Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Int J Mol Sci ; 25(2)2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38256060

ABSTRACT

Ischemic heart disease (IHD) poses a significant challenge in cardiovascular health, with current treatments showing limited success. Induced pluripotent derived-cardiomyocyte (iPSC-CM) therapy within regenerative medicine offers potential for IHD patients, although its clinical impacts remain uncertain. This study utilizes meta-analysis to assess iPSC-CM outcomes in terms of efficacy and safety in IHD animal model studies. A meta-analysis encompassing PUBMED, ScienceDirect, Web of Science, and the Cochrane Library databases, from inception until October 2023, investigated iPSC therapy effects on cardiac function and safety outcomes. Among 51 eligible studies involving 1012 animals, despite substantial heterogeneity, the iPSC-CM transplantation improved left ventricular ejection fraction (LVEF) by 8.23% (95% CI, 7.15 to 9.32%; p < 0.001) compared to control groups. Additionally, cell-based treatment reduced the left ventricle fibrosis area and showed a tendency to reduce left ventricular end-systolic volume (LVESV) and end-diastolic volume (LVEDV). No significant differences emerged in mortality and arrhythmia risk between iPSC-CM treatment and control groups. In conclusion, this meta-analysis indicates iPSC-CM therapy's promise as a safe and beneficial intervention for enhancing heart function in IHD. However, due to observed heterogeneity, the efficacy of this treatment must be further explored through large randomized controlled trials based on rigorous research design.


Subject(s)
Induced Pluripotent Stem Cells , Myocardial Ischemia , Humans , Animals , Myocytes, Cardiac , Stroke Volume , Ventricular Function, Left , Myocardial Ischemia/therapy , Disease Models, Animal
2.
Int J Mol Sci ; 24(3)2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36768243

ABSTRACT

Transthyretin cardiac amyloidosis (ATTR-CA) is characterized by high 99mTc-labeled bone tracer uptake in the heart. However, the mechanism of bone tracer uptake into the heart remains controversial. Since bone tracer uptake into metastatic bone tumors is thought to be associated with increased bone metabolism, we examined 99mTc-pyrophosphate (PYP) scintigraphy findings, endomyocardial biopsy (EMB) tissue findings, and the expression of bone metabolism-related genes in the EMB tissues in patients with ATTR-CA, amyloid light-chain cardiac amyloidosis (AL-CA), and noncardiac amyloidosis (non-CA) in this study. The uptake of 99mTc-PYP in the heart was significantly higher in the ATTR-CA patients than in the AL-CA and non-CA patients. A higher percentage of ATTR-CA EMB tissue showed von Kossa-positive microparticles: ATTR-CA, 62%; AL-CA, 33%; and non-CA, 0%. Calcified microparticles were identified using transmission electron microscopy. However, none of the osteogenic marker genes, osteoclastic marker genes, or phosphate/pyrophosphate-related genes were upregulated in the EMB samples from ATTR-CA patients compared to those from AL-CA and non-CA patients. These results suggest that active calcification-promoting mechanisms are not involved in the microcalcification observed in the heart in ATTR-CA. The mechanisms explaining bone tracer uptake in the heart, which is stronger than that in the ribs, require further investigation.


Subject(s)
Amyloidosis , Calcinosis , Cardiomyopathies , Immunoglobulin Light-chain Amyloidosis , Humans , Diphosphates , Prealbumin/genetics , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/pathology , Radiopharmaceuticals
3.
Int Heart J ; 64(3): 502-505, 2023 May 31.
Article in English | MEDLINE | ID: mdl-37197914

ABSTRACT

Fabry disease is an X-linked lysosomal storage disorder caused by defective enzyme activity of α-galactosidase A and treated with enzyme replacement therapy (ERT) with recombinant α-galactosidase. ERT reduces left ventricular mass assessed by echocardiography or magnetic resonance imaging. However, electrocardiogram changes during ERT have not been fully elucidated. In the present case, ERT with agalsidase alfa for 4 years decreased QRS voltage and negative T depth along with a reduction of left ventricular mass and wall thickness and improvement of symptoms in a female patient with Fabry disease. Long-term observation of electrocardiogram changes might be useful for determining the efficacy of ERT in this case.


Subject(s)
Fabry Disease , Humans , Female , Fabry Disease/complications , Fabry Disease/drug therapy , alpha-Galactosidase/therapeutic use , Hypertrophy, Left Ventricular/drug therapy , Hypertrophy, Left Ventricular/etiology , Enzyme Replacement Therapy , Electrocardiography , Treatment Outcome
4.
Circ J ; 86(8): 1312-1318, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35768227

ABSTRACT

The 86thAnnual Scientific Meeting of the Japanese Circulation Society was held in a web-based format on March 11-13, 2022. In accordance with the internationalization policy of the JCS, the meeting was held with the Asian Pacific Society of Cardiology Congress 2022. The main theme was "Cardiology Spreading its Wings". The number of patients with heart failure and other cardiovascular diseases is increasing dramatically, and the fields dealt with by cardiovascular medicine are also greatly expanding. This conference was both intellectually satisfying and exciting for all participants, who numbered over 14,900. The meeting was completed with great success, and the enormous amount of cooperation and support from all involved was greatly appreciated.


Subject(s)
Cardiology , Cardiovascular Diseases , Heart Failure , Animals , Humans , Japan , Societies, Medical
5.
Int J Mol Sci ; 23(7)2022 Mar 25.
Article in English | MEDLINE | ID: mdl-35408946

ABSTRACT

There is a close relationship between diabetes mellitus and heart failure, and diabetes is an independent risk factor for heart failure. Diabetes and heart failure are linked by not only the complication of ischemic heart disease, but also by metabolic disorders such as glucose toxicity and lipotoxicity based on insulin resistance. Cardiac dysfunction in the absence of coronary artery disease, hypertension, and valvular disease is called diabetic cardiomyopathy. Diabetes-induced hyperglycemia and hyperinsulinemia lead to capillary damage, myocardial fibrosis, and myocardial hypertrophy with mitochondrial dysfunction. Lipotoxicity with extensive fat deposits or lipid droplets is observed on cardiomyocytes. Furthermore, increased oxidative stress and inflammation cause cardiac fibrosis and hypertrophy. Treatment with a sodium glucose cotransporter 2 (SGLT2) inhibitor is currently one of the most effective treatments for heart failure associated with diabetes. However, an effective treatment for lipotoxicity of the myocardium has not yet been established, and the establishment of an effective treatment is needed in the future. This review provides an overview of heart failure in diabetic patients for the clinical practice of clinicians.


Subject(s)
Diabetes Mellitus , Diabetic Cardiomyopathies , Heart Failure , Insulin Resistance , Diabetes Mellitus/metabolism , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/therapy , Heart Failure/complications , Heart Failure/therapy , Humans , Hypertrophy/metabolism , Insulin Resistance/physiology , Myocardium/metabolism
6.
Int J Mol Sci ; 23(1)2021 Dec 21.
Article in English | MEDLINE | ID: mdl-35008444

ABSTRACT

Cardiac involvement has a profound effect on the prognosis of patients with systemic amyloidosis. Therapeutic methods for suppressing the production of causative proteins have been developed for ATTR amyloidosis and AL amyloidosis, which show cardiac involvement, and the prognosis has been improved. However, a method for removing deposited amyloid has not been established. Methods for reducing cytotoxicity caused by amyloid deposition and amyloid precursor protein to protect cardiovascular cells are also needed. In this review, we outline the molecular mechanisms and treatments of cardiac amyloidosis.


Subject(s)
Amyloid Neuropathies, Familial/pathology , Cardiomyopathies/pathology , Heart/physiopathology , Immunoglobulin Light-chain Amyloidosis/pathology , Animals , Humans , Prognosis
7.
J Vasc Surg ; 71(1): 229-241, 2020 01.
Article in English | MEDLINE | ID: mdl-31204215

ABSTRACT

OBJECTIVE: Intimal hyperplasia (IH) is the main cause of therapeutic failure after vascular and endovascular surgery. However, there is currently no targeted therapy for the treatment of IH. We recently reported that the inhibition of cyclic adenosine monophosphate response element (CRE) binding protein (CREB) activation is important in vein graft IH. We focused on a decoy oligodeoxynucleotide (ODN) therapeutic strategy for suppressing IH as a clinical application. The objective of this study was to confirm the therapeutic effect of a CRE decoy ODN in an animal model as a novel therapy for preventing intimal hyperplasia as the first step of the preclinical study of our strategy. METHODS: We designed two phosphorothioate CREs and two scramble decoy ODNs and screened them using a CREB transcription assay to check their ability to bind to a CRE sequence. We chose a CRE decoy ODN with high first-binding ability and transfected it into vascular smooth muscle cells (VSMCs) in vitro. Proliferation and migration were assessed using MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) assays and modified Boyden chamber assays. We examined CRE activity using a luciferase reporter gene assay. We assessed the expression of messenger RNAs by quantitative real-time polymerase chain reaction. In a wire-injury mouse model (C57BL6, n = 6), CRE decoy ODN was transfected into the injured vessel wall using an ultrasound-sonoporation method in vivo. Mitogen-activated protein kinase-activated protein kinase 3 (MAPKAPK3) and four and a half LIM domains 5 (FHL5) expression of pregrafting vein remnants were assessed by immunohistologic analyses. RESULTS: Compared with scramble decoy ODN, the selected CRE decoy ODN could significantly decrease CRE activity (mean ± standard error of the mean: 0.20 ± 0.03 vs 1.00 ± 0.16, n = 6; P < .05) as shown by a luciferase reporter gene assay, VSMC proliferation (0.73 ± 0.04 vs 0.89 ± 0.02, n = 6; P < .05) and migration (96.4 ± 6.1 vs 311.4 ± 19.1 migrated VSMCs/well, n = 6; P < .05) after 24-hour transfection. The CRE decoy ODN significantly suppressed the formation of IH at injured vessel walls in an animal model, as analyzed by pathologic staining (0.20 ± 0.02 vs 0.56 ± 0.08, area of the intima/area of the artery vs the control after 21 days' transfection, n = 6; P < .05). Furthermore, MAPKAPK3 and FHL5, which are CREB activators, were significantly expressed in pregrafting vein remnants in diabetes mellitus patients. CONCLUSIONS: CREB-CRE signaling is an important mechanism of IH formation, and CRE decoy therapy can help preventing IH. This study is the first part of the preclinical study of our strategy.


Subject(s)
Cyclic AMP/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Oligodeoxyribonucleotides/genetics , Response Elements/genetics , Vascular System Injuries/prevention & control , Animals , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Oligodeoxyribonucleotides/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular System Injuries/genetics , Vascular System Injuries/metabolism , Vascular System Injuries/pathology
8.
Ann Vasc Surg ; 66: 518-528, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32035265

ABSTRACT

BACKGROUND: Arterial reconstruction (AR) for limb ischemia may improve ambulatory function (AF) and health-related quality of life (HR-QoL). However, the efficacy of AR in terms of HR-QoL varies in studies, probably because of cohort differences in disease severity, hemodynamic outcomes, and observation duration. We assessed HR-QoL for patients with various severities of ischemia in a 3-year observational study. METHODS: We conducted a single-center 3-year observational study using Short Form 36 in patients with chronic limb ischemia. Between 2001 and 2009, 515 consecutive patients had AR, and 330 who underwent elective AR consented to the study. Of the 330 patients (claudicants 49%, critical limb ischemia [CLI] 51%), 307 underwent bypass and 23 endovascular therapy. Postal questionnaires were sent after AR, and 8 domains, the physical and mental component summary (PCS and MCS) scores, and the patient-reported AF were compared, and negative predictors were identified. RESULTS: Overall, the MCS was minimally affected, but AF and the PCS were impaired. After AR, these measures were significantly improved, and maximum recovery was attained at 6 months. In subgroup analysis, significant predictors of a negative impact on postoperative PCS included age ≥80, CLI, physical aftereffects of stroke (PAS), and previous major amputation (PMA). Of these, PMA was associated with the lowest PCS score, followed by PAS; for these patients, AR contributed minimally to HR-QoL recovery. PCS scores of claudicants attained a maximum value at 6 months; however, PCS scores of CLI patients were significantly lower than intermittent claudication patients (P < 0.0001), and patients with major tissue loss required 2 years to attain maximum PCS recovery. CONCLUSIONS: This 3-year observational study verified the efficacy of AR in improving AF and HR-QoL. Age ≥80, CLI, PAS, and PMA were definitive predictors, and for patients with the latter 2, AR contributed minimally to improving HR-QoL.


Subject(s)
Endovascular Procedures , Exercise Tolerance , Intermittent Claudication/surgery , Ischemia/surgery , Peripheral Arterial Disease/surgery , Quality of Life , Vascular Grafting , Walking , Adult , Aged , Aged, 80 and over , Chronic Disease , Endovascular Procedures/adverse effects , Female , Humans , Intermittent Claudication/diagnosis , Intermittent Claudication/physiopathology , Ischemia/diagnosis , Ischemia/physiopathology , Japan , Longitudinal Studies , Male , Middle Aged , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/physiopathology , Prospective Studies , Recovery of Function , Severity of Illness Index , Time Factors , Treatment Outcome , Vascular Grafting/adverse effects , Vascular Patency
9.
Int J Mol Sci ; 21(15)2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32751754

ABSTRACT

Arterial calcification is a hallmark of advanced atherosclerosis and predicts cardiovascular events. However, there is no clinically accepted therapy that prevents progression of arterial calcification. HMG-CoA reductase inhibitors, statins, lower low-density lipoprotein-cholesterol and reduce cardiovascular events, but coronary artery calcification is actually promoted by statins. The addition of eicosapentaenoic acid (EPA) to statins further reduced cardiovascular events in clinical trials, JELIS and REDUCE-IT. Additionally, we found that EPA significantly suppressed arterial calcification in vitro and in vivo via suppression of inflammatory responses, oxidative stress and Wnt signaling. However, so far there is a lack of evidence showing the effect of EPA on arterial calcification in a clinical situation. We reviewed the molecular mechanisms of the inhibitory effect of EPA on arterial calcification and the results of some clinical trials.


Subject(s)
Arteries/drug effects , Atherosclerosis/drug therapy , Calcinosis/drug therapy , Eicosapentaenoic Acid/metabolism , Arteries/metabolism , Arteries/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Calcinosis/metabolism , Calcinosis/pathology , Cholesterol, LDL , Clinical Trials as Topic , Eicosapentaenoic Acid/therapeutic use , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use
10.
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.
Vascular ; 27(3): 242-251, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30419804

ABSTRACT

OBJECTIVES: Laser speckle flowgraphy is a new method that enables the rapid evaluation of foot blood flow without contact with the skin. We used laser speckle flowgraphy to evaluate foot blood flow in peripheral arterial disease patients before and after surgical revascularization. MATERIALS AND METHODS: A prospective single-center study. Thirty-one patients with 33 limbs that underwent surgical revascularization for peripheral arterial disease were included. Pre- and postoperative foot blood flows were measured on the plantar surface via laser speckle flowgraphy and skin perfusion pressure. The laser speckle flowgraphy device was used to visualize the blood flow distribution of the target skin and processed the pulse wave velocity of synchronized heart beats. The mean blood flow, which was expressed as the area of the pulse wave as the beat strength of skin perfusion on laser speckle flowgraphy converted into a numerical value, was assessed as dynamic changes following surgery. Beat strength of skin perfusion was also investigated in non-peripheral arterial disease controls (23 patients/46 limbs). RESULTS: The suitability of beat strength of skin perfusion in non-peripheral arterial disease controls was achieved; the beat strength of skin perfusion value was significantly higher in every area of interest in non-peripheral arterial disease controls compared to that in peripheral arterial disease limbs at the preoperative stage (105.8 ± 8.2 vs. 26.3 ± 8.2; P < 0.01). Although the pulse wave before surgery was visually flat in peripheral arterial disease patients, the pulse wave was remarkably and immediately improved through surgical revascularization. Beat strength of skin perfusion showed a dynamic change in foot blood flow (26.3 ± 8.2 at preoperation, 98.5 ± 6.7 immediately after surgery, 107.6 ± 5.7 at seven days after surgery, P < 0.01 for each compared to preoperation) that correlated with an improvement in skin perfusion pressure. CONCLUSIONS: Laser speckle flowgraphy is a noninvasive, contact-free modality that is easy to implement, and beat strength of skin perfusion is a useful indicator of foot circulation during the perioperative period. Further analysis with a larger number of cases is necessary to establish appropriate clinical use.


Subject(s)
Foot/blood supply , Hemodynamics , Lasers, Semiconductor , Perfusion Imaging/instrumentation , Peripheral Arterial Disease/surgery , Skin/blood supply , Aged , Ankle Brachial Index , Blood Flow Velocity , Case-Control Studies , Female , Humans , Male , Perfusion Imaging/methods , Peripheral Arterial Disease/diagnostic imaging , Peripheral Arterial Disease/physiopathology , Predictive Value of Tests , Prospective Studies , Pulse Wave Analysis , Regional Blood Flow , Time Factors , Treatment Outcome
12.
Int Heart J ; 60(3): 728-735, 2019 May 30.
Article in English | MEDLINE | ID: mdl-31105148

ABSTRACT

Sodium-glucose cotransporter 2 (SGLT2) inhibitors are drugs for diabetes and might prevent heart failure. In this study, we investigated the effects of tofogliflozin, an SGLT2 inhibitor, on cardiac hypertrophy and metabolism in hypertensive rats fed a high-fat diet. Dahl salt-sensitive (DS) rats, hypertensive model rats, and Dahl salt-resistant (DR) rats, non-hypertensive model rats, were fed a high-salt and high-fat diet containing tofogliflozin (0.005%) for 9 weeks to examine the effects of this drug on cardiac hypertrophy and metabolism. Tofogliflozin tended to suppress a rise of the systolic blood pressure, relative to the control, throughout the treatment period in both DR and DS rats, and significantly suppress a rise of the systolic blood pressure, relative to the control, at the 9th week in DS rats. Tofogliflozin reduced cardiac hypertrophy (heart weight/body weight) not only in DS rats but also in DR rats. Histological analysis showed that tofogliflozin significantly decreased cardiomyocyte hypertrophy and perivascular fibrosis in both DS and DR rats. Tofogliflozin significantly decreased the expression levels of genes related to cardiac hypertrophy (encoding for natriuretic peptides A and B and interleukin-6), and to cardiac fibrosis (encoding for transforming growth factor-ß1 and collagen type IV), in DS rats. Recent studies have shown that hypertrophied and failing hearts shift to oxidizing ketone bodies as a significant fuel source. We also performed metabolome analysis for ventricular myocardial tissue. Tofogliflozin reduced 3-hydroxybutyrate, a ketone body, and significantly decreased the expression levels of ß-hydroxybutyrate dehydrogenase 1 and 3-oxoacid CoA-transferase, which are related to ketone oxidization. In conclusion, tofogliflozin ameliorated cardiac hypertrophy and fibrosis along with reduction of ketone usage in myocardial tissue.


Subject(s)
Benzhydryl Compounds/pharmacology , Blood Pressure/drug effects , Cardiomegaly/metabolism , Diet, High-Fat/adverse effects , Glucosides/pharmacology , Myocytes, Cardiac/drug effects , Animals , Benzhydryl Compounds/adverse effects , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Fibrosis/drug therapy , Glucosides/adverse effects , Interleukin-6/metabolism , Ketones/metabolism , Male , Models, Animal , Natriuretic Peptides/metabolism , Rats , Rats, Inbred Dahl , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
13.
J Vasc Surg ; 67(3): 826-837, 2018 03.
Article in English | MEDLINE | ID: mdl-28965798

ABSTRACT

OBJECTIVE: The aim of this study was to elucidate the efficacy of paramalleolar or inframalleolar bypass (PIMB) in hemodialysis-dependent (HD) patients with critical limb ischemia (CLI) and nonhemodialysis-dependent (NHD) patients in terms of clinical outcomes. METHODS: Between January 2000 and December 2013, there were 333 consecutive arteriosclerosis obliterans patients with CLI who underwent 401 PIMB procedures for limb salvage (LS). Of the 333 patients, 188 (56.5%) were HD patients. Vein grafts were exclusively used, and 172 paramalleolar and 229 inframalleolar bypasses were performed. Five-year primary and secondary cumulative graft patency, LS, and amputation-free survival (AFS) rates were compared between the two groups, and the independent determinants of these outcomes were identified in each group. RESULTS: The 5-year primary and secondary cumulative graft patency rates were 53% and 82% in HD patients and 69% and 92% in NHD patients (primary cumulative graft patency, P < .05; secondary cumulative graft patency, nonsignificant), respectively. The LS rates were 87% and 99% (P < .01) in HD patients and NHD patients, respectively. Overall, 48% and 70% of HD and NHD patients were ambulatory before PIMB (P < .01), and 73% and 85% of HD and NHD patients were ambulatory 12 months after PIMB (including 1-year survivors; nonsignificant), respectively, demonstrating drastic post-PIMB improvement in HD patients. The 5-year AFS rates in the HD and NHD groups were 27% and 69% (P < .01), respectively, demonstrating very poor AFS rates in HD patients. In HD patients, factors negatively associated with AFS were female gender (hazard ratio [HR], 2.102; 95% confidence interval [CI], 1.254-3.524), history of congestive heart failure (HR, 2.075; 95% CI, 1.395-3.085), and preoperative nonambulatory status (HR, 1.974; 95% CI, 1.305-2.986), whereas older age (HR, 2.601; 95% CI, 1.372-4.931) and history of congestive heart failure (HR, 2.928; 95% CI, 1.496-5.731) were identified as independent factors negatively associated with AFS in NHD patients. CONCLUSIONS: The use of PIMB for CLI was associated with excellent LS rates in both HD and NHD patients with low operative mortality and complications. However, the AFS rate observed in HD patients was significantly lower than that observed in NHD patients, indicating the necessity of a specific management program to improve AFS after LS in HD patients.


Subject(s)
Arm/blood supply , Arteriosclerosis Obliterans/surgery , Ischemia/surgery , Renal Dialysis , Renal Insufficiency, Chronic/surgery , Saphenous Vein/transplantation , Vascular Grafting/methods , Age Factors , Aged , Aged, 80 and over , Amputation, Surgical , Arteriosclerosis Obliterans/diagnosis , Arteriosclerosis Obliterans/mortality , Arteriosclerosis Obliterans/physiopathology , Comorbidity , Critical Illness , Disease-Free Survival , Female , Humans , Ischemia/diagnosis , Ischemia/mortality , Ischemia/physiopathology , Kaplan-Meier Estimate , Kidney/physiopathology , Limb Salvage , Male , Middle Aged , Proportional Hazards Models , Renal Dialysis/adverse effects , Renal Dialysis/mortality , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/mortality , Renal Insufficiency, Chronic/physiopathology , Retrospective Studies , Risk Factors , Saphenous Vein/physiopathology , Sex Factors , Time Factors , Treatment Outcome , Vascular Grafting/adverse effects , Vascular Grafting/mortality , Vascular Patency
14.
J Vasc Surg ; 67(5): 1556-1570.e9, 2018 05.
Article in English | MEDLINE | ID: mdl-28647196

ABSTRACT

OBJECTIVE: Venous valves are essential but are prone to injury, thrombosis, and fibrosis. We compared the behavior and gene expression of smooth muscle cells (SMCs) in the valve sinus vs nonvalve sites to elucidate biologic differences associated with vein valves. METHODS: Tissue explants of fresh human saphenous veins were prepared, and the migration of SMCs from explants of valve sinus vs nonvalve sinus areas was measured. Proliferation and death of SMCs were determined by staining for Ki67 and terminal deoxynucleotidyl transferase dUTP nick end labeling. Proliferation and migration of passaged valve vs nonvalve SMCs were determined by cell counts and using microchemotaxis chambers. Global gene expression in valve vs nonvalve intima-media was determined by RNA sequencing. RESULTS: Valve SMCs demonstrated greater proliferation in tissue explants compared with nonvalve SMCs (19.3% ± 5.4% vs 6.8% ± 2.0% Ki67-positive nuclei at 4 days, respectively; mean ± standard error of the mean, five veins; P < .05). This was also true for migration (18.2 ± 2.7 vs 7.5 ± 3.0 migrated SMCs/explant at 6 days, respectively; 24 veins, 15 explants/vein; P < .0001). Cell death was not different (39.6% ± 16.1% vs 41.5% ± 16.0% terminal deoxynucleotidyl transferase dUTP nick end labeling-positive cells, respectively, at 4 days, five veins). Cultured valve SMCs also proliferated faster than nonvalve SMCs in response to platelet-derived growth factor subunit BB (2.9 ± 0.2-fold vs 2.1 ± 0.2-fold of control, respectively; P < .001; n = 5 pairs of cells). This was also true for migration (6.5 ± 1.2-fold vs 4.4 ± 0.8-fold of control, respectively; P < .001; n = 7 pairs of cells). Blockade of fibroblast growth factor 2 (FGF2) inhibited the increased responses of valve SMCs but had no effect on nonvalve SMCs. Exogenous FGF2 increased migration of valve but not of nonvalve SMCs. Unlike in the isolated, cultured cells, blockade of FGF2 in the tissue explants did not block migration of valve or nonvalve SMCs from the explants. Thirty-seven genes were differentially expressed by valve compared with nonvalve intimal-medial tissue (11 veins). Peptide-mediated inhibition of SEMA3A, one of the differentially expressed genes, increased the number of migrated SMCs of valve but not of nonvalve explants. CONCLUSIONS: Valve compared with nonvalve SMCs have greater rates of migration and proliferation, which may in part explain the propensity for pathologic lesion formation in valves. Whereas FGF2 mediates these effects in cultured SMCs, the mediators of these stimulatory effects in the valve wall tissue remain unclear but may be among the differentially expressed genes discovered in this study. One of these genes, SEMA3A, mediates a valve-specific inhibitory effect on the injury response of valve SMCs.


Subject(s)
Cell Movement , Cell Proliferation , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Vascular System Injuries/pathology , Venous Valves/pathology , Becaplermin , Cell Death , Cells, Cultured , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation , Humans , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Neointima , Proto-Oncogene Proteins c-sis/pharmacology , Saphenous Vein/injuries , Saphenous Vein/metabolism , Saphenous Vein/pathology , Semaphorin-3A/genetics , Semaphorin-3A/metabolism , Time Factors , Vascular System Injuries/genetics , Vascular System Injuries/metabolism , Venous Valves/drug effects , Venous Valves/injuries , Venous Valves/metabolism
15.
Acta Med Okayama ; 72(1): 1-7, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29463932

ABSTRACT

The number of permanent pacemaker implantations has been increasing due to the aging of populations worldwide and the increase in the numbers of patients with heart diseases. Commercially available mechanical pacemakers are very useful but still have some problems including short battery life, a risk of infection, the absence of physiological autonomic responsiveness, metal allergy, and electronic interference. A biological pacemaker may resolve these problems and regenerate the cardiac pacemaker. Cell-based therapy and gene therapy have been addressed with the goal of solving the challenges of biological pacemaker. However, the clinical application of a biological pacemaker has not yet been realized. Here we discuss the types of cells that can be used for a biological pacemaker and the problems that remain regarding the clinical applications of cell-based therapy.


Subject(s)
Arrhythmias, Cardiac , Bioengineering , Genetic Therapy , Pacemaker, Artificial , Arrhythmias, Cardiac/therapy , Humans
16.
Int Heart J ; 59(3): 601-606, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29628472

ABSTRACT

A biological pacemaker is expected to solve the persisting problems of an artificial cardiac pacemaker including short battery life, lead breaks, infection, and electromagnetic interference. We previously reported HCN4 overexpression enhances pacemaking ability of mouse embryonic stem cell-derived cardiomyocytes (mESC-CMs) in vitro. However, the effect of these cells on bradycardia in vivo has remained unclear. Therefore, we transplanted HCN4-overexpressing mESC-CMs into bradycardia model animals and investigated whether they could function as a biological pacemaker. The rabbit Hcn4 gene was transfected into mouse embryonic stem cells and induced HCN4-overexpressing mESC-CMs. Non-cardiomyocytes were removed under serum/glucose-free and lactate-supplemented conditions. Cardiac balls containing 5 × 103 mESC-CMs were made by using the hanging drop method. One hundred cardiac balls were injected into the left ventricular free wall of complete atrioventricular block (CAVB) model rats. Heart beats were evaluated using an implantable telemetry system 7 to 30 days after cell transplantation. The result showed that ectopic ventricular beats that were faster than the intrinsic escape rhythm were often observed in CAVB model rats transplanted with HCN4-overexpressing mESC-CMs. On the other hand, the rats transplanted with non-overexpressing mESC-CMs showed sporadic single premature ventricular contraction but not sustained ectopic ventricular rhythms. These results indicated that HCN4-overexpressing mESC-CMs produce rapid ectopic ventricular rhythms as a biological pacemaker.


Subject(s)
Biological Clocks/physiology , Bradycardia/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Animals , Bradycardia/physiopathology , Cell Line , Disease Models, Animal , Fluorescent Antibody Technique , Heart Rate/physiology , Mice , Mouse Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Rabbits , Rats , Telemetry , Transfection
17.
Article in Japanese | MEDLINE | ID: mdl-28428468

ABSTRACT

OBJECTIVE: To study whether the actual radiation exposure is different between computed tomography (CT) scanners and medical centers when the same patient is scanned, we investigated the actual effective doses for a whole body (Chest-Pelvis) CT scan in a multicenter study. MATERIALS AND METHODS: Data from subjects were collected using 12 CT scanners at six medical centers in Yamagata city. Effective-dose data were acquired by scanning the same phantom (ATOM Dosimetry Phantoms Model702-B) using 120 kV tube voltage. Effective doses were calculated using corrected data from a radiophotoluminescent glass dosimeter (GD-302M). GD-302M had energy- dependent issues, which needed to be corrected. Also, differences in sensitivity based on arrangement within the phantom were insignificant. RESULTS: The mean effective energy was 48.6 keV (range, 42.5-55.4 keV), and the mean effective dose was 16.3 mSv (range, 8.9-26.0 mSv). The mean effective dose with a hybrid type iterative reconstruction was 10.7 mSv (range, 8.9-16.4 mSv), but the mean effective dose without any iterative reconstruction was 20.3 mSv (range, 16.2-26.0 mSv). We found an approximate linear correlation between dose length product (DLP) on operation consoles and the effective dose. CONCLUSION: We suggest that the actual radiation exposure was different at each medical center when the same patient is scanned.


Subject(s)
Tomography, X-Ray Computed/methods , Female , Humans , Manikins , Phantoms, Imaging , Radiation Dosage
18.
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
19.
Int Heart J ; 56(3): 354-9, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25902888

ABSTRACT

Platelet-derived growth factor (PDGF) is implicated in the pathogenesis of pulmonary arterial hypertension (PAH). Imatinib, a PDGF-receptor tyrosine kinase inhibitor, improved hemodynamics, but serious side effects and drug discontinuation are common when treating PAH. A drug delivery system using nanoparticles (NPs) enables the reduction of side effects while maintaining the effects of the drug. We examined the efficacy of imatinib-incorporated NPs (Ima-NPs) in a rat model and in human PAH-pulmonary arterial smooth muscle cells (PASMCs). Rats received a single intratracheal administration of PBS, FITC-NPs, or Ima-NPs immediately after monocrotaline injection. Three weeks after monocrotaline injection, intratracheal administration of Ima-NPs suppressed the development of pulmonary hypertension, small pulmonary artery remodeling, and right ventricular hypertrophy in the rat model of monocrotaline-induced PAH. We also examined the effects of imatinib and Ima-NPs on PDGF-induced proliferation of human PAH-PASMCs by (3)H-thymidine incorporation. Imatinib and Ima-NPs significantly inhibited proliferation after 24 hours of treatment. Ima-NPs significantly inhibited proliferation compared with imatinib at 24 hours after removal of these drugs. Delivery of Ima-NPs into lungs suppressed the development of MCT-induced PAH by sustained antiproliferative effects on PAS-MCs.


Subject(s)
Benzamides/administration & dosage , Hypertension, Pulmonary/prevention & control , Nanoparticles , Piperazines/administration & dosage , Pyrimidines/administration & dosage , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Animals , Benzamides/therapeutic use , Cells, Cultured , Disease Models, Animal , Humans , Hypertension, Pulmonary/chemically induced , Imatinib Mesylate , Male , Monocrotaline , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL