Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Funct Biomater ; 15(8)2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39194673

RESUMO

Lymphedema presents significant challenges to patients' quality of life, prompting the exploration of innovative treatments, such as collagen scaffolds, aimed at treating and reducing the risk of lymphedema. We aimed to evaluate the preventive and therapeutic efficacy and the lymphangiogenic potential of implanted aligned nanofibrillar collagen scaffolds (BioBridgeTM) following the induction of secondary lymphedema in a rabbit model. Thirty rabbits were divided into treatment (G1), prevention (G2), and control (G3) groups. Secondary lymphedema was induced in all groups. BioBridgeTM implantation was performed in G2 and G1 on days 0 and 60, respectively. Follow-ups included hindlimb circumference measurements and indocyanine green lymphography at 0, 60, and 90 days. None of the study rabbits exhibited dermal backflow on day 0 before surgery. At 60 days, the incidence rates of dermal backflow in G1, G2, and G3 were 100%, 44.4%, and 90%, respectively. Furthermore, at 90 days, the incidence rates were 22.2%, 44.4%, and 90%, respectively. New linear lymphatic observation was seen in rabbits with resolved dermal backflow. The findings of this study demonstrated the capacity of BioBridgeTM scaffolds to induce new lymphatic vessel formation and reduce dermal backflow in secondary lymphedema in a rabbit model.

2.
Biomater Sci ; 11(11): 3860-3877, 2023 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-37078624

RESUMO

Tissue engineering is a promising methodology to produce advanced therapy medicinal products (ATMPs). We have developed personalized tissue engineered veins (P-TEV) as an alternative to autologous or synthetic vascular grafts utilized in reconstructive vein surgery. Our hypothesis is that individualization through reconditioning of a decellularized allogenic graft with autologous blood will prime the tissue for efficient recellularization, protect the graft from thrombosis, and decrease the risk of rejection. In this study, P-TEVs were transplanted to vena cava in pig, and the analysis of three veins after six months, six veins after 12 months and one vein after 14 months showed that all P-TEVs were fully patent, and the tissue was well recellularized and revascularized. To confirm that the ATMP product had the expected characteristics one year after transplantation, gene expression profiling of cells from P-TEV and native vena cava were analyzed and compared by qPCR and sequencing. The qPCR and bioinformatics analysis confirmed that the cells from the P-TEV were highly similar to the native cells, and we therefore conclude that P-TEV is functional and safe in large animals and have high potential for use as a clinical transplant graft.


Assuntos
Engenharia Tecidual , Veias , Animais , Suínos , Engenharia Tecidual/métodos , Veias/transplante , Células Endoteliais , Perfilação da Expressão Gênica
3.
J Vis Exp ; (181)2022 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-35404358

RESUMO

Large animal models, specifically swine, are widely used to research cardiovascular diseases and therapies, as well as for training purposes. This paper describes two different aneurysmal swine models that may help researchers to study new therapies for aneurysmal diseases. These aneurysmal models are created by surgically adding a pouch of tissue to carotid arteries in swine. When the model is used for research, the pouch must be autologous; for training purposes, a synthetic pouch suffices. First, the right external jugular vein (EJV) and right common carotid artery (CCA) must be surgically exposed. The EJV is ligated and a vein pouch fashioned from a short segment. This pouch is then sutured to an elliptical arteriotomy performed in the CCA. Animals must be kept heparinized during model creation, and local vasodilators may be used to decrease vasospasms. Once the suture is completed, correct blood flow should be inspected, checking for bleeding from the suture line and vessel patency. Finally, the surgical incision is closed by layers and an angiography performed to image the aneurysmal model. A simplification of this aneurysmal carotid model that decreases invasiveness and surgical time is the use of a synthetic, rather than venous, pouch. For this purpose, a pouch is tailored in advance with a segment of a polytetrafluoroethylene (PTFE) prosthesis, one end of which is sutured close using polypropylene vascular suture and sterilized prior to surgery. This "sac" is then attached to an arteriotomy performed in the CCA as described. Although these models do not reproduce many of the physiopathological events related to aneurysm formation, they are hemodynamically similar to the situation found in the clinical setting. Therefore, they can be used for research or training purposes, allowing physicians to learn and practice different endovascular techniques in animal models that are close to the human system.


