRß axis offer a promising strategy for prevention and treatment of liver fibrosis. LAY SUMMARY: The liver vasculature is supposed to play a major role in the development of liver fibrosis and cirrhosis, which can lead to liver failure and liver cancer. Herein, we discovered that structural and transcriptional changes induced by genetic deletion of the transcription factor GATA4 in the hepatic endothelium were sufficient to cause liver fibrosis. Activation of the transcription factor MYC and de novo expression of the "angiocrine" growth factor PDGFB were identified as downstream drivers of fibrosis and as potential therapeutic targets for this potentially fatal disease.


Subject(s)
Endothelial Cells/metabolism , GATA4 Transcription Factor/metabolism , Liver Cirrhosis , Liver , Lymphokines , Platelet-Derived Growth Factor , Animals , Chromatin/metabolism , Drug Discovery , Gene Expression Profiling , Hepatic Stellate Cells/metabolism , Humans , Liver/blood supply , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/prevention & control , Liver Regeneration/physiology , Lymphokines/genetics , Lymphokines/metabolism , Mice , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Signal Transduction/drug effects , Zinc Fingers
18.
Sci Rep ; 10(1): 22383, 2020 12 24.
Article in English | MEDLINE | ID: mdl-33361796

ABSTRACT

Disruption of blood-brain barrier (BBB) integrity is a feature of various neurological disorders. Here we found that the BBB is differently affected during the preclinical, progression and remission phase of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). We have identified an upregulation of pro-inflammatory and pro-angiogenic factors in the BBB transcriptome and down-regulation of endothelial tight junction members coinciding with elevated BBB leakage specifically during the progression phase. These changes were antagonized by blocking PDGFRα signaling with the small tyrosine kinase inhibitor imatinib. Moreover, targeting the PDGFRα ligand PDGF-CC using a neutralizing antibody, facilitated recovery of BBB integrity and improvement of EAE symptoms. Intracerebroventricular injection of PDGF-CC induced upregulation, whereas blocking PDGF-CC during EAE led to downregulation of Tnfa and Il1a at the BBB. Our findings suggest that blocking PDGF-CC counteracts fundamental aspects of endothelial cell activation and disruption of the BBB by decreasing Tnfa and Il1a expression. We also demonstrate that both PDGF-CC and its receptor PDGFRα were upregulated in MS lesions indicating that blocking PDGF-CC may be considered a novel treatment for MS.


Subject(s)
Antibodies, Neutralizing/pharmacology , Blood-Brain Barrier/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymphokines/antagonists & inhibitors , Multiple Sclerosis/immunology , Platelet-Derived Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Blood-Brain Barrier/pathology , Down-Regulation/drug effects , Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/drug therapy , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Lymphokines/genetics , Lymphokines/immunology , Mice , Mice, Transgenic , Multiple Sclerosis/drug therapy , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
19.
PLoS One ; 15(12): e0232101, 2020.
Article in English | MEDLINE | ID: mdl-33306672

ABSTRACT

Autism susceptibility candidate 2 (AUTS2) is a neurodevelopmental regulator associated with an autosomal dominant intellectual disability syndrome, AUTS2 syndrome, and is implicated as an important gene in human-specific evolution. AUTS2 exists as part of a tripartite gene family, the AUTS2 family, which includes two relatively undefined proteins, Fibrosin (FBRS) and Fibrosin-like protein 1 (FBRSL1). Evolutionary ancestors of AUTS2 have not been formally identified outside of the Animalia clade. A Drosophila melanogaster protein, Tay bridge, with a role in neurodevelopment, has been shown to display limited similarity to the C-terminal of AUTS2, suggesting that evolutionary ancestors of the AUTS2 family may exist within other Protostome lineages. Here we present an evolutionary analysis of the AUTS2 family, which highlights ancestral homologs of AUTS2 in multiple Protostome species, implicates AUTS2 as the closest human relative to the progenitor of the AUTS2 family, and demonstrates that Tay bridge is a divergent ortholog of the ancestral AUTS2 progenitor gene. We also define regions of high relative sequence identity, with potential functional significance, shared by the extended AUTS2 protein family. Using structural predictions coupled with sequence conservation and human variant data from 15,708 individuals, a putative domain structure for AUTS2 was produced that can be used to aid interpretation of the consequences of nucleotide variation on protein structure and function in human disease. To assess the role of AUTS2 in human-specific evolution, we recalculated allele frequencies at previously identified human derived sites using large population genome data, and show a high prevalence of ancestral alleles, suggesting that AUTS2 may not be a rapidly evolving gene, as previously thought.


Subject(s)
Cytoskeletal Proteins/genetics , Evolution, Molecular , Transcription Factors/genetics , Animals , Cytoskeletal Proteins/metabolism , Drosophila melanogaster/genetics , Genome/genetics , Humans , Lymphokines/genetics , Lymphokines/metabolism , Nuclear Proteins/genetics , Phylogeny , Polycomb-Group Proteins/genetics , Sequence Homology, Nucleic Acid , Transcription Factors/metabolism
20.
Genes (Basel) ; 11(11)2020 11 11.
Article in English | MEDLINE | ID: mdl-33187088

ABSTRACT

Pulmonary arterial hypertension (PAH) describes a rare, progressive vascular disease caused by the obstruction of pulmonary arterioles, typically resulting in right heart failure. Whilst PAH most often manifests in adulthood, paediatric disease is considered to be a distinct entity with increased morbidity and often an unexplained resistance to current therapies. Recent genetic studies have substantially increased our understanding of PAH pathogenesis, providing opportunities for molecular diagnosis and presymptomatic genetic testing in families. However, the genetic architecture of childhood-onset PAH remains relatively poorly characterised. We sought to investigate a previously unsolved paediatric cohort (n = 18) using whole exome sequencing to improve the molecular diagnosis of childhood-onset PAH. Through a targeted investigation of 26 candidate genes, we applied a rigorous variant filtering methodology to enrich for rare, likely pathogenic variants. This analysis led to the detection of novel PAH risk alleles in five genes, including the first identification of a heterozygous ATP13A3 mutation in childhood-onset disease. In addition, we provide the first independent validation of BMP10 and PDGFD as genetic risk factors for PAH. These data provide a molecular diagnosis in 28% of paediatric cases, reflecting the increased genetic burden in childhood-onset disease and highlighting the importance of next-generation sequencing approaches to diagnostic surveillance.


Subject(s)
Pulmonary Arterial Hypertension/genetics , Adenosine Triphosphatases/genetics , Bone Morphogenetic Proteins/genetics , Child , Child, Preschool , Cohort Studies , Female , Genetic Predisposition to Disease/genetics , Humans , Infant , Lymphokines/genetics , Male , Membrane Transport Proteins/genetics , Mutation , Platelet-Derived Growth Factor/genetics , Risk Factors , Smad8 Protein/genetics , Sulfonylurea Receptors/genetics , Exome Sequencing/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...