Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Heart Vessels ; 38(4): 606-613, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36522555

ABSTRACT

Ischemic preconditioning (IPC) describes a phenomenon wherein brief ischemia of the heart induces a potent cardioprotective mechanism against succeeding ischemic insult. Cyclooxygenase-2 (COX-2), a rate-limiting enzyme in prostanoid biosynthesis, is upregulated in the ischemic heart and contributes to IPC. Prostaglandin E2 (PGE2) protects the heart from ischemia-reperfusion (I/R) injury via its receptor subtype EP4. We sought to clarify the role of the PGE2/EP4 system in the late phase of IPC. Mice were subjected to four IPC treatment cycles, consisting of 5 min of occlusion of the left anterior descending coronary artery (LAD). We found that COX-2 mRNA was significantly upregulated in wild-type hearts at 6 h after IPC treatment. Cardiac PGE2 levels at 24 h after IPC treatment were significantly increased in both wild-type mice and mice lacking EP4 (EP4-/-). At 24 h after IPC treatment, I/R injury was induced by 30 min of LAD occlusion followed by 2 h of reperfusion and the cardiac infarct size was determined. The infarct size was significantly reduced by IPC treatment in wild-type mice; a reduction was not observed in EP4-/- mice. AE1-329, an EP4 agonist, significantly reduced infarct size and significantly ameliorated deterioration of cardiac function in wild-type mice subjected to I/R without IPC treatment. Furthermore, AE1-329 significantly enhanced the I/R-induced activation of Akt, a pro-survival kinase. We demonstrated that the PGE2/EP4 system in the heart plays a critical role in the late phase of IPC, partly by augmenting Akt-mediated signaling. These findings clarify the mechanism of IPC and may contribute to the development of therapeutic strategies for ischemic heart disease.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocardial Infarction , Myocardial Reperfusion Injury , Mice , Animals , Proto-Oncogene Proteins c-akt/therapeutic use , Cyclooxygenase 2 , Prostaglandins/therapeutic use
3.
Sci Rep ; 11(1): 19163, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34580402

ABSTRACT

Sarcopenia is a pathophysiological malfunction induced by skeletal muscle atrophy. Several studies reported an association between sarcopenia-induced cardiac cachexia and poor prognosis in heart disease. However, due to lack of an established animal models, the underlying mechanism of disturbed cardiac repair accompanied with sarcopenia remains poorly understood. Here, we developed a novel sarcopenia-induced cardiac repair disturbance mouse model induced by tail suspension (TS) after cardiac ischemia and reperfusion (I/R). Importantly, we identified a specific exosomal-microRNA marker, miR-16-5p, in the circulating exosomes of I/R-TS mice. Of note, sarcopenia after I/R disturbed cardiac repair and raised the level of circulating-exosomal-miR-16-5p secreting from both the atrophic limbs and heart of TS mice. Likewise, miR-16-5p mimic plasmid disturbed cardiac repair in I/R mice directly. Additionally, in neonatal rat ventricular myocytes (NRVMs) cultured in vitro under hypoxic conditions in the presence of a miR-16-5p mimic, we observed increased apoptosis through p53 and Caspase3 upregulation, and also clarified that autophagosomes were decreased in NRVMs via SESN1 transcript interference-mediated mTOR activation. In conclusion, we show the pro-apoptotic effect of sarcopenia-derived miR-16-5p, which may be behind the exacerbation of myocardial infarction. Therefore, miR-16-5p can be a novel therapeutic target in the context of cardiac repair disturbances in sarcopenia-cachexia.


Subject(s)
Exosomes/genetics , MicroRNAs/genetics , Myocardial Infarction/physiopathology , Sarcopenia , Animals , Apoptosis , Disease Models, Animal , Hindlimb Suspension , Male , Mice, Inbred C57BL , Regeneration/genetics , Regeneration/physiology
4.
Am J Physiol Heart Circ Physiol ; 320(6): H2438-H2447, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33961504

ABSTRACT

Adventitial abnormalities including enhanced vasa vasorum malformation are associated with development and vulnerability of atherosclerotic plaque. However, the mechanisms of vasa vasorum malformation and its role in vascular remodeling have not been fully clarified. We recently reported that ninjurin-1 (Ninj1) is a crucial adhesion molecule for pericytes to form matured neovessels. The purpose is to examine if Ninj1 regulates adventitial angiogenesis and affects the vascular remodeling of injured vessels using pericyte-specific Ninj1 deletion mouse model. Mouse femoral arteries were injured by insertion of coiled wire. Four weeks after vascular injury, fixed arteries were decolorized. Vascular remodeling, including intimal hyperplasia and adventitial microvessel formation were estimated in a three-dimensional view. Vascular fragility, including blood leakiness was estimated by extravasation of fluorescein isothiocyanate (FITC)-lectin or FITC-dextran from microvessels. Ninj1 expression was increased in pericytes in response to vascular injury. NG2-CreER/Ninj1loxp mice were treated with tamoxifen (Tam) to induce deletion of Ninj1 in pericyte (Ninj1 KO). Tam-treated NG2-CreER or Tam-nontreated NG2-CreER/Ninj1loxp mice were used as controls. Intimal hyperplasia was significantly enhanced in Ninj1 KO compared with controls. Vascular leakiness was significantly enhanced in Ninj1 KO. In Ninj1 KO, the number of infiltrated macrophages in adventitia was increased, along with the expression of inflammatory cytokines. In conclusion, deletion of Ninj1 in pericytes induces the immature vasa vasorum formation of injured vasculature and exacerbates adventitial inflammation and intimal hyperplasia. Thus, Ninj1 contributes to the vasa vasorum maturation in response to vascular injury and to reduction of vascular remodeling.NEW & NOTEWORTHY Although abnormalities of adventitial vasa vasorum are associated with vascular remodeling such as atherosclerosis, the mechanisms of vasa vasorum malformation and its role in vascular remodeling have not been fully clarified. The present study provides a line of novel evidence that ninjurin-1 contributes to adventitial microvascular maturation during vascular injury and regulates vascular remodeling.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Femoral Artery/metabolism , Neointima/genetics , Nerve Growth Factors/genetics , Pericytes/metabolism , Vasa Vasorum/metabolism , Vascular Remodeling/genetics , Adventitia/metabolism , Adventitia/pathology , Animals , Femoral Artery/injuries , Femoral Artery/pathology , Gene Knockout Techniques , Hyperplasia/genetics , Inflammation/genetics , Inflammation/metabolism , Macrophages/pathology , Mice , Neointima/pathology , Neovascularization, Physiologic/genetics , Transcriptome , Tunica Intima/metabolism , Tunica Intima/pathology , Vasa Vasorum/pathology , Vascular System Injuries/genetics , Vascular System Injuries/metabolism , Vascular System Injuries/pathology
5.
Stem Cell Res ; 47: 101914, 2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32738632

ABSTRACT

Skeletal muscle has a capacity for muscular regeneration mediated by satellite cells (SCs) and non-SCs. Although it is proposed that non-SCs are attractive therapeutic targets for dystrophies, the biological properties of these cells remain unclear. We have recently identified novel multipotent pericytes (PCs), capillary stem cells (CapSCs) derived from the microvasculature. In the present study, we determined if CapSCs contributed to myogenic regeneration using muscular dystrophy mouse model. CapSCs were isolated as EphA7+NG2+PCs from the subcutaneous adipose tissues of GFP-transgenic mice. Co-culture with C2C12 myoblast cells showed that CapSCs effectively enhanced myogenesis as compared to controls including EphA7- PCs and adipose stromal cells (ASCs). CapSCs transplanted in cardiotoxin-injured gastrocnemius muscles were well differentiated into both muscle fibers and microvessels, as compared to controls. At three weeks after cell-transplantation into the limbs of the mdx/utrn-/-mouse, CapSCs increased the number of GFP+myofibers along with dystrophin expression and the area size of myofibers, and also enhanced the muscular mass and its performance, assessed by treadmill test as compared to controls. In conclusion, CapSCs have potent myogenic regeneration capacity and improved the pathological condition in a muscular dystrophy mouse. Thus, CapSCs are an attractive cellular source in regenerative therapy for muscular dystrophy.

6.
Stem Cells Transl Med ; 9(1): 120-130, 2020 01.
Article in English | MEDLINE | ID: mdl-31471947

ABSTRACT

The presence of pericytes (PCs) with multipotency and broad distribution along capillary suggests that microvasculature plays a role not only as a duct for blood fluid transport but also as a stem cell niche that contributes to tissue maintenance and regeneration. The lack of an appropriate marker for multipotent PCs still limits our understanding of their pathophysiological roles. We identified the novel marker EphA7 to detect multipotent PCs using microarray analysis of an immortalized PC library. PCs were isolated from microvessels of mouse subcutaneous adipose tissues, then EphA7+ PCs called capillary stem cells (CapSCs) were separated from EphA7- control PCs (ctPCs) using fluorescence-activated cell sorting system. CapSCs had highly multipotency that enabled them to differentiate into mesenchymal and neuronal lineages compared with ctPCs. CapSCs also differentiated into endothelial cells and PCs to form capillary-like structures by themselves. Transplantation of CapSCs into ischemic tissues significantly improved blood flow recovery in hind limb ischemia mouse model due to vascular formation compared with that of ctPCs and adipose stromal cells. These data demonstrate that EphA7 identifies a subpopulation of multipotent PCs that have high angiogenesis and regenerative potency and are an attractive target for regenerative therapies.


Subject(s)
Capillaries/metabolism , Ischemia/immunology , Multipotent Stem Cells/metabolism , Pericytes/metabolism , Receptor, EphA7/metabolism , Animals , Cell Differentiation , Humans , Mice
7.
Biochem Biophys Res Commun ; 519(3): 462-468, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31526566

ABSTRACT

Ninjurin 1 (Ninj1) is identified as a peripheral nerve injury-induced protein. However, the role of Ninj1 in nerve regeneration is unclear. Schwann cells (SCs) and microvasculature are critical for peripheral nerve regeneration. SCs precursors and microvascular pericytes (PCs), which are nerve/glial antigen 2 (NG2)-positive cells are observed in peripheral nervous system. In this study, we investigated the role of Ninj1 in peripheral nerve regeneration using NG2+cell-specific inducible deletion of Ninj1 mouse model. The number of NG2+cells, which were associated with and without microvessels was increased after sciatic nerve crush injury. There was a significant increase in the expression of Ninj1 and EphA7 in the injured nerve tissue. This increase was mostly observed in NG2+cells. Genetic tracing of NG2+cells was performed using tamoxifen (Tam) treatment on NG2CreERT:R26R-tdTomato mice. The sciatic nerve was injured following the Tam-treatment, then tdTomato-expressing SCs were mostly observed in regenerated SCs at 21 days after nerve injury. Ninj1 gene knockout (Ninj1 KO) in NG2+cells was induced using NG2CreERT:Ninj1loxp mice. Tam-treated-NG2CreERT or Tam-nontreated NG2CreERT:Ninj1loxp mice were used as controls. Following Tam-treatment, the sciatic nerve in each group was injured. Ninj1KO significantly attenuated the expression of the myelin binding protein (MBP) as well as the number of myelinated axons. The expression of MBP in cultured SCs was significantly reduced by SiRNA-mediated Ninj1 knockdown (KD). Ninj1KD also attenuated the differentiation of SCs by isolated EphA7+multipotent PCs. The current data indicate that Ninj1 plays a vital role in peripheral nerve regeneration. This is observed particularly in the myelination process of NG2+cells including SCs precursors and multipotent PCs.


Subject(s)
Antigens/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Nerve Growth Factors/metabolism , Nerve Regeneration/physiology , Peripheral Nerve Injuries/physiopathology , Proteoglycans/metabolism , Schwann Cells/metabolism , Animals , Antigens/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cell Line , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelin Proteins/genetics , Myelin Proteins/metabolism , Nerve Growth Factors/genetics , Pericytes/cytology , Pericytes/metabolism , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/metabolism , Proteoglycans/genetics
8.
Sci Rep ; 9(1): 7823, 2019 05 24.
Article in English | MEDLINE | ID: mdl-31127150

ABSTRACT

Apurinic/apyrimidinic endonuclease 1 (APE1) is a multifunctional protein that controls the cellular response to oxidative stress and possesses DNA-repair functions. It has important roles in the progression and outcomes of various diseases; however, its function and therapeutic prospects with respect to kidney injury are unknown. To study this, we activated APE1 during kidney injury by constructing an expression vector (pCAG-APE1), using an EGFP expression plasmid (pCAG-EGFP) as a control. We performed unilateral ureteral obstruction (UUO) as a model of tubulointerstitial fibrosis on ICR mice before each vector was administrated via retrograde renal vein injection. In this model, pCAG-APE1 injection did not produce any adverse effects and significantly reduced histological end points including fibrosis, inflammation, tubular injury, and oxidative stress, as compared to those parameters after pCAG-EGFP injection. qPCR analysis showed significantly lower expression of Casp3 and inflammation-related genes in pCAG-APE1-injected animals compared to those in pCAG-EGFP-injected UUO kidneys. RNA-Seq analyses showed that the major transcriptional changes in pCAG-APE1-injected UUO kidneys were related to immune system processes, metabolic processes, catalytic activity, and apoptosis, leading to normal kidney repair. Therefore, APE1 suppressed renal fibrosis, not only via antioxidant and DNA-repair functions, but also partly by modulating the immune system through multiple pathways including Il6, Tnf, and chemokine families. Thus, therapeutic APE1 modulation might be beneficial for the treatment of renal diseases.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/therapeutic use , Genetic Vectors/administration & dosage , Immunity/genetics , Kidney Tubules/pathology , Nephritis, Interstitial/therapy , Animals , Computational Biology , DNA Repair/immunology , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Disease Models, Animal , Fibrosis , Genetic Vectors/genetics , Humans , Injections, Intravenous , Kidney Tubules/immunology , Male , Mice , Nephritis, Interstitial/genetics , Nephritis, Interstitial/immunology , Nephritis, Interstitial/pathology , Oxidative Stress/genetics , Oxidative Stress/immunology , Plasmids/administration & dosage , Plasmids/genetics , RNA-Seq , Renal Veins
9.
Arterioscler Thromb Vasc Biol ; 38(10): 2358-2370, 2018 10.
Article in English | MEDLINE | ID: mdl-30354207

ABSTRACT

Objective- Angiogenesis, entire step from endothelial cells (ECs) sprouts to vascular maturation, is a critical response to ischemia. To form functional mature vessels, interactions between ECs and pericytes are essential. Ninj1 (ninjurin1) is an adhesion molecule that contributes to the pathogenesis of neuroinflammation. We recently demonstrated that Ninj1 is expressed in pericytes during angiogenesis. However, the role of Ninj1 in angiogenesis under pathophysiological ischemic conditions has not yet been elucidated. Approach and Results- Ninj1 was detected in microvessels, and its expression was enhanced in ischemic tissues after mouse hindlimb ischemia. Knockdown of Ninj1 was performed by injection of biodegradable microspheres releasing Ninj1-small interfering RNA into muscle tissues. Alternatively, pericyte-specific Ninj1 knockout was induced by tamoxifen treatment of NG2-CreERT/Ninj1-flox mice. Ninj1 knockdown/knockout reduced the formation of blood-circulating functional vessels among total CD31+ microvessels within ischemic tissues and subsequently attenuated color Doppler-assessed blood flow recovery. Ninj1 overexpression enhanced expression of Anpt (angiopoietin) 1, whereas Ninj1 knockdown enhanced the endogenous Anpt1 antagonist, Anpt2 expression in pericytes and inhibited the association of pericytes with ECs and subsequent formation of capillary-like structure, that is, EC tube surrounded with pericytes in 3-dimensional gel culture. Conclusions- Our data demonstrate that Ninj1 is involved in the formation of functional matured vessels through the association between pericytes and ECs, resulting in blood flow recovery from ischemia. These findings further the current our understanding of vascular maturation and may support the development of therapeutics for ischemic diseases.


Subject(s)
Cell Adhesion Molecules, Neuronal/deficiency , Endothelial Cells/metabolism , Gene Deletion , Ischemia/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Nerve Growth Factors/deficiency , Pericytes/metabolism , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Communication , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Gene Knockdown Techniques , Hindlimb , Ischemia/genetics , Ischemia/physiopathology , Male , Mice, Inbred C57BL , Nerve Growth Factors/genetics , Recovery of Function , Regional Blood Flow , Signal Transduction
11.
Stem Cells Transl Med ; 5(8): 1067-78, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27334489

ABSTRACT

UNLABELLED: : Overcoming the insufficient survival of cell grafts is an essential objective in cell-based therapy. Apurinic/apyrimidinic endonuclease/redox factor 1 (APE1) promotes cell survival and may enhance the therapeutic effect of engrafted cells. The aim of this study is to determine whether APE1 overexpression in cardiac progenitor cells (CPCs) could ameliorate the efficiency of cell-based therapy. CPCs isolated from 8- to 10-week-old C57BL/6 mouse hearts were infected with retrovirus harboring APE1-DsRed (APE1-CPC) or a DsRed control (control-CPC). Oxidative stress-induced apoptosis was then assessed in APE1-CPCs, control-CPCs, and neonatal rat ventricular myocytes (NRVMs) cocultured with these CPCs. This analysis revealed that APE1 overexpression inhibited CPC apoptosis with activation of transforming growth factor ß-activated kinase 1 (TAK1) and nuclear factor (NF)-κB. In the coculture model, NRVM apoptosis was inhibited to a greater extent in the presence of APE1-CPCs compared with control-CPCs. Moreover, the number of surviving DsRed-positive CPC grafts was significantly higher 7 days after the transplant of APE1-CPCs into a mouse myocardial infarction model, and the left ventricular ejection fraction showed greater improvement with attenuation of fibrosis 28 days after the transplant of APE1-CPCs compared with control-CPCs. Additionally, fewer inflammatory macrophages and a higher percentage of cardiac α-sarcomeric actinin-positive CPC-grafts were observed in mice injected with APE1-CPCs compared with control-CPCs after 7 days. In conclusion, antiapoptotic APE1-CPC graft, which increased TAK1-NF-κB pathway activation, survived effectively in the ischemic heart, restored cardiac function, and reduced cardiac inflammation and fibrosis. APE1 overexpression in CPCs may serve as a novel strategy to improve cardiac cell therapy. SIGNIFICANCE: Improving the survival of cell grafts is essential to maximize the efficacy of cell therapy. The authors investigated the role of APE1 in CPCs under ischemic conditions and evaluated the therapeutic efficacy of transplanted APE1-overexpressing CPCs in a mouse model of myocardial infarction. APE1 hindered apoptosis in CPC grafts subjected to oxidative stress caused in part by increased TAK1-NF-κB pathway activation. Furthermore, APE1-CPC grafts that effectively survived in the ischemic heart restored cardiac function and attenuated fibrosis through pleiotropic mechanisms that remain to be characterized. These findings suggest that APE1 overexpression in CPCs may be a novel strategy to reinforce cardiac cell therapy.


Subject(s)
Apoptosis , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Genetic Therapy/methods , Myocardial Infarction/surgery , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/transplantation , Regeneration , Stem Cell Transplantation/methods , Stem Cells/enzymology , Animals , Cell Differentiation , Cell Survival , Cells, Cultured , Coculture Techniques , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Disease Models, Animal , Enzyme Induction , Fibrosis , Graft Survival , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Oxidative Stress , Phenotype , Recovery of Function , Signal Transduction , Stem Cells/pathology , Stroke Volume , Time Factors , Transfection , Ventricular Function, Left , Ventricular Remodeling
12.
Hypertens Res ; 39(9): 633-9, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27169395

ABSTRACT

Although malnutrition indicates an unfavorable prognosis in some clinical settings, the synergistic impact of nutritional state, renal dysfunction and left ventricular hypertrophy (LVH) on cardiovascular events is unknown. Among 338 patients aged 40-80 years who underwent echocardiographic evaluation between 2003 and 2005, 161 patients who were followed for >7 years were recruited. Malnutrition was defined as a geriatric nutritional risk index (GNRI) of ⩽96. The mean patient age was 63.5±9.2 years; the mean estimated glomerular filtration rate (eGFR) was 72.9±18.7 ml min(-1) per 1.73 m(2); the mean LV mass index was 114±33 g m(-)(2); and the mean GNRI was 100.4±6.0. Among the patients, 25% (n=40) had an eGFR of <60 ml min(-1) per 1.73 m(2), 29% (n=46) exhibited chronic kidney disease (CKD) and 37% (n=59) had LVH. During the follow-up period (median: 96 months), cardiovascular events were observed in 15 patients (9%). Kaplan-Meier curves showed a significantly higher incidence of cardiovascular events in patients with an eGFR of <60 ml min(-1) per 1.73 m(2) (log-rank P=0.007), a GNRI of ⩽96 (P=0.003) or LVH (P=0.010). In a Cox regression analysis, eGFR, LVH and GNRI were independent determinants of cardiovascular event incidence after adjusting for age, gender and the presence of hypertension and diabetes. Furthermore, the combination of LVH and lower GNRI was significantly associated with a higher rate of cardiovascular events not only in all patients but also in patients with CKD. In conclusion, malnutrition, low eGFR and LVH were independent determinants of cardiovascular event incidence; they synergistically increased rates of these events in the long term. The evaluation and management of LVH progression and the improvement of nutritional status are critical for preventing cardiovascular complications even in non-dialysis patients.


Subject(s)
Acute Coronary Syndrome/epidemiology , Aortic Diseases/epidemiology , Heart Failure/epidemiology , Hypertrophy, Left Ventricular/complications , Kidney Diseases/complications , Malnutrition/complications , Stroke/epidemiology , Acute Coronary Syndrome/diagnostic imaging , Acute Coronary Syndrome/etiology , Adult , Aged , Aged, 80 and over , Aortic Diseases/diagnostic imaging , Aortic Diseases/etiology , Disease Progression , Echocardiography , Female , Glomerular Filtration Rate/physiology , Heart Failure/diagnostic imaging , Heart Failure/etiology , Humans , Hypertrophy, Left Ventricular/diagnostic imaging , Incidence , Kidney Diseases/diagnostic imaging , Male , Malnutrition/diagnostic imaging , Middle Aged , Prognosis , Risk , Stroke/diagnostic imaging , Stroke/etiology
13.
PLoS One ; 10(4): e0123733, 2015.
Article in English | MEDLINE | ID: mdl-25874702

ABSTRACT

OBJECTIVE: Relationship between microalbuminuria and worse outcome of coronary artery disease patients is discussed, but its underlying pathophysiological mechanism remains unclear. We investigated the role of microalbuminuria to the function of endothelial progenitor cells (EPCs), that might affect to outcome of acute myocardial infarction (AMI) patients. METHODS: Forty-five AMI patients were divided into two groups according to their urinary albumin excretion: normal (n = 24) and microalbuminuria (>30 mg/day, n = 21). At day-2 and day-7 after AMI onset, circulating-EPCs (CD34+ Flk1+) were quantified by flow cytometry. The number of lectin-acLDL-positive cultured-EPCs immobilized on fibronectin was determined. To assess the cellular senescence of cultured-EPCs, the expression level of sirtuin-1 mRNA and the number of SA-ß-gal positive cell were evaluated. Angiographic late in-stent loss after percutaneous coronary intervention (PCI) was evaluated at a six-month follow-up. RESULTS: No significant differences in coronary risk and the extent of myocardial damage were observed between the two groups. Late in-stent loss at the six-month follow-up was significantly higher in the microalbuminuria group (normal:microalbuminuria = 0.76±0.34:1.18±0.57 mm, p=0.021). The number of circulating-EPCs was significantly increased in microalbuminuria group at day-7, however, improved adhesion of EPCs was observed in normal group but not in microalbuminuria group from baseline to day-7 (+3.1±8.3:-1.3±4.4%: p<0.05). On the other hand, in microalbuminuria group at day-7, the level of sirtuin-1 mRNA expression of cultured-EPCs was significantly decreased (7.1±8.9:2.5±3.7 fold, p<0.05), which was based on the negative correlation between the level of sirtuin-1 mRNA expression and the extent of microalbuminuria. The ratio of SA-ß-gal-positive cells in microalbuminuria group was increased compared to that of normal group. CONCLUSIONS: Microalbuminuria in AMI patients is closely associated with functional disorder of EPCs via cellular senescence, that predicts the aggravation of coronary remodeling after PCI.


Subject(s)
Albuminuria/complications , Albuminuria/diagnosis , Cellular Senescence , Coronary Restenosis/complications , Coronary Restenosis/diagnosis , Endothelial Progenitor Cells/cytology , Myocardial Infarction/surgery , Aged , Aged, 80 and over , Angiography , Antigens, CD34/metabolism , Female , Fibronectins/metabolism , Flow Cytometry , Gene Expression Profiling , Glomerular Filtration Rate , Humans , Lectins/metabolism , Male , Middle Aged , Myocardial Infarction/complications , Risk Factors , Sirtuin 1/metabolism , Stents , Treatment Outcome
14.
Clin Exp Hypertens ; 37(5): 411-9, 2015.
Article in English | MEDLINE | ID: mdl-25856785

ABSTRACT

BACKGROUND: Combination antihypertensive therapy with an angiotensin receptor blocker (ARB) and a calcium channel blocker (CCB) or diuretics is common. This subanalysis investigated blood pressure (BP) variability in patients receiving ARB-based combination therapy. METHODS: In a prospective, randomized, open-label trial, hypertensive outpatients (≥65 years) who did not achieve their target BP with ARB monotherapy switched to losartan 50 mg/hydrochlorothiazide 12.5 mg (ARB + D) or ARB plus amlodipine 5 mg (ARB + C) for 12 months. Clinic BP and heart rate (HR), measured every 3 months, visit-to-visit variability and seasonal variation were evaluated. RESULTS: No significant between-group differences in average, maximum, or minimum systolic or diastolic BP, or HR, were found. Visit-to-visit BP variability (systolic) was significantly higher in the ARB + D group than in the ARB + C group. When each group was subdivided into two seasonal groups (summer and winter), no significant between-group differences in BP were found. Multivariate regression analyses showed a tendency toward negative correlation between outdoor temperature and urinary albumin:creatinine ratio and estimated glomerular filtration rate at 12 months in the ARB + D group. CONCLUSION: Combination therapy with an ARB plus a CCB may be preferable to that with an ARB plus diuretics for decreasing BP variability. As for seasonal variability, both treatments can be used safely regardless of season.


Subject(s)
Antihypertensive Agents/administration & dosage , Blood Pressure/physiology , Hypertension/physiopathology , Seasons , Aged , Amlodipine/administration & dosage , Blood Pressure/drug effects , Drug Therapy, Combination , Female , Humans , Hydrochlorothiazide/administration & dosage , Hypertension/drug therapy , Losartan/administration & dosage , Male , Prospective Studies , Treatment Outcome
15.
Circ J ; 79(6): 1363-71, 2015.
Article in English | MEDLINE | ID: mdl-25766274

ABSTRACT

BACKGROUND: Capillary pericytes (cPCs), the mural cells of microvessels, play an important role in the formation and maintenance of microvessels; however, little is known about the mechanisms of how cPCs regulate angiogenesis. To identify factors that modulate cPC function, genes whose levels were altered in cPCs during neovessel formation were identified through a microarray screen. METHODS AND RESULTS: Ninjurin1 (nerve injury-induced protein, Ninj1) was selected as a candidate factor for angiogenesis regulation. Ninj1 was expressed in capillary cells including endothelial cells (cECs) and was expressed at a higher level in cPCs. Hypoxia induced the gene expression of Ninj1 in addition of vascular endothelial growth factor (VEGF) in cPCs. When cPCs were co-incubated with a thoracic aorta in a three-dimensional Matrigel system, the length of the EC-tubes sprouting from the aorta was increased. Small interfering RNA-mediated downregulation of Ninj1 in cPCs enhanced these cPCs-mediated angiogenic effects, whereas overexpression of Ninj1 attenuated their effects. The production of angiogenic growth factors, such as VEGF and angiopoietin 1, by cPCs was enhanced by the downregulation of Ninj1, and reduced by the overexpression of Ninj1. CONCLUSIONS: Ninj1 is a novel regulator for the angiogenic effect of PCs. Specifically, Ninj1 negatively regulates the formation of neovessels, that is, the EC-tube, by reducing the trophic effects of cPCs.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Endothelial Cells/cytology , Neovascularization, Physiologic/physiology , Nerve Growth Factors/physiology , Pericytes/cytology , Animals , Aorta, Thoracic , Capillaries , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Cell Adhesion Molecules, Neuronal/biosynthesis , Cell Adhesion Molecules, Neuronal/genetics , Cell Culture Techniques , Cell Hypoxia , Cell Line, Transformed , Cell Lineage , Coculture Techniques , Collagen , Drug Combinations , Gene Expression Profiling , Genes, Reporter , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Ischemia/pathology , Laminin , Male , Mice , Mice, Inbred C57BL , Morphogenesis , Myocytes, Smooth Muscle , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , Organ Culture Techniques , Proteoglycans , RNA Interference , RNA, Small Interfering/pharmacology
16.
Lab Invest ; 94(12): 1340-54, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25329003

ABSTRACT

Adventitial microvessels, vasa vasorum in the vessel walls, have an active role in the vascular remodeling, although its mechanisms are still unclear. It has been reported that microvascular pericytes (PCs) possess mesenchymal plasticity. Therefore, microvessels would serve as a systemic reservoir of stem cells and contribute to the tissues remodeling. However, most aspects of the biology of multipotent PCs (mPCs), in particular of pathological microvessels are still obscure because of the lack of appropriate methods to detect and isolate these cells. In order to examine the characteristics of mPCs, we established immortalized cells residing in adventitial capillary growing at the injured vascular walls. We recently developed in vivo angiogenesis to observe adventitial microvessels using collagen-coated tube (CCT), which also can be used as an adventitial microvessel-rich tissue. By using the CCT, CD146- or NG2-positive cells were isolated from the adventitial microvessels in the injured arteries of mice harboring a temperature-sensitive SV40 T-antigen gene. Several capillary-derived endothelial cells (cECs) and PCs (cPCs) cell lines were established. cECs and cPCs maintain a number of key endothelial and PC features. Co-incubation of cPCs with cECs formed capillary-like structure in Matrigel. Three out of six cPC lines, termed capillary mPCs demonstrated both mesenchymal stem cell- and neuronal stem cell-like phenotypes, differentiating effectively into adipocytes, osteoblasts, as well as schwann cells. mPCs differentiated to ECs and PCs, and formed capillary-like structure on their own. Transplanted DsRed-expressing mPCs were resident in the capillary and muscle fibers and promoted angiogenesis and myogenesis in damaged skeletal muscle. Adventitial mPCs possess transdifferentiation potential with unique phenotypes, including the reconstitution of capillary-like structures. Their phenotype would contribute to the pathological angiogenesis associated with vascular remodeling. These cell lines also provide a reproducible cellular tool for high-throughput studies on angiogenesis, vascular remodeling, and regeneration as well.


Subject(s)
Capillaries/pathology , Pericytes/physiology , Regeneration/physiology , Vasa Vasorum/cytology , Vascular Remodeling , Animals , Antigens , Cell Differentiation , Cell Separation , Endothelial Cells/physiology , Mice , Mice, SCID , Neovascularization, Physiologic , Proteoglycans , Stem Cells/physiology , Transcriptome
18.
Biochem Biophys Res Commun ; 443(1): 150-5, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24296254

ABSTRACT

An immature vasa vasorum in the adventitia of arteries has been implicated in induction of the formation of unstable atherosclerotic plaques. Normalization/maturation of the vasa vasorum may be an attractive therapeutic approach for arteriosclerotic diseases. Nerve growth factor (NGF) is a pleotropic molecule with angiogenic activity in addition to neural growth effects. However, whether NGF affects the formation of microvessels in addition to innervation during pathological angiogenesis is unclear. In the present study, we show a new role for NGF in neovessels around injured arterial walls using a novel in vivo angiogenesis assay. The vasa vasorum around arterial walls was induced to grow using wire-mediated mouse femoral arterial injury. When collagen-coated tube (CCT) was placed beside the injured artery for 7-14 days, microvessels grew two-dimensionally in a thin layer on the CCT (CCT-membrane) in accordance with the development of the vasa vasorum. The perivascular nerve was found at not only arterioles but also capillaries in the CCT-membrane. Biodegradable hydrogels containing VEGF and NGF were applied around the injured artery/CCT. VEGF significantly increased the total length and instability of microvessels within the CCT-membrane. In contrast, NGF induced regeneration of the peripheral nerve around the microvessels and induced the maturation and stabilization of microvessels. In an ex vivo nerve-free angiogenesis assay, although NGF potentially stimulated vascular sprouting from aorta tissues, no effects of NGF on vascular maturation were observed. These data demonstrated that NGF had potent angiogenic effects on the microvessels around the injured artery, and especially induced the maturation/stabilization of microvessels in accordance with the regeneration of perivascular nerves.


Subject(s)
Femoral Artery/drug effects , Femoral Artery/injuries , Microvessels/drug effects , Neovascularization, Physiologic/drug effects , Nerve Growth Factor/pharmacology , Nerve Regeneration/drug effects , Vasa Vasorum/physiology , Vascular Endothelial Growth Factor A/pharmacology , Angiogenesis Inducing Agents/pharmacology , Animals , Femoral Artery/physiology , Male , Mice , Mice, Inbred C57BL , Microvessels/innervation , Microvessels/physiology , Neovascularization, Physiologic/physiology , Vasa Vasorum/innervation
19.
Heart Vessels ; 29(6): 867-72, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24281399

ABSTRACT

We describe three cases of J-wave syndrome in which ventricular fibrillation (VF) was probably induced by corticosteroid therapy. The patients involved were being treated with prednisolone for concomitant bronchial asthma. One of the three patients had only one episode of VF during her long follow-up period (14 years). Two patients had hypokalemia during their VF episodes. Corticosteroids have been shown to induce various types of arrhythmia and to modify cardiac potassium channels. We discuss the possible association between corticosteroid therapy and VF in J-wave syndrome based on the cases we have encountered.


Subject(s)
Anti-Arrhythmia Agents/administration & dosage , Cardiopulmonary Resuscitation/methods , Defibrillators, Implantable , Glucocorticoids/adverse effects , Hypokalemia , Ventricular Fibrillation , Adult , Asthma/drug therapy , Electrocardiography/methods , Female , Glucocorticoids/administration & dosage , Humans , Hypokalemia/chemically induced , Hypokalemia/complications , Hypokalemia/diagnosis , Male , Syndrome , Treatment Outcome , Ventricular Fibrillation/diagnosis , Ventricular Fibrillation/etiology , Ventricular Fibrillation/therapy
20.
Am J Cardiovasc Dis ; 4(4): 159-67, 2014.
Article in English | MEDLINE | ID: mdl-25628957

ABSTRACT

BACKGROUND: It remains unclear whether administration of ARB with reactive oxygen species (ROS) scavenging effects improves the prognosis of patients undergoing PCI. OBJECTIVES: This study investigated whether the pre-intervention antioxidant effect of angiotensin receptor blocker (ARB) affects long-term outcomes in patients after successful percutaneous coronary intervention (PCI) without early adverse events. METHODS: Fifty-two patients who underwent elective PCI were randomly assigned for treatment with or without ARB, which was administered within 48 hours before PCI. ROS levels in mononuclear cells (MNCs) and serum superoxide dismutase (SOD) activity were measured pre-PCI and 6 months post-PCI. After exclusion of unexpected early adverse events during angiographic follow-up period, the long-term outcome (major adverse cerebro-cardiovascular event; MACCE) was assessed in eligible patients. RESULTS: Forty-three patients (non-ARB n = 22, ARB n = 21) were followed up in this study. During angiographic follow-up period, ROS formation in MNCs was significantly increased in the non-ARB group (from 29.4 [21.6-35.2] to 37.2 [30.7-45.1] arbitrary units; p = 0.031) compared to that in the ARB group. Meanwhile, SOD activity was significantly impaired in the non-ARB group alone (from 24.0 ± 17.0 to 16.3 ± 13.8%, p = 0.004). During the follow-up period (median, 63.3 months), MACCEs were observed in 6 patients. The cumulative event ratio of MACCE was significantly higher in the non-ARB group than in the ARB group (p = 0.018). CONCLUSIONS: Concomitant administration of ARB effectively reduced ROS production of PCI patients during angiographic follow-up period. Initial ROS inhibition following ARB administration may contribute to improvement of worse outcomes in patients who have undergone successful PCI.

SELECTION OF CITATIONS
SEARCH DETAIL