Assuntos
Aneurisma , Procedimentos Endovasculares , Aneurisma/diagnóstico por imagem , Aneurisma/cirurgia , Animais , Artérias Carótidas/cirurgia , Artéria Carótida Primitiva/cirurgia , Modelos Animais de Doenças , Suínos
4.
Sci Rep ; 11(1): 22155, 2021 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-34772964

RESUMO

The epicardial administration of therapeutics via the pericardial sac offers an attractive route, since it is minimally invasive and carries no risks of coronary embolization. The aim of this study was to assess viability, safety and effectiveness of cardiosphere-derived cells (CDCs), their extracellular vesicles (EVs) or placebo administered via a mini-thoracotomy 72 h after experimental infarction in swine. The epicardial administration was completed successfully in all cases in a surgery time (knife-to-skin) below 30 min. No significant differences between groups were found in cardiac function parameters evaluated using magnetic resonance imaging before therapy and at the end of the study, despite a trend towards improved function in CDC-treated animals. Moreover, infarct size at 10 weeks was smaller in treated animals, albeit not significantly. Arrhythmia inducibility did not differ between groups. Pathological examination showed no differences, nor were there any pericardial adhesions evidenced in any case 10 weeks after surgery. These results show that the epicardial delivery of CDCs or their EVs is safe and technically easy 3 days after experimental myocardial infarction in swine, but it does not appear to have any beneficial effect on cardiac function. Our results do not support clinical translation of these therapies as implemented in this work.


Assuntos
Vesículas Extracelulares , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Animais , Modelos Animais de Doenças , Feminino , Imageamento por Ressonância Magnética , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/patologia , Miócitos Cardíacos/transplante , Pericárdio/patologia , Esferoides Celulares , Sus scrofa , Toracotomia , Transplante Autólogo
5.
Front Bioeng Biotechnol ; 9: 771400, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34805124

RESUMO

Pathologies related to the cardiovascular system are the leading causes of death worldwide. One of the main treatments is conventional surgery with autologous transplants. Although donor grafts are often unavailable, tissue-engineered vascular grafts (TEVGs) show promise for clinical treatments. A systematic review of the recent scientific literature was performed using PubMed (Medline) and Web of Science databases to provide an overview of the state-of-the-art in TEVG development. The use of TEVG in human patients remains quite restricted owing to the presence of vascular stenosis, existence of thrombi, and poor graft patency. A total of 92 original articles involving human patients and animal models were analyzed. A meta-analysis of the influence of the vascular graft diameter on the occurrence of thrombosis and graft patency was performed for the different models analyzed. Although there is no ideal animal model for TEVG research, the murine model is the most extensively used. Hybrid grafting, electrospinning, and cell seeding are currently the most promising technologies. The results showed that there is a tendency for thrombosis and non-patency in small-diameter grafts. TEVGs are under constant development, and research is oriented towards the search for safe devices.

6.
Pharmaceutics ; 13(8)2021 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-34452230

RESUMO

The use of allogeneic adipose-derived mesenchymal stromal cells (alloADSCs) represents an attractive approach for treating myocardial infarction (MI). Furthermore, adding a natural support improves alloADSCs engraftment and survival in heart tissues, leading to a greater therapeutic effect. We aimed to examine the safety and immunological reaction induced by epicardial implantation of a clinical-grade collagen scaffold (CS) seeded with alloADSCs for its future application in humans. Thus, cellularized scaffolds were myocardially or subcutaneously implanted in immunosuppressed rodent models. The toxicological parameters were not significantly altered, and tumor formation was not found over the short or long term. Furthermore, biodistribution analyses in the infarcted immunocompetent rats displayed cell engraftment in the myocardium but no migration to other organs. The immunogenicity of alloADSC-CS was also evaluated in a preclinical porcine model of chronic MI; no significant humoral or cellular alloreactive responses were found. Moreover, CS cellularized with human ADSCs cocultured with human allogeneic immune cells produced no alloreactive response. Interestingly, alloADSC-CS significantly inhibited lymphocyte responses, confirming its immunomodulatory action. Thus, alloADSC-CS is likely safe and does not elicit any alloreactive immunological response in the host. Moreover, it exerts an immunomodulatory action, which supports its translation to a clinical setting.

