95 results on '"Frank J. Giordano"'
Search Results
2. Low-dose Dasatinib Ameliorates Hypertrophic Cardiomyopathy in Noonan Syndrome with Multiple Lentigines
- Author
-
Kana Mizuno, Sravan K. Perla, Jae Sung Yi, Anton M. Bennett, Alexander A. Vinks, Frank J. Giordano, and Yan Huang
- Subjects
0301 basic medicine ,Cardiac fibrosis ,medicine.drug_class ,Dasatinib ,030204 cardiovascular system & hematology ,Pharmacology ,Tyrosine-kinase inhibitor ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Pharmacokinetics ,hemic and lymphatic diseases ,medicine ,LEOPARD Syndrome ,Animals ,Pharmacology (medical) ,business.industry ,Hypertrophic cardiomyopathy ,General Medicine ,Cardiomyopathy, Hypertrophic ,medicine.disease ,PTPN11 ,Disease Models, Animal ,030104 developmental biology ,Mutation ,Cardiology and Cardiovascular Medicine ,business ,Noonan Syndrome with Multiple Lentigines ,Proto-oncogene tyrosine-protein kinase Src ,medicine.drug - Abstract
Purpose Noonan syndrome with multiple lentigines (NSML) is an autosomal dominant disorder presenting with hypertrophic cardiomyopathy (HCM). Up to 85% of NSML cases are caused by mutations in the PTPN11 gene that encodes for the Src homology 2 (SH2) domain-containing protein tyrosine phosphatase 2 (SHP2). We previously showed that low-dose dasatinib protects from the development of cardiac fibrosis in a mouse model of NSML harboring a Ptpn11Y279C mutation. This study is performed to determine the pharmacokinetic (PK) and pharmacodynamic (PD) properties of a low-dose of dasatinib in NSML mice and to determine its effectiveness in ameliorating the development of HCM. Methods Dasatinib was administered intraperitoneally into NSML mice with doses ranging from 0.05 to 0.5 mg/kg. PK parameters of dasatinib in NSML mice were determined. PD parameters were obtained for biochemical analyses from heart tissue. Dasatinib-treated NSML mice (0.1 mg/kg) were subjected to echocardiography and assessment of markers of HCM by qRT-PCR. Transcriptome analysis was performed from the heart tissue of low-dose dasatinib-treated mice. Results Low-dose dasatinib exhibited PK properties that were linear across doses in NSML mice. Dasatinib treatment of between 0.05 and 0.5 mg/kg in NSML mice yielded an exposure-dependent inhibition of c-Src and PZR tyrosyl phosphorylation and inhibited AKT phosphorylation. We found that doses as low as 0.1 mg/kg of dasatinib prevented HCM in NSML mice. Transcriptome analysis identified differentially expressed HCM-associated genes in the heart of NSML mice that were reverted to wild type levels by low-dose dasatinib administration. Conclusion These data demonstrate that low-dose dasatinib exhibits desirable therapeutic PK properties that is sufficient for effective target engagement to ameliorate HCM progression in NSML mice. These data demonstrate that low-dose dasatinib treatment may be an effective therapy against HCM in NSML patients.
- Published
- 2021
3. Abstract 16205: MKP-5 Deficiency Attenuates Pressure Overload-induced Cardiac Hypertrophy
- Author
-
Yan Huang, Anton M. Bennett, Frank J. Giordano, Sudip Bajpeyi, and Kisuk Min
- Subjects
Pressure overload ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Hypertrophic cardiomyopathy ,medicine.disease ,Fibrosis ,Physiology (medical) ,Internal medicine ,Cardiac hypertrophy ,Heart failure ,medicine ,Cardiology ,Cardiology and Cardiovascular Medicine ,business ,Pathological ,Electrocardiography - Abstract
Introduction: Cardiac remodeling occurs in response to pathological stimuli including chronic pressure overload, subsequently leading to heart failure. Despite considerable research efforts, the molecular mechanisms responsible for heart failure have yet to be fully elucidated. One of the prominent signaling pathways involved in the development of pathological cardiac hypertrophy is the mitogen-activated protein kinases (MAPKs) pathways. The MAPKs are inactivated by the MAPK phosphatases (MKPs) through direct dephosphorylation. Growing evidence suggests the importance of MKP-5 signaling mechanisms in physiological and pathological processes. However, the role of MKP-5 has not been explored in cardiac muscle. The objective of this study is to investigate how MKP-5-mediated MAPK activity contributes to mechanisms responsible for pressure overload-induced cardiac hypertrophy. Hypothesis: We tested the hypothesis that MKP-5 serves as a central regulator of MAPKs in pressure overload-induced cardiac hypertrophy. Methods: To investigate the role of MKP-5 in cardiac muscle, we caused pressure overload-induced cardiac hypertrophy in wild type (mkp-5 +/+ ) mice and MKP-5 deficient mice (mkp-5 -/- ) through transverse aortic constriction (TAC). Cardiac function was evaluated by echocardiographic analysis at 4 weeks after TAC. Cardiac hypertrophy was measured by heart-to-body weight ratio. Interstitial myocardial fibrosis was evaluated by Sirius red stains and expression of fibrogenic genes was determined by quantitative PCR. Results: Echocardiographic analysis showed that the ejection fraction and fractional shortening of mkp-5 +/+ mice significantly decreased by at 4 weeks after TAC. Heart-to-body weight ratio increased in mkp-5 +/+ mice. However, MKP-5-deficient heart was protected from cardiac dysfunction and cardiac hypertrophy induced by TAC. Importantly, the fibrogenic genes were markedly reduced in mkp-5 -/- mice as compared with mkp-5 +/+ mice at 4 weeks after TAC. Conclusions: Collectively, our study demonstrates that MKP-5 deficiency prevents the heart from pressure overload-induced cardiac hypertrophy and suggests that MKP-5 may serve as a novel therapeutic target for treatment of heart disease.
- Published
- 2020
- Full Text
- View/download PDF
4. Cover Image, Volume 58, Issue 3‐4
- Author
-
Qun Lin, Yan Huang, Frank J. Giordano, and Zhong Yun
- Subjects
Endocrinology ,Genetics ,Cell Biology - Published
- 2020
- Full Text
- View/download PDF
5. Loss of skeletal muscle HIF-1alpha results in altered exercise endurance.
- Author
-
Steven D Mason, Richard A Howlett, Matthew J Kim, I Mark Olfert, Michael C Hogan, Wayne McNulty, Reed P Hickey, Peter D Wagner, C Ronald Kahn, Frank J Giordano, and Randall S Johnson
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The physiological flux of oxygen is extreme in exercising skeletal muscle. Hypoxia is thus a critical parameter in muscle function, influencing production of ATP, utilization of energy-producing substrates, and manufacture of exhaustion-inducing metabolites. Glycolysis is the central source of anaerobic energy in animals, and this metabolic pathway is regulated under low-oxygen conditions by the transcription factor hypoxia-inducible factor 1alpha (HIF-1alpha). To determine the role of HIF-1alpha in regulating skeletal muscle function, we tissue-specifically deleted the gene encoding the factor in skeletal muscle. Significant exercise-induced changes in expression of genes are decreased or absent in the skeletal-muscle HIF-1alpha knockout mice (HIF-1alpha KOs); changes in activities of glycolytic enzymes are seen as well. There is an increase in activity of rate-limiting enzymes of the mitochondria in the muscles of HIF-1alpha KOs, indicating that the citric acid cycle and increased fatty acid oxidation may be compensating for decreased flow through the glycolytic pathway. This is corroborated by a finding of no significant decreases in muscle ATP, but significantly decreased amounts of lactate in the serum of exercising HIF-1alpha KOs. This metabolic shift away from glycolysis and toward oxidation has the consequence of increasing exercise times in the HIF-1alpha KOs. However, repeated exercise trials give rise to extensive muscle damage in HIF-1alpha KOs, ultimately resulting in greatly reduced exercise times relative to wild-type animals. The muscle damage seen is similar to that detected in humans in diseases caused by deficiencies in skeletal muscle glycogenolysis and glycolysis. Thus, these results demonstrate an important role for the HIF-1 pathway in the metabolic control of muscle function.
- Published
- 2004
- Full Text
- View/download PDF
6. Generation of a hypoxia-sensing mouse model
- Author
-
Zhong Yun, Qun Lin, Yan Huang, and Frank J. Giordano
- Subjects
Recombinant Fusion Proteins ,Ischemia ,Cre recombinase ,Mice, Transgenic ,Biology ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Endocrinology ,Genes, Reporter ,Genetics ,medicine ,Animals ,Hypoxia ,030304 developmental biology ,0303 health sciences ,Embryogenesis ,Reproducibility of Results ,Embryo ,Cell Biology ,Hypoxia (medical) ,medicine.disease ,Embryonic stem cell ,Cell biology ,Oxygen ,Hypoxia-inducible factors ,Gene Expression Regulation ,Models, Animal ,medicine.symptom ,030217 neurology & neurosurgery ,Homeostasis ,Signal Transduction - Abstract
Oxygen (O2 ) homeostasis is essential to the metazoan life. O2 -sensing or hypoxia-regulated molecular pathways are intimately involved in a wide range of critical cellular functions and cell survival from embryogenesis to adulthood. In this report, we have designed an innovative hypoxia sensor (O2 CreER) based on the O2 -dependent degradation domain of the hypoxia-inducible factor-1α and Cre recombinase. We have further generated a hypoxia-sensing mouse model, R26-O2 CreER, by targeted insertion of the O2 CreER-coding cassette in the ROSA26 locus. Using the ROSAmTmG mouse strain as a reporter, we have found that this novel hypoxia-sensing mouse model can specifically identify hypoxic cells under the pathological condition of hind-limb ischemia in adult mice. This model can also label embryonic cells including vibrissal follicle cells in E13.5-E15.5 embryos. This novel mouse model offers a valuable genetic tool for the study of hypoxia and O2 sensing in mammalian systems under both physiological and pathological conditions.
- Published
- 2019
7. Smooth Muscle Hypoxia-Inducible Factor 1α Links Intravascular Pressure and Atherosclerosis—Brief Report
- Author
-
Dinggang Liu, Li Lei, Frank J. Giordano, Jacob Cleman, Yan Huang, Weidong Jiang, Carlos Fernández-Hernando, William C. Sessa, Annarita Di Lorenzo, and Matthew Desir
- Subjects
0301 basic medicine ,Apolipoprotein E ,medicine.medical_specialty ,Time Factors ,Vascular smooth muscle ,Aortic Diseases ,Inflammation ,030204 cardiovascular system & hematology ,Biology ,Diet, High-Fat ,Muscle, Smooth, Vascular ,Pathogenesis ,03 medical and health sciences ,Apolipoproteins E ,0302 clinical medicine ,Internal medicine ,medicine.artery ,medicine ,Animals ,Arterial Pressure ,Ligation ,Aorta ,Mice, Knockout ,Atherosclerosis ,Hypoxia-Inducible Factor 1, alpha Subunit ,Plaque, Atherosclerotic ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Blood pressure ,Gene Expression Regulation ,Hypoxia-inducible factors ,Regional Blood Flow ,Cardiology ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Blood Flow Velocity ,Signal Transduction - Abstract
Objective— We hypothesized that the hypoxia-inducible factor (HIF) 1α in vascular smooth muscle contributes to the development of atherosclerosis, and links intravascular pressure to this process. Approach and Results— Transverse aortic constriction was used to create high-pressure vascular segments in control, apolipoprotein E ( ApoE ) −/− , smooth muscle- HIF1α −/− , and ApoE −/− ×smooth muscle- HIF1α −/− double-knockout mice. Transverse aortic constriction selectively induced atherosclerosis in high-pressure vascular segments in young ApoE −/− mice on normal chow, including coronary plaques within 1 month. Concomitant deletion of HIF1α from smooth muscle significantly reduced vascular inflammation, and attenuated atherosclerosis. Conclusions— HIF1α in vascular smooth muscle plays an important role in the pathogenesis of atherosclerosis, and may provide a mechanistic link between blood pressure, vascular inflammation, and lipid deposition.
- Published
- 2016
- Full Text
- View/download PDF
8. Nogo-B regulates endothelial sphingolipid homeostasis to control vascular function and blood pressure
- Author
-
Frank J. Giordano, Hideru Obinata, Timothy Hla, Sylvain Galvani, Yi Zhang, Annarita Di Lorenzo, Milankumar Kothiya, Xian-Cheng Jiang, Mariarosaria Bucci, Anna Cantalupo, Cantalupo, A, Zhang, Ying, Kothiya, M, Galvani, S, Obinata, H, Bucci, M, Giordano, Fj, Jiang, Xc, Solin, HEIKKI LAURI ABEL, and Di Lorenzo, A
- Subjects
Male ,medicine.medical_specialty ,Nitric Oxide Synthase Type III ,Endothelium ,Nogo Proteins ,Blood Pressure ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Mice ,chemistry.chemical_compound ,Sphingosine ,Myriocin ,Internal medicine ,Chlorocebus aethiops ,medicine ,Animals ,Homeostasis ,Humans ,Endothelial dysfunction ,endothelium dysfunction, lipid mediators, blood pressure ,Autocrine signalling ,Sphingolipids ,Serine C-palmitoyltransferase ,General Medicine ,Lipid signaling ,medicine.disease ,Sphingolipid ,Cell biology ,Receptors, Lysosphingolipid ,HEK293 Cells ,Endocrinology ,medicine.anatomical_structure ,chemistry ,COS Cells ,Endothelium, Vascular ,Lysophospholipids ,Myelin Proteins - Abstract
Endothelial dysfunction is a critical factor in many cardiovascular diseases, including hypertension. Although lipid signaling has been implicated in endothelial dysfunction and cardiovascular disease, specific molecular mechanisms are poorly understood. Here we report that Nogo-B, a membrane protein of the endoplasmic reticulum, regulates endothelial sphingolipid biosynthesis with direct effects on vascular function and blood pressure. Nogo-B inhibits serine palmitoyltransferase, the rate-limiting enzyme of the de novo sphingolipid biosynthetic pathway, thereby controlling production of endothelial sphingosine 1-phosphate and autocrine, G protein-coupled receptor-dependent signaling by this metabolite. Mice lacking Nogo-B either systemically or specifically in endothelial cells are hypotensive, resistant to angiotensin II-induced hypertension and have preserved endothelial function and nitric oxide release. In mice that lack Nogo-B, pharmacological inhibition of serine palmitoyltransferase with myriocin reinstates endothelial dysfunction and angiotensin II-induced hypertension. Our study identifies Nogo-B as a key inhibitor of local sphingolipid synthesis and shows that autocrine sphingolipid signaling within the endothelium is critical for vascular function and blood pressure homeostasis.
- Published
- 2015
- Full Text
- View/download PDF
9. Endothelial Uncoupling Protein 2 Regulates Mitophagy and Pulmonary Hypertension During Intermittent Hypoxia
- Author
-
Michael J. Jurczak, Patty J. Lee, Frank J. Giordano, Serpil C. Erzurum, Yi Zhang, Iva Dostanic, Kerry S. Russell, Maria Haslip, Yan Huang, and Praveen Mannam
- Subjects
medicine.medical_specialty ,Endothelium ,Hypertension, Pulmonary ,Biology ,Ion Channels ,Article ,Mitochondrial Proteins ,Mice ,Random Allocation ,Reference Values ,Internal medicine ,Mitophagy ,Autophagy ,medicine ,Animals ,Humans ,Uncoupling Protein 2 ,Hypoxia ,Cells, Cultured ,Mice, Knockout ,Hypertrophy, Right Ventricular ,Endothelial Cells ,Intermittent hypoxia ,Hypoxia (medical) ,medicine.disease ,Pulmonary hypertension ,Endothelial stem cell ,Disease Models, Animal ,medicine.anatomical_structure ,Endocrinology ,Mitochondrial biogenesis ,Biochemistry ,Knockout mouse ,Endothelium, Vascular ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Protein Kinases - Abstract
Objectives— Pulmonary hypertension (PH) is a process of lung vascular remodeling, which can lead to right heart dysfunction and significant morbidity. The underlying mechanisms leading to PH are not well understood, and therapies are limited. Using intermittent hypoxia (IH) as a model of oxidant-induced PH, we identified an important role for endothelial cell mitophagy via mitochondrial uncoupling protein 2 (Ucp2) in the development of IH-induced PH. Approach and Results— Ucp2 endothelial knockout (VE-KO) and Ucp2 Flox (Flox) mice were subjected to 5 weeks of IH. Ucp2 VE-KO mice exhibited higher right ventricular systolic pressure and worse right heart hypertrophy, as measured by increased right ventricle weight/left ventricle plus septal weight (RV/LV+S) ratio, at baseline and after IH. These changes were accompanied by increased mitophagy. Primary mouse lung endothelial cells transfected with Ucp2 siRNA and subjected to cyclic exposures to CoCl 2 (chemical hypoxia) showed increased mitophagy, as measured by PTEN-induced putative kinase 1 and LC3BII/I ratios, decreased mitochondrial biogenesis, and increased apoptosis. Similar results were obtained in primary lung endothelial cells isolated from VE-KO mice. Moreover, silencing PTEN-induced putative kinase 1 in the endothelium of Ucp2 knockout mice, using endothelial-targeted lentiviral silencing RNA in vivo, prevented IH-induced PH. Human pulmonary artery endothelial cells from people with PH demonstrated changes similar to Ucp2-silenced mouse lung endothelial cells. Conclusions— The loss of endothelial Ucp2 leads to excessive PTEN-induced putative kinase 1–induced mitophagy, inadequate mitochondrial biosynthesis, and increased apoptosis in endothelium. An endothelial Ucp2–PTEN-induced putative kinase 1 axis may be effective therapeutic targets in PH.
- Published
- 2015
- Full Text
- View/download PDF
10. Thioredoxin-2 Inhibits Mitochondrial Reactive Oxygen Species Generation and Apoptosis Stress Kinase-1 Activity to Maintain Cardiac Function
- Author
-
George Tellides, Ford Hinojosa-Kirschenbaum, Frank J. Giordano, Qunhua Huang, Haifeng Zhang, Huanjiao Jenny Zhou, Wang Min, Yan Huang, Grant R. Budas, Peidong Fan, Luiz Belardinelli, and Lina Yao
- Subjects
Pathology ,medicine.medical_specialty ,Cardiomegaly ,Mitochondrion ,Biology ,MAP Kinase Kinase Kinase 5 ,medicine.disease_cause ,Mitochondria, Heart ,Article ,Mice ,Thioredoxins ,Physiology (medical) ,medicine ,Animals ,Humans ,ASK1 ,Ventricular remodeling ,Cells, Cultured ,Mice, Knockout ,chemistry.chemical_classification ,Reactive oxygen species ,Dilated cardiomyopathy ,medicine.disease ,Cell biology ,chemistry ,Heart failure ,Thioredoxin ,Reactive Oxygen Species ,Cardiology and Cardiovascular Medicine ,Oxidative stress - Abstract
Background— Thioredoxin 2 (Trx2) is a key mitochondrial protein that regulates cellular redox and survival by suppressing mitochondrial reactive oxygen species generation and by inhibiting apoptosis stress kinase-1 (ASK1)–dependent apoptotic signaling. To date, the role of the mitochondrial Trx2 system in heart failure pathogenesis has not been investigated. Methods and Results— Western blot and histological analysis revealed that Trx2 protein expression levels were reduced in hearts from patients with dilated cardiomyopathy, with a concomitant increase in ASK1 phosphorylation/activity. Cardiac-specific Trx2 knockout mice develop spontaneous dilated cardiomyopathy at 1 month of age with increased heart size, reduced ventricular wall thickness, and a progressive decline in left ventricular contractile function, resulting in mortality due to heart failure by ≈4 months of age. The progressive decline in cardiac function observed in cardiac-specific Trx2 knockout mice was accompanied by the disruption of mitochondrial ultrastructure, mitochondrial membrane depolarization, increased mitochondrial reactive oxygen species generation, and reduced ATP production, correlating with increased ASK1 signaling and increased cardiomyocyte apoptosis. Chronic administration of a highly selective ASK1 inhibitor improved cardiac phenotype and reduced maladaptive left ventricular remodeling with significant reductions in oxidative stress, apoptosis, fibrosis, and cardiac failure. Cellular data from Trx2-deficient cardiomyocytes demonstrated that ASK1 inhibition reduced apoptosis and reduced mitochondrial reactive oxygen species generation. Conclusions— Our data support an essential role for mitochondrial Trx2 in preserving cardiac function by suppressing mitochondrial reactive oxygen species production and ASK1-dependent apoptosis. Inhibition of ASK1 represents a promising therapeutic strategy for the treatment of dilated cardiomyopathy and heart failure.
- Published
- 2015
- Full Text
- View/download PDF
11. HIF-1α represses the expression of the angiogenesis inhibitor thrombospondin-2
- Author
-
Themis R. Kyriakides, Nicole E. Calabro, Tara Bancroft, Tanuj Sharma, Yan Huang, Meenakshi Krishna, Frank J. Giordano, Jun Yu, Susan MacLauchlan, and William C. Sessa
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Transcription, Genetic ,Angiogenesis ,Myocytes, Smooth Muscle ,Matrix metalloproteinase ,Biology ,Article ,Muscle, Smooth, Vascular ,Nitric oxide ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,In vivo ,Internal medicine ,medicine ,Animals ,Humans ,Muscle, Skeletal ,Promoter Regions, Genetic ,Molecular Biology ,Messenger RNA ,Thrombospondin ,Hypoxia (medical) ,Hypoxia-Inducible Factor 1, alpha Subunit ,Cell Hypoxia ,Angiogenesis inhibitor ,Cell biology ,Amino Acids, Dicarboxylic ,030104 developmental biology ,Endocrinology ,HEK293 Cells ,chemistry ,030220 oncology & carcinogenesis ,NIH 3T3 Cells ,medicine.symptom ,Thrombospondins - Abstract
Thrombospondin-2 (TSP2) is a potent inhibitor of angiogenesis whose expression is dynamically regulated following injury. In the present study, it is shown that HIF-1α represses TSP2 transcription. Specifically, in vitro studies demonstrate that the prolyl hydroxylase inhibitor DMOG or hypoxia decrease TSP2 expression in fibroblasts. This effect is shown to be via a transcriptional mechanism as hypoxia does not alter TSP2 mRNA stability and this effect requires the TSP2 promoter. In addition, the documented repressive effect of nitric oxide (NO) on TSP2 is shown to be non-canonical and involves stabilization of hypoxia inducible factor-1a (HIF-1α). The regulation of TSP2 by hypoxia is supported by the in vivo observation that TSP2 has spatiotemporal expression distinct from regions of hypoxia in gastrocnemius muscle following murine hindlimb ischemia (HLI). A role for TSP2 regulation by HIF-1α is supported by the dysregulation of TSP2 expression in SM22α-cre HIF-1α KO mice following HLI. Indeed, there is a reduction in blood flow recovery in the SM22a-cre HIF-1α KO mice compared to littermate controls following HLI surgery, associated with impaired recovery and increased TSP2 levels. Moreover, SM22α-cre HIF-1α KO smooth muscle cells mice have increased TSP2 mRNA levels that persist in hypoxia. These findings identify a novel, ischemia-induced pro-angiogenic mechanism involving the transcriptional repression of TSP2 by HIF-1α.
- Published
- 2017
12. Polycystin 2-dependent cardio-protective mechanisms revealed by cardiac stress
- Author
-
Frank J. Giordano, Yan Huang, Esther Giehl, Barbara E. Ehrlich, Ivana Y. Kuo, and Fernanda O. Lemos
- Subjects
0301 basic medicine ,Cardiac function curve ,medicine.medical_specialty ,endocrine system ,Heterozygote ,TRPP Cation Channels ,Physiology ,medicine.drug_class ,Heart Ventricles ,Clinical Biochemistry ,Autosomal dominant polycystic kidney disease ,030204 cardiovascular system & hematology ,Biology ,urologic and male genital diseases ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Physiology (medical) ,Internal medicine ,Renin–angiotensin system ,medicine ,Natriuretic peptide ,Animals ,RNA, Messenger ,education ,education.field_of_study ,Aldosterone ,PKD1 ,urogenital system ,Heart ,medicine.disease ,Polycystic Kidney, Autosomal Dominant ,female genital diseases and pregnancy complications ,Up-Regulation ,Mice, Inbred C57BL ,030104 developmental biology ,Polycystin 2 ,Endocrinology ,Proprotein Convertase 2 ,chemistry ,Heart failure ,cardiovascular system ,Signal Transduction - Abstract
Although autosomal dominant polycystic kidney disease (ADPKD) is characterized by the development of multiple kidney cysts, the most frequent cause of death in ADPKD patients is cardiovascular disease. ADPKD is linked to mutations in PKD1 or pkd2, the genes that encode for the proteins polycystin 1 and polycystin 2 (PC1 and PC2, respectively). The cardiovascular complications have been assumed to be a consequence of renal hypertension and activation of renin/angiotensin/aldosterone (RAAS) pathway. However, the expression of PC1 and PC2 in cardiac tissue suggests additional direct effects of these proteins on cardiac function. We previously reported that zebrafish lacking PC2 develop heart failure, and that heterozygous Pkd2+/− mice are hypersensitive to acute β-adrenergic receptor (βAR) stimulation. Here, we investigate the effect of cardiac stress (prolonged continuous βAR stimulus) on Pkd2+/− mice. After βAR stimulation for 7 days, wild-type (WT) mice had increased left ventricular mass and natriuretic peptide (ANP and BNP) mRNA levels. The WT mice also had upregulated levels of PC2 and chromogranin B (CGB, an upstream regulator of BNP). Conversely, Pkd2+/− mice had increased left ventricular mass, but natriuretic peptide and CGB expression levels remained constant. Reversal of the increased cardiac mass was observed in WT mice 3 days after cessation of the βAR stimulation, but not in Pkd2+/− mice. We suggest that cardiac stress leads to upregulation of the PC2-CGB-BNP signaling axis, and this pathway regulates the production of cardio-protective natriuretic peptides. The lack of a PC2-dependent cardio-protective function may contribute to the severity of cardiac dysfunction in Pkd2+/− mice and in ADPKD patients.
- Published
- 2017
13. Hypoxia-Inducible Factor-1α in Vascular Smooth Muscle Regulates Blood Pressure Homeostasis Through a Peroxisome Proliferator–Activated Receptor-γ–Angiotensin II Receptor Type 1 Axis
- Author
-
Weidong Jiang, Frank J. Giordano, Annarita Di Lorenzo, Yan Huang, Anna Cantalupo, and William C. Sessa
- Subjects
Angiotensin receptor ,medicine.medical_specialty ,Angiotensin II receptor type 1 ,Vascular smooth muscle ,Angiotensin-converting enzyme ,Biology ,medicine.disease ,Angiotensin II ,Endocrinology ,Internal medicine ,Pathophysiology of hypertension ,Renin–angiotensin system ,Internal Medicine ,medicine ,biology.protein ,Telmisartan ,medicine.drug - Abstract
Hypertension is a major worldwide health issue for which only a small proportion of cases have a known mechanistic pathogenesis. Of the defined causes, none have been directly linked to heightened vasoconstrictor responsiveness, despite the fact that vasomotor tone in resistance vessels is a fundamental determinant of blood pressure. Here, we reported a previously undescribed role for smooth muscle hypoxia-inducible factor-1α (HIF-1α) in controlling blood pressure homeostasis. The lack of HIF-1α in smooth muscle caused hypertension in vivo and hyperresponsiveness of resistance vessels to angiotensin II stimulation ex vivo. These data correlated with an increased expression of angiotensin II receptor type I in the vasculature. Specifically, we show that HIF-1α, through peroxisome proliferator–activated receptor-γ, reciprocally defined angiotensin II receptor type I levels in the vessel wall. Indeed, pharmacological blockade of angiotensin II receptor type I by telmisartan abolished the hypertensive phenotype in smooth muscle cell-HIF-1α-KO mice. These data revealed a determinant role of a smooth muscle HIF-1α/peroxisome proliferator–activated receptor-γ/angiotensin II receptor type I axis in controlling vasomotor responsiveness and highlighted an important pathway, the alterations of which may be critical in a variety of hypertensive-based clinical settings.
- Published
- 2013
- Full Text
- View/download PDF
14. Endothelial HIF-1α Enables Hypothalamic Glucose Uptake to Drive POMC Neurons
- Author
-
Frank J. Giordano, Marya Shanabrough, Tamas L. Horvath, Yan Huang, Xiao-Bing Gao, Matthias H. Tschöp, Luis Varela, and Shigetomo Suyama
- Subjects
0301 basic medicine ,medicine.medical_specialty ,endocrine system ,Patch-Clamp Techniques ,Pro-Opiomelanocortin ,Endocrinology, Diabetes and Metabolism ,Glucose uptake ,Blotting, Western ,Hypothalamus ,Carbohydrate metabolism ,Hyperphagia ,Real-Time Polymerase Chain Reaction ,03 medical and health sciences ,Mice ,Proopiomelanocortin ,Downregulation and upregulation ,Internal medicine ,Internal Medicine ,medicine ,Premovement neuronal activity ,Animals ,Endothelium ,Neurons ,Gene knockdown ,biology ,Behavior, Animal ,digestive, oral, and skin physiology ,Hypoxia-Inducible Factor 1, alpha Subunit ,Immunohistochemistry ,Mitochondria ,Microscopy, Electron ,030104 developmental biology ,Endocrinology ,Glucose ,Metabolism ,nervous system ,Gene Knockdown Techniques ,biology.protein ,Energy Metabolism ,Food Deprivation ,Ex vivo ,hormones, hormone substitutes, and hormone antagonists - Abstract
Glucose is the primary driver of hypothalamic proopiomelanocortin (POMC) neurons. We show that endothelial hypoxia-inducible factor 1α (HIF-1α) controls glucose uptake in the hypothalamus and that it is upregulated in conditions of undernourishment, during which POMC neuronal activity is decreased. Endothelium-specific knockdown of HIF-1α impairs the ability of POMC neurons to adapt to the changing metabolic environment in vivo, resulting in overeating after food deprivation in mice. The impaired functioning of POMC neurons was reversed ex vivo or by parenchymal glucose administration. These observations indicate an active role for endothelial cells in the central control of metabolism and suggest that central vascular impairments may cause metabolic disorders.
- Published
- 2016
15. Abstract 87: Endothelial Nogo-B Regulates Sphingolipid Biosynthesis to Promote the Onset of Pathological Hypertrophy During Chronic Pressure Overload
- Author
-
Yan Huang, Anna Cantalupo, Annarita Di Lorenzo, Paula S. Azevedo, Frank J. Giordano, Yi Zhang, Mauro Siragusa, and Jacek Bielawski
- Subjects
Pressure overload ,medicine.medical_specialty ,Endocrinology ,Physiology ,business.industry ,Internal medicine ,medicine ,Cardiology and Cardiovascular Medicine ,Sphingolipid biosynthesis ,business ,Pathological ,Muscle hypertrophy - Abstract
Chronic pressure overload leads to an initial compensatory cardiac hypertrophy, and eventually to heart failure. The mechanisms regulating the transition from adaptive to pathological cardiac hypertrophy remain elusive. We recently discovered that endothelial Nogo-B, a membrane protein of the ER, regulates vascular functions by inhibiting the rate-limiting enzyme in de novo sphingolipid biosynthesis, serine palmitoyltransferase (SPT). Here, we show that sphingolipids produced by the vasculature, particularly S1P, protect the heart function during pressure overload, through a paracrine mode of action. SPT activity is upregulated in banded hearts in vivo , as well as in TNF-α-activated endothelium in vitro , and loss of Nogo-mediated brake on SPT increases the production of S1P, which enhances the coronary vasculature compliance to high pressure and endothelial barrier. Hence, mice lacking Nogo-B, systemically or specifically in the endothelium, are resistant to the onset of pathological hypertrophy. Furthermore, pharmacological inhibition of SPT with myriocin restores permeability, inflammation, and heart dysfunction in Nogo-A/B-deficient mice to wild-type levels; whereas SEW2871, an S1P 1 receptor agonist, prevents myocardial inflammation and dysfunction in WT banded mice. Our study identifies a critical role of endothelial sphingolipid biosynthesis and its regulation by Nogo-B in the development of pathological cardiac hypertrophy, and proposes a potential new therapeutic target for the attenuation or reversal of this clinical condition.
- Published
- 2016
- Full Text
- View/download PDF
16. Endothelial Nogo-B regulates sphingolipid biosynthesis to promote pathological cardiac hypertrophy during chronic pressure overload
- Author
-
Yan Huang, Anna Cantalupo, Paula S. Azevedo, Annarita Di Lorenzo, Frank J. Giordano, Mauro Siragusa, Yi Zhang, and Jacek Bielawski
- Subjects
0301 basic medicine ,Pressure overload ,medicine.medical_specialty ,Endothelium ,Serine C-palmitoyltransferase ,Inflammation ,General Medicine ,Biology ,medicine.disease ,Sphingolipid ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Downregulation and upregulation ,chemistry ,Fibrosis ,Internal medicine ,Myriocin ,mental disorders ,medicine ,medicine.symptom ,Research Article - Abstract
We recently discovered that endothelial Nogo-B, a membrane protein of the ER, regulates vascular function by inhibiting the rate-limiting enzyme, serine palmitoyltransferase (SPT), in de novo sphingolipid biosynthesis. Here, we show that endothelium-derived sphingolipids, particularly sphingosine-1-phosphate (S1P), protect the heart from inflammation, fibrosis, and dysfunction following pressure overload and that Nogo-B regulates this paracrine process. SPT activity is upregulated in banded hearts in vivo as well as in TNF-α–activated endothelium in vitro, and loss of Nogo removes the brake on SPT, increasing local S1P production. Hence, mice lacking Nogo-B, systemically or specifically in the endothelium, are resistant to the onset of pathological cardiac hypertrophy. Furthermore, pharmacological inhibition of SPT with myriocin restores permeability, inflammation, and heart dysfunction in Nogo-A/B–deficient mice to WT levels, whereas SEW2871, an S1P1 receptor agonist, prevents myocardial permeability, inflammation, and dysfunction in WT banded mice. Our study identifies a critical role of endothelial sphingolipid biosynthesis and its regulation by Nogo-B in the development of pathological cardiac hypertrophy and proposes a potential therapeutic target for the attenuation or reversal of this clinical condition.
- Published
- 2016
- Full Text
- View/download PDF
17. Cellularity and structure of fresh human coronary thrombectomy specimens; presence of cells with markers of progenitor cells
- Author
-
Li Lei, Michael W. Cleman, John F. Setaro, Steven Pfau, Zhengrong Hao, Jude Clancy, Henry S. Cabin, Michael S. Remetz, Frank J. Giordano, Joseph Brennan, Yan Huang, Ion S. Jovin, Christopher J. Howes, and Jeptha P. Curtis
- Subjects
Pathology ,medicine.medical_specialty ,Myocardial Infarction ,CD34 ,Lewis X Antigen ,Antigens, CD34 ,Coronary Disease ,CD15 ,acute coronary syndrome ,Desmin ,von Willebrand Factor ,medicine ,Humans ,Platelet ,Progenitor cell ,coronary ,Cells, Cultured ,endothelial progenitor cells ,business.industry ,Coronary Thrombosis ,Stem Cells ,Endothelial Cells ,Original Articles ,Cell Biology ,Plaque, Atherosclerotic ,Endothelial stem cell ,Haematopoiesis ,thrombus ,thrombectomy ,platelets ,Molecular Medicine ,Stem cell ,business ,Biomarkers - Abstract
Acute coronary syndromes and acute myocardial infarctions are often related to plaque rupture and the formation of thrombi at the site of the rupture. We examined fresh coronary thrombectomy specimens from patients with acute coronary syndromes and assessed their structure and cellularity. The thrombectomy specimens consisted of platelets, erythrocytes and inflammatory cells. Several specimens contained multiple cholesterol crystals. Culture of thrombectomy specimens yielded cells growing in various patterns depending on the culture medium used. Culture in serum-free stem cell enrichment medium yielded cells with features of endothelial progenitor cells which survived in culture for a year. Immunohistochemical analysis of the thrombi revealed cells positive for CD34, cells positive for CD15 and cells positive for desmin in situ, whereas cultured cell from thrombi was desmin positive but pancytokeratin negative. Cells cultured in endothelial cell medium were von Willebrand factor positive. The content of coronary thrombectomy specimens is heterogeneous and consists of blood cells but also possibly cells from the vascular wall and cholesterol crystals. The culture of cells contained in the specimens yielded multiplying cells, some of which demonstrated features of haematopoietic progenitor cells and which differentiated into various cell-types.
- Published
- 2012
- Full Text
- View/download PDF
18. Normal glucose uptake in the brain and heart requires an endothelial cell-specific HIF-1α–dependent function
- Author
-
Yan Huang, Li Lei, Dinggang Liu, Frank J. Giordano, Ion S. Jovin, Randall S. Johnson, Annarita Di Lorenzo, and Raymond R. Russell
- Subjects
Blood Glucose ,Male ,medicine.medical_specialty ,Endothelium ,Glucose uptake ,Carbohydrate metabolism ,Mice ,Internal medicine ,medicine ,Animals ,Humans ,Mice, Knockout ,Glucose Transporter Type 1 ,Multidisciplinary ,biology ,Myocardium ,Glucose transporter ,Brain ,Endothelial Cells ,Biological Sciences ,Hypoxia-Inducible Factor 1, alpha Subunit ,Endothelial stem cell ,medicine.anatomical_structure ,Endocrinology ,Knockout mouse ,biology.protein ,GLUT1 - Abstract
Although intimately positioned between metabolic substrates in the bloodstream and the tissue parenchymal cells that require these substrates, a major role of the vascular endothelium in the regulation of tissue metabolism has not been widely appreciated. We hypothesized that via control of transendothelial glucose transport and contributing paracrine mechanisms the endothelium plays a major role in regulating organ and tissue glucose metabolism. We further hypothesized that the hypoxia-inducible factor -1α (HIF-1α) plays an important role in coordinating these endothelial functions. To test these hypotheses, we generated mice with endothelial cell-specific deletion of HIF-1α. Loss of HIF in the endothelium resulted in significantly increased fasting blood glucose levels, a blunted insulin response with delayed glucose clearance from the blood after i.v. loading, and significantly decreased glucose uptake into the brain and heart. Endothelial HIF-1α knockout mice also exhibited a reduced cerebrospinal fluid/blood glucose ratio, a finding consistent with reduced transendothelial glucose transport and a diagnostic criterion for the Glut1 deficiency genetic syndrome. Endothelial cells from these mice demonstrated decreased Glut1 levels and reduced glucose uptake that was reversed by forced expression of Glut1. These data strongly support an important role of the vascular endothelium in determining whole-organ glucose metabolism and indicate that HIF-1α is a critical mediator of this function.
- Published
- 2012
- Full Text
- View/download PDF
19. A Designed Zinc-finger Transcriptional Repressor of Phospholamban Improves Function of the Failing Heart
- Author
-
H Steve, Zhang, Dingang, Liu, Yan, Huang, Stefan, Schmidt, Reed, Hickey, Dmitry, Guschin, Haili, Su, Ion S, Jovin, Mike, Kunis, Sarah, Hinkley, Yuxin, Liang, Linda, Hinh, S Kaye, Spratt, Casey C, Case, Edward J, Rebar, Barbara E, Ehrlich, Barbara, Ehrlich, Philip D, Gregory, and Frank J, Giordano
- Subjects
Blotting, Western ,Druggability ,Repressor ,030204 cardiovascular system & hematology ,Biology ,Adenoviridae ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Calcium-binding protein ,Drug Discovery ,Genetics ,Animals ,Humans ,Myocytes, Cardiac ,Molecular Biology ,Gene ,Psychological repression ,Transcription factor ,030304 developmental biology ,Pharmacology ,Zinc finger ,Heart Failure ,0303 health sciences ,Reverse Transcriptase Polymerase Chain Reaction ,Calcium-Binding Proteins ,Zinc Fingers ,3. Good health ,Cell biology ,Phospholamban ,Rats ,Kinetics ,030220 oncology & carcinogenesis ,Molecular Medicine ,Original Article ,Corrigendum ,Transcription Factors - Abstract
Selective inhibition of disease-related proteins underpins the majority of successful drug–target interactions. However, development of effective antagonists is often hampered by targets that are not druggable using conventional approaches. Here, we apply engineered zinc-finger protein transcription factors (ZFP TFs) to the endogenous phospholamban (PLN) gene, which encodes a well validated but recalcitrant drug target in heart failure. We show that potent repression of PLN expression can be achieved with specificity that approaches single-gene regulation. Moreover, ZFP-driven repression of PLN increases calcium reuptake kinetics and improves contractile function of cardiac muscle both in vitro and in an animal model of heart failure. These results support the development of the PLN repressor as therapy for heart failure, and provide evidence that delivery of engineered ZFP TFs to native organs can drive therapeutically relevant levels of gene repression in vivo. Given the adaptability of designed ZFPs for binding diverse DNA sequences and the ubiquity of potential targets (promoter proximal DNA), our findings suggest that engineered ZFP repressors represent a powerful tool for the therapeutic inhibition of disease-related genes, therefore, offering the potential for therapeutic intervention in heart failure and other poorly treated human diseases.
- Published
- 2012
20. miR-1 mediated suppression of Sorcin regulates myocardial contractility through modulation of Ca2+ signaling
- Author
-
Richard W. Kim, Stephen E. Maher, Yan Huang, George Tellides, Frank J. Giordano, Rahmat Ali, and Arnar Geirsson
- Subjects
Male ,Ribonuclease III ,medicine.medical_specialty ,Mice, 129 Strain ,Cardiac Volume ,education ,Negative control ,Cell Line ,DEAD-box RNA Helicases ,Contractility ,Mice ,Internal medicine ,microRNA ,medicine ,Animals ,Humans ,Calcium Signaling ,RNA, Small Interfering ,Molecular Biology ,health care economics and organizations ,Calcium signaling ,Mice, Knockout ,Base Sequence ,biology ,Myocardium ,Calcium-Binding Proteins ,Heart ,Myocardial Contraction ,Up-Regulation ,Mice, Inbred C57BL ,MicroRNAs ,Endocrinology ,biology.protein ,Cancer research ,RNA Interference ,Cardiomyopathies ,Cardiology and Cardiovascular Medicine ,Ca2 signaling ,Dicer - Abstract
MicroRNAs are negative gene regulators and play important roles in cardiac development and disease. As evident by cardiomyopathy following cardiac-specific Dicer knockdown they also are required for maintaining normal cardiac contractile function but the specific role of miR-1 in the process is poorly understood. To characterize the role of miR-1 in particular and to identify its specific targets we created a tamoxifen-inducible, cardiac-specific Dicer knockdown mouse and demonstrated that Dicer downregulation results in a dramatic and rapid decline in cardiac function concurrent with significantly reduced levels of miR-1. The importance of miR-1 was established by miR-1 antagomir treatment of wild-type mice, which replicated the cardiac-specific Dicer knockdown phenotype. Down-regulation of miR-1 was associated with up-regulation of its predicted target Sorcin, an established modulator of calcium signaling and excitation-contraction coupling, subsequently verified as a miR-1 target with luciferase constructs. siRNA-mediated knockdown of Sorcin effectively rescued the cardiac phenotypes after Dicer or miR-1 knockdown affirming Sorcin as a critical mediator of the acute cardiomyopathy observed. The regulatory relationship between miR-1 and Sorcin was further confirmed in cultured mouse cardiomyocytes where modulation of miR-1 was associated with discordant Sorcin levels and dysregulation of calcium signaling. Pathological relevance of our findings included decreased miR-1 and increased Sorcin expression in end-stage cardiomyopathy. These findings demonstrate the importance of miR-1 in cardiac function and in the pathogenesis of heart failure via Sorcin-dependent calcium homeostasis.
- Published
- 2012
- Full Text
- View/download PDF
21. Inhibition of MicroRNA-29 Enhances Elastin Levels in Cells Haploinsufficient for Elastin and in Bioengineered Vessels—Brief Report
- Author
-
Pei Zhang, Jay D. Humphrey, Robert P. Mecham, William C. Sessa, Frank J. Giordano, Angela Huang, Laura E. Niklason, Jacopo Ferruzzi, George Tellides, and Barry Starcher
- Subjects
Extracellular matrix ,Downregulation and upregulation ,Tissue engineering ,Blood vessel prosthesis ,RNA interference ,microRNA ,Cancer research ,biology.protein ,Transfection ,Anatomy ,Biology ,Cardiology and Cardiovascular Medicine ,Elastin - Abstract
Objective— The goal of this study was to determine whether antagonizing microRNA (miR)-29 enhances elastin (ELN) levels in cells and tissues lacking ELN. Methods and Results— miR-29 mimics reduced ELN levels in fibroblasts and smooth muscle cells, whereas miR-29 inhibition increased ELN levels. Antagonism of miR-29 also increased ELN levels in cells from patients haploinsufficient for ELN and in bioengineered human vessels. Conclusion— miR-29 antagonism may promote increased ELN levels during conditions of ELN deficiencies.
- Published
- 2012
- Full Text
- View/download PDF
22. Inhibition of intimal hyperplasia after stenting by over-expression of p15: A member of the INK4 family of cyclin-dependent kinase inhibitors
- Author
-
John D. Sparkes, Jack Gauldie, Beiping Qiang, Michelle Ladouceur-Wodzak, Frank J. Giordano, Ronen Jaffe, Nafiseh Nili, Ashley R. Fraser, Jagdish Butany, Amit Segev, Azriel B. Osherov, and Bradley H. Strauss
- Subjects
Intimal hyperplasia ,Population ,Cell Growth Processes ,Retinoblastoma Protein ,Muscle, Smooth, Vascular ,Adenoviridae ,Transduction, Genetic ,Cyclin-dependent kinase ,medicine ,Animals ,Humans ,Transgenes ,Phosphorylation ,education ,Molecular Biology ,Cells, Cultured ,Cyclin-Dependent Kinase Inhibitor p15 ,education.field_of_study ,Hyperplasia ,biology ,DNA synthesis ,Cell growth ,G1 Phase ,Retinoblastoma protein ,Genetic Therapy ,Cell cycle ,beta-Galactosidase ,medicine.disease ,Rats ,Carotid Arteries ,biology.protein ,Cancer research ,Stents ,Rabbits ,Tunica Intima ,Cardiology and Cardiovascular Medicine - Abstract
We evaluated the role of p15(Ink4), a member of the INK4 family of CDK inhibitors on vascular smooth muscle cells (VSMCs) proliferation, cell cycle progression and intimal hyperplasia after stenting. Aortic VSMCs transduced with either adenovirus encoding for p15(Ink4) or β-galactosidase were assessed for DNA synthesis, cell cycle progression, and pRb phosphorylation. Rabbit carotid arteries were stented and treated with peri-adventitial delivery of saline or adenovirus encoding for p15(Ink4) or β-galactosidase. p15(Ink4) transgene and protein expression were evaluated at 24 h and 72 h, respectively. In-stent cell proliferation was evaluated by BrdU at day 7. Histomorphometric analysis of in-stent intimal hyperplasia was performed at 10 weeks. Human p15(Ink4) DNA was detected in transduced VSMCs at 24h. p15(Ink4) over-expression reduced VSMCs DNA synthesis by 60%. Cell cycle progression was inhibited, with a 30% increase in G1 population accompanied by inhibition of pRb phosphorylation. Human p15(Ink4) transgene was identified in transduced stented arteries but not in control arteries. p15(Ink4) immunostaining was increased and cell proliferation significantly reduced by 50% in p15(Ink4) transduced arteries. Intimal cross-sectional area (CSA) of p15(Ink4)-treated group was significantly lower than the β-gal treated and non-transduced groups (p=0.008). There were no differences in the intimal or medial inflammatory response between groups. p15(Ink4) over-expression blocks cell cycle progression leading to inhibition of VSMCs proliferation. Peri-adventitial delivery of p15(Ink4) significantly inhibits in-stent intimal hyperplasia.
- Published
- 2011
- Full Text
- View/download PDF
23. Hypoxia-Inducible Factor-Dependent Degeneration, Failure, and Malignant Transformation of the Heart in the Absence of the von Hippel-Lindau Protein
- Author
-
Dinggang Liu, Carolyn Marks, Li Lei, Frank J. Giordano, Ion S. Jovin, Randall S. Johnson, Marc Pypaert, Reed Hickey, Yan Huang, and Steven D. Mason
- Subjects
medicine.medical_specialty ,Neovascularization, Physiologic ,Biology ,Malignant transformation ,Heart Neoplasms ,Mice ,Fibrosis ,Internal medicine ,medicine ,Animals ,Myocyte ,Phosphorylation ,Hypoxia ,Molecular Biology ,Heart Failure ,Mice, Knockout ,Myocardium ,Gene Transfer Techniques ,Cardiac muscle ,Articles ,Cell Biology ,Proto-Oncogene Proteins c-met ,Hypoxia (medical) ,Hypoxia-Inducible Factor 1, alpha Subunit ,Lipid Metabolism ,medicine.disease ,Lipids ,Capillaries ,Ubiquitin ligase ,ErbB Receptors ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Endocrinology ,Hypoxia-inducible factors ,Von Hippel-Lindau Tumor Suppressor Protein ,Heart failure ,ras Proteins ,biology.protein ,medicine.symptom ,Gene Deletion - Abstract
Hypoxia-inducible transcription factor 1 (HIF-1) and HIF-2alpha regulate the expression of an expansive array of genes associated with cellular responses to hypoxia. Although HIF-regulated genes mediate crucial beneficial short-term biological adaptations, we hypothesized that chronic activation of the HIF pathway in cardiac muscle, as occurs in advanced ischemic heart disease, is detrimental. We generated mice with cardiac myocyte-specific deletion of the von Hippel-Lindau protein (VHL), an essential component of an E3 ubiquitin ligase responsible for suppressing HIF levels during normoxia. These mice were born at expected frequency and thrived until after 3 months postbirth, when they developed severe progressive heart failure and premature death. VHL-null hearts developed lipid accumulation, myofibril rarefaction, altered nuclear morphology, myocyte loss, and fibrosis, features seen for various forms of human heart failure. Further, nearly 50% of VHL(-/-) hearts developed malignant cardiac tumors with features of rhabdomyosarcoma and the capacity to metastasize. As compelling evidence for the mechanistic contribution of HIF-1alpha, the concomitant deletion of VHL and HIF-1alpha in the heart prevented this phenotype and restored normal longevity. These findings strongly suggest that chronic activation of the HIF pathway in ischemic hearts is maladaptive and contributes to cardiac degeneration and progression to heart failure.
- Published
- 2008
- Full Text
- View/download PDF
24. Urocortin2 inhibits tumor growth via effects on vascularization and cell proliferation
- Author
-
Jake Cleman, Ion S. Jovin, Frank J. Giordano, Zhengrong Hao, Wylie Vale, Tracy L. Bale, and Yan Huang
- Subjects
Male ,medicine.medical_specialty ,Cell ,Biology ,Receptors, Corticotropin-Releasing Hormone ,Neovascularization ,Carcinoma, Lewis Lung ,Mice ,In vivo ,Internal medicine ,medicine ,Animals ,Receptor ,Urocortins ,Cell Proliferation ,Urocortin ,Multidisciplinary ,Neovascularization, Pathologic ,Cell growth ,Lewis lung carcinoma ,Biological Sciences ,In vitro ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Endocrinology ,Cancer research ,medicine.symptom - Abstract
The corticotropin-releasing factor (CRF) receptor CRFR2 is expressed widely in peripheral tissues and in the vasculature, although its functional roles in those tissues have only recently begun to be elucidated. Previously we found that genetic deletion of CRFR2 resulted in profound postnatal hypervascularization in mice, characterized by both an increase in total vessel number and a dramatic increase in vessel diameter. These data strongly suggested that ligands for CRFR2 act to limit tissue vascularity, perhaps as a counterbalance to factors that promote neovascularization. Urocortin 2 (Ucn2) is a specific ligand for the CRFR2. We hypothesized that activation of CRFR2 by Ucn2 might thus suppress tumor vascularization and consequently limit tumor growth. Here, we show that viral-mediated expression of Ucn2 strikingly inhibits the growth and vascularization of Lewis Lung Carcinoma Cell (LLCC) tumors in vivo . Further, we found that this effect on tumor growth inhibition was independent of whether exposure to Ucn2 occurred before or after establishment of measurable tumors. In vitro , Ucn2 directly inhibited the proliferation of LLCC, suggesting that the tumor-suppressing effects of CRFR2 activation involve a dual mechanism of both a direct inhibition of tumor cell cycling and the suppression of tumor vascularization. These results establish that Ucn2 inhibits tumor growth, suggesting a potential therapeutic role for CRFR2 ligands in clinical malignancies.
- Published
- 2008
- Full Text
- View/download PDF
25. Therapeutic Gene Regulation
- Author
-
Frank J. Giordano
- Subjects
Regulation of gene expression ,TBX1 ,Genetics ,Exon ,Physiology (medical) ,Alternative splicing ,Coding region ,Human genome ,Biology ,Cardiology and Cardiovascular Medicine ,Genome ,Gene - Abstract
In this issue of Circulation , Rajagopolan et al1 report the first clinical results of a gene therapy approach predicated on transcriptional activation of a patient’s own genes. This represents a second-generation gene therapy methodology for cardiovascular disease and is important for a number of reasons. We have witnessed in the past decade the primary sequencing of the human genome.2,3 One of the initial reactions to this milestone accomplishment was surprise at the relatively small number of definitive genes that are encoded by human DNA. Although the exact number is still uncertain, estimates as low as 23 299 have been made. In comparison, the genome of the worm Caenorhabditis elegans encodes approximately 19 000 genes, and the genome of the common fruit fly encodes approximately 18 000 genes, raising the question of how such significant differences in biological complexity and diversity are engendered by so few genes. A complete set of answers to this question is not currently in our grasp, but some crucial aspects are understood and are relevant to the clinical trial discussed here. Article p 1234 One manner in which the biological effect of a finite number of genes is amplified is by alternative splicing. This is the process whereby a single gene encodes a number of alternative proteins by simply including or excluding specific exons within the coding sequence of that gene during mRNA transcription and maturation. The number of alternative splice variants that exist as transcriptional products of the human genome is not known, but this process alone likely increases the number of unique proteins that can be encoded by the genome 2- to 3-fold, possibly more. Interestingly, there are known human cardiovascular diseases caused by inappropriate splicing, including a cardiomyopathy and sudden death syndrome in children caused by deficient …
- Published
- 2007
- Full Text
- View/download PDF
26. Endothelial-Specific Expression of Mitochondrial Thioredoxin Improves Endothelial Cell Function and Reduces Atherosclerotic Lesions
- Author
-
Yun He, Frank J. Giordano, Yan Huang, Wei Zhang, Shengchuan Dai, Rong Zhang, Yan Luo, Pascal Bernatchez, Gerald S. Shadel, Wang Min, William C. Sessa, and Haifeng Zhang
- Subjects
chemistry.chemical_classification ,Reactive oxygen species ,Endothelium ,Vasodilation ,Mitochondrion ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,Nitric oxide ,Cell biology ,Endothelial stem cell ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Immunology ,medicine ,Thioredoxin ,Oxidative stress - Abstract
The function of the mitochondrial antioxidant system thioredoxin (Trx2) in vasculature is not understood. By using endothelial cell (EC)-specific transgenesis of the mitochondrial form of the thioredoxin gene in mice (Trx2 TG), we show the critical roles of Trx2 in regulating endothelium functions. Trx2 TG mice have increased total antioxidants, reduced oxidative stress, and increased nitric oxide (NO) levels in serum compared with their control littermates. Consistently, aortas from Trx2 TG mice show reduced vasoconstriction and enhanced vasodilation. By using ECs isolated from Trx2 TG mice, we further show that Trx2 increases the capacities of ECs in scavenging reactive oxygen species generated from mitochondria, resulting in increases in NO bioavailability in ECs. More importantly, Trx2 improves EC function and reduces atherosclerotic lesions in the apolipoprotein E-deficient mouse model. Our data provide the first evidence that Trx2 plays a critical role in preserving vascular EC function and prevention of atherosclerosis development, in part by reducing oxidative stress and increasing NO bioavailability.
- Published
- 2007
- Full Text
- View/download PDF
27. The Spatial Distribution of Inositol 1,4,5-Trisphosphate Receptor Isoforms Shapes Ca2+ Waves
- Author
-
Oscar Bruna-Romero, Emma A. Kruglov, Frank J. Giordano, Dawidson Assis Gomes, Michele Angela Rodrigues, Erick Hernandez, M. Fatima Leite, Michael H. Nathanson, Jonathan A. Dranoff, and Mateus T. Guerra
- Subjects
Gene isoform ,Vasopressins ,Molecular Sequence Data ,Receptors, Cytoplasmic and Nuclear ,Biology ,Biochemistry ,Article ,chemistry.chemical_compound ,Animals ,Inositol 1,4,5-Trisphosphate Receptors ,Protein Isoforms ,Inositol ,Calcium Signaling ,Receptor ,Molecular Biology ,Cells, Cultured ,Calcium signaling ,Membrane Glycoproteins ,Base Sequence ,Voltage-dependent calcium channel ,Cell Biology ,Inositol trisphosphate receptor ,Molecular biology ,Rats ,Cell biology ,chemistry ,Cell culture ,Second messenger system ,Hepatocytes ,Calcium Channels - Abstract
Cytosolic Ca(2+) is a versatile second messenger that can regulate multiple cellular processes simultaneously. This is accomplished in part through Ca(2+) waves and other spatial patterns of Ca(2+) signals. To investigate the mechanism responsible for the formation of Ca(2+) waves, we examined the role of inositol 1,4,5-trisphosphate receptor (InsP3R) isoforms in Ca(2+) wave formation. Ca(2+) signals were examined in hepatocytes, which express the type I and II InsP3R in a polarized fashion, and in AR4-2J cells, a nonpolarized cell line that expresses type I and II InsP3R in a ratio similar to what is found in hepatocytes but homogeneously throughout the cell. Expression of type I or II InsP3R was selectively suppressed by isoform-specific DNA antisense in an adenoviral delivery system, which was delivered to AR4-2J cells in culture and to hepatocytes in vivo. Loss of either isoform inhibited Ca(2+) signals to a similar extent in AR4-2J cells. In contrast, loss of the basolateral type I InsP3R decreased the sensitivity of hepatocytes to vasopressin but had little effect on the initiation or spread of Ca(2+) waves across hepatocytes. Loss of the apical type II isoform caused an even greater decrease in the sensitivity of hepatocytes to vasopressin and resulted in Ca(2+) waves that were much slower and delayed in onset. These findings provide evidence that the apical concentration of type II InsP3Rs is essential for the formation of Ca(2+) waves in hepatocytes. The subcellular distribution of InsP3R isoforms may critically determine the repertoire of spatial patterns of Ca(2+) signals.
- Published
- 2007
- Full Text
- View/download PDF
28. Interferon-γ-Mediated Allograft Rejection Exacerbates Cardiovascular Disease of Hyperlipidemic Murine Transplant Recipients
- Author
-
Rahmat Ali, Jing Zhou, Mehran M. Sadeghi, Frank J. Giordano, Yan Huang, Lingfeng Qin, Jordan S. Pober, Tai Yi, George Tellides, Zuzana Tobiasova, Jiasheng Zhang, Guangxin Li, James Yun, Yang Jiao, and Qingle Li
- Subjects
Graft Rejection ,Male ,medicine.medical_specialty ,Apolipoprotein B ,Physiology ,medicine.medical_treatment ,Aortic Diseases ,Hyperlipidemias ,Disease ,Lymphocyte Activation ,Organ transplantation ,Article ,Ventricular Function, Left ,Interferon-gamma ,Ventricular Dysfunction, Left ,Apolipoproteins E ,Antigen ,medicine.artery ,medicine ,Animals ,Mice, Knockout ,Aorta ,Mice, Inbred BALB C ,biology ,business.industry ,Hemodynamics ,Histocompatibility Antigens Class II ,Immunosuppression ,Th1 Cells ,Allografts ,Atherosclerosis ,Transplantation ,Mice, Inbred C57BL ,Disease Models, Animal ,Allograft rejection ,Cardiovascular Diseases ,Immunology ,biology.protein ,Heart Transplantation ,Female ,Inflammation Mediators ,Cardiology and Cardiovascular Medicine ,business ,Cardiomyopathies ,Signal Transduction - Abstract
Rationale: Transplantation, the most effective therapy for end-stage organ failure, is markedly limited by early-onset cardiovascular disease (CVD) and premature death of the host. The mechanistic basis of this increased CVD is not fully explained by known risk factors. Objective: To investigate the role of alloimmune responses in promoting CVD of organ transplant recipients. Methods and Results: We established an animal model of graft-exacerbated host CVD by combining murine models of atherosclerosis (apolipoprotein E–deficient recipients on standard diet) and of intra-abdominal graft rejection (heterotopic cardiac transplantation without immunosuppression). CVD was absent in normolipidemic hosts receiving allogeneic grafts and varied in severity among hyperlipidemic grafted hosts according to recipient–donor genetic disparities, most strikingly across an isolated major histocompatibility complex class II antigen barrier. Host disease manifested as increased atherosclerosis of the aorta that also involved the native coronary arteries and new findings of decreased cardiac contractility, ventricular dilatation, and diminished aortic compliance. Exacerbated CVD was accompanied by greater levels of circulating cytokines, especially interferon-γ and other Th1-type cytokines, and showed both systemic and intralesional activation of leukocytes, particularly T-helper cells. Serological neutralization of interferon-γ after allotransplantation prevented graft-related atherosclerosis, cardiomyopathy, and aortic stiffening in the host. Conclusions: Our study reveals that sustained activation of the immune system because of chronic allorecognition exacerbates the atherogenic diathesis of hyperlipidemia and results in de novo cardiovascular dysfunction in organ transplant recipients.
- Published
- 2015
29. Smad2 and Smad3 Play Different Roles in Rat Hepatic Stellate Cell Function and α-Smooth Muscle Actin Organization
- Author
-
Marianna D. A. Gaça, Rebecca G. Wells, Michael Reiss, Frank J. Giordano, E. Scott Swenson, and Masayuki Uemura
- Subjects
Male ,Genetic Vectors ,Biology ,Chronic liver disease ,Adenoviridae ,Rats, Sprague-Dawley ,Extracellular matrix ,Pathogenesis ,Transduction, Genetic ,medicine ,Animals ,Molecular Biology ,Cells, Cultured ,Actin ,Cell Proliferation ,Extracellular Matrix Proteins ,Cell growth ,Chemotaxis ,Stem Cells ,Articles ,Cell Biology ,medicine.disease ,Actins ,Rats ,Up-Regulation ,Cell biology ,Liver ,Hepatic stellate cell ,Stem cell ,Myofibroblast - Abstract
Hepatic stellate cells (HSC) play a central role in the pathogenesis of liver fibrosis, transdifferentiating in chronic liver disease from "quiescent" HSC to fibrogenic myofibroblasts. Transforming growth factor-beta (TGF-beta), acting both directly and indirectly, is a critical mediator of this process. To characterize the function of the TGF-beta signaling intermediates Smad2 and Smad3 in HSC, we infected primary rat HSC in culture with adenoviruses expressing wild-type and dominant negative Smads 2 and 3. Smad3-overexpressing cells exhibited increased deposition of fibronectin and type 1 collagen, increased chemotaxis, and decreased proliferation compared with uninfected cells and those infected with Smad2 or either dominant negative, demonstrating different biological functions for the two Smads. Additionally, coinfection experiments suggested that Smad2 and Smad3 signal via independent pathways. Smad3-overexpressing cells as well as TGF-beta-treated cells demonstrated more focal adhesions and increased alpha-smooth muscle actin (alpha-SMA) organization in stress fibers, although all cells reached the same level of alpha-SMA expression, indicating that Smad3 also regulates cytoskeletal organization in HSC. We suggest that TGF-beta, signaling via Smad3, plays an important role in the morphological and functional maturation of hepatic myofibroblasts.
- Published
- 2005
- Full Text
- View/download PDF
30. Stromal Cell–Derived Factor-1α Plays a Critical Role in Stem Cell Recruitment to the Heart After Myocardial Infarction but Is Not Sufficient to Induce Homing in the Absence of Injury
- Author
-
Dingang Liu, Reed Hickey, Yan Huang, Frank J. Giordano, J. Dawn Abbott, and Diane S. Krause
- Subjects
Vascular Endothelial Growth Factor A ,Benzylamines ,Receptors, CXCR4 ,Stromal cell ,Recombinant Fusion Proteins ,Myocardial Infarction ,Vascular Cell Adhesion Molecule-1 ,Bone Marrow Cells ,Mice, SCID ,Gene delivery ,Cyclams ,CXCR4 ,Andrology ,Mice ,Cell Movement ,Heterocyclic Compounds ,Mice, Inbred NOD ,Transduction, Genetic ,Physiology (medical) ,Animals ,Medicine ,Cell Lineage ,Myocardial infarction ,Receptor ,Bone Marrow Transplantation ,Cell adhesion molecule ,business.industry ,Gene Expression Profiling ,Myocardium ,Stem Cells ,Genetic Therapy ,Intercellular Adhesion Molecule-1 ,medicine.disease ,Chemokine CXCL12 ,Real-time polymerase chain reaction ,Gene Expression Regulation ,Matrix Metalloproteinase 9 ,Immunology ,Female ,Stem cell ,Cardiology and Cardiovascular Medicine ,business ,Chemokines, CXC ,Stem Cell Transplantation - Abstract
Background— After myocardial infarction (MI), bone marrow–derived cells (BMDCs) are found within the myocardium. The mechanisms determining BMDC recruitment to the heart remain unclear. We investigated the role of stromal cell–derived factor-1α (SDF-1) in this process. Methods and Results— MI produced in mice by coronary ligation induced SDF-1 mRNA and protein expression in the infarct and border zone and decreased serum SDF-1 levels. By quantitative polymerase chain reaction, 48 hours after intravenous infusion of donor-lineage BMDCs, there were 80.5±15.6% more BDMCs in infarcted hearts compared with sham-operated controls ( P P P Conclusion— SDF-1/CXCR4 interactions play a crucial role in the recruitment of BMDCs to the heart after MI and can further increase homing in the presence, but not in the absence, of injury.
- Published
- 2004
- Full Text
- View/download PDF
31. Loss of HIF-1α in endothelial cells disrupts a hypoxia-driven VEGF autocrine loop necessary for tumorigenesis
- Author
-
Frank J. Giordano, Lianchun Wang, Hans-Peter Gerber, Randall S. Johnson, Napoleone Ferrara, Jeffrey D. Esko, Nan Tang, and Yan Huang
- Subjects
Vascular Endothelial Growth Factor A ,Cancer Research ,Angiogenesis ,Biology ,Neovascularization ,Extracellular matrix ,Mice ,Neoplasms ,Thrombospondin 1 ,medicine ,Animals ,Autocrine signalling ,Transcription factor ,Cells, Cultured ,Cell Proliferation ,Wound Healing ,Neovascularization, Pathologic ,Chemotaxis ,Endothelial Cells ,Cell Biology ,Hypoxia-Inducible Factor 1, alpha Subunit ,Vascular Endothelial Growth Factor Receptor-2 ,Cell Hypoxia ,Cell biology ,Gene Expression Regulation ,Oncology ,Immunology ,medicine.symptom ,Wound healing ,Transcription Factors - Abstract
We deleted the hypoxia-responsive transcription factor HIF-1alpha in endothelial cells (EC) to determine its role during neovascularization. We found that loss of HIF-1alpha inhibits a number of important parameters of EC behavior during angiogenesis: these include proliferation, chemotaxis, extracellular matrix penetration, and wound healing. Most strikingly, loss of HIF-1alpha in EC results in a profound inhibition of blood vessel growth in solid tumors. These phenomena are all linked to a decreased level of VEGF expression and loss of autocrine response of VEGFR-2 in HIF-1alpha null EC. We thus show that a HIF-1alpha-driven, VEGF-mediated autocrine loop in EC is an essential component of solid tumor angiogenesis.
- Published
- 2004
- Full Text
- View/download PDF
32. AMP-activated protein kinase mediates ischemic glucose uptake and prevents postischemic cardiac dysfunction, apoptosis, and injury
- Author
-
Christoph Peter Gerhard Zechner, Lawrence H. Young, Frank J. Giordano, Raymond R. Russell, Ji Li, James Mu, Marc Pypaert, David L. Coven, Monica Palmeri, and Morris J. Birnbaum
- Subjects
medicine.medical_specialty ,Glucose uptake ,Myocardial Ischemia ,Ischemia ,Apoptosis ,Mice, Transgenic ,Myocardial Reperfusion Injury ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,Carbohydrate metabolism ,Mice ,AMP-activated protein kinase ,Multienzyme Complexes ,Internal medicine ,medicine ,Animals ,Glycolysis ,Protein kinase A ,biology ,Glucose transporter ,AMPK ,General Medicine ,medicine.disease ,Mice, Inbred C57BL ,Glucose ,Endocrinology ,Commentary ,biology.protein - Abstract
AMP-activated protein kinase (AMPK) is an important regulator of diverse cellular pathways in the setting of energetic stress. Whether AMPK plays a critical role in the metabolic and functional responses to myocardial ischemia and reperfusion remains uncertain. We examined the cardiac consequences of long-term inhibition of AMPK activity in transgenic mice expressing a kinase dead (KD) form of the enzyme. The KD mice had normal fractional shortening and no heart failure, cardiac hypertrophy, or fibrosis, although the in vivo left ventricular (LV) dP/dt was lower than that in WT hearts. During low-flow ischemia and postischemic reperfusion in vitro, KD hearts failed to augment glucose uptake and glycolysis, although glucose transporter content and insulin-stimulated glucose uptake were normal. KD hearts also failed to increase fatty acid oxidation during reperfusion. Furthermore, KD hearts demonstrated significantly impaired recovery of LV contractile function during postischemic reperfusion that was associated with a lower ATP content and increased injury compared with WT hearts. Caspase-3 activity and TUNEL-staining were increased in KD hearts after ischemia and reperfusion. Thus, AMPK is responsible for activation of glucose uptake and glycolysis during low-flow ischemia and plays an important protective role in limiting damage and apoptotic activity associated with ischemia and reperfusion in the heart.
- Published
- 2004
- Full Text
- View/download PDF
33. Decorin Inhibition of PDGF-Stimulated Vascular Smooth Muscle Cell Function
- Author
-
Asim N. Cheema, Mohammad R. Eskandarian, Gerard Pasterkamp, Nafiseh Nili, Frank J. Giordano, Alan Barolet, Jagdish Butany, Mirjam B. Smeets, Saeid Babaei, Reed Hickey, and Bradley H. Strauss
- Subjects
Platelet-derived growth factor ,Vascular smooth muscle ,Intimal hyperplasia ,biology ,Decorin ,Growth factor ,medicine.medical_treatment ,medicine.disease ,Pathology and Forensic Medicine ,carbohydrates (lipids) ,chemistry.chemical_compound ,Proteoglycan ,chemistry ,Immunology ,cardiovascular system ,Cancer research ,biology.protein ,medicine ,Platelet-derived growth factor receptor ,Transforming growth factor - Abstract
Decorin is a small proteoglycan that binds to transforming growth factor-β (TGF-β) and inhibits its activity. However, its interaction with platelet-derived growth factor (PDGF), involved in arterial repair after injury, is not well characterized. The objectives of this study were to assess decorin-PDGF and decorin-PDGF receptor (PDGFR) interactions, the in vitro effects of decorin on PDGF-stimulated smooth muscle cell (SMC) functions and the in vivo effects of decorin overexpression on arterial repair in a rabbit carotid balloon-injury model. Decorin binding to PDGF was demonstrated by solid-phase binding and affinity cross-linking assays. Decorin potently inhibited PDGF-stimulated PDGFR phosphorylation. Pretreatment of rabbit aortic SMC with decorin significantly inhibited PDGF-stimulated cell migration, proliferation, and collagen synthesis. Decorin overexpression by adenoviral-mediated gene transfection in balloon-injured carotid arteries significantly decreased intimal cross-sectional area and collagen content by ∼50% at 10 weeks compared to β-galactosidase-transfected or balloon-injured, non-transfected controls. This study shows that decorin binds to PDGF and inhibits its stimulatory activity on SMCs by preventing PDGFR phosphorylation. Decorin overexpression reduces intimal hyperplasia and collagen content after arterial injury. Decorin may be an effective therapy for the prevention of intimal hyperplasia after balloon angioplasty.
- Published
- 2003
- Full Text
- View/download PDF
34. A New Role for Corticotropin-Releasing Factor Receptor-2 Suppression of Vascularization
- Author
-
Frank J. Giordano, Tracy L. Bale, and Wylie Vale
- Subjects
Agonist ,medicine.medical_specialty ,medicine.drug_class ,Angiogenesis ,Neovascularization, Physiologic ,Blood Pressure ,Biology ,Receptors, Corticotropin-Releasing Hormone ,Muscle, Smooth, Vascular ,Neovascularization ,chemistry.chemical_compound ,Heart Rate ,Internal medicine ,medicine ,Animals ,Humans ,Growth factor receptor inhibitor ,Receptor ,Dose-Response Relationship, Drug ,Cell biology ,Vasodilation ,Vascular endothelial growth factor ,Vascular endothelial growth factor B ,Endocrinology ,chemistry ,Vascular endothelial growth factor C ,medicine.symptom ,Cardiology and Cardiovascular Medicine - Abstract
The corticotropin-releasing factor (CRF) family of receptors and ligands are known to have potent effects on vasculature function. Our recent data has described a novel role of corticotropin-releasing factor receptor-2 (CRFR2) as a tonic inhibitor of neovascularization. CRFR2 is found in both endothelial and smooth muscle cells (SMC) in the vasculature, where its function has been elusive. From investigation into the role of CRFR2 as a determinant of tissue vascularization, it was discovered that mice deficient for CRFR2 become hypervascularized postnatally and express increased levels of vascular endothelial growth factor (VEGF). Mechanistically, CRFR2 activation in vitro was found to result in reduced VEGF release from SMCs, an inhibition of SMC proliferation, and an inhibition of capillary tube formation in collagen gels. Treatment of a subcutaneously injected gel matrix with a CRFR2 agonist inhibited growth factor-induced vascularization in vivo. Western blots for the cell-cycle retinoblastoma protein (Rb)—essential for cell-cycle progression, showed decreased levels of phosphorylated Rb following CRFR2 agonist treatment in SMCs—supporting a role for CRFR2 in regulation of SMC proliferation. These results suggest that CRFR2 is a critical component of a novel pathway necessary for tonic inhibition of adult neovascularization, and that CRFR2 may be a potential target for therapeutic modulation of angiogenesis.
- Published
- 2003
- Full Text
- View/download PDF
35. Induction of angiogenesis in a mouse model using engineered transcription factors
- Author
-
Andrew C. Jamieson, Lei Zhang, Casey C. Case, Reed Hickey, David F. Meoli, Anjali K. Nath, Frank J. Giordano, Bingliang Chen, Edward J. Rebar, Yan Huang, Lei Xu, S. Kaye Spratt, Alan P. Wolffe, Yuxin Liang, and Sameer K. Nath
- Subjects
Vascular Endothelial Growth Factor A ,Angiogenesis ,Molecular Sequence Data ,Artificial transcription factor ,Neovascularization, Physiologic ,Endogeny ,Biology ,Protein Engineering ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Complementary DNA ,Animals ,Amino Acid Sequence ,Transcription factor ,Regulation of gene expression ,Zinc Fingers ,3T3 Cells ,Genetic Therapy ,General Medicine ,Molecular biology ,Recombinant Proteins ,Cell biology ,Vascular endothelial growth factor A ,Gene Expression Regulation ,Drug Design ,Models, Animal ,Angiogenesis Inducing Agents ,Transcription Factors - Abstract
The relationship between the structure of zinc-finger protein (ZFP) transcription factors and DNA sequence binding specificity has been extensively studied. Advances in this field have made it possible to design ZFPs de novo that will bind to specific targeted DNA sequences. It has been proposed that such designed ZFPs may eventually be useful in gene therapy. A principal advantage of this approach is that activation of an endogenous gene ensures expression of the natural array of splice variants. Preliminary studies in tissue culture have validated the feasibility of this approach. The studies reported here were intended to test whether engineered transcription factors are effective in a whole-organism model. ZFPs were designed to regulate the endogenous gene encoding vascular endothelial growth factor-A (Vegfa). Expression of these new ZFPs in vivo led to induced expression of the protein VEGF-A, stimulation of angiogenesis and acceleration of experimental wound healing. In addition, the neovasculature resulting from ZFP-induced expression of Vegfa was not hyperpermeable as was that produced by expression of murine Vegfa(164) cDNA. These data establish, for the first time, that specifically designed transcription factors can regulate an endogenous gene in vivo and evoke a potentially therapeutic biophysiologic effect.
- Published
- 2002
- Full Text
- View/download PDF
36. Corticotropin-releasing factor receptor 2 is a tonic suppressor of vascularization
- Author
-
Reed Hickey, Wylie Vale, Tracy L. Bale, Yan Huang, Kirk L. Peterson, Frank J. Giordano, Kuo-Fen Lee, and Anjali K. Nath
- Subjects
Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,Angiogenesis ,Neovascularization, Physiologic ,Angiogenesis Inhibitors ,Endothelial Growth Factors ,Receptors, Corticotropin-Releasing Hormone ,Muscle, Smooth, Vascular ,Neovascularization ,Mice ,chemistry.chemical_compound ,Internal medicine ,medicine ,Animals ,Receptor ,Cells, Cultured ,Mice, Knockout ,Lymphokines ,Multidisciplinary ,biology ,Vascular Endothelial Growth Factors ,Cell growth ,Retinoblastoma protein ,Biological Sciences ,Cell cycle ,Capillaries ,Cell biology ,Vascular endothelial growth factor ,Vascular endothelial growth factor A ,Endocrinology ,Gene Expression Regulation ,chemistry ,biology.protein ,Endothelium, Vascular ,medicine.symptom ,Cell Division - Abstract
Angiogenesis is regulated by means of a balance between activators and inhibitors. However, little is known regarding the regulation of the quiescent state of adult vessels. Corticotropin-releasing factor receptor 2 (CRFR2) is found in both endothelial and smooth muscle cells (SMCs) in the vasculature, where its function has remained elusive. We have investigated the role of CRFR2 as a determinant of tissue vascularization by comparing control and CRFR2-deficient mice with immunohistological and morphometric techniques. To define the mechanisms responsible for CRFR2 inhibition of angiogenesis, we have also examined in vitro the effect of ligand activation on cell proliferation, cell cycle protein phosphorylation, and capillary tube formation. Our results demonstrate that mice deficient for CRFR2 become hypervascularized postnatally. Activation of this receptor in vitro results in reduced vascular endothelial growth factor (VEGF) release from SMCs, an inhibition of SMC proliferation, and inhibition of capillary tube formation in collagen gels. Treatment of a subcutaneously injected gel matrix with a CRFR2 agonist inhibits growth factor-induced vascularization. Western blots show that cell cycle retinoblastoma protein, which is essential for cell cycle progression, is decreased by CRFR2 agonist treatment in SMCs. These results suggest that CRFR2 is a critical component of a pathway necessary for tonic inhibition of adult neovascularization. CRFR2 may be a potential target for therapeutic modulation of angiogenesis in cancer and ischemic cardiovascular disease.
- Published
- 2002
- Full Text
- View/download PDF
37. The expression of epidermal growth factor-like domain 7 regulated by oxygen tension via hypoxia inducible factor (HIF)-1α activity
- Author
-
Ai-qiong Qin, Yu-Sheng Liu, Qing-Hua Lu, Frank J. Giordano, Wei-dong Jiang, Yang Huang, and Zhi-Wei Huang
- Subjects
EGF Family of Proteins ,Angiogenesis ,Mice, Transgenic ,Hyperoxia ,Mice ,Epidermal growth factor ,medicine ,Human Umbilical Vein Endothelial Cells ,Animals ,Hypoxia ,Gene knockdown ,business.industry ,Calcium-Binding Proteins ,Proteins ,General Medicine ,Hypoxia (medical) ,Hypoxia-Inducible Factor 1, alpha Subunit ,Immunohistochemistry ,Reverse transcriptase ,Oxygen tension ,Cell biology ,Oxygen ,Hypoxia-inducible factors ,Animals, Newborn ,Gene Expression Regulation ,Immunology ,Endothelium, Vascular ,medicine.symptom ,business - Abstract
Hypoxia inducible factor-1α (HIF-1α) regulates many genes involved in angiogenesis during embryonic development. Epidermal growth factor-like domain 7 (Egfl7) is a specific marker for human arterial endothelial cells that are in an activated state of proliferation, migration, and remodeling. This study evaluates the intricate relationship between HIF-1α and Egfl7 under both hyperoxia and hypoxia states.The neonatal mice were exposed to either hyperoxia or hypoxia in order to detect the pulmonary and cardiac Egfl7 messenger RNA (mRNA) or protein expression regulated by oxygen tension in vivo by reverse transcriptase polymerase chain reaction or immunohistochemistry staining. Egfl7 expression in HIF-1α null pulmonary endothelial cells in hypoxia conditions and effects of overexpression or knockdown of HIF-1α on Egfl7 expression in human umbilical vein endothelial cells would be clarified in vitro by reverse transcriptase polymerase chain reaction and Western blot, respectively.Hyperoxia exposure significantly reduced Egfl7 expression in neonatal mice lungs by 36% compared with age-matched normoxia control mice (P0.05, n = 6). The pulmonary Egfl7 transcription levels were increased by 1.7- and 1.9-fold in 24 hours and by day 8 in hypoxia groups compared with the normoxia control values (P0.05, n = 6). The cardiac Egfl7 mRNA expression was significantly increased by 4.5-fold in the day 8 group compared with the normoxia control values (P0.05, n = 6). The expression of Egfl7 decreased significantly in the HIF-1α(-/-) endothelial cells (ECs), which was only 26% of wild-type HIF-1α(+/+) ECs (P0.05, n = 3). Hypoxia caused a mild but significant increase of Egfl7 expression in HIF-1α(+/+) ECs (P0.05). In vitro, overexpression of HIF-1α enhanced Egfl7 expression, whereas knockdown of HIF-1α reduced Egfl7 expression.Overexpression of HIF-1α enhanced Egfl7 expression, whereas knockdown of HIF-1α reduced Egfl7 expression. Egfl7 could be a HIF-1α responsive gene regulated by oxygen tension.
- Published
- 2014
38. Abstract 155: Sustained CD4+ T-Cell Activation Promotes Transplant-Associated Recipient Cardiovascular Disease
- Author
-
Mehran M. Sadeghi, Jing Zhou, Yang Jiao, Jordan S. Pober, George Tellides, Rahmat Ali, Jiasheng Zhang, Zuzana Tobiasova, Yan Huang, Qingle Li, Guangxin Li, Tai Yi, Frank J. Giordano, and Lingfeng Qin
- Subjects
Apolipoprotein E ,medicine.medical_treatment ,T cell ,Biology ,medicine.disease ,Major histocompatibility complex ,Proinflammatory cytokine ,Transplant rejection ,Transplantation ,Cytokine ,medicine.anatomical_structure ,Antigen ,Immunology ,medicine ,biology.protein ,Cardiology and Cardiovascular Medicine - Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in transplant patients. The mechanisms underlying increased recipient CVD are poorly understood. We have established an animal model of transplant-associated recipient CVD by combining murine models of atherosclerosis (apolipoprotein E-deficient recipients, ApoE -/- ) and of transplant rejection (heterotopic cardiac transplantation). Mice were placed on a normal chow diet for 3 months after transplantation and then analyzed. ApoE -/- recipients of rejecting allografts developed more severe aortic atherosclerosis compared to syngeneic controls, most strikingly across a major histocompatibility complex (MHC) class II antigen barrier (Fig. A). Atherosclerotic lesions were associated with increased CD4 + T cell infiltration and sustained CD4 + T cell activation as assessed by flow cytometry. Total cholesterol and triglyceride levels were unchanged. Cardiac function and aortic distention by echocardiography demonstrated significant impairment in recipients of MHC class II mismatched grafts (Fig. B). In addition, quantitative assessment of proinflammatory cytokines demonstrated significantly elevated plasma levels of interferon (IFN)-γ, the signature Th1 cytokine, as well as IFN-γ inducer cytokine IL-12 in mice with MHC class II mismatched grafts. Our study provides an experimental model of transplant-associated recipient CVD, reveals the critical role of sustained CD4 + T cell activation by donor-recipient immunological crosstalk, and has the potential for identifying novel therapeutic interventions for transplant-associated recipient CVD.
- Published
- 2014
- Full Text
- View/download PDF
39. A cardiac myocyte vascular endothelial growth factor paracrine pathway is required to maintain cardiac function
- Author
-
Hans-Peter Gerber, Nicholas VanBruggen, Reed Hickey, Yan Huang, Frank J. Giordano, Kirk L. Peterson, Pilar Ruiz-Lozano, Simon-Peter Williams, John Ross, Napoleone Ferrara, Stuart Bunting, Kenneth R. Chien, Anjali K. Nath, Yusu Gu, and Nancy D. Dalton
- Subjects
Vascular Endothelial Growth Factor A ,Cardiac function curve ,medicine.medical_specialty ,Endothelial Growth Factors ,Biology ,Mice ,chemistry.chemical_compound ,Paracrine signalling ,Mediator ,Internal medicine ,medicine ,Animals ,In Situ Hybridization ,Mice, Knockout ,Lymphokines ,Multidisciplinary ,Vascular Endothelial Growth Factors ,Gene Expression Profiling ,Myocardium ,Cardiac myocyte ,Heart ,Biological Sciences ,Immunohistochemistry ,Cardiovascular physiology ,Vascular endothelial growth factor B ,Vascular endothelial growth factor ,Vascular endothelial growth factor A ,Endocrinology ,chemistry ,Models, Animal ,cardiovascular system - Abstract
The role of the cardiac myocyte as a mediator of paracrine signaling in the heart has remained unclear. To address this issue, we generated mice with cardiac myocyte-specific deletion of the vascular endothelial growth factor gene, thereby producing a cardiomyocyte-specific knockout of a secreted factor. The hearts of these mice had fewer coronary microvessels, thinned ventricular walls, depressed basal contractile function, induction of hypoxia-responsive genes involved in energy metabolism, and an abnormal response to β-adrenergic stimulation. These findings establish the critical importance of cardiac myocyte-derived vascular endothelial growth factor in cardiac morphogenesis and determination of heart function. Further, they establish an adult murine model of hypovascular nonnecrotic cardiac contractile dysfunction.
- Published
- 2001
- Full Text
- View/download PDF
40. Angiogenesis: the role of the microenvironment in flipping the switch
- Author
-
Frank J. Giordano and Randall S. Johnson
- Subjects
Cell signaling ,Cell division ,Angiogenesis ,Cell Communication ,Biology ,Neovascularization ,Genetics ,Transcriptional regulation ,medicine ,Animals ,Humans ,G alpha subunit ,Neovascularization, Pathologic ,Nuclear Proteins ,Hypertrophy ,Hypoxia (medical) ,Hypoxia-Inducible Factor 1, alpha Subunit ,Cell Hypoxia ,Extracellular Matrix ,Cell biology ,DNA-Binding Proteins ,Oxygen ,Cell Transformation, Neoplastic ,Immunology ,Hypoxia-Inducible Factor 1 ,medicine.symptom ,Cell Division ,Transcription Factors ,Developmental Biology ,Morphogen - Abstract
The initiating factors in angiogenesis during development and disease are often microenvironmental changes, which induce signaling to the vasculature from affected tissues. Among these, lowered oxygen pressure, hypoxia, is one of the most potent inducers/initiators of an angiogenic response. Significant evidence indicates that hypoxia acts as a morphogen during vascularization - inducing and shaping the recruitment and formation of new vascular beds through critical transcriptional control pathways. Recent advances indicate that extensive interactions occur between developing blood vessels, the tissues that they vascularize, and the interstitial environment to control and shape the establishment of new capillary beds. Identification of the processes that control the hypoxic response intracellularly has allowed an increasingly sophisticated understanding of angiogenesis as a process that is very closely tied to the microenvironment that it occurs in. Further understanding of these processes may present powerful therapeutic opportunities for disease intervention.
- Published
- 2001
- Full Text
- View/download PDF
41. Antisense fos B RNA Inhibits Thrombin-Induced Hypertrophy in Cultured Pulmonary Arterial Smooth Muscle Cells
- Author
-
Xuping Bao, Chuanyi M. Lu, Frank J. Giordano, Kemberlyn C. Morris, and Abraham Rothman
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Basic fibroblast growth factor ,Pulmonary Artery ,Biology ,Muscle, Smooth, Vascular ,Cell Line ,Muscle hypertrophy ,chemistry.chemical_compound ,Thrombin ,Bacterial Proteins ,Physiology (medical) ,Internal medicine ,Sense (molecular biology) ,medicine ,Growth Substances ,Growth factor ,Hypertrophy ,Molecular biology ,Angiotensin II ,Antisense RNA ,Antisense Elements (Genetics) ,Endocrinology ,chemistry ,Calcium-Calmodulin-Dependent Protein Kinases ,RNA ,Cardiology and Cardiovascular Medicine ,Cell Division ,FOSB ,medicine.drug - Abstract
Background —We have previously reported that fos B mRNA is induced by hypertrophic stimuli (thrombin, angiotensin II) but not proliferative stimuli (platelet-derived growth factor, basic fibroblast growth factor) in pulmonary arterial smooth muscle cells (PASMCs) ( J Biol Chem . 1994;9:6399–6404). Our aim in the present study was to investigate the potential role of FosB in PASMC hypertrophy. Methods and Results —Adenoviruses carrying sense or antisense fos B RNA expression cassettes were used to infect cultured PASMCs with the aim of increasing or inhibiting fos B expression, respectively. We examined whether fos B expression modification affected the growth of quiescent PASMCs, thrombin-induced hypertrophy, or platelet-derived growth factor–induced proliferation. PASMC growth was assessed by daily cell number count, determination of [ 3 H]leucine incorporation, and quantification of total cellular protein. Neither an increase nor a decrease in FosB protein expression caused a significant change in the growth of quiescent PASMCs over a period of 96 hours, indicating that FosB alone is not sufficient to induce hypertrophy. Modification of FosB levels did not affect platelet-derived growth factor–induced PASMC proliferation. An increase in FosB expression did not augment thrombin-induced hypertrophy; however, inhibition of FosB expression resulted in a diminution of thrombin-induced hypertrophy by 58±6% ( P Conclusions —These results suggest that FosB is necessary but not sufficient for thrombin-induced hypertrophy in cultured PASMCs.
- Published
- 1998
- Full Text
- View/download PDF
42. An engineered VEGF‐activating zinc finger protein transcription factor improves blood flow and limb salvage in advanced‐age mice
- Author
-
Casey C. Case, Yuxin Liang, Dinggang Liu, William C. Sessa, Reed Hickey, Edward J. Rebar, Jun Yu, Kaye Spratt, Li Lei, Yan Huang, Frank J. Giordano, Jennifer L. Yeh, and Linda Hinh
- Subjects
Vascular Endothelial Growth Factor A ,Aging ,Angiogenesis ,Genetic Vectors ,Molecular Sequence Data ,Neovascularization, Physiologic ,Hindlimb ,Biology ,Protein Engineering ,Biochemistry ,Adenoviridae ,Neovascularization ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,Ischemia ,Gene expression ,Genes, Synthetic ,Laser-Doppler Flowmetry ,Genetics ,medicine ,Animals ,Amino Acid Sequence ,RNA, Messenger ,Molecular Biology ,Transcription factor ,Zinc finger ,Zinc Fingers ,Genetic Therapy ,Molecular biology ,Recombinant Proteins ,Cell biology ,Mice, Inbred C57BL ,Vascular endothelial growth factor ,Vascular endothelial growth factor A ,Gene Expression Regulation ,chemistry ,Feasibility Studies ,medicine.symptom ,Blood Flow Velocity ,Transcription Factors ,Biotechnology - Abstract
Advances in understanding the relationship between protein structure and DNA binding specificity have made it possible to engineer zinc finger protein (ZFP) transcription factors to specifically activate or repress virtually any gene. To evaluate the potential clinical utility of this approach for peripheral vascular disease, we investigated the ability of an engineered vascular endothelial growth factor (VEGFa)-activating ZFP (MVZ+426b) to induce angiogenesis and rescue hindlimb ischemia in a murine model. Hindlimb ischemia was surgically induced in advanced-age C57/BL6 mice. Adenovirus (Ad) encoding either MVZ+426b or the fluorescent marker dsRed was delivered to the adducter muscle of the ischemic hindlimb, and the effects on blood flow, limb salvage, and vascularization were assessed. Ad-MVZ+426b induced expression of VEGFa at the mRNA and protein levels and stimulated a significant increase in vessel counts in the ischemic limb. This was accompanied by significantly increased blood flow and limb salvage as measured serially for 4 wk. These data demonstrate that activation of the endogenous VEGFa gene by an engineered ZFP can induce angiogenesis in a clinically relevant model and further document the feasibility of designing ZFPs to therapeutically regulate gene expression in vivo.
- Published
- 2006
- Full Text
- View/download PDF
43. Activation of hypoxia-inducible factor-2 in adipocytes results in pathological cardiac hypertrophy
- Author
-
M. Celeste Simon, Frank J. Giordano, Carmen J. Booth, Qun Lin, Zhong Yun, Yan Huang, Volker H. Haase, and Randall S. Johnson
- Subjects
obesity ,Time Factors ,Adipose tissue ,030204 cardiovascular system & hematology ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Adipocyte ,Adipocytes ,Basic Helix-Loop-Helix Transcription Factors ,Myocytes, Cardiac ,Original Research ,Mice, Knockout ,0303 health sciences ,HIF‐2 alpha ,medicine.anatomical_structure ,Phenotype ,Hypoxia-inducible factors ,Von Hippel-Lindau Tumor Suppressor Protein ,Cytokines ,medicine.symptom ,Inflammation Mediators ,Cardiology and Cardiovascular Medicine ,Signal Transduction ,medicine.medical_specialty ,Inflammation ,Cardiomegaly ,Mice, Transgenic ,Proinflammatory cytokine ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Genetic Predisposition to Disease ,Transcription factor ,030304 developmental biology ,Heart Failure ,business.industry ,hypoxia ,Monocyte ,Editorials ,Hypoxia-Inducible Factor 1, alpha Subunit ,Mice, Inbred C57BL ,Disease Models, Animal ,Endocrinology ,HIF1A ,chemistry ,Gene Expression Regulation ,inflammation ,business ,cardiomyopathy - Abstract
Background Obesity can cause structural and functional abnormalities of the heart via complex but largely undefined mechanisms. Emerging evidence has shown that obesity results in reduced oxygen concentrations, or hypoxia, in adipose tissue. We hypothesized that the adipocyte hypoxia‐signaling pathway plays an essential role in the development of obesity‐associated cardiomyopathy. Methods and Results Using a mouse model in which the hypoxia‐inducible factor ( HIF ) pathway is activated by deletion of the von Hippel–Lindau gene specifically in adipocytes, we found that mice with adipocyte–von Hippel–Lindau deletion developed lethal cardiac hypertrophy. HIF activation in adipocytes results in overexpression of key cardiomyopathy‐associated genes in adipose tissue, increased serum levels of several proinflammatory cytokines including interleukin‐1β and monocyte chemotactic protein‐1, and activation of nuclear factor–κB and nuclear factor of activated T cells in the heart. Interestingly, genetic deletion of Hif2a , but not Hif1a , was able to rescue cardiac hypertrophy and abrogate adipose inflammation. Conclusion We have discovered a previously uncharacterized mechanism underlying a critical and direct role of the adipocyte HIF ‐2 transcription factor in the development of adipose inflammation and pathological cardiac hypertrophy.
- Published
- 2013
44. Rapamycin inhibits smooth muscle cell proliferation and obstructive arteriopathy attributable to elastin deficiency
- Author
-
Yibing Qyang, Qingle Li, Wei Li, Lingfeng Qin, George Tellides, William C. Sessa, Barbara R. Pober, Frank J. Giordano, May Tassabehji, and Rahmat Ali
- Subjects
Adult ,Male ,Williams Syndrome ,medicine.medical_specialty ,Myocytes, Smooth Muscle ,Arterial Occlusive Diseases ,Biology ,Gene mutation ,Muscle, Smooth, Vascular ,Mice ,Internal medicine ,medicine.artery ,medicine ,Myocyte ,Animals ,Humans ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Sirolimus ,Aorta ,Cell growth ,TOR Serine-Threonine Kinases ,Hyperplasia ,Middle Aged ,medicine.disease ,Elastin ,Aortic Stenosis, Supravalvular ,Mice, Inbred C57BL ,Endocrinology ,cardiovascular system ,biology.protein ,Female ,Cardiology and Cardiovascular Medicine ,Supravalvular aortic stenosis ,Signal Transduction - Abstract
Objective— Patients with elastin deficiency attributable to gene mutation (supravalvular aortic stenosis) or chromosomal microdeletion (Williams syndrome) are characterized by obstructive arteriopathy resulting from excessive smooth muscle cell (SMC) proliferation, mural expansion, and inadequate vessel size. We investigated whether rapamycin, an inhibitor of the cell growth regulator mammalian target of rapamycin (mTOR) and effective against other SMC proliferative disorders, is of therapeutic benefit in experimental models of elastin deficiency. Approach and Results— As previously reported, Eln −/− mice demonstrated SMC hyperplasia and severe stenosis of the aorta, whereas Eln +/− mice exhibited a smaller diameter aorta with more numerous but thinner elastic lamellae. Increased mTOR signaling was detected in elastin-deficient aortas of newborn pups that was inhibited by maternal administration of rapamycin. mTOR inhibition reduced SMC proliferation and aortic obstruction in Eln −/− pups and prevented medial hyperlamellation in Eln +/− weanlings without compromising aortic size. However, rapamycin did not prolong the survival of Eln −/− pups, and it retarded the somatic growth of juvenile Eln +/− and Eln +/+ mice. In cell cultures, rapamycin inhibited prolonged mTOR activation and enhanced proliferation of SMC derived from patients with supravalvular aortic stenosis and with Williams syndrome. Conclusions— mTOR inhibition may represent a pharmacological strategy to treat diffuse arteriopathy resulting from elastin deficiency.
- Published
- 2013
45. Adenovirus-mediated Gene Transfer of a Heat Shock Protein 70 (hsp70i) Protects Against Simulated Ischemia
- Author
-
Wolfgang H. Dillmann, Frank J. Giordano, Andre G. Conde, and Ruben Mestril
- Subjects
Genetically modified mouse ,Recombinant Fusion Proteins ,Genetic Vectors ,Ischemia ,Gene Expression ,Biology ,medicine.disease_cause ,Adenoviridae ,Cell Line ,Viral vector ,Heat shock protein ,medicine ,Animals ,Humans ,HSP70 Heat-Shock Proteins ,Creatine Kinase ,Molecular Biology ,Cells, Cultured ,L-Lactate Dehydrogenase ,Myocardium ,Gene Transfer Techniques ,medicine.disease ,Molecular biology ,Rats ,Hsp70 ,Cell culture ,Cardiology and Cardiovascular Medicine ,Reperfusion injury - Abstract
We have recently shown that the overexpression of a heat shock protein 70 (hsp 70) in a rat myogenic cell line confers protection against simulated ischemia. We also developed and demonstrated that overexpression of this protein, in the hearts of transgenic mice, protects against ischemia/reperfusion injury. We have now inserted the hsp70 gene in an adenoviral vector and show that we are able to transfer and achieve overexpression of this protein in neonatal cardiomyocytes and in the rat myogenic cell line H9c2. We find that cells infected with the adenoviral-hsp70i construct are rendered tolerant to simulated ischemia as compared to cells infected with a control recombinant adenoviral construct. In conclusion, our results demonstrate the feasibility of using adenoviral vectors to overexpress the hsp70 in myogenic cells, specially in cardiomyocytes, and the efficiency of this approach for providing protection against myocardial ischemia.
- Published
- 1996
- Full Text
- View/download PDF
46. Endothelial Nogo-B regulates sphingolipid biosynthesis to promote the transition from hypertrophy to heart failure during chronic pressure overload
- Author
-
Yan Huang, Anna Cantalupo, Paula S. Azevedo, Yi Zhang, Frank J. Giordano, Annarita Di Lorenzo, and Mauro Siragusa
- Subjects
0301 basic medicine ,Pressure overload ,medicine.medical_specialty ,Transition (genetics) ,business.industry ,Sphingolipid biosynthesis ,medicine.disease ,Muscle hypertrophy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,030220 oncology & carcinogenesis ,Internal medicine ,Heart failure ,Internal Medicine ,medicine ,Cardiology and Cardiovascular Medicine ,business - Published
- 2016
- Full Text
- View/download PDF
47. Retrograde coronary perfusion: a superior route to deliver therapeutics to the heart?**Editorials published in the Journal of the American College of Cardiologyreflect the views of the authors and do not necessarily represent the views of JACCor the American College of Cardiology
- Author
-
Frank J. Giordano
- Subjects
Pathology ,medicine.medical_specialty ,business.industry ,medicine ,Genomics ,business ,Bioinformatics ,Proteomics ,Cardiology and Cardiovascular Medicine ,Perfusion - Abstract
As the genomics and proteomics revolutions progress and the molecular biology of various cardiovascular pathologies is better understood, an increasing number of potential molecular therapies are being proposed. The ultimate success of any of these therapies depends upon many factors, including the
- Published
- 2003
- Full Text
- View/download PDF
48. Engineered Zinc-Finger Proteins Can Compensate Genetic Haploinsufficiency by Transcriptional Activation of the Wild-Type Allele: Application to Willams-Beuren Syndrome and Supravalvular Aortic Stenosis
- Author
-
Pei Zhang, Barry Starcher, William C. Sessa, Frank J. Giordano, Yan Huang, Angela Huang, Manuel Morales-Ruiz, and Laura E. Niklason
- Subjects
Transcriptional Activation ,Williams Syndrome ,Gene Expression ,Haploinsufficiency ,Biology ,medicine.disease_cause ,Protein Engineering ,Cell Line ,Mutant protein ,Cell Movement ,Dosage Compensation, Genetic ,Genetics ,medicine ,Humans ,Allele ,Molecular Biology ,Research Articles ,Alleles ,Cell Proliferation ,Regulation of gene expression ,Mutation ,Wild type ,Zinc Fingers ,Elastin ,Nonsense Mediated mRNA Decay ,Aortic Stenosis, Supravalvular ,Gene Expression Regulation ,Organ Specificity ,biology.protein ,Molecular Medicine ,Supravalvular aortic stenosis - Abstract
Williams-Beuren syndrome (WBS) and supravalvular aortic stenosis (SVAS) are genetic syndromes marked by the propensity to develop severe vascular stenoses. Vascular lesions in both syndromes are caused by haploinsufficiency of the elastin gene. We used these distinct genetic syndromes as models to evaluate the feasibility of using engineered zinc-finger protein transcription factors (ZFPs) to achieve compensatory expression of haploinsufficient genes by inducing augmented expression from the remaining wild-type allele. For complex genes with multiple splice variants, this approach could have distinct advantages over cDNA-based gene replacement strategies. Targeting the elastin gene, we show that transcriptional activation by engineered ZFPs can induce compensatory expression from the wild-type allele in the setting of classic WBS and SVAS genetic mutations, increase elastin expression in wild-type cells, induce expression of the major elastin splice variants, and recapitulate their natural stoichiometry. Further, we establish that transcriptional activation of the mutant allele in SVAS does not overcome nonsense-mediated decay, and thus ZFP-mediated transcriptional activation is not likely to induce production of a mutant protein, a crucial consideration. Finally, we show in bioengineered blood vessels that ZFP-mediated induction of elastin expression is capable of stimulating functional elastogenesis. Haploinsufficiency is a common mechanism of genetic disease. These findings have significant implications for WBS and SVAS, and establish that haploinsufficiency can be overcome by targeted transcriptional activation without inducing protein expression from the mutant allele.
- Published
- 2012
49. Integrin ß-1 Regulates Eph-B4–Mediated Vein Graft Adaptation
- Author
-
Clinton D. Protack, Kenneth R. Ziegler, Michael J. Collins, Caroline C. Jadlowiec, Sammy D.D. Eghbalieh, Frank J. Giordano, Akihito Muto, Alan Dardik, Xin Li, and Lynn S. Model
- Subjects
biology ,Graft patency ,business.industry ,Integrin ,technology, industry, and agriculture ,Erythropoietin-producing hepatocellular (Eph) receptor ,Vein graft ,macromolecular substances ,Adsorption ,biology.protein ,Biophysics ,Medicine ,Surgery ,business ,Cardiology and Cardiovascular Medicine - Abstract
increased when siRNA was complexed with PEI. The amount of siRNA adsorbed generally appeared greater in ePET than in PTFE fabrics. Repeated dipping of ePET in siGLO Red/PEI solution significantly increased siRNA adsorption vs single dipping. Conclusions: siRNA adsorption to ePET and PTFE surfaces is significantly increased if the siRNA is complexed with PEI. Preliminary data in this study suggest that siRNA adsorbs better to ePET than PTFE. Further, repeated dipping of ePET additionally increased adsorption of siRNA to fabric. Thus, coating of ePET with siRNA/PEI may be a method to improve graft patency.
- Published
- 2011
- Full Text
- View/download PDF
50. Renalase deficiency aggravates ischemic myocardial damage
- Author
-
Dinggang Liu, Frank J. Giordano, Raymond R. Russell, Janete Quelhas-Santos, Kerry S. Russell, Jianchao Xu, Manuel Pestana, Peili Wang, Yanling Wu, Gary V. Desir, Richard A. Flavell, Benedita Sampaio-Maia, Heino Velazquez, and Guoyong Li
- Subjects
Male ,medicine.medical_specialty ,renal failure ,amine oxidase ,Cardiotonic Agents ,Epinephrine ,Monoamine oxidase ,Myocardial Ischemia ,chemistry.chemical_compound ,Mice ,Norepinephrine ,Ventricular hypertrophy ,Internal medicine ,Medicine ,Animals ,Humans ,Renal Insufficiency, Chronic ,Blood urea nitrogen ,Monoamine Oxidase ,Renalase ,DNA Primers ,sympathetic nervous system ,Mice, Knockout ,Aldosterone ,Base Sequence ,business.industry ,medicine.disease ,NAD ,Recombinant Proteins ,Mice, Inbred C57BL ,Disease Models, Animal ,Endocrinology ,chemistry ,Nephrology ,NADH oxidase ,Knockout mouse ,Hypertension ,Female ,business ,catecholamines ,medicine.drug ,Kidney disease - Abstract
Chronic kidney disease (CKD) leads to an 18-fold increase in cardiovascular complications not fully explained by traditional risk factors. Levels of renalase, a recently discovered oxidase that metabolizes catecholamines, are decreased in CKD. Here we show that renalase deficiency in a mouse knockout model causes increased plasma catecholamine levels and hypertension. Plasma blood urea nitrogen, creatinine, and aldosterone were unaffected. However, knockout mice had normal systolic function and mild ventricular hypertrophy but tolerated cardiac ischemia poorly and developed myocardial necrosis threefold more severe than that found in wild-type mice. Treatment with recombinant renalase completely rescued the cardiac phenotype. To gain insight into the mechanisms mediating this cardioprotective effect, we tested if gene deletion affected nitrate and glutathione metabolism, but found no differences between hearts of knockout and wild-type mice. The ratio of oxidized (NAD) to reduced (NADH) nicotinamide adenine dinucleotide in cardiac tissue, however, was significantly decreased in the hearts of renalase knockout mice, as was plasma NADH oxidase activity. In vitro studies confirmed that renalase metabolizes NADH and catecholamines. Thus, renalase plays an important role in cardiovascular pathology and its replacement may reduce cardiac complications in renalase-deficient states such as CKD.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.