Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
NPJ Aging ; 8(1): 10, 2022 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-35927427

RESUMO

Age-related differences in stem-cell potency contribute to variable outcomes in clinical stem cell trials. To help understand the effect of age on stem cell potency, bone marrow-derived mesenchymal stem cells (MSCs) were isolated from young (6 weeks) and old (18-24 months) mice. HUVEC tubule formation (TF) induced by the old and young MSCs and ELISA of conditioned media were compared to one another, and to old MSCs after 7 d in indirect co-culture with young MSCs. Old MSCs induced less TF than did young (1.56 ± 0.11 vs 2.38 ± 0.17, p = 0.0003) and released lower amounts of VEGF (p = 0.009) and IGF1 (p = 0.037). After 7 d in co-culture with young MSCs, TF by the old MSCs significantly improved (to 2.09 ± 0.18 from 1.56 ± 0.11; p = 0.013), and was no longer different compared to TF from young MSCs (2.09 ± 0.18 vs 2.38 ± 0.17; p = 0.27). RNA seq of old MSCs, young MSCs, and old MSCs following co-culture with young MSCs revealed that the age-related differences were broadly modified by co-culture, with the most significant changes associated with lysosomal pathways. These results indicate that the age-associated decreased paracrine-mediated effects of old MSCs are improved following indirect co-culture with young MSC. The observed effect is associated with broad transcriptional modification, suggesting potential targets to both assess and improve the therapeutic potency of stem cells from older patients.

2.
Cureus ; 14(1): e21159, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35165609

RESUMO

We present a case of a 45-year-old female who presented to a community hospital with an anterior ST-elevation myocardial infarction (STEMI) that subsequently developed prolonged ventricular fibrillation (VF) refractory to repeated defibrillation and antiarrhythmic medications. Primary percutaneous coronary intervention was performed in the patient with VF but supported only by the Lund University Cardiac Arrest System (LUCAS). Despite a total VF time of 127 minutes, the patient was eventually discharged neurologically intact with a normal left ventricular function. For the right patient, this case illustrates the utility of the LUCAS device, especially at community hospitals without immediate venoarterial extracorporeal membrane oxygenation or ventricular assist device capability.

3.
J Am Coll Cardiol ; 77(16): 1994-2003, 2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33888249

RESUMO

BACKGROUND: The coronavirus disease 2019 (COVID-19) pandemic has impacted many aspects of ST-segment elevation myocardial infarction (STEMI) care, including timely access to primary percutaneous coronary intervention (PPCI). OBJECTIVES: The goal of the NACMI (North American COVID-19 and STEMI) registry is to describe demographic characteristics, management strategies, and outcomes of COVID-19 patients with STEMI. METHODS: A prospective, ongoing observational registry was created under the guidance of 3 cardiology societies. STEMI patients with confirmed COVID+ (group 1) or suspected (person under investigation [PUI]) (group 2) COVID-19 infection were included. A group of age- and sex-matched STEMI patients (matched to COVID+ patients in a 2:1 ratio) treated in the pre-COVID era (2015 to 2019) serves as the control group for comparison of treatment strategies and outcomes (group 3). The primary outcome was a composite of in-hospital death, stroke, recurrent myocardial infarction, or repeat unplanned revascularization. RESULTS: As of December 6, 2020, 1,185 patients were included in the NACMI registry (230 COVID+ patients, 495 PUIs, and 460 control patients). COVID+ patients were more likely to have minority ethnicity (Hispanic 23%, Black 24%) and had a higher prevalence of diabetes mellitus (46%) (all p < 0.001 relative to PUIs). COVID+ patients were more likely to present with cardiogenic shock (18%) but were less likely to receive invasive angiography (78%) (all p < 0.001 relative to control patients). Among COVID+ patients who received angiography, 71% received PPCI and 20% received medical therapy (both p < 0.001 relative to control patients). The primary outcome occurred in 36% of COVID+ patients, 13% of PUIs, and 5% of control patients (p < 0.001 relative to control patients). CONCLUSIONS: COVID+ patients with STEMI represent a high-risk group of patients with unique demographic and clinical characteristics. PPCI is feasible and remains the predominant reperfusion strategy, supporting current recommendations.


Assuntos
COVID-19/epidemiologia , Intervenção Coronária Percutânea/estatística & dados numéricos , SARS-CoV-2 , Infarto do Miocárdio com Supradesnível do Segmento ST/epidemiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Canadá/epidemiologia , Feminino , Mortalidade Hospitalar , Hospitalização/estatística & dados numéricos , Humanos , Masculino , Pessoa de Meia-Idade , América do Norte/epidemiologia , Estudos Prospectivos , Recidiva , Sistema de Registros/estatística & dados numéricos , Reoperação/estatística & dados numéricos , Infarto do Miocárdio com Supradesnível do Segmento ST/diagnóstico , Infarto do Miocárdio com Supradesnível do Segmento ST/mortalidade , Infarto do Miocárdio com Supradesnível do Segmento ST/terapia , Acidente Vascular Cerebral/epidemiologia , Acidente Vascular Cerebral/etiologia , Estados Unidos/epidemiologia , Adulto Jovem
4.
J Am Heart Assoc ; 8(15): e012351, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31340693

RESUMO

Background Prevention of adverse remodeling after myocardial infarction (MI) is an important goal of stem cell therapy. Clinical trial results vary, however, and poor cell retention and survival after delivery likely limit the opportunity to exert beneficial effects. To overcome these limitations, we built an implantable intravascular bioreactor (IBR) designed to protect contained cells from washout, dilution, and immune attack while allowing sustained release of beneficial paracrine factors. Methods and Results IBRs were constructed using semipermeable membrane adhered to a clinical-grade catheter shaft. Mesenchymal stem cell (MSC) viability in and paracrine factor release from IBRs were assessed in vitro and IBR biocompatibility and immune protection confirmed in vivo. In a porcine anterior MI model, IBRs containing 25 million allogeneic MSCs (IBR-MSCs) were compared with IBRs containing media alone (IBR-Placebo; n=8 per group) with adverse remodeling assessed by magnetic resonance imaging. Four weeks after MI, IBR-MSCs had no significant change in end-diastolic volume (+0.33±4.32 mL; P=0.89), end-systolic volume (+2.14±4.13 mL; P=0.21), and left ventricular ejection fraction (-2.27±2.94; P=0.33) while IBR-Placebo had significant increases in end-diastolic volume (+10.37±3.84 mL; P=0.01) and ESV (+11.35±2.88 mL; P=0.01), and a significant decrease in left ventricular ejection fraction (-5.78±1.70; P=0.025). Eight weeks after MI, adherent pericarditis was present in 0 of 8 IBR-MSCs versus 4 of 8 IBR-Placebo (P=0.02), suggesting an anti-inflammatory effect. In a separate study, 25 million allogeneic pig MSCs directly injected in the peri-infarct zone 3 days after MI (n=6) showed no significant benefit in adverse remodeling at 4 weeks compared with IBR-MSCs. Conclusions MSCs deployed inside an implantable, removable, and potentially rechargeable bioreactor in a large animal model remain viable, are immunoprotected, and attenuate adverse remodeling 4 weeks after MI.


Assuntos
Reatores Biológicos , Transplante de Células-Tronco Mesenquimais/métodos , Infarto do Miocárdio/complicações , Próteses e Implantes , Remodelação Ventricular , Animais , Procedimentos Endovasculares , Desenho de Equipamento , Feminino , Suínos
5.
Am Heart J ; 197: 18-26, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29447780

RESUMO

BACKGROUND: The CPORT-E trial showed the noninferiority of nonprimary percutaneous coronary intervention (PCI) at hospitals without cardiac surgery on-site (SoS) compared with hospitals with SoS for 6-week mortality and 9-month major adverse cardiac events (MACE). However, target vessel revascularization (TVR) was increased at non-SoS hospitals. Therefore, we aimed to determine the consistency of the CPORT-E trial findings across the spectrum of enrolled patients. METHODS: Post hoc subgroup analyses of 6-week mortality and 9-month MACE, defined as the composite of death, Q-wave myocardial infarction, or TVR, were performed. Patients with and without 9-month TVR and rates of related outcomes were compared. RESULTS: There was no interaction between SoS status and clinically relevant subgroups for 6-week mortality or 9-month MACE (P for any interaction=.421 and .062, respectively). In addition to increased 9-month rates of TVR and diagnostic catheterization at hospitals without SoS, non-TVR was also increased (2.7% vs 1.9%, P=.002); there was no difference in myocardial infarction-driven TVR, non-TVR, or diagnostic catheterization. Predictors of 9-month TVR included intra-aortic balloon pump use, any index PCI complication, and 3-vessel PCI, whereas predictors of freedom from TVR included SoS, discharge on a P2Y12 inhibitor, and stent implantation. CONCLUSIONS: The noninferiority of nonprimary PCI at non-SoS hospitals was consistent across clinically relevant subgroups. Elective PCI at an SoS hospital conferred a TVR benefit which may be related to a lower rate of referral for diagnostic catheterization for reasons other than myocardial infarction.


Assuntos
Cateterismo Cardíaco , Procedimentos Cirúrgicos Cardíacos , Doença da Artéria Coronariana , Vasos Coronários , Hospitais , Infarto do Miocárdio , Revascularização Miocárdica , Idoso , Cateterismo Cardíaco/métodos , Cateterismo Cardíaco/estatística & dados numéricos , Procedimentos Cirúrgicos Cardíacos/métodos , Procedimentos Cirúrgicos Cardíacos/estatística & dados numéricos , Doença da Artéria Coronariana/complicações , Doença da Artéria Coronariana/diagnóstico , Vasos Coronários/diagnóstico por imagem , Vasos Coronários/cirurgia , Procedimentos Cirúrgicos Eletivos/métodos , Procedimentos Cirúrgicos Eletivos/estatística & dados numéricos , Feminino , Hospitais/classificação , Hospitais/normas , Hospitais/estatística & dados numéricos , Humanos , Balão Intra-Aórtico/estatística & dados numéricos , Masculino , Pessoa de Meia-Idade , Mortalidade , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/mortalidade , Infarto do Miocárdio/terapia , Revascularização Miocárdica/efeitos adversos , Revascularização Miocárdica/métodos , Revascularização Miocárdica/estatística & dados numéricos , Avaliação de Resultados em Cuidados de Saúde , Índice de Gravidade de Doença
6.
Circulation ; 137(3): 307-309, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29046319
7.
Catheter Cardiovasc Interv ; 90(3): 366-377, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28160375

RESUMO

OBJECTIVES: To compare bivalirudin to heparin during non-primary percutaneous coronary intervention (PCI). BACKGROUND: The optimal anticoagulant to support PCI remains uncertain. METHODS: We performed a propensity score-based analysis comparing clinical outcomes of patients receiving heparin to those receiving bivalirudin during non-primary PCI. RESULTS: Of 18,867 patients in the Cardiovascular Patient Outcomes Research Team Non-Primary PCI (CPORT-E) trial, we selected 7,913 patients undergoing non-staged PCI of whom 57.3% received heparin and 42.7% received bivalirudin. In-hospital myocardial infarction occurred in 4.4% of patients receiving bivalirudin and 3.0% of patients receiving heparin (relative risk [RR] 1.5, 95% confidence interval [CI] 1.1-2.1, P = 0.022); this difference persisted at 6 weeks (5.0% vs. 3.6%, RR 1.4, 95% CI 1.0-1.8, P = 0.041). There was no difference in all-cause mortality either in-hospital (0.2% vs. 0.1% for heparin vs. bivalirudin, P = 0.887) or at 6 weeks (0.5% vs. 0.7%, P = 0.567). In-hospital bleeding requiring transfusion occurred in 0.9% of patients receiving bivalirudin and 1.9% of patients receiving heparin (RR 0.4, 95% CI 0.3-0.7, P <0.001), but there was no difference at 6 weeks (2.7% for heparin vs. 1.9% for bivalirudin, RR 0.7, 95% CI 0.5-1.0, P = 0.062). CONCLUSIONS: In patients undergoing non-primary PCI at hospitals without on-site cardiac surgery, bivalirudin was associated with a decreased risk of in-hospital bleeding requiring transfusion and an increased risk of in-hospital MI compared to heparin. © 2017 Wiley Periodicals, Inc.


Assuntos
Anticoagulantes/uso terapêutico , Antitrombinas/uso terapêutico , Doença das Coronárias/terapia , Heparina/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Intervenção Coronária Percutânea , Idoso , Anticoagulantes/efeitos adversos , Antitrombinas/efeitos adversos , Transfusão de Sangue , Doença das Coronárias/sangue , Doença das Coronárias/diagnóstico por imagem , Doença das Coronárias/mortalidade , Feminino , Hemorragia/induzido quimicamente , Hemorragia/terapia , Heparina/efeitos adversos , Hirudinas/efeitos adversos , Mortalidade Hospitalar , Humanos , Estimativa de Kaplan-Meier , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/etiologia , Razão de Chances , Fragmentos de Peptídeos/efeitos adversos , Intervenção Coronária Percutânea/efeitos adversos , Intervenção Coronária Percutânea/mortalidade , Pontuação de Propensão , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/uso terapêutico , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
8.
Am J Cardiol ; 119(7): 1053-1060, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28185634

RESUMO

The high rate of re-hospitalization for heart failure might be reduced by improving noninvasive techniques for identifying elevated left ventricular (LV) filling pressure. We previously showed that changes in a finger photoplethysmography (PPG) waveform during the Valsalva maneuver (VM) reflect invasively measured LV end-diastolic pressure (LVEDP). We have since developed a hand-held device that analyzes PPG while guiding the expiratory effort of a VM. Here we assessed the sensitivity and specificity of this device for identifying elevated LVEDP in patients. We tested 82 participants (28 women), aged 40 to 85 years, before a clinically indicated left heart catheterization. Each performed a VM between 18 and 25 mm Hg for 10 seconds into a pressure transducer. PPG was recorded continuously before and during the VM. LVEDP was measured during the catheterization. An equation for calculating LVEDP was derived using (1) ratio of signal amplitudes: minimum during VM to average at baseline, (2) ratio of peak-to-peak time intervals: minimum during VM to average at baseline, and (3) mean blood pressure. Calculated and measured LVEDP were compared. The range of measured LVEDP was 4 to 35 mm Hg. Calculated LVEDP correlated with measured LVEDP (p <0.0001, r = 0.56). A calculated LVEDP >20 mm Hg had a 70% sensitivity and 86% specificity for identifying measured LVEDP >20 mm Hg (area under receiver-operating characteristic curve 0.83). In conclusion, a hand-held device for assessing LV filling pressure had high specificity and good sensitivity for identifying LVEDP >20 mm Hg, a clinically meaningful threshold in heart failure.


Assuntos
Volume Sanguíneo/fisiologia , Dedos/irrigação sanguínea , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/fisiopatologia , Fotopletismografia/instrumentação , Manobra de Valsalva/fisiologia , Disfunção Ventricular Esquerda/fisiopatologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Pressão Sanguínea/fisiologia , Cateterismo Cardíaco , Desenho de Equipamento , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Sensibilidade e Especificidade , Transdutores de Pressão
9.
Biomaterials ; 52: 318-26, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25818438

RESUMO

Stem cell therapies for atherosclerotic diseases are promising, but benefits remain modest with present cell delivery devices in part due to cell washout and immune attack. Many stem cell effects are believed mediated by paracrine factors (PFs) secreted by the stem cells which potentiate tissue repair via activation and enhancement of intrinsic host repair mechanisms We therefore sought to create an "intravascular paracrine factor factory" by harnessing stem cells on a stent using a nanofiber (NF) stent sleeve, and thus providing a sheltered milieu for cells to continuously produce PFs on-stent. The NF sleeve acts as a substrate on which stem cells grow, and as a semi-permeable barrier that protects cells from washout and host immune response while allowing free outward passage of PFs. NF stent sleeves were created by covering stents with electrospun poly-lactic-co-glycolic acid nanofibers and were then uniformly coated with mesenchymal stem cells (MSCs). NF sleeves blocked cell passage but did not hamper MSC attachment or proliferation, and did not alter MSC morphology or surface markers. NF sleeve MSCs continued to secrete PFs that were biologically active and successfully induced tubulogenesis in human endothelial cells. NF stent sleeves seeded with allogeneic MSCs implanted in pigs remained patent at 7 days without thrombotic occlusion or immune rejection. Our results demonstrate the feasibility of creating an intravascular PF factory using a stem cell impregnated NF stent sleeve, and pave the way for animal studies to assess the efficacy of local PF production to treat ischemic artery disease.


Assuntos
Sistemas de Liberação de Medicamentos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Nanofibras/química , Stents , Angiografia , Animais , Adesão Celular , Proliferação de Células , Sobrevivência Celular , Materiais Revestidos Biocompatíveis/química , Desenho de Equipamento , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Isquemia/patologia , Permeabilidade , Suínos , Transplante Homólogo
11.
Circulation ; 119(22): 2928-35, 2009 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-19470891

RESUMO

BACKGROUND: The challenge of angiogenesis science is that stable sustained vascular regeneration in humans has not been realized despite promising preclinical findings. We hypothesized that angiogenic therapies powerfully self-regulate by dynamically altering tissue characteristics. Induced neocapillaries increase drug clearance and limit tissue retention and subsequent angiogenesis even in the face of sustained delivery. METHODS AND RESULTS: We quantified how capillary flow clears fibroblast growth factor after local epicardial delivery. Fibroblast growth factor spatial loading was significantly reduced with intact coronary perfusion. Penetration and retention decreased with transendothelial permeability, a trend diametrically opposite to intravascular delivery, in which factor delivery depends on vascular leak, but consistent with a continuum model of drug transport in perfused tissues. Model predictions of fibroblast growth factor sensitivity to manipulations of its diffusivity and transendothelial permeability were validated by conjugation to sucrose octasulfate. Induction of neocapillaries adds pharmacokinetic complexity. Sustained local fibroblast growth factor delivery in vivo produced a burst of neovascularization in ischemic myocardium but was followed by drug washout and a 5-fold decrease in fibroblast growth factor penetration depth. CONCLUSIONS: The very efficacy of proangiogenic compounds enhances their clearance and abrogates their pharmacological benefit. This self-limiting property of angiogenesis may explain the failures of promising proangiogenic therapies.


Assuntos
Fatores de Crescimento de Fibroblastos/sangue , Microcirculação/fisiologia , Microvasos/fisiologia , Neovascularização Fisiológica , Farmacocinética , Animais , Permeabilidade Capilar , Difusão , Endotélio Vascular , Fatores de Crescimento de Fibroblastos/metabolismo , Isquemia , Modelos Teóricos , Ratos , Ratos Sprague-Dawley
12.
J Control Release ; 109(1-3): 236-43, 2005 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-16289420

RESUMO

The efficacy of drug-eluting stents (DES) requires delivery of potent compounds directly to the underlying arterial tissue. The commercially available DES drugs rapamycin and paclitaxel bind specifically to their respective therapeutic targets, FKBP12 and polymerized microtubules, while also associating in a more general manner with other tissue elements. As it is binding that provides biological effect the question arises as to whether other locally released or systemically circulating drugs can displace DES drugs from their tissue binding domains. Specific and general binding sites for both drugs are distributed across the media and adventitia with higher specific binding associated with the higher specific binding site densities in the media. The ability of rapamycin and paclitaxel to compete for specific protein binding and general tissue deposition was assessed for both compounds simultaneously and in the presence of other commonly administered cardiac drugs. Drugs classically used to treat standard cardiovascular diseases, such as hypertension and hypercoaguability, displace rapamycin and paclitaxel from general binding sites, possibly decreasing tissue reserve capacity for locally delivered drugs. Paclitaxel and rapamycin do not affect the other's binding to their biologically relevant specific protein targets, but can generally displace each other from tissue at three log order molar excess, decreasing arterials loads by greater than 50%. Local competitive binding therefore should not limit the placement of rapamycin and paclitaxel eluting stents in close proximity.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Stents , Animais , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/uso terapêutico , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/uso terapêutico , Artérias/efeitos dos fármacos , Artérias/metabolismo , Ligação Competitiva , Bovinos , Relação Dose-Resposta a Droga , Técnicas In Vitro , Paclitaxel/administração & dosagem , Paclitaxel/farmacocinética , Paclitaxel/uso terapêutico , Ligação Proteica , Sirolimo/administração & dosagem , Sirolimo/farmacocinética , Sirolimo/uso terapêutico
13.
Circulation ; 111(13): 1619-26, 2005 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-15795325

RESUMO

BACKGROUND: Drug-eluting stents deliver potent compounds directly to arterial segments but can become clot laden when deployed. The question arises as to whether thrombi affect drug elution and arterial uptake. METHODS AND RESULTS: Paclitaxel transport and retention were assessed in clots of different blood components. Diffusivity, affected by clot organization, is fastest in fibrin (approximately 347 microm2/s), slower in fibrin-red blood cell clots (34.98 microm2/s), and slowest in whole-blood clots (3.55 microm2/s). Blood cells bind and retain paclitaxel such that levels in clot increase linearly with red cell fraction. At physiological hematocrit, clot retains 3 times the amount of paclitaxel in surrounding solutions. Computational models predict that the potential of thrombus to absorb, retain, and release drug or to act as a barrier to drug delivery depends on clot geometry and strut position in clot relative to the vessel wall. Clot between artery and stent can reduce uptake 10-fold, whereas clot overlying the stent can shield drug from washout, increasing uptake. Model assumptions were confirmed and predictions were validated in a novel rat model that introduces thrombosis within stented aortas where nonocclusive thrombus acts as capacitive space for drug and shifts drug levels to decrease tissue uptake 2-fold. CONCLUSIONS: Thrombus apposed on stents creates large variations in drug uptake and can act to either increase or decrease wall deposition according to the clot and stent geometry. Arterial deposition of drug from stents deployed in clots will be highly variable and unpredictable unless the clot can be adequately controlled or removed.


Assuntos
Artérias/metabolismo , Paclitaxel/farmacocinética , Stents , Trombose/fisiopatologia , Animais , Coagulação Sanguínea , Simulação por Computador , Difusão , Humanos , Masculino , Paclitaxel/administração & dosagem , Farmacocinética , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
14.
Plast Reconstr Surg ; 114(5): 1086-96; discussion 1097-8, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15457017

RESUMO

The mechanism of action of the Vacuum Assisted Closure Therapy (VAC; KCI, San Antonio, Texas), a recent novel innovation in the care of wounds, remains unknown. In vitro studies have revealed that cells allowed to stretch tend to divide and proliferate in the presence of soluble mitogens, whereas retracted cells remain quiescent. The authors hypothesize that application of micromechanical forces to wounds in vivo can promote wound healing through this cell shape-dependent, mechanical control mechanism. The authors created a computer model (finite element) of a wound and simulated VAC application. Finite element modeling is commonly used to engineer complex systems by breaking them down into simple discrete elements. In this model, the authors altered the pressure, pore diameter, and pore volume fraction to study the effects of vacuum-induced material deformations. The authors compared the morphology of deformation of this wound model with histologic sections of wounds treated with the VAC. The finite element model showed that most elements stretched by VAC application experienced deformations of 5 to 20 percent strain, which are similar to in vitro strain levels shown to promote cellular proliferation. Importantly, the deformation predicted by the model also was similar in morphology to the surface undulations observed in histologic cross-sections of the wounds. The authors hypothesize that this tissue deformation stretches individual cells, thereby promoting proliferation in the wound microenvironment. The application of micromechanical forces may be a useful method with which to stimulate wound healing through promotion of cell division, angiogenesis, and local elaboration of growth factors. Finite element modeling of the VAC device is consistent with this mechanism of action.


Assuntos
Proliferação de Células , Vácuo , Cicatrização/fisiologia , Ferimentos e Lesões/fisiopatologia , Ferimentos e Lesões/cirurgia , Simulação por Computador , Análise de Elementos Finitos , Humanos , Estresse Mecânico , Ferimentos e Lesões/patologia
15.
Proc Natl Acad Sci U S A ; 101(25): 9463-7, 2004 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-15197278

RESUMO

Endovascular drug-eluting stents have changed the practice of medicine, and yet it is unclear how they so dramatically reduce restenosis and how to distinguish between the different formulations available. Biological drug potency is not the sole determinant of biological effect. Physicochemical drug properties also play important roles. Historically, two classes of therapeutic compounds emerged: hydrophobic drugs, which are retained within tissue and have dramatic effects, and hydrophilic drugs, which are rapidly cleared and ineffective. Researchers are now questioning whether individual properties of different drugs beyond lipid avidity can further distinguish arterial transport and distribution. In bovine internal carotid segments, tissue-loading profiles for hydrophobic paclitaxel and rapamycin are indistinguishable, reaching load steady state after 2 days. Hydrophilic dextran reaches equilibrium in several hours at levels no higher than surrounding solution concentrations. Both paclitaxel and rapamycin bind to the artery at 30-40 times bulk concentration. Competitive binding assays confirm binding to specific tissue elements. Most importantly, transmural drug distribution profiles are markedly different for the two compounds, reflecting, perhaps, different modes of binding. Rapamycin, which binds specifically to FKBP12 binding protein, distributes evenly through the artery, whereas paclitaxel, which binds specifically to microtubules, remains primarily in the subintimal space. The data demonstrate that binding of rapamycin and paclitaxel to specific intracellular proteins plays an essential role in determining arterial transport and distribution and in distinguishing one compound from another. These results offer further insight into the mechanism of local drug delivery and the specific use of existing drug-eluting stent formulations.


Assuntos
Proteínas Sanguíneas/metabolismo , Artéria Carótida Interna/metabolismo , Paclitaxel/sangue , Sirolimo/sangue , Animais , Bovinos , Dextranos/sangue , Dextranos/farmacocinética , Cinética , Músculo Liso Vascular/metabolismo , Paclitaxel/farmacocinética , Sirolimo/farmacocinética
16.
Med Phys ; 30(10): 2622-8, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14596298

RESUMO

Radiolabeled drug-eluting stents have been proposed recently as a novel method to potentially reduce restenosis in coronary arteries. A P-32 labeled oligonucleotide (ODN) loaded on a polymer coated stent is slowly released in the arterial wall to deliver a therapeutic dose to the target tissue. However, the relatively low proportion of drugs transferred to the arterial wall (<2%-5% typically) raises questions about the degree to which radiolabeled drugs eluted from the stent can contribute to the total radiation dose delivered to tissues. A three-dimensional diffusion-convection transport model is used to model the transport of a hydrophilic drug released from the surface of a stent to the arterial media. Large drug concentration gradients are observed near the stent struts giving rise to a nonuniform radiation activity distribution for the drug in the tissues as a function of time. A voxel-based kernel convolution method is used to calculate the radiation dose rate resulting from this activity build-up in the arterial wall based on the medical internal radiation dose formalism. Measured residence time for the P-32 ODN in the arterial wall and at the stent surface obtained from animal studies are used to normalize the results in terms of absolute dose to tissue. The results indicate that radiation due to drug eluted from the stent contributes only a small fraction of the total radiation delivered to the arterial wall, the main contribution coming from the activity that remains embedded in the stent coating. For hydrophilic compounds with rapid transit times in arterial tissue and minimal binding interactions, the activity build-up in the arterial wall contributes only a small fraction to the total dose delivered by the P-32 ODN stent. For these compounds, it is concluded that radiolabeled drug-eluting stent will not likely improve the performance of radioactive stents for the treatment of restenosis. Also, variability in the delivery efficacy of drug delivery devices makes accurate dosimetry difficult and the drug washout in the systemic circulatory system may yield an unnecessary activity build-up and dose to healthy organs.


Assuntos
Reestenose Coronária/radioterapia , Vasos Coronários/patologia , Artérias/patologia , Constrição Patológica/terapia , Difusão , Heparina/química , Humanos , Modelos Estatísticos , Oligonucleotídeos/farmacologia , Oligonucleotídeos/uso terapêutico , Radioisótopos de Fósforo/farmacologia , Polímeros/química , Radiometria , Stents , Fatores de Tempo
17.
Int J Cardiovasc Intervent ; 5(1): 7-12, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12623559

RESUMO

Drugs released from stents are driven by physiological transport forces, principally solvent-driven flow (convection) and random molecular agitation (diffusion). The relative strength of these two forces determines drug penetration and distribution in the arterial wall. Drug physicochemical factors can induce critical modulations to the primary distribution, both transiently and at steady state. Hydrophobic interactions and nonspecific binding, for example, can both result in tissue drug concentrations severalfold above administered concentration. Drug interaction with native proteins may also interfere with drug transfer at the stent-artery interface. These transport forces and tissue interactions can induce local drug concentrations even at steady state to vary by one or more orders of magnitude over the span of a few cells. To account for significant local variations in drug concentrations following stent-based delivery, rational design of vascular delivery systems requires consideration of drug distribution and tissue interactions on a local, continuum basis. Continuum analysis adapts traditional pharmacokinetics to the local environment by supplementing discrete global parameters of drug content with continuous local values of concentration, transport and binding. The interplay of these parameters with local flux conditions and drug and tissue properties defines the local drug distribution in space and over time. This type of analysis may well become increasingly relevant given the trend toward stent-based drug therapy in cardiovascular care.


Assuntos
Angioplastia com Balão/métodos , Sistemas de Liberação de Medicamentos/métodos , Preparações Farmacêuticas/metabolismo , Stents , Animais , Transporte Biológico/fisiologia , Fármacos Cardiovasculares/metabolismo , Artérias Carótidas/metabolismo , Bovinos , Ensaios Clínicos como Assunto , Difusão , Humanos , Modelos Teóricos
18.
Circ Res ; 90(7): 826-32, 2002 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-11964377

RESUMO

An incomplete understanding of the transport forces and local tissue structures that modulate drug distribution has hampered local pharmacotherapies in many organ systems. These issues are especially relevant to arteries, where stent-based delivery allows fine control of locally directed drug release. Local delivery produces tremendous drug concentration gradients and although these are in part derived from transport forces, differences in deposition from tissue to tissue imply that tissue ultrastructure also plays an important role. We measured the equilibrium drug uptake and the penetration and diffusivity of dextrans (a model hydrophilic drug similar to heparin) and albumin in orthogonal planes in arteries explanted from different vascular beds. We found significant variations in drug distribution with geometric orientation and arterial connective tissue content. Drug diffusivities parallel to the connective tissue sheaths were one to two orders of magnitude greater than across these sheaths. This diffusivity difference remained relatively constant for drugs up to 70 kDa before decreasing for larger drugs. Drugs also distributed better into elastic arteries, especially at lower molecular weights, with almost 66% greater transfer into the thoracic aorta than into the carotid artery. Arterial drug transport is thus highly anisotropic and dependent on arterial tissue content. The role of the local composition and geometric organization of arterial tissue in influencing vascular pharmacokinetics is likely to become a critical consideration for local vascular drug delivery.


Assuntos
Artérias/metabolismo , Artérias/ultraestrutura , Transporte Biológico/fisiologia , Dextranos/farmacocinética , Albumina Sérica/farmacocinética , Animais , Anisotropia , Artérias/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Bovinos , Tecido Conjuntivo/metabolismo , Difusão/efeitos dos fármacos , Elasticidade , Elastina/metabolismo , Técnicas In Vitro , Modelos Biológicos , Peso Molecular , Músculo Liso Vascular/metabolismo , Perfusão , Distribuição Tecidual/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...