7.
Int J Pharm ; 599: 120454, 2021 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-33676988

RESUMO

Myocardial infarction is caused by an interruption of coronary blood flow, leading to one of the main death causes worldwide. Current therapeutic approaches are palliative and not able to solve the loss of cardiac tissue. Cardiosphere derived cells (CDCs) reduce scarring, and increase viable myocardium, with safety and adequate biodistribution, but show a low rate engraftment and survival after implantation. In order to solve the low retention, we propose the encapsulation of CDCs within three-dimensional alginate-poly-L-lysine-alginate matrix as therapy for cardiac regeneration. In this work, we demonstrate the encapsulation of CDCs in alginate matrix, with no decrease in viability over a month, and showing the preservation of CDCs phenotype, differentiation potential, gene expression profile and growth factor release after encapsulation, moving a step forward to clinical translation of CDCs therapy in regeneration in heart failure.


Assuntos
Miocárdio , Transplante de Células-Tronco , Alginatos , Animais , Diferenciação Celular , Coração , Miócitos Cardíacos , Suínos , Distribuição Tecidual
8.
Stem Cell Res Ther ; 10(1): 152, 2019 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-31151405

RESUMO

BACKGROUND: Allogeneic cardiac-derived progenitor cells (CPC) without immunosuppression could provide an effective ancillary therapy to improve cardiac function in reperfused myocardial infarction. We set out to perform a comprehensive preclinical feasibility and safety evaluation of porcine CPC (pCPC) in the infarcted porcine model, analyzing biodistribution and mid-term efficacy, as well as safety in healthy non-infarcted swine. METHODS: The expression profile of several pCPC isolates was compared with humans using both FACS and RT-qPCR. ELISA was used to compare the functional secretome. One week after infarction, female swine received an intracoronary (IC) infusion of vehicle (CON), 25 × 106 pCPC (25 M), or 50 × 106 pCPC (50 M). Animals were followed up for 10 weeks using serial cardiac magnetic resonance imaging to assess functional and structural remodeling (left ventricular ejection fraction (LVEF), systolic and diastolic volumes, and myocardial salvage index). Statistical comparisons were performed using Kruskal-Wallis and Mann-Whitney U tests. Biodistribution analysis of 18F-FDG-labeled pCPC was also performed 4 h after infarction in a different subset of animals. RESULTS: Phenotypic and functional characterization of pCPC revealed a gene expression profile comparable to their human counterparts as well as preliminary functional equivalence. Left ventricular functional and structural remodeling showed significantly increased LVEF 10 weeks after IC administration of 50 M pCPC, associated to the recovery of left ventricular volumes that returned to pre-infarction values (LVEF at 10 weeks was 42.1 ± 10.0% in CON, 46.5 ± 7.4% in 25 M, and 50.2 ± 4.9% in 50 M, p < 0.05). Infarct remodeling was also improved following pCPC infusion with a significantly higher myocardial salvage index in both treated groups (0.35 ± 0.20 in CON; 0.61 ± 0.20, p = 0.04, in 25 M; and 0.63 ± 0.17, p = 0.01, in 50 M). Biodistribution studies demonstrated cardiac tropism 4 h after IC administration, with substantial myocardial retention of pCPC-associated tracer activity (18% of labeled cells in the heart), and no obstruction of coronary flow, indicating their suitability as a cell therapy product. CONCLUSIONS: IC administration of allogeneic pCPC at 1 week after acute myocardial infarction is feasible, safe, and associated with marked structural and functional benefit. The robust cardiac tropism of pCPC and the paracrine effects on left ventricle post-infarction remodeling established the preclinical bases for the CAREMI clinical trial (NCT02439398).


Assuntos
Miócitos Cardíacos/transplante , Doença Aguda , Animais , Modelos Animais de Doenças , Infarto do Miocárdio , Suínos , Transplante Homólogo
9.
Tissue Eng Part C Methods ; 23(11): 736-744, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28699384

RESUMO

The combination of biomatrices and induced pluripotent stem cell (iPSC) derivatives to aid repair and myocardial scar formation may soon become a reality for cardiac regenerative medicine. However, the tumor risk associated with residual undifferentiated cells remains an important safety concern of iPSC-based therapies. This concern is not satisfactorily addressed in xenotransplantation, which requires immune suppression of the transplanted animal. In this study, we assessed the safety of transplanting undifferentiated iPSCs in an allogeneic setting. Given that swine are commonly used as large animal models in cardiac medicine, we used porcine iPSCs (p-iPSCs) in conjunction with bioengineered constructs that support recovery after acute myocardial infarction. Histopathology analyses found no evidence of p-iPSCs or p-iPSC-derived cells within the host myocardium or biomatrices after 30 and 90 days of follow-up. Consistent with the disappearance of the implanted cells, we could not observe functional benefit of these treatments in terms of left ventricular ejection fraction, cardiac output, ventricular volumes, or necrosis. We therefore conclude that residual undifferentiated iPSCs should pose no safety concern when used on immune-competent recipients in an allogeneic setting, at least in the context of cardiac regenerative medicine.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Transplante de Células-Tronco , Animais , Modelos Animais de Doenças , Testes de Função Cardíaca , Inflamação/patologia , Imageamento por Ressonância Magnética , Infarto do Miocárdio/fisiopatologia , Neovascularização Fisiológica , Reação em Cadeia da Polimerase , Sus scrofa , Engenharia Tecidual , Alicerces Teciduais/química , Transplante Homólogo , Cicatrização
10.
Stem Cells Transl Med ; 6(2): 647-655, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28191775

RESUMO

Cardiac tissue engineering, which combines cells and biomaterials, is promising for limiting the sequelae of myocardial infarction (MI). We assessed myocardial function and scar evolution after implanting an engineered bioactive impedance graft (EBIG) in a swine MI model. The EBIG comprises a scaffold of decellularized human pericardium, green fluorescent protein-labeled porcine adipose tissue-derived progenitor cells (pATPCs), and a customized-design electrical impedance spectroscopy (EIS) monitoring system. Cardiac function was evaluated noninvasively by using magnetic resonance imaging (MRI). Scar healing was evaluated by using the EIS system within the implanted graft. Additionally, infarct size, fibrosis, and inflammation were explored by histopathology. Upon sacrifice 1 month after the intervention, MRI detected a significant improvement in left ventricular ejection fraction (7.5% ± 4.9% vs. 1.4% ± 3.7%; p = .038) and stroke volume (11.5 ± 5.9 ml vs. 3 ± 4.5 ml; p = .019) in EBIG-treated animals. Noninvasive EIS data analysis showed differences in both impedance magnitude ratio (-0.02 ± 0.04 per day vs. -0.48 ± 0.07 per day; p = .002) and phase angle slope (-0.18° ± 0.24° per day vs. -3.52° ± 0.84° per day; p = .004) in EBIG compared with control animals. Moreover, in EBIG-treated animals, the infarct size was 48% smaller (3.4% ± 0.6% vs. 6.5% ± 1%; p = .015), less inflammation was found by means of CD25+ lymphocytes (0.65 ± 0.12 vs. 1.26 ± 0.2; p = .006), and a lower collagen I/III ratio was detected (0.49 ± 0.06 vs. 1.66 ± 0.5; p = .019). An EBIG composed of acellular pericardium refilled with pATPCs significantly reduced infarct size and improved cardiac function in a preclinical model of MI. Noninvasive EIS monitoring was useful for tracking differential scar healing in EBIG-treated animals, which was confirmed by less inflammation and altered collagen deposit. Stem Cells Translational Medicine 2017;6:647-655.


Assuntos
Infarto do Miocárdio/cirurgia , Miócitos Cardíacos/transplante , Regeneração , Transplante de Células-Tronco/métodos , Células-Tronco , Engenharia Tecidual/métodos , Alicerces Teciduais , Função Ventricular Esquerda , Remodelação Ventricular , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Espectroscopia Dielétrica , Modelos Animais de Doenças , Humanos , Imageamento por Ressonância Magnética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fenótipo , Recuperação de Função Fisiológica , Células-Tronco/metabolismo , Volume Sistólico , Sus scrofa , Fatores de Tempo
11.
Stem Cell Res Ther ; 7(1): 94, 2016 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-27423905

RESUMO

BACKGROUND: Insulin-like growth factor 1 (IGF-1) and hepatocyte growth factor (HGF) are among the most promising growth factors for promoting cardiorepair. Here, we evaluated the combination of cell- and gene-based therapy using mesenchymal stem cells (MSC) genetically modified to overexpress IGF-1 or HGF to treat acute myocardial infarction (AMI) in a porcine model. METHODS: Pig MSC from adipose tissue (paMSC) were genetically modified for evaluation of different therapeutic strategies to improve AMI treatment. Three groups of infarcted Large White pigs were compared (I, control, non-transplanted; II, transplanted with paMSC-GFP (green fluorescent protein); III, transplanted with paMSC-IGF-1/HGF). Cardiac function was evaluated non-invasively using magnetic resonance imaging (MRI) for 1 month. After euthanasia and sampling of the animal, infarcted areas were studied by histology and immunohistochemistry. RESULTS: Intramyocardial transplant in a porcine infarct model demonstrated the safety of paMSC in short-term treatments. Treatment with paMSC-IGF-1/HGF (1:1) compared with the other groups showed a clear reduction in inflammation in some sections analyzed and promoted angiogenic processes in ischemic tissue. Although cardiac function parameters were not significantly improved, cell retention and IGF-1 overexpression was confirmed within the myocardium. CONCLUSIONS: The simultaneous administration of IGF-1- and HGF-overexpressing paMSC appears not to promote a synergistic effect or effective repair. The combined enhancement of neovascularization and fibrosis in paMSC-IGF-1/HGF-treated animals nonetheless suggests that sustained exposure to high IGF-1 + HGF levels promotes beneficial as well as deleterious effects that do not improve overall cardiac regeneration.


Assuntos
Fator de Crescimento de Hepatócito/genética , Fator de Crescimento Insulin-Like I/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Infarto do Miocárdio/terapia , Neovascularização Fisiológica , Animais , Terapia Baseada em Transplante de Células e Tecidos , Modelos Animais de Doenças , Feminino , Fibrose , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Células-Tronco Mesenquimais/citologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Regeneração/fisiologia , Suínos , Transgenes
12.
PLoS One ; 11(2): e0149001, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26866919

RESUMO

INTRODUCTION: The intrapericardial delivery has been defined as an efficient method for pharmacological agent delivery. Here we hypothesize that intrapericardial administration of cardiosphere-derived cells (CDCs) may have an immunomodulatory effect providing an optimal microenvironment for promoting cardiac repair. To our knowledge, this is the first report studying the effects of CDCs for myocardial repair using the intrapericardial delivery route. MATERIAL AND METHODS: CDCs lines were isolated, expanded and characterized by flow cytometry and PCR. Their differentiation ability was determined using specific culture media and differential staining. 300,000 CDCs/kg were injected into the pericardial space of a swine myocardial infarcted model. Magnetic resonance imaging, biochemical analysis of pericardial fluid and plasma, cytokine measurements and flow cytometry analysis were performed. RESULTS: Our results showed that, phenotype and differentiation behavior of porcine CDCs were equivalent to previously described CDCs. Moreover, the intrapericardial administration of CDCs fulfilled the safety aspects as non-adverse effects were reported. Finally, the phenotypes of resident lymphocytes and TH1 cytokines in the pericardial fluid were significantly altered after CDCs administration. CONCLUSIONS: The pericardial fluid could be considered as a safe and optimal vehicle for CDCs administration. The observed changes in the studied immunological parameters could exert a modulation in the inflammatory environment of infarcted hearts, indirectly benefiting the endogenous cardiac repair.


Assuntos
Infarto do Miocárdio/imunologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Pericárdio/metabolismo , Adipócitos/citologia , Animais , Diferenciação Celular , Transplante de Células , Condrócitos/citologia , Citocinas/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Imageamento por Ressonância Magnética , Miocárdio/patologia , Miócitos Cardíacos/imunologia , Osteogênese , Fenótipo , Reação em Cadeia da Polimerase , Regeneração , Transplante de Células-Tronco/métodos , Suínos
13.
Cardiovasc Intervent Radiol ; 39(2): 161-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26563245

RESUMO

Rationale of prostatic artery embolization (PAE) in the treatment of symptomatic benign prostatic hyperplasia is conventionally believed to include two parts: shrinkage of the enlarged prostate gland as a result of PAE-induced ischemic infarction and potential effects to relax the increased prostatic smooth muscle tone by reducing the number and density of α1-adrenergic receptor in the prostate stroma. This review describes new insights into the likely mechanisms behind PAE, such as ischemia-induced apoptosis, apoptosis enhanced by blockage of androgens circulation to the embolized prostate, secondary denervation following PAE, and potential effect of nitric oxide pathway immediately after embolization. Studies on therapeutic mechanisms in PAE may shed light on potentially new treatment strategies and development of novel techniques.


Assuntos
Embolização Terapêutica/métodos , Próstata/irrigação sanguínea , Hiperplasia Prostática/terapia , Humanos , Masculino , Próstata/patologia , Hiperplasia Prostática/patologia , Resultado do Tratamento
14.
Cardiovasc Intervent Radiol ; 39(1): 1-7, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26581418

RESUMO

Pathological features of benign prostatic hyperplasia (BPH) dictate various responses to prostatic artery embolization (PAE). Typically, BPH originates in the transition zone and periurethral region, where should be considered the primary target area in PAE procedures. Given that histological heterogeneity of components in hyperplasia nodules, epithelial or stromal, identifying the more responsive nodules to PAE will have clinical implications. Since some lower urinary tract symptoms (LUTS) in patients with BPH are usually related to bladder outlet obstruction-induced changes in bladder function rather than to outflow obstruction directly, proper selection of candidate patients prior to PAE is of great clinical importance. BPH is a typical chronic progressive condition, suggesting PAE could aim not only to relieve LUTS but also to delay or prevent the clinical progression. Awareness of the pathological background of BPH is essential for interventional radiologists to improve clinical outcomes and develop new treatment strategies in clinical practice of PAE.


Assuntos
Embolização Terapêutica , Hiperplasia Prostática/patologia , Hiperplasia Prostática/terapia , Humanos , Masculino , Próstata/patologia , Resultado do Tratamento
15.
J Transl Med ; 13: 156, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25964098

RESUMO

BACKGROUND: The optimal timing of cardiac stem cells administration is still unclear. We assessed the safety of same-day and delayed (one week) delivery and the possible influence of the timing on the therapeutic outcomes of allogeneic porcine cardiac stem cells administration after acute myocardial infarction in a closed-chest ischemia-reperfusion model. METHODS: Female swine surviving 90 min occlusion of the mid left anterior descending coronary artery received an intracoronary injection of 25x10(6) porcine cardiac stem cells either two hours (n = 5, D0) or 7 days (n = 6, D7) after reperfusion. Controls received intracoronary injection of vehicle on day 7 (n = 6, CON). Safety was defined in terms of absence of major cardiac events, changes to the ECG during injection, post-administration coronary flow assessed using the TIMI scale and cardiac troponin I determination after the intervention. Cardiac Magnetic Resonance was performed for morphological and functional assessment prior to infarction, before injection (D7 and CON groups only), at one and 10 weeks. Samples were taken from the infarct and transition areas for pathological examination. RESULTS: No major adverse cardiac events were seen during injection in any group. Animals receiving the therapy on the same day of infarction (D0 group) showed mild transient ST changes during injection (n = 4) and, in one case, slightly compromised coronary flow (TIMI 2). Cardiac function parameters and infarct sizes were not significantly different between groups, with a trend towards higher ejection fraction in the treated groups. Ventricular volumes indexed to body surface area increased over time in control animals, and decreased by the end of the study in animals receiving the therapy, significantly so when comparing End Diastolic Volume between CON and D7 groups (CON: 121.70 ml/m(2) ± 26.09 ml/m(2), D7: 98.71 ml/m(2) ± 8.30 ml/m(2), p = 0.037). The treated groups showed less organization of the collagenous scar, and a significantly (p = 0.019) higher amount of larger, more mature vessels at the infarct border. CONCLUSIONS: The intracoronary injection of 25x10(6) allogeneic cardiac stem cells is generally safe, both early and 7 days after experimental infarction, and alleviates myocardial dysfunction, with a greater limitation of left ventricular remodeling when performed at one week.


Assuntos
Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Transplante de Células-Tronco , Células-Tronco/citologia , Remodelação Ventricular , Animais , Feminino , Testes de Função Cardíaca , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Humanos , Imageamento por Ressonância Magnética , Infarto do Miocárdio/patologia , Líquido Pericárdico , Sus scrofa , Fatores de Tempo , Transplante Homólogo , Troponina/metabolismo , Cromossomo Y/metabolismo
16.
PLoS One ; 10(3): e0122377, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25816232

RESUMO

The appropriate administration route for cardiovascular cell therapy is essential to ensure the viability, proliferative potential, homing capacity and implantation of transferred cells. At the present, the intrapericardial administration of pharmacological agents is considered an efficient method for the treatment of cardiovascular diseases. However, only a few reports have addressed the question whether the intrapericardial delivery of Mesenchymal Stem Cells (MSCs) could be an optimal administration route. This work firstly aimed to analyze the pericardial fluid as a cell-delivery vehicle. Moreover, the in vivo biodistribution pattern of intrapericardially administered MSCs was evaluated in a clinically relevant large animal model. Our in vitro results firstly showed that, MSCs viability, proliferative behavior and phenotypic profile were unaffected by exposure to pericardial fluid. Secondly, in vivo cell tracking by magnetic resonance imaging, histological examination and Y-chromosome amplification clearly demonstrated the presence of MSCs in pericardium, ventricles (left and right) and atrium (left and right) when MSCs were administered into the pericardial space. In conclusion, here we demonstrate that pericardial fluid is a suitable vehicle for MSCs and intrapericardial route provides an optimal retention and implantation of MSCs.


Assuntos
Rastreamento de Células/métodos , Imageamento por Ressonância Magnética/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Líquido Pericárdico/citologia , Animais , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Masculino , Modelos Animais , Suínos , Distribuição Tecidual
17.
Expert Rev Cardiovasc Ther ; 13(3): 285-99, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25653187

RESUMO

Myocardial infarction, even after reperfusion, leads to significant loss of cardiomyocytes and to a maladaptive remodeling process. A possibility gaining attention as an ancillary therapy is the use of cardiac-derived cell products, with early stage clinical trials reporting highly promising results with autologous cells. However, an autologous therapy presents limitations, such as timeframe of therapy, cell processing and culture costs, risks posed to the patient by the tissue harvesting, etc. Allogeneic cells may represent an answer, providing an off-the-shelf product that could be used in the acute stage, before the myocardial damage is irrevocable. To date, allogeneic cardiac-derived cell products are being tested extensively, but the questions of their immunogenicity (and therefore safety), efficacy, cost-effectiveness, etc. are only partially elucidated. Small Phase I/II clinical trials (ALLSTAR, CAREMI) have started and their results will shed the much needed light on the feasibility and safety of a much needed therapy.


Assuntos
Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Transplante de Células-Tronco/métodos , Animais , Humanos , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia , Transplante de Células-Tronco/efeitos adversos , Fatores de Tempo , Transplante Homólogo
18.
Eur J Pharm Biopharm ; 84(1): 29-39, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23266493

RESUMO

Adipose tissue-derived stem cells (ASCs) have properties of self-renewal, pluripotency and high proliferative capability that make them useful for the treatment of cardiac ventricular function following ischaemic injury. However, their therapeutic use is limited due to the low retention of the cells at the targeted site. To address this issue, we developed semipermeable membrane microcapsules labelled with Endorem (magnetocapsules) that provide mechanical and immunological immune protection to the cells while maintaining internal cell microenvironment. In addition, the particles allow tracking the presence and migration of injected cells in vivo by Magnetic Resonance Imaging (MRI). Results indicate that after 21 days in culture, the cells encapsulated in the magnetocapsules showed similar viabilities than cells encapsulated in conventional microcapsules. MRI confirmed a gradual loss of the intensity of the iron oxide label in the non-encapsulated Endorem labelled cells, while magnetocapsules were detected throughout the study period, suggesting that cell retention in the myocardium is improved when cells are enclosed within the magnetocapsules. To further evaluate treatment's effect on global cardiac function, MRI determination of infarct size and left ventricular ejection fraction (LVEF) was performed. In vivo results showed no statistically significant differences in heart rate and cardiac output between treatment groups. In conclusion, cells enclosed within magnetocapsules have shown suitable viability and have been detected in vivo throughout the study period. Further studies will evaluate whether increasing cell loading with the particles may help to improve the therapeutic results.


Assuntos
Tecido Adiposo/transplante , Alginatos/administração & dosagem , Modelos Animais de Doenças , Microesferas , Infarto do Miocárdio/cirurgia , Transplante de Células-Tronco/métodos , Tecido Adiposo/citologia , Alginatos/química , Animais , Células Cultivadas , Ácido Glucurônico/administração & dosagem , Ácido Glucurônico/química , Ácidos Hexurônicos/administração & dosagem , Ácidos Hexurônicos/química , Imageamento por Ressonância Magnética/métodos , Infarto do Miocárdio/diagnóstico , Distribuição Aleatória , Suínos
19.
AJR Am J Roentgenol ; 197(2): 495-501, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21785100

RESUMO

OBJECTIVE: The purpose of this study was to prospectively evaluate pathologic responses to transarterial prostatic embolization and its technical safety in a canine model. MATERIALS AND METHODS: Ten adult male beagle dogs were surgically castrated and given hormonal therapy for 4 months to induce prostatic hyperplasia. After three months of hormonal therapy, the dogs were randomly assigned to a transarterial prostatic embolization group (n = 7) or a control group (n = 3). Dogs in the transarterial prostatic embolization group were subjected to embolization with microspheres 300-500 µm in diameter. Four months after the study was begun, all dogs were sacrificed for pathologic study. Transrectal ultrasound and MRI were performed to evaluate pathologic responses. The data on prostate size acquired with transrectal ultrasound were processed for statistical analysis by paired Student t test. RESULTS: The canine prostatic hyperplasia model was successfully established in 10 dogs. The increase in mean prostate size being as great as 572% after 3 months of hormonal therapy. An intraprostatic cavity was detected 1 month after transarterial prostatic embolization in all seven dogs. Four dogs had significant shrinkage of the prostate, and the other three had an increase in prostate size. Imaging examinations and necropsy revealed a huge cavity occupying almost the entire prostate in the three dogs with increased prostate size. No complications associated with transarterial prostatic embolization were encountered. CONCLUSION: Transarterial prostatic embolization is a safe procedure that can induce prostatic infarction and ablate the prostate. The findings suggest the procedure has potential clinical applications in the care of patients with benign prostatic hyperplasia.


Assuntos
Embolização Terapêutica/métodos , Hiperplasia Prostática/terapia , Angiografia , Animais , Modelos Animais de Doenças , Cães , Imageamento por Ressonância Magnética , Masculino , Microesferas , Estudos Prospectivos , Hiperplasia Prostática/diagnóstico por imagem , Hiperplasia Prostática/patologia , Distribuição Aleatória , Segurança , Ultrassonografia
20.
Ann Vasc Surg ; 23(4): 506-18, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19375889

RESUMO

Our aim was to study the physiological repercussions of varying ischemic times during laparoscopic aortic surgery. After quarantine, laparoscopy was performed in 24 healthy pigs, which were randomly allocated to four study groups: group I (n=6), laparoscopic infrarenal abdominal aortic surgery with 30 min of crossclamping; group II (n=6), laparoscopic infrarenal abdominal aortic surgery with 60 min of cross-clamping; group III (n=6), laparoscopic infrarenal abdominal aortic surgery with 120 min of cross-clamping; group IV (n=6), 120 min pneumoperitoneum in the absence of aortic cross-clamping (control group). Hematological, biochemical, hormone, hemodynamic, and ventilatory studies were conducted during and after surgery; and a postoperative neurological evaluation was performed 10 days after surgery. Group III evidenced an increase in arterial blood pressure and heart rate that was significantly higher than those present in the other groups. Significant decreases in pH were observed in groups II and III, whereas no changes in this parameter were seen in groups I and IV. Catecholamine levels during surgery were similar in all groups (a significant [p<0.001] increase in plasmatic adrenaline and noradrenaline was seen immediately after pneumoperitoneum creation in all groups). A positive association was found between the duration of aortic clamping and hormone values at 30 and 60 min after declamping but not after 24hr. A significant increase in the renal resistive index (RRI) and a significant decrease in urine output were evidenced during laparoscopy, with significantly lower RRI values seen in group IV immediately after surgery. Thus, a synergic effect of pneumoperitoneum and aortic cross-clamping was seen in this study. These two factors together cause a hemodynamic compromise, with decreased renal perfusion and acidosis, thus negatively affecting the patient's general state during this type of surgery. Despite being well tolerated in healthy pigs, a laparoscopic aortic cross-clamping time over 60min produces significant hemodynamic, metabolic, and hormonal changes. Careful patient selection is mandatory to avoid any severe complications.


Assuntos
Acidose/etiologia , Aorta Abdominal/cirurgia , Hemodinâmica , Nefropatias/etiologia , Laparoscopia/efeitos adversos , Pneumoperitônio Artificial/efeitos adversos , Acidose/sangue , Animais , Aorta Abdominal/fisiopatologia , Pressão Sanguínea , Débito Cardíaco , Constrição , Epinefrina/sangue , Feminino , Frequência Cardíaca , Concentração de Íons de Hidrogênio , Nefropatias/sangue , Nefropatias/fisiopatologia , Modelos Animais , Norepinefrina/sangue , Recuperação de Função Fisiológica , Circulação Renal , Suínos , Fatores de Tempo , Resistência Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA