Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Int J Mol Sci ; 22(10)2021 May 13.
Article in English | MEDLINE | ID: mdl-34068392

ABSTRACT

Myocardial infarction (MI) remains the leading cause of death in the western world. Despite advancements in interventional revascularization technologies, many patients are not candidates for them due to comorbidities or lack of local resources. Non-invasive approaches to accelerate revascularization within ischemic tissues through angiogenesis by providing Vascular Endothelial Growth Factor (VEGF) in protein or gene form has been effective in animal models but not in humans likely due to its short half-life and systemic toxicity. Here, we tested the hypothesis that PR1P, a small VEGF binding peptide that we developed, which stabilizes and upregulates endogenous VEGF, could be used to improve outcome from MI in rodents. To test this hypothesis, we induced MI in mice and rats via left coronary artery ligation and then treated animals with every other day intraperitoneal PR1P or scrambled peptide for 14 days. Hemodynamic monitoring and echocardiography in mice and echocardiography in rats at 14 days showed PR1P significantly improved multiple functional markers of heart function, including stroke volume and cardiac output. Furthermore, molecular biology and histological analyses of tissue samples showed that systemic PR1P targeted, stabilized and upregulated endogenous VEGF within ischemic myocardium. We conclude that PR1P is a potential non-invasive candidate therapeutic for MI.


Subject(s)
AC133 Antigen/metabolism , Disease Models, Animal , Ischemia/complications , Myocardial Infarction/prevention & control , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Animals , Female , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Inbred C57BL , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Ventricular Function, Left/drug effects
2.
Am J Transplant ; 20(6): 1527-1537, 2020 06.
Article in English | MEDLINE | ID: mdl-31991042

ABSTRACT

Inflammatory responses associated with ischemia/reperfusion injury (IRI) play a central role in alloimmunity and transplant outcomes. A key event driving these inflammatory responses is the burst of reactive oxygen species (ROS), with hydrogen peroxide (H2 O2 ) as the most abundant form that occurs as a result of surgical implantation of the donor organ. Here, we used a syngeneic rat renal transplant and IRI model to evaluate the therapeutic properties of APP-103, a polyoxalate-based copolymer molecule containing vanillyl alcohol (VA) that exhibits high sensitivity and specificity toward the production of H2 O2 . We show that APP-103 is safe, and that it effectively promotes kidney function following IRI and survival of renal transplants. APP-103 reduces tissue injury and IRI-associated inflammatory responses in models of both warm ischemia (kidney clamping) and prolonged cold ischemia (syngeneic renal transplant). Mechanistically, we demonstrate that APP-103 exerts protective effects by specifically targeting the production of ROS. Our data introduce APP-103 as a novel, nontoxic, and site-activating therapeutic approach that effectively ameliorates the consequences of IRI in solid organ transplantation.


Subject(s)
Kidney Transplantation , Reperfusion Injury , Animals , Ischemia , Kidney Transplantation/adverse effects , Polymers , Rats , Reactive Oxygen Species , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control
3.
Am J Physiol Heart Circ Physiol ; 317(5): H891-H922, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31418596

ABSTRACT

Cell death is a fundamental process in cardiac pathologies. Recent studies have revealed multiple forms of cell death, and several of them have been demonstrated to underlie adverse cardiac remodeling and heart failure. With the expansion in the area of myocardial cell death and increasing concerns over rigor and reproducibility, it is important and timely to set a guideline for the best practices of evaluating myocardial cell death. There are six major forms of regulated cell death observed in cardiac pathologies, namely apoptosis, necroptosis, mitochondrial-mediated necrosis, pyroptosis, ferroptosis, and autophagic cell death. In this article, we describe the best methods to identify, measure, and evaluate these modes of myocardial cell death. In addition, we discuss the limitations of currently practiced myocardial cell death mechanisms.


Subject(s)
Biomedical Research/standards , Cardiovascular Diseases/pathology , Cell Death , Guidelines as Topic/standards , Myocytes, Cardiac/pathology , Animals , Biomarkers/metabolism , Cardiovascular Diseases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Myocytes, Cardiac/metabolism , Reproducibility of Results , Signal Transduction
4.
Am J Pathol ; 187(10): 2337-2347, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28736316

ABSTRACT

Tumors induce their heterogeneous vasculature by secreting vascular endothelial growth factor (VEGF)-A. Anti-VEGF/VEGF receptor (VEGFR) drugs treat cancer, but the underlying mechanisms remain unclear. An adenovirus expressing VEGF-A (Ad-VEGF-A164) replicates the tumor vasculature in mice without tumor cells. Mother vessels (MV) are the first angiogenic vessel type to form in tumors and after Ad-VEGF-A164. Multiday treatments with a VEGF trap reverted MV back to normal microvessels. We now show that, within hours, a single dose of several anti-VEGF drugs collapsed MV to form glomeruloid microvascular proliferations (GMP), accompanied by only modest endothelial cell death. GMP, common in many human cancers but of uncertain origin, served as an intermediary step in MV reversion to normal microvessels. The vasodisruptive drug combretastatin CA4 also targeted MV selectively but acted differently, extensively killing MV endothelium. Antivascular changes were quantified with a novel Evans blue dye assay that measured vascular volumes. As in tumors, Ad-VEGF-A164 strikingly increased endothelial nitric oxide synthase (eNOS) expression. The eNOS inhibitor N(G)-Nitro-l-arginine methyl ester mimicked anti-VEGF/VEGFR drugs, rapidly collapsing MV to GMP. Inhibition of eNOS reduces synthesis of its vasodilatory product, nitric oxide, leading to arterial contraction. Patients and mice receiving anti-VEGF/VEGFR drugs develop hypertension, reflecting systemic arterial contraction. Together, anti-VEGF/VEGFR drugs act in part by inhibiting eNOS, causing vasocontraction, MV collapse to GMP, and subsequent reversion of GMP to normal microvessels, all without extensive vascular killing.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Blood Vessels/drug effects , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adenoviridae/metabolism , Animals , Bibenzyls/pharmacology , Cell Death/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Hypertension/pathology , Mice, Inbred C57BL , Mice, Nude , Microvessels/drug effects , Microvessels/pathology , Models, Biological , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Am J Physiol Heart Circ Physiol ; 311(6): H1392-H1408, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27694217

ABSTRACT

The HDL receptor SR-BI mediates the transfer of cholesteryl esters from HDL to cells and controls HDL abundance and structure. Depending on the genetic background, loss of SR-BI causes hypercholesterolemia, anemia, reticulocytosis, splenomegaly, thrombocytopenia, female infertility, and fatal coronary heart disease (CHD). The carboxy terminus of SR-BI (505QEAKL509) must bind to the cytoplasmic adaptor PDZK1 for normal hepatic-but not steroidogenic cell-expression of SR-BI protein. To determine whether SR-BI's carboxy terminus is also required for normal protein levels in steroidogenic cells, we introduced into SR-BI's gene a 507Ala/STOP mutation that produces a truncated receptor (SR-BIΔCT). As expected, the dramatic reduction of hepatic receptor protein in SR-BIΔCT mice was similar to that in PDZK1 knockout (KO) mice. Unlike SR-BI KO females, SR-BIΔCT females were fertile. The severity of SR-BIΔCT mice's hypercholesterolemia was intermediate between those of SR-BI KO and PDZK1 KO mice. Substantially reduced levels of the receptor in adrenal cortical cells, ovarian cells, and testicular Leydig cells in SR-BIΔCT mice suggested that steroidogenic cells have an adaptor(s) functionally analogous to hepatic PDZK1. When SR-BIΔCT mice were crossed with apolipoprotein E KO mice (SR-BIΔCT/apoE KO), pathologies including hypercholesterolemia, macrocytic anemia, hepatic and splenic extramedullary hematopoiesis, massive splenomegaly, reticulocytosis, thrombocytopenia, and rapid-onset and fatal occlusive coronary arterial atherosclerosis and CHD (median age of death: 9 wk) were observed. These results provide new insights into the control of SR-BI in steroidogenic cells and establish SR-BIΔCT/apoE KO mice as a new animal model for the study of CHD.


Subject(s)
Adrenal Cortex/metabolism , Hypercholesterolemia/genetics , Leydig Cells/metabolism , Liver/metabolism , Ovary/metabolism , Scavenger Receptors, Class B/genetics , Anemia, Macrocytic/genetics , Animals , Apolipoproteins E/genetics , Coronary Artery Disease/genetics , Coronary Artery Disease/mortality , Coronary Disease/genetics , Coronary Disease/mortality , Coronary Occlusion/genetics , Coronary Occlusion/mortality , Female , Gene Knock-In Techniques , Hematopoiesis, Extramedullary/genetics , Immunoblotting , Lipoproteins, HDL/genetics , Male , Mice , Mutation , Polymerase Chain Reaction , Receptors, Lipoprotein/genetics , Reticulocytosis/genetics , Splenomegaly/genetics , Thrombocytopenia/genetics , Transcriptome
6.
Am J Pathol ; 185(2): 325-34, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25478810

ABSTRACT

We previously characterized the Mdr2(Abcb4)(-/-) mouse as a reproducible model of chronic biliary liver disease. However, it demonstrates relatively slow fibrosis progression, possibly due to its fibrosis-resistant genetic background. We aimed to improve the model by moving it onto a fibrosis-susceptible background. We generated novel BALB/c.Mdr2(-/-) mouse via genetic backcross onto highly fibrosis-susceptible BALB/c substrain, identified in inbred mouse strain screening. Liver fibrosis, portal pressure, and hepatic tumor burden in BALB/c.Mdr2(-/-) mice were studied up to 1 year of age in direct comparison to parental strain FVB.Mdr2(-/-). BALB/c.Mdr2(-/-) mice developed periductular onion-skin type fibrotic lesions and pronounced ductular reaction starting from 4 weeks of age. Compared to parental strain, BALB/c.Mdr2(-/-) mice demonstrated dramatically accelerated liver fibrosis, with threefold increase in collagen deposition and bridging fibrosis/early signs of cirrhosis at 12 weeks. This was accompanied by early-onset severe portal hypertension and twofold to fourfold increase in profibrogenic transcripts Col1a1 [procollagen α1(I)], Tgfb1, and Timp1. Primary liver cancers in BALB/c.Mdr2(-/-) developed earlier, with greater tumor burden compared to FVB.Mdr2(-/-). BALB/c.Mdr2(-/-) mice have unprecedented degree and rapidity of hepatic fibrosis progression and clinically relevant cirrhosis complications, such as early-onset portal hypertension and primary liver cancers. This new model will facilitate development of antifibrotic drugs and studies into mechanisms of biliary fibrosis progression.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/deficiency , Cholangitis, Sclerosing , Hypertension, Portal , Liver Cirrhosis , Liver Neoplasms , Animals , Cholangitis, Sclerosing/genetics , Cholangitis, Sclerosing/metabolism , Cholangitis, Sclerosing/pathology , Disease Models, Animal , Hypertension, Portal/genetics , Hypertension, Portal/metabolism , Hypertension, Portal/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , ATP-Binding Cassette Sub-Family B Member 4
7.
Circ Res ; 115(2): 238-251, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24874427

ABSTRACT

RATIONALE: Forkhead box-O transcription factors (FoxOs) transduce a wide range of extracellular signals, resulting in changes in cell survival, cell cycle progression, and several cell type-specific responses. FoxO1 is expressed in many cell types, including endothelial cells (ECs). Previous studies have shown that Foxo1 knockout in mice results in embryonic lethality at E11 because of impaired vascular development. In contrast, somatic deletion of Foxo1 is associated with hyperproliferation of ECs. Thus, the precise role of FoxO1 in the endothelium remains enigmatic. OBJECTIVE: To determine the effect of endothelial-specific knockout and overexpression of FoxO1 on vascular homeostasis. METHODS AND RESULTS: We show that EC-specific disruption of Foxo1 in mice phenocopies the full knockout. Although endothelial expression of FoxO1 rescued otherwise Foxo1-null animals, overexpression of constitutively active FoxO1 resulted in increased EC size, occlusion of capillaries, elevated peripheral resistance, heart failure, and death. Knockdown of FoxO1 in ECs resulted in marked inhibition of basal and vascular endothelial growth factor-induced Akt-mammalian target of rapamycin complex 1 (mTORC1) signaling. CONCLUSIONS: Our findings suggest that in mice, endothelial expression of FoxO1 is both necessary and sufficient for embryonic development. Moreover, FoxO1-mediated feedback activation of Akt maintains growth factor responsive Akt/mTORC1 activity within a homeostatic range.


Subject(s)
Endothelial Cells/metabolism , Forkhead Transcription Factors/physiology , Heart Failure/genetics , Multiprotein Complexes/physiology , Neovascularization, Physiologic/physiology , Proto-Oncogene Proteins c-akt/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Enzyme Induction , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Heart Failure/physiopathology , Homeostasis , Human Umbilical Vein Endothelial Cells , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Mice, Transgenic , Neovascularization, Physiologic/genetics , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Organ Specificity , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins , Signal Transduction/physiology , Yolk Sac/blood supply
8.
J Am Soc Nephrol ; 25(4): 717-25, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24335973

ABSTRACT

Soluble fms-like tyrosine kinase 1 (sFlt1), a circulating antiangiogenic protein, is elevated in kidney diseases and contributes to the development of preeclampsia. Hydrogen sulfide is a vasorelaxant and proangiogenic gas with therapeutic potential in several diseases. Therefore, we evaluated the potential therapeutic effect and mechanisms of action of hydrogen sulfide in an animal model of sFlt1-induced hypertension, proteinuria, and glomerular endotheliosis created by adenovirus-mediated overexpression of sFlt1 in Sprague-Dawley rats. We injected sFlt1-overexpressing animals intraperitoneally with the hydrogen sulfide-donor sodium hydrosulfide (NaHS) (50 µmol/kg, twice daily) or vehicle (n=7 per group). Treatment with NaHS for 8 days significantly reduced sFlt1-induced hypertension, proteinuria, and glomerular endotheliosis. Measurement of plasma protein concentrations with ELISA revealed a reduction of free plasma sFlt1 and an increase of free plasma vascular endothelial growth factor (VEGF) after treatment with NaHS. Renal VEGF-A mRNA expression increased significantly with NaHS treatment. In vitro, NaHS was proangiogenic in an endothelial tube assay and attenuated the antiangiogenic effects of sFlt1. Stimulation of podocytes with NaHS resulted in both short-term VEGF release (120 minutes) and upregulation of VEGF-A mRNA levels (24 hours). Furthermore, pretreatment of mesenteric vessels with a VEGF receptor 2-neutralizing antibody significantly attenuated NaHS-induced vasodilation. These results suggest that hydrogen sulfide ameliorates sFlt1-induced hypertension, proteinuria, and glomerular endotheliosis in rats by increasing VEGF expression. Further studies are warranted to evaluate the role of hydrogen sulfide as a novel therapeutic agent for vascular disorders such as preeclampsia.


Subject(s)
Hydrogen Sulfide/pharmacology , Hypertension/drug therapy , Proteinuria/drug therapy , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor Receptor-1/physiology , Animals , Female , Kidney Glomerulus/drug effects , Podocytes/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vasodilation/drug effects
9.
Biomacromolecules ; 14(5): 1618-26, 2013 May 13.
Article in English | MEDLINE | ID: mdl-23590189

ABSTRACT

Oxidative stress is induced by accumulation of hydrogen peroxide (H2O2), and therefore, H2O2 could serve as a potential biomarker of various oxidative stress-associated inflammatory diseases. Vanillin is one of the major components of natural vanilla and has potent antioxidant and anti-inflammatory activities. In this work, we developed a novel inflammation-responsive antioxidant polymeric prodrug of vanillin, termed poly(vanillin oxalate) (PVO). In design, PVO incorporates H2O2-reacting peroxalate ester bonds and bioactive vanillin via acid-responsive acetal linkages in its backbone. Therefore, in cells undergoing damages by oxidative stress, PVO readily degrades into three nontoxic components, one of which is antioxidant and anti-inflammatory vanillin. PVO nanoparticles exhibit potent antioxidant activities by scavenging H2O2 and inhibiting the generation of ROS (reactive oxygen species) and also reduce the expression of pro-inflammatory cytokines in activated macrophages in vitro and in vivo. We, therefore, anticipate that PVO nanoparticles have great potential as novel antioxidant therapeutics and drug delivery systems for ROS-associated inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/chemical synthesis , Antioxidants/chemical synthesis , Benzaldehydes/chemistry , Dioxanes/chemical synthesis , Hydrogen Peroxide/chemistry , Nanoparticles/chemistry , Polyesters/chemical synthesis , Prodrugs/chemical synthesis , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacokinetics , Antioxidants/pharmacology , Biomarkers/chemistry , Dioxanes/pharmacokinetics , Dioxanes/pharmacology , Inflammation/prevention & control , Macrophages/drug effects , Macrophages/metabolism , Mice , Nanoparticles/therapeutic use , Oxidative Stress , Polyesters/pharmacokinetics , Polyesters/pharmacology , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Reactive Oxygen Species/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
10.
J Exp Med ; 203(6): 1447-58, 2006 Jun 12.
Article in English | MEDLINE | ID: mdl-16702604

ABSTRACT

Sepsis, the systemic inflammatory response to infection, is a leading cause of morbidity and mortality. The mechanisms of sepsis pathophysiology remain obscure but are likely to involve a complex interplay between mediators of the inflammatory and coagulation pathways. An improved understanding of these mechanisms should provide an important foundation for developing novel therapies. In this study, we show that sepsis is associated with a time-dependent increase in circulating levels of vascular endothelial growth factor (VEGF) and placental growth factor (PlGF) in animal and human models of sepsis. Adenovirus-mediated overexpression of soluble Flt-1 (sFlt-1) in a mouse model of endotoxemia attenuated the rise in VEGF and PlGF levels and blocked the effect of endotoxemia on cardiac function, vascular permeability, and mortality. Similarly, in a cecal ligation puncture (CLP) model, adenovirus-sFlt-1 protected against cardiac dysfunction and mortality. When administered in a therapeutic regimen beginning 1 h after the onset of endotoxemia or CLP, sFlt peptide resulted in marked improvement in cardiac physiology and survival. Systemic administration of antibodies against the transmembrane receptor Flk-1 but not Flt-1 protected against sepsis mortality. Adenovirus-mediated overexpression of VEGF but not PlGF exacerbated the lipopolysaccharide-mediated toxic effects. Together, these data support a pathophysiological role for VEGF in mediating the sepsis phenotype.


Subject(s)
Sepsis/blood , Vascular Endothelial Growth Factor A/blood , Animals , Cecum/microbiology , Disease Models, Animal , Endotoxemia/blood , Humans , Inflammation/blood , Lipopolysaccharides/toxicity , Mice , Placenta Growth Factor , Pregnancy Proteins/blood , Sepsis/mortality
11.
Basic Res Cardiol ; 106(3): 397-407, 2011 May.
Article in English | MEDLINE | ID: mdl-21387140

ABSTRACT

Heat shock protein 70 (Hsp70) has been shown to have an anti-apoptotic function, but its mechanism is not clear in heart. In this study, we examined the effect of Hsp70 deletion on AIF-induced apoptosis during ischemia/reperfusion (I/R) in vivo. Although Hsp70 KO and WT mice demonstrated similar amounts of AIF released from mitochondria after I/R surgery, Hsp70 KO mice showed a significantly greater increase in apoptosis, larger infarct size, and decreased cardiac output. There was also a significant fourfold increase in the nuclear accumulation of AIF in Hsp70 KO mice compared with WT mice. Treatment with 4-AN (4-amino-1,8-napthalimide, 3 mg/kg), a potent inhibitor of PARP-1, which is a critical regulator of AIF-induced apoptosis, significantly blocked the release of AIF from mitochondria and the translocation of AIF into the nuclei after I/R in both WT and Hsp70 KO mice. In addition, 4-AN treatment resulted in a significant inhibition of apoptosis, a reduction of infarct size, and attenuated cardiac dysfunction in both WT and Hsp70 KO mice after I/R. The anti-apoptotic function of Hsp70 occurs through the inhibition of AIF-induced apoptosis by blocking the mitochondria to nucleus translocation of AIF. PARP-1 inhibition improves cardiac function by blocking AIF-induced apoptosis.


Subject(s)
Apoptosis Inducing Factor/metabolism , Cell Nucleus/metabolism , HSP70 Heat-Shock Proteins/metabolism , Mitochondria/metabolism , Myocardial Reperfusion Injury/metabolism , Animals , Apoptosis/physiology , Enzyme Activation/physiology , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Transport/physiology
12.
J Card Fail ; 17(12): 1051-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22123370

ABSTRACT

BACKGROUND: Activated vitamin D analog, paricalcitol, has been shown to attenuate the development of cardiac hypertrophy in Dahl salt sensitive (DSS) rats. To determine whether an antihypertrophic effect is class specific, we tested if doxercalciferol (a pro-hormone vitamin D2 analog) could also attenuate the development of cardiac hypertrophy in DSS rats. METHODS AND RESULTS: Male DSS rats were fed a high salt (HS) diet for 6 weeks beginning at 6 weeks of age. Doxercalciferol was administered intraperitoneally at 150 ng, 3 times per week (Monday, Wednesday, Friday) for 6 weeks. Pathological and echocardiographic findings demonstrated that rats on HS diet with doxercalciferol administration had significant decrease in cardiac hypertrophy and improved cardiac function compared to the HS + vehicle. In addition, there was a significant decrease in plasma brain natriuretic peptide (BNP) level and tissue atrial natriuretic factor (ANF) mRNA level with doxercalciferol treatment. Doxercalciferol also significantly reduced the level of protein kinase C-α (PKCα) suggesting that PKC-mediated cardiac hypertrophy may be associated with vitamin D deficiency. CONCLUSIONS: Administration of doxercalciferol attenuated the development of HS diet induced cardiac hypertrophy and cardiac dysfunction in DSS rats.


Subject(s)
Ergocalciferols/pharmacology , Heart Failure/drug therapy , Hypertrophy, Left Ventricular/drug therapy , Vitamins/pharmacology , Animals , Atrial Natriuretic Factor , Disease Models, Animal , Ergocalciferols/therapeutic use , Heart Failure/diagnostic imaging , Hypertrophy, Left Ventricular/diagnostic imaging , Male , Natriuretic Peptide, Brain , RNA, Messenger , Rats , Rats, Inbred Dahl , Signal Transduction , Ultrasonography , Vitamins/therapeutic use
13.
J Am Heart Assoc ; 10(20): e021212, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34622671

ABSTRACT

Background Ischemia/reperfusion (I/R) injury causes overproduction of reactive oxygen species, which are the major culprits of oxidative stress that leads to inflammation, apoptosis, myocardial damage, and dysfunction. Bilirubin acts as a potent endogenous antioxidant that is capable of scavenging various reactive oxygen species. We have previously generated bilirubin nanoparticles (BRNPs) consisting of polyethylene glycol-conjugated bilirubin. In this study, we examined the therapeutic effects of BRNPs on myocardial I/R injury in mice. Methods and Results In vivo imaging using fluorophore encapsulated BRNPs showed BRNPs preferentially targeted to the site of I/R injury in the heart. Cardiac I/R surgery was performed by first ligating the left anterior descending coronary artery. After 45 minutes, reperfusion was achieved by releasing the ligation. BRNPs were administered intraperitoneally at 5 minutes before and 24 hours after reperfusion. Mice that received BRNPs showed significant improvements in their cardiac output, assessed by echocardiogram and pressure volume loop measurements, compared with the ones that received vehicle treatment. BRNPs treatment also significantly reduced the myocardial infarct size in mice that underwent cardiac I/R, compared with the vehicle-treatment group. In addition, BRNPs effectively suppressed reactive oxygen species and proinflammatory factor levels, as well as the amount of cardiac apoptosis. Conclusions Taken together, BRNPs could exert their therapeutic effects on cardiac I/R injury through attenuation of oxidative stress, apoptosis, and inflammation, providing a novel therapeutic modality for myocardial I/R injury.


Subject(s)
Myocardial Infarction , Myocardial Reperfusion Injury , Nanoparticles , Animals , Apoptosis , Bilirubin , Inflammation , Mice , Myocardial Reperfusion Injury/prevention & control , Oxidative Stress , Reactive Oxygen Species
14.
J Cardiovasc Pharmacol Ther ; 26(3): 279-288, 2021 05.
Article in English | MEDLINE | ID: mdl-33111565

ABSTRACT

Mortality and morbidity after cardiac arrest remain high due to ischemia/reperfusion (I/R) injury causing multi-organ damages, even after successful return of spontaneous circulation. We previously generated H2O2-activatable antioxidant nanoparticles formulated with copolyoxalate containing vanillyl alcohol (PVAX) to prevent I/R injury. In this study, we examined whether PVAX could effectively reduce organ damages in a rat model of whole-body ischemia/reperfusion injury (WBIR). To induce a cardiac arrest, 70µl/100 g body weight of 1 mmol/l potassium chloride was administered via the jugular venous catheter. The animals in both the vehicle and PVAX-treated groups had similar baseline blood pressure. After 5.5 minutes of cardiac arrest, animals were resuscitated via intravenous epinephrine followed by chest compressions. PVAX or vehicle was injected after the spontaneous recovery of blood pressure was noted, followed by the same dose of second injection 10 minutes later. After 24 hours, multiple organs were harvested for pathological, biochemical, molecular analyses. No significant difference on the restoration of spontaneous circulation was observed between vehicle and PVAX groups. Analysis of organs harvested 24 hours post procedure showed that whole body I/R significantly increased reactive oxygen species (ROS) generation, inflammatory markers, and apoptosis in multiple organs (heart, brain, and kidney). PVAX treatment effectively blocked ROS generation, reduced the elevation of pro-inflammatory cytokines, and decreased apoptosis in these organs. Taken together, our results suggest that PVAX has potent protective effect against WBIR induced multi-organ injury, possibly by blocking ROS-mediated cell damage.


Subject(s)
Antioxidants/pharmacology , Hydrogen Peroxide/pharmacology , Nanoparticles/chemistry , Reperfusion Injury/prevention & control , Animals , Antioxidants/administration & dosage , Benzyl Alcohols/chemistry , Disease Models, Animal , Female , Hydrogen Peroxide/administration & dosage , Inflammation Mediators , Male , Multiple Organ Failure/prevention & control , Nanoparticles/administration & dosage , Polymers/chemistry , Random Allocation , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors
15.
Am J Physiol Heart Circ Physiol ; 299(5): H1374-81, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20833960

ABSTRACT

Although caspase activation is generally thought to be necessary to induce apoptosis, recent evidence suggests that apoptosis can be activated in the setting of caspase inhibition. In this study, we tested the hypothesis that caspase-independent apoptotic pathways contribute to the development of heart failure in the absence of caspase activation. Acute cardiomyopathy was induced using a single dose of doxorubicin (Dox, 20 mg/kg) injected into male wild-type (WT) and transgenic (Tg) mice with a cardiac-specific expression of cytokine response modifier A (CrmA), a known caspase inhibitor. Early (6 day) survival was significantly better in CrmA Tg (81%) than WT (38%) mice. Twelve days after Dox injection, however, the mortality benefit had dissipated, and increased cardiac apoptosis was observed in both groups. There was, however, a significantly greater release of apoptosis-inducing factor (AIF) from mitochondria to cytosol in CrmA Tg compared with WT mice, which suggests that an enhancement of activation in caspase-independent apoptotic pathways had occurred. The administration of a poly(ADP-ribose) polymerase-1 inhibitor, 4-amino-1,8-naphthalimide (4-AN), to Dox-treated mice resulted in significantly improved cardiac function, a significant blockade of AIF released from mitochondria, and decreased cardiac apoptosis. There were also significantly improved survival in WT (18% without 4-AN vs. 89% with 4-AN) and CrmA Tg (13% without 4-AN vs. 93% with 4-AN) mice 12 days after Dox injection. In conclusion, these findings suggest that apoptosis can be induced in the heart lacking caspase activation via caspase-independent pathways and that enabling the inhibition of AIF activation may provide a significant cardiac benefit.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Heart Failure/metabolism , Serpins/metabolism , Viral Proteins/metabolism , 1-Naphthylamine/analogs & derivatives , 1-Naphthylamine/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Inducing Factor/metabolism , Caspase Inhibitors , Disease Models, Animal , Doxorubicin/adverse effects , Heart Failure/chemically induced , Male , Mice , Mice, Transgenic , Mitochondria, Heart/metabolism , Naphthalimides/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Quinolones/pharmacology , Serpins/genetics , Viral Proteins/genetics
16.
Can J Physiol Pharmacol ; 88(5): 568-75, 2010 May.
Article in English | MEDLINE | ID: mdl-20555426

ABSTRACT

Exposure to tobacco smoke is known to have deleterious cardiovascular effects. In this study, we tested whether exposure to tobacco smoke exacerbates the severity of viral myocarditis in mice. Viral myocarditis was generated in 4-week-old male BALB/c mice by injection of Encephalomyocarditis virus (EMCV). Four groups were studied: (1) control (C, no smoke and no virus); (2) smoke only (S, exposure to cigarette smoke for 90 min/day for 15 days); (3) virus only (V); and (4) exposure to smoke for 5 days before plus 10 days following virus injection (S+V). We found that viral inoculation preceded by smoke exposure increased mortality more than twofold compared with virus inoculation alone. In addition, the mRNA level of atrial natriuretic factor was significantly higher in S+V than among any of the other 3 groups. Virus injection significantly decreased cardiac function compared with controls, with further deterioration observed in the S+V group. We also observed a significantly increased rate of apoptosis, with an increased activation of apoptosis-inducing factor in hearts exposed to S+V compared with those exposed to V alone. Our results suggest that preexposure to smoke significantly exacerbates the severity of viral myocarditis, likely through increased viral load and increased cardiomyocyte cell death.


Subject(s)
Cardiovirus Infections/virology , Heart/virology , Myocarditis/virology , Tobacco Smoke Pollution/adverse effects , Viral Load , Animals , Apoptosis , Apoptosis Inducing Factor/biosynthesis , Atrial Natriuretic Factor/biosynthesis , Blotting, Western , Cardiovirus Infections/metabolism , Cardiovirus Infections/pathology , Cardiovirus Infections/physiopathology , Encephalomyocarditis virus , Hemodynamics/physiology , Male , Mice , Mice, Inbred BALB C , Myocarditis/metabolism , Myocarditis/pathology , Myocarditis/physiopathology , Myocardium/metabolism , Myocardium/pathology , Reverse Transcriptase Polymerase Chain Reaction
17.
Antioxidants (Basel) ; 9(12)2020 Dec 17.
Article in English | MEDLINE | ID: mdl-33348578

ABSTRACT

Oxidative stress plays a key role in many physiological and pathological conditions. The intracellular oxidative homeostasis is tightly regulated by the reactive oxygen species production and the intracellular defense mechanisms. Increased oxidative stress could alter lipid, DNA, and protein, resulting in cellular inflammation and programmed cell death. Evidences show that oxidative stress plays an important role in the progression of various cardiovascular diseases, such as atherosclerosis, heart failure, cardiac arrhythmia, and ischemia-reperfusion injury. There are a number of therapeutic options to treat oxidative stress-associated cardiovascular diseases. Well known antioxidants, such as nutritional supplements, as well as more novel antioxidants have been studied. In addition, novel therapeutic strategies using miRNA and nanomedicine are also being developed to treat various cardiovascular diseases. In this article, we provide a detailed description of oxidative stress. Then, we will introduce the relationship between oxidative stress and several cardiovascular diseases. Finally, we will focus on the clinical implications of oxidative stress in cardiovascular diseases.

18.
Nanomaterials (Basel) ; 10(4)2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32244653

ABSTRACT

BACKGROUND: Nanomedicine is a field of science that uses nanoscale materials for the diagnosis and treatment of human disease. It has emerged as an important aspect of the therapeutics, but at the same time, also raises concerns regarding the safety of the nanomaterials involved. Recent applications of functionalized biodegradable nanomaterials have significantly improved the safety profile of nanomedicine. OBJECTIVE: Our goal is to evaluate different types of biodegradable nanomaterials that have been functionalized for their biomedical applications. METHOD: In this review, we used PubMed as our literature source and selected recently published studies on biodegradable nanomaterials and their applications in nanomedicine. RESULTS: We found that biodegradable polymers are commonly functionalized for various purposes. Their property of being naturally degraded under biological conditions allows these biodegradable nanomaterials to be used for many biomedical purposes, including bio-imaging, targeted drug delivery, implantation and tissue engineering. The degradability of these nanoparticles can be utilized to control cargo release, by allowing efficient degradation of the nanomaterials at the target site while maintaining nanoparticle integrity at off-target sites. CONCLUSION: While each biodegradable nanomaterial has its advantages and disadvantages, with careful design and functionalization, biodegradable nanoparticles hold great future in nanomedicine.

19.
Eur J Pharmacol ; 882: 173261, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32534073

ABSTRACT

Neuropeptide-Y (NPY) leads to angiogenesis and remodeling of the ischemic myocardium. The objective of this study is to assess the therapeutic potential of NPY in a model of acute myocardial ischemia using a nanoparticles delivery system targeted to tissue with oxidative stress. NPY3-36 was loaded onto copolyoxalate containing vanillyl alcohol (PVAX) using a double emulsification strategy. Adult C57BL/J6 mice (n = 49) were randomly divided into PVAX-NPY3-36 (n = 22), Vehicle (Saline) (n = 16), and Sham (n = 11) groups. The ischemia to left anterior descending artery was induced in PVAX-NPY3-36 or vehicle groups. The tissue was collected at the end of two weeks after assessing the functional and echocardiographic data. There was a significant decrease in infarction size and mortality in PVAX-NPY3-36 group compared to the Vehicle group (P = 0.01 and P = 0.05). On echocardiography, there was significant improvement in contractility and diastolic parameters (P = 0.01). On pressure-volume loop there was significant increase in stroke volume (P = 0.01), cardiac output (P = 0.01) and ventricular stroke work (P = 0.01) in the PVAX-NPY3-36 group. On Western blot analysis, there was a significant increase in pro-angiogenic factors Ang-1, TGF-ß, PDGF- ß and its receptors and VEGF in the ischemic tissue treated with PVAX-NPY3-36 as compared to Vehicle ischemic tissue (P = 0.01, P = 0.0003, and P < 0.05 respectively). It may be possible to have targeted delivery of labile neurotransmitters NPY3-36 to the ischemic myocardium using nanoparticle PVAX and achieving angiogenesis and significant functional improvement.


Subject(s)
Benzyl Alcohols/administration & dosage , Cardiotonic Agents/administration & dosage , Myocardial Ischemia/drug therapy , Nanoparticles/administration & dosage , Neovascularization, Physiologic/drug effects , Neuropeptide Y/administration & dosage , Oxalates/administration & dosage , Peptide Fragments/administration & dosage , Polymers/administration & dosage , Animals , Disease Models, Animal , Hemodynamics/drug effects , Male , Mice, Inbred C57BL , Myocardial Contraction/drug effects , Myocardial Ischemia/physiopathology , Ventricular Function, Left/drug effects
20.
Circulation ; 117(11): 1423-35, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18316486

ABSTRACT

BACKGROUND: Heart failure is the leading cause of death in the United States. By delineating the pathways that regulate cardiomyocyte function, we can better understand the pathogenesis of cardiac disease. Many cardiomyocyte signaling pathways activate protein tyrosine kinases. However, the role of specific protein tyrosine phosphatases (PTPs) in these pathways is unknown. METHODS AND RESULTS: Here, we show that mice with muscle-specific deletion of Ptpn11, the gene encoding the SH2 domain-containing PTP Shp2, rapidly develop a compensated dilated cardiomyopathy without an intervening hypertrophic phase, with signs of cardiac dysfunction appearing by the second postnatal month. Shp2-deficient primary cardiomyocytes are defective in extracellular signal-regulated kinase/mitogen-activated protein kinase (Erk/MAPK) activation in response to a variety of soluble agonists and pressure overload but show hyperactivation of the RhoA signaling pathway. Treatment of primary cardiomyocytes with Erk1/2- and RhoA pathway-specific inhibitors suggests that both abnormal Erk/MAPK and RhoA activities contribute to the dilated phenotype of Shp2-deficient hearts. CONCLUSIONS: Our results identify Shp2 as the first PTP with a critical role in adult cardiac function, indicate that in the absence of Shp2 cardiac hypertrophy does not occur in response to pressure overload, and demonstrate that the cardioprotective role of Shp2 is mediated via control of both the Erk/MAPK and RhoA signaling pathways.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Myocytes, Cardiac/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency , Signal Transduction/physiology , rho GTP-Binding Proteins/physiology , Animals , Cardiomegaly/enzymology , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/physiopathology , LEOPARD Syndrome/enzymology , LEOPARD Syndrome/genetics , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Models, Animal , Noonan Syndrome/enzymology , Noonan Syndrome/genetics , Organ Specificity , Phenotype , Pressure , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , rho GTP-Binding Proteins/antagonists & inhibitors , rhoA GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL