Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
ESC Heart Fail ; 10(2): 895-906, 2023 04.
Article in English | MEDLINE | ID: mdl-36460605

ABSTRACT

AIMS: The timely selection of severe heart failure (HF) patients for cardiac transplantation and advanced HF therapy is challenging. Peak oxygen consumption (VO2 ) values obtained by the cardiopulmonary exercise testing are used to determine the transplant recipient list. This study reassessed the prognostic predictability of peak VO2 and compared it with the Heart Failure Survival Score (HFSS) in the modern optimized guideline-directed medical therapy (GDMT) era. METHODS AND RESULTS: We retrospectively selected 377 acute HF patients discharged from the hospital. The primary outcome was a composite of all-cause mortality, or urgent cardiac transplantation. We divided these patients into the more GDMT (two or more types of GDMT) and less GDMT groups (fewer than two types of GDMT) and compared the performance of their peak VO2 and HFSS in predicting primary outcomes. The median follow-up period was 3.3 years. The primary outcome occurred in 57 participants. Peak VO2 outperformed HFSS when predicting 1 year (0.81 vs. 0.61; P = 0.017) and 2 year (0.78 vs. 0.58; P < 0.001) major outcomes. The cutoff peak VO2 for predicting a 20% risk of a major outcome within 2 years was 10.2 (11.8-7.0) for the total cohort. Multivariate Cox regression analyses showed that peak VO2 , sodium, previous implantable cardioverter defibrillator (ICD) implantation, and estimated glomerular filtration rate were significant predictors of major outcomes. CONCLUSIONS: Optimizing the cutoff value of peak VO2 is required in the current GDMT era for advanced HF therapy. Other clinical factors such as ICD use, hyponatraemia, and chronic kidney disease could also be used to predict poor prognosis. The improvement of resource allocation and patient outcomes could be achieved by careful selection of appropriate patients for advanced HF therapies, such as cardiac transplantation.


Subject(s)
Exercise Test , Heart Failure , Humans , Retrospective Studies , Oxygen Consumption , Heart Failure/diagnosis , Heart Failure/therapy , Prognosis , Risk Assessment
2.
Biomed Pharmacother ; 146: 112551, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34923336

ABSTRACT

This study tested the hypothesis that valsartan (Val) and melatonin (Mel)-assisted adipose-derived mesenchymal stem cells (ADMSCs) preserved the residual renal function in chronic kidney disease (CKD) rat through promoting cellular-prior-protein (PrPC) to upregulate PI3K/Akt/mTOR signaling and cell proliferation. In vitro study demonstrated that as compared with CKD-derived-ADMSCs, Val/Mel/overexpression of PrPC-treated CKD derived-ADMSCs significantly upregulated cell proliferation and protein expressions of PrPC and phosphorylated (p)-PI3K/p-Akt/p-mTOR, and downregulated oxidative stress (all p < 0.001). Rats (n = 42) were categorized into group 1 (sham-operated-control), group 2 (CKD), group 3 (CKD + ADMSCs/1.2 ×106 cells) + Mel/20 mg/kg/day), group 4 (CKD + siRNA-PrPC-ADMSCs/1.2 ×106 cells), group 5 (CKD + ADMSCs/1.2 ×106 cells + Val/20 mg/kg/day) and group 6 (CKD + Val + Mel). By day 35, the kidney specimens were harvested and the result showed that the protein expression of PrPC was highest in group 1, lowest in groups 2/4 and significantly lower in group 6 than in groups 3/5, but it was similar in groups 3/5 (all p < 0.0001). The protein expressions of cell-stress-signaling (p-PI3K/p-Akt/p-mTOR) and cell-cycle activity (cyclin-D1/clyclin-E2/Cdk2/Cdk4) exhibited an identical pattern, whereas the protein expressions of oxidative-stress (NOX-1/NOX-2)/mitochondrial fission (PINK1/DRP1)/apoptosis (cleaved-capsase3/cleaved-PARP) and fibrosis (TFG-ß/Smad3) as well as creatinine/BUN levels, ratio of urine-protein to urine-creatine and kidney-injured score exhibited an opposite pattern of PrPC among the groups (all p < 0.0001). In conclusion, Mel/Val facilitated-ADMSCs preserved renal architecture and function in CKD rat through promoting PrPC to regulate the cell proliferation/oxidative-stress/cell-stress signalings.


Subject(s)
Melatonin/pharmacology , Mesenchymal Stem Cells/metabolism , Renal Insufficiency, Chronic/pathology , Valsartan/pharmacology , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Kidney/drug effects , Male , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Prion Proteins/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/drug effects , Up-Regulation
3.
Ann Vasc Surg ; 72: 617-626, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33249131

ABSTRACT

BACKGROUND: Ascending aortic thrombus (AAT) in a nonaneurysmal aorta is an extremely rare event and has potentially catastrophic complications, with a life-threatening risk of myocardial infarction and cerebral embolization. This systematic review aims to elucidate the clinical manifestations and to compare the outcomes of anticoagulation therapy versus open aortic surgery for AAT. METHODS: The MEDLINE/PubMed databases were extensively searched between 1995 and 2019. All relevant publications on AAT in adults were reviewed, and individual patient data were pooled in this meta-analysis. The primary outcome was AAT resolution. The adverse outcome variables were recurrent arterial embolic events, complications related to open aortic surgery, and mortality during the study period. Chi-squared test and logistic regression analysis were used to compare groups and identify any predictors of mortality. RESULTS: Overall, 107 patients from 101 articles were included, of whom 29 patients who received anticoagulation therapy and 59 who underwent open aortic surgery were included in the outcome analysis. Among 29 patients treated with initial anticoagulation therapy, the persistence of AAT was observed in 11 patients (38%) and recurrent arterial embolization was developed in 6 patients (21%). All 11 patients in the anticoagulation group underwent secondary aortic surgery for the persistence of AAT with uneventful postoperative course. Compared with patients treated with primary aortic surgery, patients treated with initial anticoagulation therapy had higher risk of recurrent embolization (P = 0.002). No significant difference existed in the mortality rates between the groups (P = 0.106). Hemodynamic instability was an independent predictor of mortality (P = 0.008). CONCLUSIONS: Anticoagulation therapy and open aortic surgery for AAT show similar results; however, open aortic surgery reliably removes AAT and reduces the risk of recurrent embolization compared with anticoagulation therapy. Furthermore, the preoperative hemodynamic status significantly influences the clinical outcome and is a strong predictor of prognosis.


Subject(s)
Anticoagulants/therapeutic use , Aorta/surgery , Aortic Diseases/therapy , Fibrinolytic Agents/therapeutic use , Thrombosis/therapy , Vascular Surgical Procedures , Adult , Aged , Aged, 80 and over , Anticoagulants/adverse effects , Aorta/diagnostic imaging , Aorta/physiopathology , Aortic Diseases/diagnostic imaging , Aortic Diseases/mortality , Aortic Diseases/physiopathology , Female , Fibrinolytic Agents/adverse effects , Hemodynamics , Humans , Male , Middle Aged , Postoperative Complications/etiology , Recurrence , Risk Assessment , Risk Factors , Thrombosis/diagnostic imaging , Thrombosis/mortality , Thrombosis/physiopathology , Treatment Outcome , Vascular Surgical Procedures/adverse effects , Vascular Surgical Procedures/mortality
4.
J Cardiothorac Surg ; 15(1): 295, 2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33008478

ABSTRACT

BACKGROUND: Veno-arterial extracorporeal membrane oxygenation (ECMO) is increasingly being utilized in patients with massive pulmonary embolism (PE). However, the efficacy and the safety remain uncertain. This study aimed to investigate clinical courses and outcomes in ECMO-treated patients with acute PE. METHODS: Twenty-one patients with acute PE rescued by ECMO from January 2012 to December 2019 were retrospectively analysed. Clinical features, laboratory biomarkers, and imaging findings of these patients were reviewed, and the relationship with immediate outcome and clinical course was investigated. RESULTS: Sixteen patients (76.2%) experienced refractory circulatory collapse requiring cardiopulmonary resuscitation (CPR) or ECMO support within 2 h after the onset of cardiogenic shock, and none could receive definitive reperfusion therapy before ECMO initiation. Before or during ECMO support, more than 90% of patients had imaging signs of right ventricular (RV) dysfunction. In normotension patients, the computed tomography (CT) value was a valuable predictor of rapid disease progression compared with cardiac troponin I level. Ultimately, in-hospital death occurred in ten patients (47.6%) and 90% of them died of prolonged CPR-related brain death. Cardiac arrest was a significant predictor of poor prognosis (p = 0.001). CONCLUSIONS: ECMO appears to be a safe and effective circulatory support in patients with massive PE. Close monitoring in intensive care unit is recommended in patients with RV dysfunction and aggressive use of ECMO may reduce the risk of sudden cardiac arrest and improve clinical outcome.


Subject(s)
Extracorporeal Membrane Oxygenation , Pulmonary Embolism/therapy , Acute Disease , Adult , Aged , Aged, 80 and over , Brain Death , Cardiopulmonary Resuscitation , Female , Heart Arrest/etiology , Heart Arrest/therapy , Humans , Male , Middle Aged , Pulmonary Embolism/complications , Reperfusion , Retrospective Studies , Shock, Cardiogenic/therapy , Tomography, X-Ray Computed , Ventricular Dysfunction, Right/etiology
5.
Cell Transplant ; 29: 963689720954140, 2020.
Article in English | MEDLINE | ID: mdl-33050736

ABSTRACT

This study tested the hypothesis that both allogenic adipose-derived mesenchymal stem cells (ADMSCs) and human inducible pluripotent stem cell-derived MSCs (iPS-MSCs) offered a comparable effect for protecting the lung against ischemia-reperfusion (IR) injury in rodent through downregulating the inflammatory, oxidative stress, and autophagic signaling pathways. Adult male Sprague-Dawley rats (n = 32) were categorized into group 1 (sham-operated control), group 2 (IRI), group 3 [IRI + ADMSCs (1.0 × 106 cells)/tail-vein administration at 0.5/18/36 h after IR], and group 4 [IRI + iPS-MSCs (1.0 × 106 cells)/tail-vein administration at 0.5/18/36 h after IR], and lungs were harvested at 72 h after IR procedure. In vitro study demonstrated that protein expressions of three signaling pathways in inflammation (TLR4/MyD88/TAK1/IKK/I-κB/NF-κB/Cox-2/TNF-α/IL-1ß), mitochondrial damage/cell apoptosis (cytochrome C/cyclophilin D/DRP1/ASK1/APAF-1/mitochondrial-Bax/caspase3/8/9), and autophagy/cell death (ULK1/beclin-1/Atg5,7,12, ratio of LCB3-II/LC3B-I, p-AKT/m-TOR) were significantly higher in lung epithelial cells + 6h hypoxia as compared with the control, and those were significantly reversed by iPS-MSC treatment (all P < 0.001). Flow cytometric analysis revealed that percentages of the inflammatory cells in bronchioalveolar lavage fluid and circulation, and immune cells in circulation/spleen as well as circulatory early and late apoptotic cells were highest in group 2, lowest in group 1, and significantly higher in group 3 than in group 4 (all P < 0.0001). Microscopy showed the lung injury score and numbers of inflammatory cells and Western blot analysis showed the signaling pathways of inflammation, mitochondrial damage/cell apoptosis, autophagy, and oxidative stress exhibited an identical pattern of flow cytometric results among the four groups (all P < 0.0001). Both xenogeneic and allogenic MSCs protected the lung against IRI via suppressing the inflammatory, oxidative stress, and autophagic signaling.


Subject(s)
Autophagy , Down-Regulation , Inflammation/pathology , Lung/pathology , Mesenchymal Stem Cells/cytology , Oxidative Stress , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Animals , Apoptosis , Biomarkers/metabolism , Cell Death , Cell Proliferation , DNA Damage , Induced Pluripotent Stem Cells/cytology , Male , Mesenchymal Stem Cell Transplantation , Mitochondria/pathology , Models, Biological , Oxygen , Rats, Sprague-Dawley , Signal Transduction , Transplantation, Heterologous , Transplantation, Homologous
6.
Stem Cell Res Ther ; 11(1): 324, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32727585

ABSTRACT

BACKGROUND: Treating patients with end-stage diffuse coronary artery disease (EnD-CAD) unsuitable for coronary intervention remains a clinical challenge. They usually express refractory angina and have a high risk of mortality. Although growing data have indicated cell therapy is an alternative solution to medical or invasive therapy, there are still lacking useful markers to predict whether heart function will improve in the EnD-CAD patients who underwent circulatory-derived CD34+ cell therapy. By utilizing the baseline variables and results from our previous phase I/II clinical trials, the aim of this study tried to elucidate the variables predictive of the "good response" to CD34+ cell therapy. METHODS: This retrospective study included 38 patients in phase I clinical trial (2011-2014), and 30 patients in phase II clinical trial (2013-2017). These patients were categorized into "good responders" and "non-responders" according to their 1-year improvement of LVEF ≥ 7.0% or < 7.0% after intracoronary CD34+ cell therapy. Univariate and multivariate logistic regression models were performed to identify potential independent predictors of a good responder to cell therapy, followed by Hosmer-Lemeshow (H-L) test for goodness of fit and prediction power. RESULTS: Among baseline data, multivariate analysis demonstrated that the history of a former smoker was independently predictive of good responders (p = 0.006). On the other hand, male gender, the baseline Canadian Cardiovascular Society angina score ≥ 3, and grades of LV diastolic dysfunction ≥ 2 were significantly negative predictors of good responders (all p < 0.01). After administration of subcutaneous granulocyte-colony stimulating factor (G-CSF), a higher post-G-CSF neutrophil count in addition to the above four baseline variables also played crucial roles in early prediction of good response to CD34+ cell therapy for EnD-CAD (all p < 0.03). The H-L test displayed a good prediction power with sensitivity 83.3%, specificity 85.3%, and accuracy 84.4%. CONCLUSIONS: Using the results of our phase I/II clinical trials, previous smoking habit, female sex, lower grades of angina score, and diastolic dysfunction were identified to be independently predictive of "good response" to CD34+ cell therapy in the patients with EnD-CAD. TRIAL REGISTRATION: This is a retrospective analysis based on phase I ( ISRCTN72853206 ) and II ( ISRCTN26002902 ) clinical trials.


Subject(s)
Coronary Artery Disease , Canada , Cell- and Tissue-Based Therapy , Coronary Artery Disease/therapy , Female , Granulocyte Colony-Stimulating Factor , Humans , Male , Retrospective Studies
7.
J Cell Mol Med ; 24(15): 8379-8390, 2020 08.
Article in English | MEDLINE | ID: mdl-32639656

ABSTRACT

Increased circulating level of uraemic solute p-cresyl sulphate (PCS) in patients with chronic kidney disease (CKD) is known to increase myocardial burden relevant to mitochondrial abnormalities. This study aimed at investigating mitochondrial response to PCS in H9C2 cardiomyoblasts. H9C2 cardiomyoblasts were treated with four different concentrations of PCS (3.125, 6.25, 12.5 and 25.0 µg/mL) to study the changes in cell proliferation, cell size and mitochondrial parameters including morphology, respiration, biogenesis and membrane potential. The lowest effective dose of PCS (6.25 µg/mL) induced mitochondrial hyperfusion with enhanced mitochondrial connectivity, mitochondrial oxygen consumption rates, mitochondrial mass, mitochondrial DNA copy number and increased volume of cardiomyoblasts. After PCS treatment, phosphorylation of energy-sensing adenosine monophosphate-activated protein kinase (AMPK) was increased without induction of apoptosis. In contrast, mitochondrial mass was recovered after AMPK silencing. Additionally, mitochondrial hyperfusion and cell volume were reversed after cessation of PCS treatment. The results of the present study showed that low-level PCS not only caused AMPK-dependent mitochondrial hyperfusion but also induced cell enlargement in H9C2 cardiomyoblasts in vitro.


Subject(s)
Cresols/pharmacology , Mitochondria/drug effects , Myocytes, Cardiac/drug effects , Sulfuric Acid Esters/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Oxygen Consumption/drug effects , Rats
8.
J Clin Med ; 9(4)2020 Apr 07.
Article in English | MEDLINE | ID: mdl-32272750

ABSTRACT

This phase II randomized controlled trial tested whether intracoronary autologous CD34+ cell therapy could further improve left ventricular (LV) systolic function in patients with diffuse coronary artery disease (CAD) with relatively preserved LV ejection fraction (defined as LVEF >40%) unsuitable for coronary intervention. Between December 2013 and November 2017, 60 consecutive patients were randomly allocated into group 1 (CD34+ cells, 3.0 × 107/vessel/n = 30) and group 2 (optimal medical therapy; n = 30). All patients were followed for one year, and preclinical and clinical parameters were compared between two groups. Three-dimensional echocardiography demonstrated no significant difference in LVEF between groups 1 and 2 (54.9% vs. 51.0%, respectively, p = 0.295) at 12 months. However, compared with baseline, 12-month LVEF was significantly increased in group 1 (p < 0.001) but not in group 2 (p = 0.297). From baseline, there were gradual increases in LVEF in group 1 compared to those in group 2 at 1-month, 3-months, 6-months and 12 months (+1.6%, +2.2%, +2.9% and +4.6% in the group 1 vs. -1.6%, -1.5%, -1.4% and -0.9% in the group 2; all p < 0.05). Additionally, one-year angiogenesis (2.8 ± 0.9 vs. 1.3 ± 1.1), angina (0.4 ± 0.8 vs. 1.8 ± 0.9) and HF (0.7 ± 0.8 vs. 1.8 ± 0.6) scores were significantly improved in group 1 compared to those in group 2 (all p < 0.001). In conclusion, autologous CD34+ cell therapy gradually and effectively improved LV systolic function in patients with diffuse CAD and preserved LVEF who were non-candidates for coronary intervention (Trial registration: ISRCTN26002902 on the website of ISRCTN registry).

9.
Crit Care Med ; 48(5): e391-e399, 2020 05.
Article in English | MEDLINE | ID: mdl-32187077

ABSTRACT

OBJECTIVES: To investigate the safety, feasibility, and possible adverse events of single-dose human umbilical cord-derived mesenchymal stem cells in patients with moderate-to-severe acute respiratory distress syndrome. DESIGN: Prospective phase I clinical trial. SETTING: Medical center in Kaohsiung, Taiwan. PATIENTS: Moderate-to-severe acute respiratory distress syndrome with a PaO2/FIO2 ratio less than 200. INTERVENTIONS: Scaling for doses was required by Taiwan Food and Drug Administration as follows: the first three patients received low-dose human umbilical cord-derived mesenchymal stem cells (1.0 × 10 cells/kg), the next three patients with intermediate dose (5.0 × 10 cells/kg), and the final three patients with high dose (1.0 × 10 cells/kg) between December 2017 and August 2019. MEASUREMENTS AND MAIN RESULTS: Nine consecutive patients were enrolled into the study. In-hospital mortality was 33.3% (3/9), including two with recurrent septic shock and one with ventilator-induced severe pneumomediastinum and subcutaneous emphysema. No serious prespecified cell infusion-associated or treatment-related adverse events was identified in any patient. Serial flow-cytometric analyses of circulating inflammatory biomarkers (CD14CD33/CD11b+CD16+/CD16+MPO+/CD11b+MPO+/CD14CD33+) and mesenchymal stem cell markers (CD26+CD45-/CD29+CD45-/CD34+CD45-/CD44+CD45-/CD73+CD45-/CD90+CD45-/CD105+CD45-/CD26+CD45-) were notably progressively reduced (p for trend < 0.001), whereas the immune cell markers (Helper-T-cell/Cytotoxity-T-cell/Regulatory-T-cell) were notably increased (p for trend < 0.001) after cell infusion. CONCLUSIONS: The result of this phase I clinical trial showed that a single-dose IV infusion of human umbilical cord-derived mesenchymal stem cells was safe with favorable outcome in nine acute respiratory distress syndrome patients.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Respiratory Distress Syndrome/therapy , Umbilical Cord/physiology , Adult , Aged , Drug Dosage Calculations , Female , Hospital Mortality/trends , Humans , Male , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cell Transplantation/mortality , Mesenchymal Stem Cells/classification , Middle Aged , Prospective Studies , Respiratory Distress Syndrome/mortality , Severity of Illness Index
10.
Am J Transl Res ; 11(9): 5375-5389, 2019.
Article in English | MEDLINE | ID: mdl-31632517

ABSTRACT

This study tested the hypothesis that early administration with cold water (CW)-assisted adipose-derived mesenchymal stem cell (ADMSC)-derived exosome (Exo) therapy was superior to either one on protecting the heart against ischemia-reperfusion (IR) (i.e., by ligation of 50 minutes and relieved by day 5 prior to euthanizing the animals) injury. Adult-male SD rats (n=30) were equally categorized into groups 1 (sham-operated control), 2 (IR), 3 (IR + CW), 4 (IR + Exo) and 5 (IR + CW-Exo). The left ventricular ejection fraction (LVEF) was highest in group 1, lowest in group 2, and significantly higher in group 5 than in groups 3 and 4, but no difference between groups 3 and 4 (all P<0.001). The protein expressions of oxidative-stress (NOX-1/NOX-2/NOX-4/oxidized protein), apoptotic/mitochondrial-damaged (mitochondrial-Bax/caspase 3/PARP/p53/cytosolic-cytochrome-C) and inflammatory (IL-1ß/TNF-α/NF-κB/MMP-9) biomarkers, and cellular-stress response signaling (PI3K/Akt/GSK3ß and p-m-TOR) showed an opposite pattern, whereas the anti-oxidants (SIRT1/SIRT3), anti-inflammation (IL-10) and IKB-α/p-AMKP/mitochondrial-cytochrome-C exhibited an identical pattern to the LVEF among the five groups (all P<0.0001). The cellular expressions of inflammation (CD68), total cellular ROS (i.e., stained by H2DCFDA) and the LV infarct/fibrotic/collagen-deposition areas displayed an opposite pattern, whereas the cell gap junction (coonexin 43) and sarcomere length exhibited an identical pattern of LVEF among the five groups (all P<0.0001). Conclusion: Combined CW-exosome therapy markedly protected the heart against IR injury.

11.
Biomed Pharmacother ; 118: 109296, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31545254

ABSTRACT

BACKGROUND: This study tested the hypothesis that intramuscular injections of erythropoietin (EPO) and endothelial progenitor cells (EPC) to critical limb ischemia (CLI; primary treatment site) could also improve heart function in rat after acute myocardial infarction (AMI; remote ischemic organ). METHOD: Adult-male SD rats (n = 40) were equally categorized into group 1 (sham-operated control), group 2 (CLI-AMI), group 3 [CLI-AMI + EPO (10 mg/kg)], group 4 [CLI-AMI + EPCs (1.2 × 106)] and group 5 (CLI-AMI + EPCs + EPO). RESULTS: By day 21 (end of study period), 2-D echo and Laser doppler showed that left-ventricular injection fraction (LVEF) and the ratio of ischemic to normal blood flow were highest in group 1, lowest in group 2, significantly higher in group 5 than in groups 3 and 4, but not different in the latter two groups (all p < 0.0001). Flow cytometry and ELISA demonstrated that circulating angiogenesis factors were significantly progressively increased from groups 1 to 5 (all p < 0.001). The number of small vessels and protein (CD31/eNOS)/cellular (vWF) expressions reflecting integrity of endothelium exhibited an identical pattern to LVEF whereas protein (VEGF/SDF-1α)/cellular (VEGF) expressions were significantly progressively increased from groups 1 to 5 in quadriceps and heart tissues (all p < 0.0001). Protein expressions of apoptotic (Bax/caspase-3/PARP)/inflammatory (MMP-9) and microscopic findings of ischemic/fibrotic/collagen-deposition areas and DNA-damage marker (γ-H2AX+) were lowest in group 1 and significantly progressively decreased from groups 2 to 5 in quadriceps and heart tissues (all p < 0.0001). CONCLUSIONS: Direct injection of EPO-EPC into CLI effectively restored blood flow in the CLI area and also preserved remote heart function.


Subject(s)
Endothelial Progenitor Cells/transplantation , Erythropoietin/pharmacology , Heart Ventricles/physiopathology , Ischemia/therapy , Myocardial Infarction/physiopathology , Regional Blood Flow/drug effects , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Blood Vessels/drug effects , Blood Vessels/pathology , Collagen/metabolism , DNA Damage , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Heart Ventricles/drug effects , Male , Myocardium/pathology , Neovascularization, Physiologic/drug effects , Rats, Sprague-Dawley , Stroke Volume/drug effects
12.
Am J Transl Res ; 11(3): 1282-1298, 2019.
Article in English | MEDLINE | ID: mdl-30972162

ABSTRACT

Endothelial cell (EC) dysfunction plays a crucial role for arterial obstructive disease. This study tested the therapeutic role of autologous endothelial progenitor cells (EPCs)/rosuvastatin-(Rosu)/valsartan-(Val) on repair of injured carotid ECs. Male Sprague-Dawley rats (n = 60) were categorized into five groups [sham-control (SC), left common carotid artery injury induced by balloon denudation (LCABD), LCABD + Rosu (10 mg/kg/day), LCABD + Val (20 mg/kg/day), and LCABD + EPC (1.2 × 106)]. By day 5, the LCA was harvested from each rat (n = 6/each time interval in group) after the procedure. Carotid-ring angiogenesis was significantly lower in LCABD than the other groups (all P < 0.001). Compared with LCABD, the number of EC was significantly higher in LCABD treated with adipose-derived mesenchymal stem cells (ADMSCs) and more significantly higher in LCABD treated with EPCs (all P < 0.001). Gene expression of EC (CD31/vWF), EPC (SDF-1α/CXCR4) and angiogenesis (VEGF/VEGF-receptor/angiopoietin/eNOS) and EC intercellular junction (VE-cadherin) biomarkers were significantly lower in LCABD than in groups LCABD + Rosu to LCABD + EPC (all P < 0.001). Conversely, the gene expression of inflammatory (VCAM-1/MMP-9/TNF-α), oxidative-stress (NOX-1/NOX-2), apoptosis (cleaved caspase-3/PARP) and thrombin cofactor (thrombomodulin) biomarkers were significantly higher in LCABD than in other groups (all P < 0.001). By day 14, the neointimal-layer area and cellular expressions of (CD40+/CD68+) were highest in LCABD, lowest in SC, significantly higher in LCABD + Val than in LCABD + Rosu and LCABD + EPC (all P < 0.001). In conclusion, EPCs were comparable to rosuvastatin and valsartan in upregulation of angiogenesis and repair of injured carotid ECs.

13.
Biomed Pharmacother ; 113: 108737, 2019 May.
Article in English | MEDLINE | ID: mdl-30852418

ABSTRACT

This study tested the hypothesis that exposure to ambient fine particulate matter (PM2.5) pollution increased susceptibility of rat lung to damage from acute ischemia-reperfusion (IR) injury that was reversed by melatonin (Mel) treatment. Male-adult SD rats (n = 30) were categorized into group 1 (normal control), group 2 (PM2.5 only), group 3 (IR only at day 8 after PM2.5 exposure), group 4 (PM2.5 + IR) and group 5 (PM2.5 + IR + Mel), and all animals were sacrificed by day 10 after PM2.5 exposure. Oxygen saturation (%) was significantly higher in group 1 than in other groups and significantly lower in group 4 than in groups 2, 3 and 5 but it did not differ among the latter three groups (p < 0.01). Pulmonary protein expressions of inflammation (MMP-9/TNF-α/NF-kB), oxidative stress (NOX-1/NOX-2/oxidized protein), apoptosis (mitochondrial-Bax/caspase-3/PARP) and fibrosis were lowest in group 1, highest in group 4, significantly higher in group 5 than in groups 2 and 3 (all p < 0.0001), but they did not differ between groups 2 and 3. Inflammatory cell infiltration in lung parenchyma, specific inflammatory cell surface markers (CD14+, F4/88+), allergic inflammatory cells (IgE+, eosinophil+), number of goblet cells, thickness of tracheal epithelial layer and fibrotic area exhibited an identical pattern of protein expressions to inflammation among the five groups (all p < 0.0001). In conclusion, lung parenchymal damage and a rigorous inflammatory response were identified in rodent even with short-term PM2.5 exposure.


Subject(s)
Melatonin/pharmacology , Oxidative Stress/drug effects , Particulate Matter/toxicity , Reperfusion Injury/prevention & control , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Disease Models, Animal , Inflammation/etiology , Inflammation/prevention & control , Lung/drug effects , Lung/pathology , Male , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Oxygen/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Tumor Necrosis Factor-alpha/metabolism
14.
Am J Transl Res ; 11(1): 67-86, 2019.
Article in English | MEDLINE | ID: mdl-30787970

ABSTRACT

We tested the hypothesis that adipose-derived fresh stromal vascular fraction (SVF) is non-inferior to conventional adipose-derived mesenchymal stem cell (ADMSC) therapy for improving left-ventricular ejection fraction (LVEF) in rat after acute myocardial infarction (AMI). Male-adult SD rats (n = 48) were categorized into group 1 (sham control), AMI, AMI + ADMSCs (1.2 × 106) cells] and AMI + SVF (1.2 × 106) cells]. Flow cytometric and qPCR analyses showed that the expressions of surface biomarkers for endothelial progenitor cells, and cardiac-stem cells were significantly higher in the SVF population than in the ADMSC population, whereas MSCs showed a reversed pattern between these two groups (all P < 0.001). By day-42 after AMI, LVEF was highest in SC, lowest in AMI, and significantly higher in AMI + SVF than in AMI + ADMSCs (P < 0.0001). Protein expression indicating angiogenesis, anti-inflammatory/anti-apoptotic, mitochondrial/bioenergy-integrity and antifibrotic biomarkers showed an identical pattern, whereas protein expressions for inflammatory, apoptotic and pressure-overload/heart failure biomarkers exhibited an opposite pattern to LVEF among the four groups (all P < 0.001). Histopathology displayed that LV infarction/fibrotic area/collagen-deposition areas, cellular expressions of DNA-damage, and inflammatory biomarkers exhibited an opposite pattern, whereas cellular expressions of endothelial/gap-junction biomarkers showed an identical pattern to LVEF among the four groups (all P < 0.0001). Cellular expression of angiogenesis biomarkers significantly and progressively increased from groups 1 to 4 (all P < 0.0001). In conclusion, SVF may be better than ADMSC at improving LVEF in rat after AMI.

16.
J Neuropathol Exp Neurol ; 78(1): 65-77, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30481326

ABSTRACT

This study tested the hypothesis that combined adipose-derived mesenchymal stem cell (ADMSC) and low-energy extracorporeal shock wave (ECSW) therapy could protect brain from brain death (BD)-induced injury. Adult male Sprague Dawley rats were categorized into group 1 (sham control), group 2 (BD), group 3 (BD + ECSW [0.15 mJ/mm2/300 impulses] applied to the skull surface 3 hours after BD induction), group 4 (BD + ADMSC [1.2 × 106 cell] by intravenous injection 3 hours after BD induction) and group 5 (BD + ECSW + ADMSC). By 6 hours after BD induction, circulating/spleen levels of immune cells (CD3/CD4+, CD8/CD4+, Treg+) and circulating levels of inflammatory cells (MPO/Ly6G/CD11a/b) and soluble mediators (TNF-α/IL-6) were lowest in group 1 and significantly progressively reduced from groups 2 to 5 (all p < 0.0001). Brain protein expressions of inflammatory (TNF-α/NF-κB/MMP-9/IL-1ß), apoptotic (caspase-3/PARP/mitochondrial-BAX), oxidative stress/DNA-damage (NOX-1/NOX-2/oxidized protein/γ-H2AX) biomarkers exhibited an identical pattern, whereas anti-oxidant (SIRT1/SIRT3) and mitochondrial-integrity (mitochondrial-cytochrome-C) biomarkers exhibited an opposite pattern to inflammatory biomarkers among the 5 groups (all p < 0.0001). The cellular expressions of inflammatory/brain-edema (F4/80/CD14+/GFAP/AQP4) biomarkers exhibited an identical pattern to inflammation among the 5 groups (all p < 0.0001). In conclusion, ECSW-ADMSC therapy is superior to either alone for attenuating brain from BD-induced damage.


Subject(s)
Brain Death/pathology , Brain Injuries/prevention & control , Extracorporeal Shockwave Therapy/methods , Mesenchymal Stem Cell Transplantation/methods , Adipose Tissue/cytology , Animals , Brain Injuries/etiology , Male , Rats , Rats, Sprague-Dawley
17.
Oxid Med Cell Longev ; 2018: 7518920, 2018.
Article in English | MEDLINE | ID: mdl-30416645

ABSTRACT

This study tested the hypothesis that extracorporeal shock wave- (ECSW-) assisted adipose-derived stromal vascular fraction (SVF) therapy could preserve left ventricular ejection fraction (LVEF) and inhibit LV remodeling in a rat after acute myocardial infarction (AMI). Adult male SD rats were categorized into group 1 (sham control), group 2 (AMI induced by left coronary artery ligation), group 3 [AMI + ECSW (280 impulses at 0.1 mJ/mm2, applied to the chest wall at 3 h, days 3 and 7 after AMI), group 4 [AMI + SVF (1.2 × 106) implanted into the infarct area at 3 h after AMI], and group 5 (AMI + ECSW-SVF). In vitro, SVF protected H9C2 cells against menadione-induced mitochondrial damage and increased fluorescent intensity of mitochondria in nuclei (p < 0.01). By day 42 after AMI, LVEF was highest in group 1, lowest in group 2, significantly higher in group 5 than in groups 3 and 4, and similar between the latter two groups (all p < 0.0001). LV remodeling and infarcted, fibrotic, and collagen deposition areas as well as apoptotic nuclei exhibited an opposite pattern to LVEF among the groups (all p < 0.0001). Protein expressions of CD31/vWF/eNOS/PGC-1α/α-MHC/mitochondrial cytochrome C exhibited an identical pattern, whilst protein expressions of MMP-9/TNF-α/IL-1ß/NF-κB/caspase-3/PARP/Samd3/TGF-ß/NOX-1/NOX-2/oxidized protein/ß-MHC/BNP exhibited an opposite pattern to LVEF among five groups (all p < 0.0001). Cellular expressions of CXCR4/SDF-1α/Sca-1/c-Kit significantly and progressively increased from groups 1 to 5 (all p < 0.0001). Cellular expression of γ-H2AX/CD68 displayed an opposite pattern to LVEF among the five groups (all p < 0.0001). In conclusion, ECSW-SVF therapy effectively preserved LVEF and inhibited LV remodeling in rat AMI.


Subject(s)
Adipose Tissue/metabolism , Extracorporeal Shockwave Therapy , Myocardial Infarction/physiopathology , Ventricular Function, Left/physiology , Ventricular Remodeling/physiology , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Coculture Techniques , Collagen/metabolism , DNA Damage , Echocardiography , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mitochondrial Dynamics/drug effects , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Stromal Cells/drug effects , Stromal Cells/metabolism , Time Factors , Tumor Suppressor p53-Binding Protein 1/metabolism , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Vitamin K 3/pharmacology , X-ray Repair Cross Complementing Protein 1/metabolism
18.
Am J Transl Res ; 10(9): 2975-2989, 2018.
Article in English | MEDLINE | ID: mdl-30323884

ABSTRACT

This phase I clinical trial tested the hypothesis that circulatory CD34+ cell therapy might be safe for old ischemic stroke (IS) (defined as IS>6 months) patients and also to evaluate the neurological function after the therapy. Nine old IS patients (with mean IS interval: 8.6 ± 6.4 years) were consecutively enrolled and received intra-carotid artery transfusion of circulatory-derived autologous CD34+ cells (3.0×107 cells/patient) into the ipsilateral brain infarct area at catheterization room by Catheter Looping Technique, after subcutaneous G-CSF injection (5 µg/kg twice a day for 4 days). The results showed that procedural safety was 100% with all patients uneventfully discharged. The circulating number of EPCs and angiogenesis (i.e., by Matrigel assay) were significantly higher at post than at prior to G-CSF treatment (all P<0.001). Time courses (0/5/10/30 minutes) of blood samplings from right-internal jugular vein exhibited significantly increased in levels of SDF-1α and EPCs numbers in time points of 5/10/30 minutes than in the baseline (0 minute) (all P<0.05). Barthel index was increased (defined as ≥5 scores) in 44.4% (4/9) and CASI score was notably improved (all P<0.01) at 6-month follow-up after the cell therapy as compared to the baseline. No recurrent IS or any tumorigenesis was found in these patients with a mean follow-up time interval of 16.5 ± 6.2 months. All of these patients remain survive and are followed up at outpatient department. In conclusion, CD34+ cell therapy is safe and might offer some benefit to old IS patients.

19.
Am J Transl Res ; 10(8): 2290-2305, 2018.
Article in English | MEDLINE | ID: mdl-30210671

ABSTRACT

This study tested the hypothesis that entresto therapy effectively protected heart and lung against cardiopulmonary syndrome (CPS) caused by transverse aortic constriction (TAC) in rat. Adult-Male SD rats (n = 36) were equally categorized into group 1 [sham-operated control (SC)], group 2 [SC + enalapril (7 mg/kg/day) since day-28 after TAC induction], group 3 [SC + entresto (30 mg/kg/day) since day-14 after TAC induction], group 4 (TAC only), group 5 (TAC + enalapril) and group 6 (TAC + entresto) and euthanized at day 60 after TAC induction. By day 60, the left-ventricular (LV) ejection fraction was significantly lower in group 4 than in other groups and significantly lower in groups 5 and 6 than in groups 1 to 3, whereas the ratios of heart and lung weights to tibial-length as well as the right-ventricular-systolic blood pressure exhibited an opposite pattern among the groups (all P<0.001). The sarcomere-length (SL), LV fibrotic area, cardiomyocyte size, and lung injury score were highest in group 4, lowest in groups 1 to 3 and significantly lower in group 6 than in group 5 (all P<0.0001). The protein expressions of fibrotic (Smad3/TGF-ß), apoptotic (mitochondrial-Bax/cleaved-caspase3/PARP) and DNA-damaged (γ-H2AX) markers in lung and LV myocardium as well as oxidative (NOX-1/NOX2/oxidized protein) in LV myocardium exhibited an identical pattern of SL (all P<0.0001). The protein expressions of pressure/volume overload (BNP/MHC-ß) mitochondrial-damaged (cytosolic cytochrome-C) of LV myocardium exhibited an identical pattern of SL (all P<0.001). In conclusion, Entresto is non-inferior to enalapril for protecting the heart-lung against CPS.

20.
Int J Mol Sci ; 19(9)2018 Sep 15.
Article in English | MEDLINE | ID: mdl-30223594

ABSTRACT

Myocardial ischemia-reperfusion (IR) injury contributes to adverse cardiac outcomes after myocardial ischemia, cardiac surgery, or circulatory arrest. In this study, we evaluated the ability of combined SS31-mitochondria (Mito) therapy to protect heart cells from myocardial IR injury. Adult male SD rats (n = 8/each group) were randomized: group 1 (sham-operated control), group 2 (IR, 30-min ischemia/72 h reperfusion), group 3 (IR-SS31 (2 mg intra-peritoneal injection at 30 min/24 h/48 h after IR)), group 4 (IR-mitochondria (2 mg/derived from donor liver/intra-venous administration/30 min after IR procedure)), and group 5 (IR-SS31-mitochondria). In H9C2 cells, SS31 suppressed menadione-induced oxidative-stress markers (NOX-1, NOX-2, oxidized protein) while it increased SIRT1/SIRT3 expression and ATP levels. In adult male rats 72 h after IR, left ventricular ejection fraction (LVEF) was highest in sham-operated control animals and lowest in the IR group. LVEF was also higher in IR rats treated with SS31-Mito than untreated IR rats or those treated with Mito or SS31 alone. Areas of fibrosis/collagen-deposition showed the opposite pattern. Likewise, levels of oxidative-stress markers (NOX-1, NOX-2, oxidized protein), inflammatory markers (MMP-9, CD11, IL-1ß, TNF-α), apoptotic markers (mitochondrial-Bax, cleaved-caspase-3, PARP), fibrosis markers (p-Smad3, TGF-ß), DNA-damage (γ-H2AX), sarcomere-length, and pressure/volume overload markers (BNP, ß-MHC) all showed a pattern opposite that of LVEF. Conversely, anti-apoptotic (BMP-2, Smad1/5) and energy integrity (PGC-1α/mitochondrial cytochrome-C) markers exhibited a pattern identical to that of LVEF. This study demonstrates that the combined SS31-Mito therapy is superior to either therapy alone for protecting myocardium from IR injury and indicates that the responsible mechanisms involved increased SIRT1/SIRT3 expression, which suppresses inflammation and oxidative stress and protects mitochondrial integrity.


Subject(s)
Mitochondria/drug effects , Mitochondria/metabolism , Myocardial Reperfusion Injury/metabolism , Oligopeptides/pharmacology , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cell Line , Collagen/metabolism , DNA Copy Number Variations , DNA Damage/drug effects , Disease Models, Animal , Echocardiography , Inflammation Mediators/metabolism , Male , Mitochondria/genetics , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/drug therapy , Myocardium/metabolism , Oxidative Stress/drug effects , Oxygen Consumption , Rats , Sirtuin 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...