Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Leukemia ; 37(9): 1842-1849, 2023 09.
Article in English | MEDLINE | ID: mdl-37507426

ABSTRACT

Intensive induction chemotherapy achieves complete remissions (CR) in >60% of patients with acute myeloid leukemia (AML) but overall survival (OS) is poor for relapsing patients not eligible for allogeneic hematopoietic stem cell transplantation (allo-HSCT). Oral azacytidine may be used as maintenance treatment in AML in first remission, but can be associated with substantial side effects, and less toxic strategies should be explored. Twenty AML patients in first CR (CR1) ineligible for allo-HSCT were treated with FDC101, an autologous RNA-loaded mature dendritic cell (mDC) vaccine expressing two leukemia-associated antigens (LAAs). Each dose consisted of 2.5-5 × 106 mDCs per antigen, given weekly until week 4, at week 6, and then monthly, during the 2-year study period. Patients were followed for safety and long-term survival. Treatment was well tolerated, with mild and transient injection site reactions. Eleven of 20 patients (55%) remained in CR, while 4 of 6 relapsing patients achieved CR2 after salvage therapy and underwent allo-HSCT. OS at five years was 75% (95% CI: 50-89), with 70% of patients ≥60 years of age being long-term survivors. Maintenance therapy with this DC vaccine was well tolerated in AML patients in CR1 and was accompanied by encouraging 5-year long-term survival.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Humans , Induction Chemotherapy , Transplantation, Homologous , Leukemia, Myeloid, Acute/therapy , Remission Induction , Recurrence , Dendritic Cells , Retrospective Studies , Antigens, Neoplasm , WT1 Proteins/genetics
2.
Cell Cycle ; 10(14): 2281-6, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21654195

ABSTRACT

Potentially, adult stem cell-based therapy provides a new therapeutic option for myocardial regeneration. However, to date, with regard to the benefits seen, the mechanisms involved in stem cell-based therapy are not well understood. Suggested pathways proposed so far include fusion of stem cells with cardiomyocytes, transdifferentiation into cardiac and vascular cells and secretion of paracrine factors. In a recent study, our group examined the fate of human adipose tissue-derived stem cells (hASCs) fused with rat cardiomyocytes after treatment with fusion-inducing hemagglutinating virus of Japan (HVJ). In this study, we demonstrated that cells of fused hASC cardiomyocytes display a cardiomyocyte phenotype and spontaneous rhythmic contraction and generate an action potential in vitro. As part of the work underlying this paper, we co-cultured rat neonatal cardiomyocytes with hASCs or pig bone marrow-derived mesenchymal stem cells (MSCs), where ASCs or MSCs had previously been transduced with a lentivirus encoding eGFP. Our data evidence early cardiac contractile proteins, such as Titin and MF20, identified in eGFP-positive cells, suggesting a cardiomyogenic phenotype. Recent work by others has shown that the myogenic conversion increased when BMSCs were cultured with apoptotic cells. In this Extra View article, we review the current understanding of stem cell-derived factors, fusion/partial fusion and the manner in which the exchange of cellular contents between stem cells and cardiomyocytes might contribute to the reprogramming of fully differentiated cardiomyocytes based on recently published literature.


Subject(s)
Cell Transdifferentiation , Mitochondria/physiology , Stem Cells/cytology , Animals , Cell Fusion , Heart/physiology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Paracrine Communication , Regeneration , Stem Cells/metabolism
3.
Kidney Int ; 79(11): 1161-3, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21566637

ABSTRACT

Yeagy and colleagues present long-term data from a preclinical model of cystinosis after hematopoietic stem cell transplantation. The results suggest a therapeutic benefit independent of target tissue differentiation but dependent on the level of bone marrow chimerism. The mode of action remains mysterious, but positive effects are seen. Although the work presents a potential therapeutic option for an otherwise dismal disease, the search for the mechanism of action in cellular therapies continues.


Subject(s)
Bone Marrow Transplantation , Cystinosis/surgery , Hematopoietic Stem Cell Transplantation , Kidney Diseases/prevention & control , Kidney/pathology , Animals , Biomarkers/blood , Biomarkers/urine , Cell Differentiation , Chronic Disease , Cysteine/metabolism , Cystinosis/complications , Cystinosis/metabolism , Cystinosis/pathology , Cystinosis/physiopathology , Disease Models, Animal , Humans , Kidney/metabolism , Kidney/physiopathology , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Mice , Proteinuria/genetics , Proteinuria/metabolism , Proteinuria/prevention & control , Time Factors , Transplantation Chimera
4.
Nephrol Dial Transplant ; 25(12): 3874-84, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20921297

ABSTRACT

BACKGROUND: Acute kidney injury (AKI) represents a major clinical problem with high mortality and limited causal treatments. The use of cell therapy has been suggested as a potential modality to improve the course and outcome of AKI. METHODS: We investigated the possible renoprotection of freshly isolated, uncultured adipose tissue-derived stem and regenerative cells (ADRCs) before and after cryopreservation in a rat ischemia-reperfusion (I-R) model of AKI. RESULTS: We demonstrated that ADRC therapy drastically reduced mortality (survival 100% vs. 57%, ADRC vs. controls, respectively) and significantly reduced serum creatinine (sCr on Day 3: 3.03 ± 1.58 vs. 7.37 ± 2.32 mg/dL, ADRC vs. controls, respectively). Histological analysis further validated a significantly reduced intratubular cast formation, ameliorated acute tubular epithelial cell necrosis and mitigated macrophage infiltration. Furthermore, a reduced RNA expression of CXCL2 and IL-6 was found in the ADRC group which could explain the reduced macrophage recruitment. Use of cryopreserved ADRCs resulted in an equally high survival (90% vs. 33% in the control group) and similarly improved renal function (sCr on Day 3: 4.64 ± 2.43 vs. 7.24 ± 1.40 mg/dL in controls). CONCLUSIONS: Collectively, these results suggest a potential clinical role for ADRC therapy in patients with AKI. Importantly, cryopreservation of ADRCs could offer an autologous treatment strategy for patients who are at high risk for AKI during planned interventions.


Subject(s)
Acute Kidney Injury/prevention & control , Adipose Tissue/cytology , Cell- and Tissue-Based Therapy/methods , Cryopreservation , Mesenchymal Stem Cell Transplantation , Reperfusion Injury/complications , Stem Cells/cytology , Acute Kidney Injury/metabolism , Acute Kidney Injury/physiopathology , Animals , Cell Movement/physiology , Cell Proliferation , Chemokine CXCL2/metabolism , Interleukin-6/metabolism , Kidney Tubules/metabolism , Kidney Tubules/pathology , Kidney Tubules/physiopathology , Macrophages/pathology , Models, Animal , Necrosis , Rats , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Stem Cells/physiology
5.
Ann Plast Surg ; 64(2): 222-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20098110

ABSTRACT

Current practice of autologous fat transfer for soft tissue augmentation is limited by poor long-term graft retention. Adipose-derived regenerative cells (ADRCs) contain several types of stem and regenerative cells, which may help improve graft retention through multiple mechanisms. Using a murine fat transplantation model, ADRCs were added to transplanted fat to test whether ADRCs could improve the long-term retention of the grafts. This study showed, at both 6 and 9 months after transplantation, ADRCs not only increased graft retention by 2-fold but also improved the quality of the grafts. ADRC-supplemented grafts had a higher capillary density, indicating ADRCs could promote neovascularization. Further cell tracking and gene expression studies suggest ADRCs may promote angiogenesis and adipocyte differentiation and prevent apoptosis through the expression of various growth factors, including VEGFA and IGF-1. Taken together, these results suggest a potential clinical utility of ADRCs in facilitating autologous fat transfer for soft tissue augmentation.


Subject(s)
Adipocytes/cytology , Adipose Tissue/transplantation , Neovascularization, Physiologic/physiology , Stem Cell Transplantation/methods , Adipocytes/physiology , Adipose Tissue/cytology , Animals , Apoptosis/physiology , Cell Differentiation , Female , Immunohistochemistry , Ischemia/physiopathology , Mice , Mice, Inbred Strains , Models, Animal , Regeneration/physiology
6.
Int J Cardiol ; 144(1): 26-35, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-19443059

ABSTRACT

BACKGROUND: The aim of this study was to investigate the effect of intracoronary administration of freshly isolated, uncultured autologous tissue-derived stromal cells on cardiac function and perfusion after acute infarction in pigs. METHODS: A transmural myocardial infarction in a porcine model was induced by occlusion of the mid LAD with an angioplasty balloon for 3 h. Upon reperfusion, freshly isolated, uncultured autologous stromal cells (1.5×106 cells/kg) or control solution was injected into the infarct artery. Cardiac function and area at risk were determined by (99m)Tc-SPECT. RESULTS: Eight weeks after infarction, cell treated pigs showed a 20% smaller myocardial perfusion defect compared to control animals (35±9% vs. 44±5% of LV, treated vs. control, respectively, p<0.05). The reduction of the perfusion defect was associated with a significantly higher myocardial salvage index in the cell group as well as a significant increase in ejection fraction compared to control (EF at 8 weeks 43±7% vs. 35±3%, treated vs. control, respectively, p<0.05). This functional improvement was reflected by an increased wall thickness of the infarct and border zone in the treated group (11.2±2.2 mm) compared to control (8.6±1.6 mm, p<0.05) as well as an increased capillary density in the border zone (treated vs. control; 41.6±17.9 vs. 32.9±12.6 capillaries per 0.1 mm², p<0.05). CONCLUSIONS: This study demonstrates for the first time that recovery and intracoronary delivery of uncultured autologous tissue derived stromal cells at time of vessel reperfusion is feasible and improves ventricular function.


Subject(s)
Atrial Function/physiology , Coronary Circulation/physiology , Myocardial Infarction/surgery , Recovery of Function , Stromal Cells/transplantation , Ventricular Function/physiology , Animals , Disease Models, Animal , Flow Cytometry , Follow-Up Studies , Myocardial Infarction/diagnosis , Myocardial Infarction/physiopathology , Stromal Cells/cytology , Swine , Tomography, Emission-Computed, Single-Photon , Transplantation, Autologous
7.
Future Cardiol ; 4(3): 285-98, 2008 May.
Article in English | MEDLINE | ID: mdl-19804333

ABSTRACT

Cell therapy, particularly with stem cells, has created great interest as a solution to the fact that there are limited treatments for postischemic heart disease and none that can regenerate damaged heart cells to strengthen cardiac performance. From the first efforts with myoblasts to recent clinical trials with bone marrow-derived stem cells, early reports of cell therapy suggest improvement in cardiac performance as well as other clinical end points. Based on these exciting but tentative results, other stem cell types are being explored for their particular advantages as a source of adult stem cells. Autologous adipose-derived stem cells are multilinear and can be obtained relatively easily in large quantities from patients; cardiac-derived stem cells are highly appropriate for engraftment in their natural niche, the heart. Human umbilical cord blood cells are potentially forever young and allogenic adult mesenchymal stem cells appear not to evoke the graft versus host reaction. Human embryonic stem cells are effective and can be scaled up for supply purposes. The recent discovery of induced pluripotentcy in human adult stem cells, with only three transcription factor genes, opens a whole new approach to making autologous human pluripotent stem cells from skin or other available tissues. Despite the excitement, stem cells may have to be genetically modified with heme oxygenase, Akt or other genes to survive transplantation in a hypoxic environment. Homing factors and hormones secreted from transplanted stem cells may be more important than cells if they provide the necessary stimulus to trigger cardiac regrowth to replace scar tissue. As we await results from larger and more prolonged clinical trials, the science of stem cell therapy in cardiac disease keeps progressing.

8.
Eur Heart J ; 28(21): 2667-77, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17933755

ABSTRACT

AIMS: This study was designed to assess whether intracoronary application of adipose tissue-derived stem cells (ADSCs) compared with bone marrow-derived stem cells (BMSCs) and control could improve cardiac function after 30 days in a porcine acute myocardial infarction/reperfusion model. METHODS AND RESULTS: An acute transmural porcine myocardial infarction was induced by inflating an angioplasty balloon for 180 min in the mid-left anterior descending artery. Two million cultured autologous stem cells were intracoronary injected through the central lumen of the inflated balloon catheter. Analysis of scintigraphic data obtained after 28 +/- 3 days showed that both absolute and relative perfusion defect decreased significantly after intracoronary administration of ADSCs or BMSCs (relative 30 or 31%, respectively), compared with carrier administration alone (12%, P = 0.048). Left ventricular ejection fraction after 4 weeks increased significantly more after ADSC and BMSC administration than after carrier administration: 11.39 +/- 4.62 and 9.59 +/- 7.95%, respectively vs. 1.95 +/- 4.7%, P = 0.02). The relative thickness of the ventricular wall in the infarction area after cell administration was significantly greater than that after carrier administration. The vascular density of the border zone also improved. The grafted cells co-localized with von Willebrand factor and alpha-smooth muscle actin and incorporated into newly formed vessels. CONCLUSION: This is the first study to show that not only bone marrow-derived cells but also ADSCs engrafted in the infarct region 4 weeks after intracoronary cell transplantation and improved cardiac function and perfusion via angiogenesis.


Subject(s)
Adipocytes/transplantation , Bone Marrow Transplantation/methods , Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/therapy , Ventricular Remodeling/physiology , Adipocytes/diagnostic imaging , Angioplasty, Balloon, Coronary/methods , Animals , Cardiac Catheterization/methods , Myocardial Infarction/diagnostic imaging , Myocardial Reperfusion/methods , Radionuclide Imaging , Random Allocation , Stroke Volume/physiology , Swine
9.
Biochem Biophys Res Commun ; 354(4): 999-1003, 2007 Mar 23.
Article in English | MEDLINE | ID: mdl-17276389

ABSTRACT

Cardiomyocyte regeneration is limited in adult life and is not sufficient to compensate for cell loss with myocardial infarction. Hence, the identification of a useful source of cardiomyocyte progenitors is of great interest for possible use in regenerative therapy. In this study, we isolated stem cells derived from human subcutaneous adipose tissue. The expression of Nkx2.5 and GATA-4 can be observed by PCR directly after extraction and during cultivation in some of these cells. Cardiac Troponin T and myosin light chain-2v become positive after 12 days of cultivation. To define respective factors responsible for spontaneous differentiation, we measured VEGF level in ADSC conditioned medium. Our data showed that ADSC secrete significant amount of VEGF (283.5pg per microgram DNA) and that anti-VEGF receptor antibodies blocked the cardiac differentiation. In conclusion, we demonstrated the spontaneous differentiation of human subcutaneous adipose-derived stem cells into a cardiomyocyte phenotype under standard culturing conditions.


Subject(s)
Cell Differentiation/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Stem Cells/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Cardiac Myosins/biosynthesis , Cells, Cultured , GATA4 Transcription Factor/biosynthesis , Homeodomain Proteins/biosynthesis , Humans , Myosin Light Chains/biosynthesis , Rats , Subcutaneous Fat/cytology , Transcription Factors/biosynthesis , Troponin T/biosynthesis
10.
Biochem Biophys Res Commun ; 353(3): 665-71, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17196165

ABSTRACT

In the present study, the potential of human adipose-derived stem cells to differentiate into cells with characteristics of cardiomyocytes was investigated. Adipose tissue-derived stem cells (ADSCs) were transduced with two different lentiviral vectors simultaneously: (1) a lentiviral vector expressing eGFP controlled by the Nkx2.5 promoter and (2) a lentiviral vector expressing DsRed2 controlled by the myosin light chain-2v promoter (MLC-2v). Nkx2.5-eGFP and MLC-2v-DsRed2 dual positive cells were isolated by FACS. Immunostaining and RT-PCR analysis of the dual positive cells revealed that these cells are positive for Nkx2.5, cardiac troponin I, and L-type calcium channel alpha-1c subunit. Electrophysiology studies demonstrated the presence of functional voltage-dependent calcium and potassium channels. These observations confirm that cardiac progenitor cells can be isolated and enriched from human adipose-derived stem cells using lentiviral selection, and they might represent a new source for cell therapy for myocardial infarction and heart failure.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation , Myocytes, Cardiac/cytology , Stem Cells/metabolism , Calcium Channels, L-Type/physiology , Flow Cytometry , Green Fluorescent Proteins/biosynthesis , Humans , Lentivirus/genetics , Luminescent Proteins/biosynthesis , Myocytes, Cardiac/metabolism , Potassium Channels, Voltage-Gated/physiology , Transduction, Genetic
12.
J Interv Cardiol ; 18(1): 9-10, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15788047
13.
Basic Res Cardiol ; 99(4): 272-8, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15221345

ABSTRACT

Autocrine stimulation and paracrine interaction between coronary smooth muscle cells (cSMC) and endothelial cells (EC) act as regulators of the vascular angiogenesis. Basic fibroblast growth factor (bFGF), its receptor FGF-R1, and coreceptor heparansulfate proteoglycan (HSPG) are important components involved in this angiogenic process. We investigated the influence of angiotensin (Ang) II on this trimolecular bFGF complex, the underlying signaling and the proliferative process in human cSMC. Ang II induces an AT1 receptor-dependent expression of bFGF and also upregulates the FGF-R1 and HSPG expression which is suppressed by losartan, the AT1 receptor blocker. AT1 receptor signaling which is characterized by phosphorylation of p42-mitogen-activated protein kinase (MAPK) is involved in Ang II-induced bFGF, FGF-R1 and HSPG upregulation and DNA synthesis in human cSMC. In contrast, inhibition of the AT2 receptor by PD123,319 has no influence on these Ang II-stimulated and via the MAPK cascade-mediated proangiogenic effects. Finally, our data show that the Ang II-induced DNA synthesis in cSMC is mediated via the bFGF expression. In conclusion, our results suggest that the Ang II-induced angiogenic effects in the vessel wall are supported by the AT1 receptor-stimulated and MAPK pathway-mediated upregulation of the autocrine/paracrine trimolecular bFGF complex in cSMC.


Subject(s)
Angiotensin II/metabolism , Coronary Vessels , Endothelial Cells/metabolism , Fibroblast Growth Factor 2/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Angiogenesis Inducing Agents/metabolism , Autocrine Communication , Blotting, Northern , Blotting, Western , Cells, Cultured , Coronary Vessels/metabolism , Heparan Sulfate Proteoglycans/metabolism , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Paracrine Communication , Receptor, Fibroblast Growth Factor, Type 1 , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...