Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Crohns Colitis 360 ; 4(3): otac016, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36777426

RESUMEN

Background: Bowel urgency is commonly experienced by patients with ulcerative colitis (UC) and is associated with reduced health-related quality of life (QoL). Mirikizumab, a humanized monoclonal antibody directed against the p19 subunit of IL-23, significantly reduced bowel urgency in a double-blind, randomized, placebo-controlled Phase 2 clinical trial in patients with moderate-to-severe UC (NCT02589665). Methods: All patients (N = 249) reported symptoms including absence or presence of bowel urgency. Absence of urgency was defined as no urgency for the 3 consecutive days prior to each scheduled visit. Missing urgency data were imputed as present. After 12 weeks of induction treatment, patients who achieved clinical response continued maintenance mirikizumab treatment through Week 52. We assessed the relationship of urgency with QoL, clinical outcomes, and inflammatory biomarkers at Weeks 12 and 52. Results: Patients with absence of urgency demonstrated significantly greater improvement in Inflammatory Bowel Disease Questionnaire (IBDQ) scores even after adjusting for rectal bleeding (RB) and stool frequency (SF), significantly higher rates of all clinical outcomes at Weeks 12 and 52, and a greater decrease in inflammatory biomarkers C-reactive protein and fecal calprotectin compared to those with presence of urgency. Absence of urgency at Week 12 was associated with improved IBDQ scores at Week 52, while Week 12 RB or SF status was not. Conclusions: Absence of urgency is strongly associated with improvement in QoL as well as clinical measures of UC disease activity. These findings suggest urgency may be a useful surrogate marker of disease activity and an important treatment target for UC.

2.
Vascular ; 26(6): 608-614, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29871586

RESUMEN

INTRODUCTION: The protective effect of diabetes mellitus on abdominal aortic aneurysm formation and growth has been repeatedly observed in population studies but continues to be poorly understood. However, recent investigations have suggested that metformin, a staple antihyperglycemic medication, may be independently protective against abdominal aortic aneurysm formation and growth. Therefore, we describe the effect of metformin in abdominal aortic aneurysm and at-risk patients on markers of inflammation, the driver of early abdominal aortic aneurysm formation and growth. METHODS: Peripheral blood was collected from patients previously diagnosed with abdominal aortic aneurysm or presenting for their U.S. Preventive Task Force-recommended abdominal aortic aneurysm screening. Plasma and circulating peripheral blood mononuclear cells were isolated using Ficoll density centrifugation. Circulating plasma inflammatory and regulatory cytokines were assessed with enzyme-linked immunosorbent assays. CD4+ cell phenotyping was performed using flow cytometric analysis and expressed as a proportion of total CD4+ cells. To determine the circulating antibody to self-antigen response, a modified enzyme-linked immunosorbent assay was performed against antibodies to collagen type V and elastin fragments. RESULTS: Peripheral blood was isolated from 266 patients without diabetes mellitus ( n=182), with diabetes mellitus not treated with metformin ( n=34), and with diabetes mellitus actively taking metformin ( n=50) from 2015 to 2017. We found no differences in the expression of Tr1, Th17, and Treg CD4+ fractions within diabetics ± metformin. When comparing inflammatory cytokines, we detected no differences in IL-1ß, IL-6, IL-17, IL-23, IFN-γ, and TNF-α. Conversely, no differences were observed pertaining to the expression to regulatory cytokines IL-4, IL-10, IL-13, TSG-6, or TGF-ß. Lastly, no differences in expression of collagen type V and elastin fragment antigen and/or antibodies were detected with metformin use in diabetics. CONCLUSION: Metformin in diabetics at-risk for abdominal aortic aneurysm or diagnosed with abdominal aortic aneurysm does not seem to alter the peripheral inflammatory environment.


Asunto(s)
Aneurisma de la Aorta Abdominal/sangre , Citocinas/sangre , Diabetes Mellitus/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Mediadores de Inflamación/sangre , Metformina/uso terapéutico , Aneurisma de la Aorta Abdominal/diagnóstico por imagen , Aneurisma de la Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/prevención & control , Biomarcadores/sangre , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus/sangre , Diabetes Mellitus/diagnóstico , Humanos , Masculino , Factores Protectores , Estudios Retrospectivos , Factores de Riesgo
3.
Surgery ; 164(2): 354-358, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29716755

RESUMEN

BACKGROUND: The pathogenesis driving the formation of abdominal aortic aneurysms continues to be poorly understood. Therefore, we systemically define the cytokine and circulating immune cell environment observed in human abdominal aortic aneurysm compared with risk-factor matched controls. METHODS: From 2015 to 2017, a total of 274 patients donated blood to the Indiana University Center for Aortic Disease. Absolute concentrations of circulating cytokines were determined, using enzyme-linked immunosorbent assays while the expression of circulating immune cell phenotypes were assayed via flow cytometric analysis. RESULTS: Human abdominal aortic aneurysm is characterized by a significant depletion of the antigen-specific, CD4+ Tr1 regulatory lymphocyte that corresponds to an upregulation of the antigen-specific, inflammatory Th17 cell. We found no differences in the incidence of Treg, B10, and myeloid-derived suppressor regulatory cells. Similarly, no disparities were noted in the following inflammatory cytokines: IL-1ß, C-reactive protein, tumor necrosis factor α, interferon γ, and IL-23. However, significant upregulation of the inflammatory cytokines osteopontin, IL-6, and IL-17 were noted. Additionally, no changes were observed in the regulatory cytokines IL-2, IL-4, IL-13, TNF-stimulated gene 6 protein, and prostaglandin E2, but we did observe a significant decrease in the essential regulatory cytokine IL-10. CONCLUSION: In this investigation, we systematically characterize the abdominal aortic aneurysm-immune environment and present preliminary evidence that faulty immune regulation may also contribute to aneurysm formation and growth.


Asunto(s)
Aneurisma de la Aorta Abdominal/inmunología , Linfocitos T CD4-Positivos , Citocinas/sangre , Anciano , Aneurisma de la Aorta Abdominal/sangre , Estudios de Casos y Controles , Colágeno Tipo V/inmunología , Elastina/inmunología , Femenino , Humanos , Macrófagos , Masculino , Persona de Mediana Edad , Factores de Riesgo
4.
Physiol Rep ; 6(8): e13668, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29696811

RESUMEN

Abdominal aortic aneurysms are pathological dilations that can suddenly rupture, causing more than 15,000 deaths in the U.S. annually. Current treatment focuses on observation until an aneurysm's size warrants surgical intervention. Thus, there is a need for therapeutic intervention to inhibit growth of smaller aneurysms. An experimental aneurysm model that infuses angiotensin II into apolipoprotein E-deficient mice is widely used to investigate underlying pathological mechanisms and potential therapeutics, but this model has two caveats: (1) aneurysms do not always form, and (2) aneurysm severity and growth is inconsistent among animals. Here we use high-frequency ultrasound to collect data from angiotensin II-induced aneurysms to develop prediction models of both aneurysm formation and growth. Baseline measurements of aortic diameter, volume/length, and strain were used with animal mass and age in a quadratic discriminant analysis and logistic regression to build two statistical models to predict disease status. Longitudinal ultrasound data were also acquired from mice with aneurysms to quantify aneurysm diameter, circumferential strain, blood flow velocity, aneurysm volume/length, and thrombus and open-false lumen volumes over 28 days. Measurements taken at aneurysm diagnosis were used with branching artery information to produce a multiple linear regression model to predict final aneurysm volume/length. All three statistical models could be useful in future aneurysm therapeutic studies to better delineate the effects of preventative and suppressive treatments from normal variations in the angiotensin II aneurysm model.


Asunto(s)
Angiotensina II , Aneurisma de la Aorta Abdominal/diagnóstico por imagen , Disección Aórtica/diagnóstico por imagen , Disección Aórtica/inducido químicamente , Animales , Aneurisma de la Aorta Abdominal/inducido químicamente , Apolipoproteínas E/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Masculino , Ratones , Ultrasonografía
5.
J Vasc Surg ; 68(2): 560-566, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29503004

RESUMEN

OBJECTIVE: Ethnic minorities (nonwhites) with critical limb ischemia (CLI) have historically performed worse compared with whites with regard to major amputation risk reduction and amputation-free survival (AFS) after peripheral vascular intervention. This post hoc analysis was completed to determine whether this precedent also extended to treatment of CLI without a suitable revascularization option with intramuscular injections of concentrated bone marrow aspirate (cBMA). METHODS: The treatment arm of the randomized, double-blind, multicenter MarrowStim PAD Kit for the Treatment of Critical Limb Ischemia in Subjects with Severe Peripheral Arterial Disease (MOBILE) trial was stratified by ethnicity and evaluated for demographics, comorbidities, and outcomes. The primary and therapeutic end point was 1-year AFS and major amputation, respectively. Noninferiority analysis was performed with the margin set at historically reported hazard ratios. RESULTS: Thirty-seven minority (African American, Hispanic, other) CLI patients (9 placebo, 28 cBMA) with no suitable revascularization option were randomized to cBMA or placebo at a 3:1 ratio during the MOBILE trial. At 1-year follow-up for the treatment group, overall AFS was 80%. Of the 28 minority patients randomized to cBMA intervention, an 89% AFS rate was observed compared with 77% in whites. Specifically, 22 of 24 (92%) African Americans survived amputation free at 1-year follow-up. Noninferiority testing confirmed no difference between whites and the ethnic minority treated with cBMA with respect to major amputation reduction; however, noninferiority could not be confirmed with regard to AFS. No significant differences favoring whites treated with cBMA were noted in the secondary end points of vascular quality of life, limb pain, ankle-brachial index, toe-brachial index, transcutaneous oximetry, and 6-minute walk testing. CONCLUSIONS: This post hoc analysis of the MOBILE trial demonstrates noninferiority of cBMA intervention in minorities with no-option CLI for the therapeutic end point of major amputation prevention. cBMA represents a novel treatment paradigm and should be explored for minorities with poor revascularization options who face impending amputation secondary to progressive CLI.


Asunto(s)
Amputación Quirúrgica , Trasplante de Médula Ósea/efectos adversos , Etnicidad , Isquemia/cirugía , Grupos Minoritarios , Enfermedad Arterial Periférica/cirugía , Población Blanca , Anciano , Enfermedad Crítica , Supervivencia sin Enfermedad , Método Doble Ciego , Femenino , Disparidades en el Estado de Salud , Humanos , Isquemia/diagnóstico , Isquemia/etnología , Estimación de Kaplan-Meier , Recuperación del Miembro , Masculino , Persona de Mediana Edad , Enfermedad Arterial Periférica/diagnóstico , Enfermedad Arterial Periférica/etnología , Factores de Riesgo , Factores de Tiempo , Trasplante Autólogo , Resultado del Tratamiento
6.
J Vasc Surg ; 68(6S): 22S-29S, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29402664

RESUMEN

OBJECTIVE: Previous in vitro and animal studies have suggested that osteopontin (OPN), an inflammatory extracellular matrix protein, is involved in the formation and growth of abdominal aortic aneurysms (AAAs). However, the mechanism by which this occurs continues to be nebulous. The relationship between OPN and inflammation-suppressing lymphocytes present in the human AAA condition was investigated and presented herein. METHODS: Serum OPN concentrations were measured in healthy, risk factor-matched non-AAA and AAA patients by enzyme-linked immunosorbent assay (ELISA). Immunohistochemistry was used to determine the source of OPN secretion using aortic tissue collected from multiorgan donors and AAA patients undergoing open surgical repair. Vascular smooth muscle cells (VSMCs) were exposed to various inflammatory mediators, and OPN expression was evaluated by quantitative reverse transcriptase-polymerase chain reaction and ELISA. The inflammatory nature of OPN and the aortic wall was determined using a TR1 suppressor cell induction assay as a surrogate and characterized by ELISA and fluorescence-activated cell sorting. RESULTS: OPN was found to be elevated in both the plasma and aortic homogenate of AAA patients compared with controls. On immunohistochemistry, OPN localized to the tunica media of the diseased aorta but was minimally expressed in healthy aorta. In vitro, cigarette smoke extract was the most potent stimulator of OPN secretion by VSMCs and increased both messenger RNA and supernatant concentrations. OPN demonstrated an ability to inhibit the induction of interleukin 10-secreting TR1 lymphocytes, a depleted population in the AAA patient, from naive precursors. Last, neutralizing receptor targets of OPN in the setting of AAA homogenate coincubation abrogated the inhibition of TR1 induction. CONCLUSIONS: OPN, secreted by the VSMCs of the tunica media, is elevated in the circulating plasma and aortic wall of patients with AAA. It can inhibit the induction of the TR1 suppressor cell, leading to an overall proinflammatory state contributing to progressive aortic wall breakdown and dilation.


Asunto(s)
Aneurisma de la Aorta Abdominal/sangre , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Osteopontina/sangre , Aorta Abdominal/inmunología , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/patología , Estudios de Casos y Controles , Células Cultivadas , Dilatación Patológica , Humanos , Interleucina-10/metabolismo , Activación de Linfocitos , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/patología , Osteopontina/genética , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Regulación hacia Arriba , Remodelación Vascular
7.
J Vasc Surg ; 68(1): 176-181.e1, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29395424

RESUMEN

OBJECTIVE: Currently, there are no accepted nonsurgical therapies that improve the delivery of blood-derived nutrients to patients with critical limb ischemia. Here, we describe the ongoing phase 1/2 Clinical and Histologic Analysis of Mesenchymal Stromal Cells in AmPutations (CHAMP) trial, which will provide crucial evidence of the safety profile of mesenchymal stromal cells (MSCs) and explore their therapeutic mechanisms in the setting of critical limb ischemia requiring below-knee amputation (BKA). METHODS: In the CHAMP and the parallel marrowCHAMP trials (hereafter grouped together as CHAMP), a total of 32 extremities with rest pain or tissue loss requiring BKA will be enrolled to receive intramuscular injections of allogeneic MSCs (CHAMP; n = 16) or autogenous concentrated bone marrow aspirate (marrowCHAMP; n = 16) along the distribution of the BKA myocutaneous flap and proximal tibialis anterior. After treatment, subjects are randomized to BKA at four time points after injection (days 3, 7, 14, and 21). At the time of amputation, skeletal muscle is collected at 2-cm increments from the tibialis injection site and used to determine proangiogenic cytokine description, MSC retention, quantification of proangiogenic hematopoietic progenitor cells, and histologic description. Clinical limb perfusion before and after treatment will be quantified using transcutaneous oximetry, toe-brachial index, ankle-brachial index, and indocyanine angiography. Additional clinical end points include all-cause mortality, need for amputation revision, and gangrene incidence during the 6-month post-treatment follow-up. RESULTS: Enrollment is under way, with 10 patients treated per protocol thus far. We anticipate full conclusion of follow-up within the next 24 months. CONCLUSIONS: CHAMP will be pivotal in characterizing the safety, efficacy, and, most important, therapeutic mechanism of allogeneic MSCs and autogenous concentrated bone marrow aspirate in ischemic skeletal muscle.


Asunto(s)
Amputación Quirúrgica , Isquemia/cirugía , Trasplante de Células Madre Mesenquimatosas , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/cirugía , Enfermedad Arterial Periférica/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Amputación Quirúrgica/efectos adversos , Proteínas Angiogénicas/metabolismo , Células Cultivadas , Protocolos Clínicos , Enfermedad Crítica , Citocinas/metabolismo , Femenino , Humanos , Indiana , Isquemia/diagnóstico , Isquemia/fisiopatología , Extremidad Inferior , Masculino , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Enfermedad Arterial Periférica/diagnóstico , Enfermedad Arterial Periférica/fisiopatología , Proyectos de Investigación , Factores de Tiempo , Resultado del Tratamiento
8.
Ann Vasc Surg ; 47: 230-237, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28916304

RESUMEN

BACKGROUND: Abdominal aortic aneurysms (AAAs) are a major source of morbidity and mortality despite continuing advances in surgical technique and care. Although the inciting factors for AAA development continue to be elusive, accumulating evidence suggests a significant periaortic inflammatory response leading to degradation and dilation of the aortic wall. Previous human trials have demonstrated safety and efficacy of mesenchymal stem cells (MSCs) in the treatment of inflammation-related pathologies such as rheumatoid arthritis, graft versus host disease, and transplant rejection. Therefore, herein, we describe the Aortic Aneurysm Repression with Mesenchymal Stem Cells (ARREST) trial, a phase I investigation into the safety of MSC infusion for patients with small AAA and the cells' effects on modulation of AAA-related inflammation. METHODS: ARREST is a phase I, single-center, double-blind, randomized controlled trial (RCT) investigating infusion both dilute and concentrated MSCs compared to placebo in 36 small AAA (35-45 mm) patients. Subjects will be followed by study personnel for 12 months to ascertain incidence of adverse events, immune cell phenotype expression, peripheral cytokine profile, and periaortic inflammation. Maximum transverse aortic diameter will be assessed regularly for 5 years by a combination of computed tomography and duplex sonography. RESULTS: Four patients have thus far been enrolled, randomized, and treated per protocol. We anticipate the conclusion of the treatment phase within the next 24 months with ongoing long-term follow-up. CONCLUSIONS: ARREST will be pivotal in assessing the safety of MSC infusion and provide preliminary data on the ability of MSCs to favorably modulate the pathogenic AAA host immune response. The data gleaned from this phase I trial will provide the groundwork for a larger, phase III RCT which may provide the first pharmaceutical intervention for AAA.


Asunto(s)
Aneurisma de la Aorta Abdominal/cirugía , Trasplante de Células Madre Mesenquimatosas , Aneurisma de la Aorta Abdominal/sangre , Aneurisma de la Aorta Abdominal/diagnóstico por imagen , Aneurisma de la Aorta Abdominal/fisiopatología , Aortografía/métodos , Biomarcadores/sangre , Protocolos Clínicos , Angiografía por Tomografía Computarizada , Citocinas/sangre , Dilatación Patológica , Método Doble Ciego , Humanos , Indiana , Mediadores de Inflamación/sangre , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Proyectos de Investigación , Factores de Tiempo , Resultado del Tratamiento , Ultrasonografía Doppler Dúplex , Remodelación Vascular
9.
J Surg Res ; 220: 311-319, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29180197

RESUMEN

BACKGROUND: The formation of abdominal aortic aneurysms (AAA) is characterized by a dominance of proinflammatory forces that result in smooth muscle cell apoptosis, extracellular matrix degradation, and progressive diameter expansion. Additional defects in the antiinflammatory response may also play a role but have yet to be fully characterized. TSG-6 (TNF-stimulated gene-6) is a potent antiinflammatory protein involved in extracellular matrix stabilization and cell migration active in many pathological conditions. Here, we describe its role in AAA formation. METHODS: Blood and/or aortic tissue samples were collected from organ donors, subjects undergoing elective AAA screening, and open surgical AAA repair. Aortic specimens collected were preserved for IHC or immediately assayed after tissue homogenization. Protein concentrations in tissue and plasma were assayed by ELISA. All immune cell populations were assayed using FACS. In vitro, macrophage polarization from monocytes was performed with young, healthy donor PBMCs. RESULTS: TSG-6 was found to be abnormally elevated in both the plasma and aortic wall of patients with AAA compared with healthy and risk-factor matched non-AAA donors. We observed the highest tissue concentration of TSG-6 in the less-diseased proximal and distal shoulders compared with the central aspect of the aneurysm. IHC localized most TSG-6 to the tunica media with minor expression in the tunica adventitia of the aortic wall. Higher concentrations of both M1 and M2 macrophages where also observed, however M1/M2 ratios were unchanged from healthy controls. We observed no difference in M1/M2 ratios in the peripheral blood of risk-factor matched non-AAA and AAA patients. Interesting, TSG-6 inhibited the polarization of the antiinflammatory M2 phenotype in vitro. CONCLUSIONS: AAA formation results from an imbalance of inflammatory forces causing aortic wall infiltration of mononuclear cells leading to the vessel breakdown. In the AAA condition, we report an elevation of TSG-6 expression in both the aortic wall and the peripheral circulation.


Asunto(s)
Aneurisma de la Aorta Abdominal/metabolismo , Moléculas de Adhesión Celular/sangre , Anciano , Aorta Abdominal/inmunología , Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/inmunología , Estudios de Casos y Controles , Humanos , Macrófagos/fisiología , Masculino , Músculo Liso Vascular/metabolismo
10.
J Vasc Surg ; 66(6): 1854-1863, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28655551

RESUMEN

OBJECTIVE: Postnatal resident endothelium of blood vessels has been proposed to represent terminally differentiated tissue that does not replicate. We previously isolated endothelial colony-forming cells (ECFCs) from human umbilical cord blood (CB) and term placenta by using colony-forming assays and immunocytochemistry. We showed that ECFCs are highly proliferative and form functioning vessels in vivo, the defining characteristics of a true endothelial progenitor cell. This exploratory investigation was conducted to determine whether the endothelium of healthy adult blood vessels contained resident ECFCs. METHODS: The endothelium of great saphenous vein (GSV) obtained from vein stripping procedures was collected with mechanical scraping, and ECFCs were isolated according to established protocols. RESULTS: GSV ECFCs incorporated acetylated low-density lipoprotein, formed tubules in Matrigel (BD Biosciences, San Jose, Calif) at 24 hours, and expressed endothelial antigens cluster of differentiation (CD) 144, CD31, CD105, and kinase insert domain receptor but not hematopoietic antigen CD45. Using cumulative population doublings and single-cell assays, we demonstrated that GSV ECFCs exhibited comparable proliferative capacities compared with CB ECFCs, including similar numbers of highly proliferative cells. When injected in collagen/fibronectin gels implanted in nonobese diabetic/severe combined immune deficiency mice, GSV ECFCs formed blood vessels with circulating murine red blood cells, demonstrating their vasculogenic potential. CONCLUSIONS: The ECFCs of the GSV contain a hierarchy of progenitor cells with a comparable number of highly proliferative clones as ECFCs of CB. The results of this investigation demonstrate that the adult endothelium contains resident progenitor cells that may have a critical role in vascular homeostasis and repair and could potentially be used as a source of autologous cells for cell therapies focusing on vasculogenesis.


Asunto(s)
Células Madre Adultas/fisiología , Proliferación Celular , Células Progenitoras Endoteliales/fisiología , Neovascularización Fisiológica , Vena Safena/citología , Nicho de Células Madre , Células Madre Adultas/metabolismo , Células Madre Adultas/trasplante , Animales , Biomarcadores/metabolismo , Separación Celular/métodos , Células Cultivadas , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/trasplante , Humanos , Cinética , Ratones Endogámicos NOD , Ratones SCID , Fenotipo
11.
J Vasc Surg ; 65(6): 1850-1857.e2, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28390770

RESUMEN

OBJECTIVE: Critical limb ischemia (CLI) continues to place a significant encumbrance on patients and the health care system as it progresses to limb loss and long-term disability. Traditional methods of revascularization offer a significant benefit; however, for one-third of CLI patients, these surgical options are not technically possible or patency is severely limited by disease burden (deemed "poor-option" for revascularization). In a previous phase I trial, we demonstrated intramuscular injection of concentrated bone marrow aspirate (cBMA) via MarrowStim (Zimmer Biomet, Warsaw, Ind) harvest is safe and may decrease major amputation in patients with CLI unfit for surgical revascularization. Therefore, we describe and rationalize the MarrowStim PAD Kit for the Treatment of Critical Limb Ischemia in Subjects with Severe Peripheral Arterial Disease (MOBILE) trial, a study geared to provide the pivotal proof of efficacy of cBMA in CLI. METHODS: MOBILE is a multicenter, randomized, double-blind, placebo-controlled trial designed to assess the efficacy of intramuscular injections of cBMA in promoting amputation-free survival in patients with poor-option CLI. Patients (aged >21 years) with rest pain or tissue loss resulting from advanced peripheral arterial disease, as characterized by ankle-brachial index (<0.6), toe-brachial index (<0.4), or transcutaneous pressure of oxygen (<50 mm Hg), were eligible for inclusion if surgical revascularization was not possible secondary to advanced disease. RESULTS: Treatment and 1-year follow-up of 152 patients enrolled in MOBILE are completed. Long-term follow-up is ongoing. Currently, we are in the process of unblinding the initial results for preliminary data analysis. CONCLUSIONS: If successful, MOBILE could add definitive, high-quality evidence in support of cBMA for the treatment of poor-option CLI patients and provide an additional modality for patients who face amputation secondary to advanced limb ischemia.


Asunto(s)
Trasplante de Médula Ósea/métodos , Separación Celular/instrumentación , Isquemia/cirugía , Extremidad Inferior/irrigación sanguínea , Enfermedad Arterial Periférica/cirugía , Índice Tobillo Braquial , Monitoreo de Gas Sanguíneo Transcutáneo , Trasplante de Médula Ósea/efectos adversos , Protocolos Clínicos , Enfermedad Crítica , Método Doble Ciego , Diseño de Equipo , Humanos , Inyecciones Intramusculares , Isquemia/diagnóstico , Isquemia/fisiopatología , Enfermedad Arterial Periférica/diagnóstico , Enfermedad Arterial Periférica/fisiopatología , Proyectos de Investigación , Índice de Severidad de la Enfermedad , Factores de Tiempo , Trasplante Autólogo , Resultado del Tratamiento
12.
Circ Res ; 118(10): 1512-24, 2016 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-27076598

RESUMEN

RATIONALE: Transmembrane tumor necrosis factor-α (tmTNF-α) is the prime ligand for TNF receptor 2, which has been shown to mediate angiogenic and blood vessel repair activities in mice. We have previously reported that the angiogenic potential of highly proliferative endothelial colony-forming cells (ECFCs) can be explained by the absence of senescent cells, which in mature endothelial cells occupy >30% of the population, and that exposure to a chronic inflammatory environment induced premature, telomere-independent senescence in ECFCs. OBJECTIVE: The goal of this study was to determine the role of tmTNF-α in the proliferation of ECFCs. METHODS AND RESULTS: Here, we show that tmTNF-α expression on ECFCs selects for higher proliferative potential and when removed from the cell surface promotes ECFC senescence. Moreover, the induction of premature senescence by chronic inflammatory conditions is blocked by inhibition of tmTNF-α cleavage. Indeed, the mechanism of chronic inflammation-induced premature senescence involves an abrogation of tmTNF/TNF receptor 2 signaling. This process is mediated by activation of the tmTNF cleavage metalloprotease TNF-α-converting enzyme via p38 MAP kinase activation and its concurrent export to the cell surface by means of increased iRhom2 expression. CONCLUSIONS: Thus, we conclude that tmTNF-α on the surface of highly proliferative ECFCs plays an important role in the regulation of their proliferative capacity.


Asunto(s)
Proliferación Celular , Senescencia Celular , Células Progenitoras Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Anciano , Arterias/citología , Células Cultivadas , Células Progenitoras Endoteliales/citología , Endotelio Vascular/citología , Humanos , Masculino , Metaloproteasas/metabolismo , Persona de Mediana Edad , Proteolisis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Cardiovasc Pathol ; 24(5): 279-82, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26233281

RESUMEN

With effective antiretroviral therapy (ART), many HIV-infected people die of diseases other than acquired immune deficiency syndrome (AIDS). In particular, coronary artery disease has emerged as one of most critical complications of HIV infection and a major cause of morbidity and mortality. Although reportedly antiretroviral combination therapy itself may accelerate atherosclerosis by enhancing dyslipidemia, most recent epidemiological studies support the notion that HIV infection itself contributes to cardiovascular disease. However, it is still a mystery how the virus can contribute to cardiovascular disease development even while suppressed by ARTs. This review discusses the current understanding of interactions between HIV infection and cardiovascular diseases in both clinical and experimental studies with special focus on those viral proteins that are still produced by HIV. This will help infectious disease/vascular biology experts to gain insights into the pathophysiological mechanisms of HIV-associated cardiovascular disease and new trends to treat and prevent cardiovascular disease in the HIV-infected population.


Asunto(s)
Fármacos Anti-VIH/efectos adversos , Enfermedades Cardiovasculares/virología , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Enfermedades Cardiovasculares/etiología , Humanos , Factores de Riesgo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
14.
AIDS Res Hum Retroviruses ; 31(2): 217-20, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25062432

RESUMEN

We report here the novel finding that HIV-negative factor (Nef) protein is present in considerable numbers of peripheral blood mononuclear cells (PBMCs) from viremic HIV-infected patients not on antiretroviral therapy (ART) and also in patients receiving virologically suppressive ART, though to a smaller degree. Interestingly, these Nef-positive PBMCs constitute predominantly uninfected bystander cells. These results may explain systemic pathology in HIV patients, even in those receiving ART.


Asunto(s)
Sangre/virología , Infecciones por VIH/virología , Leucocitos Mononucleares/química , Leucocitos Mononucleares/virología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/análisis , Antirretrovirales/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Humanos
15.
PLoS One ; 9(6): e99545, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24911518

RESUMEN

HIV-1 infection enhances HCV replication and as a consequence accelerates HCV-mediated hepatocellular carcinoma (HCC). However, the precise molecular mechanism by which this takes place is currently unknown. Our data showed that infectious HIV-1 failed to replicate in human hepatocytic cell lines. No discernible virus replication was observed, even when the cell lines transfected with HIV-1 proviral DNA were co-cultured with Jurkat T cells, indicating that the problem of liver deterioration in the co-infected patient is not due to the replication of HIV-1 in the hepatocytes of the HCV infected host. Instead, HIV-1 Nef protein was transferred from nef-expressing T cells to hepatocytic cells through conduits, wherein up to 16% (average 10%) of the cells harbored the transferred Nef, when the hepatocytic cells were co-cultured with nef-expressing Jurkat cells for 24 h. Further, Nef altered the size and numbers of lipid droplets (LD), and consistently up-regulated HCV replication by 1.5∼2.5 fold in the target subgenomic replicon cells, which is remarkable in relation to the initially indolent viral replication. Nef also dramatically augmented reactive oxygen species (ROS) production and enhanced ethanol-mediated up-regulation of HCV replication so as to accelerate HCC. Taken together, these data indicate that HIV-1 Nef is a critical element in accelerating progression of liver pathogenesis via enhancing HCV replication and coordinating modulation of key intra- and extra-cellular molecules for liver decay.


Asunto(s)
Hepacivirus/fisiología , Hepatocitos/metabolismo , Hepatocitos/virología , Linfocitos T/metabolismo , Replicación Viral , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Línea Celular , Técnicas de Cocultivo , Coinfección , Genes Reporteros , Infecciones por VIH , VIH-1 , Humanos , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/virología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
16.
PLoS One ; 9(3): e91063, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24608713

RESUMEN

With effective antiretroviral therapy (ART), cardiovascular diseases (CVD) are emerging as a major cause of morbidity and death in the aging HIV-infected population. To address whether HIV-Nef, a viral protein produced in infected cells even when virus production is halted by ART, can lead to endothelial activation and dysfunction, we tested Nef protein transfer to and activity in endothelial cells. We demonstrated that Nef is essential for major endothelial cell activating effects of HIV-infected Jurkat cells when in direct contact with the endothelium. In addition, we found that Nef protein in endothelial cells is sufficient to cause apoptosis, ROS generation and release of monocyte attractant protein-1 (MCP-1). The Nef protein-dependent endothelial activating effects can be best explained by our observation that Nef protein rapidly transfers from either HIV-infected or Nef-transfected Jurkat cells to endothelial cells between these two cell types. These results are of in vivo relevance as we demonstrated that Nef protein induces GFP transfer from T cells to endothelium in CD4.Nef.GFP transgenic mice and Nef is present in chimeric SIV-infected macaques. Analyzing the signal transduction effects of Nef in endothelial cells, we found that Nef-induced apoptosis is mediated through ROS-dependent mechanisms, while MCP-1 production is NF-kB dependent. Together, these data indicate that inhibition of Nef-associated pathways may be promising new therapeutic targets for reducing the risk for cardiovascular disease in the HIV-infected population.


Asunto(s)
Endotelio/metabolismo , Endotelio/fisiopatología , Espacio Intracelular/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Comunicación Celular , Muerte Celular , Colorantes/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , Humanos , Células Jurkat , Ratones Transgénicos , Nanotubos , Transducción de Señal
17.
Am J Physiol Lung Cell Mol Physiol ; 306(4): L372-82, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24318111

RESUMEN

Chronic lung diseases, such as pulmonary emphysema, are increasingly recognized complications of infection with the human immunodeficiency virus (HIV). Emphysema in HIV may occur independent of cigarette smoking, via mechanisms that are poorly understood but may involve lung endothelial cell apoptosis induced by the HIV envelope protein gp120. Recently, we have demonstrated that lung endothelial apoptosis is an important contributor to the development of experimental emphysema, via upregulation of the proinflammatory cytokine endothelial monocyte-activating polypeptide II (EMAP II) in the lung. Here we investigated the role of EMAP II and its receptor, CXCR3, in gp120-induced lung endothelial cell apoptosis. We could demonstrate that gp120 induces a rapid and robust increase in cell surface expression of EMAP II and its receptor CXCR3. This surface expression occurred via a mechanism involving gp120 signaling through its CXCR4 receptor and p38 MAPK activation. Both EMAP II and CXCR3 were essentially required for gp120-induced apoptosis and exposures to low gp120 concentrations enhanced the susceptibility of endothelial cells to undergo apoptosis when exposed to soluble cigarette smoke extract. These data indicate a novel mechanism by which HIV infection causes endothelial cell loss involved in lung emphysema formation, independent but potentially synergistic with smoking, and suggest therapeutic targets for emphysema prevention and/or treatment.


Asunto(s)
Apoptosis , Citocinas/metabolismo , Células Endoteliales/fisiología , Proteína gp120 de Envoltorio del VIH/fisiología , Proteínas de Neoplasias/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores CXCR3/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Células Endoteliales/virología , Infecciones por VIH/complicaciones , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , Humanos , Pulmón/irrigación sanguínea , Microvasos/patología , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/patología , Enfisema Pulmonar/virología , Receptores CXCR4/metabolismo , Transducción de Señal , Fumar/efectos adversos , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Am J Respir Cell Mol Biol ; 47(6): 807-14, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22936405

RESUMEN

Endothelial monocyte-activating polypeptide II (EMAP II) and interferon-inducible protein (IP)-10 are proinflammatory mediators, which in addition to their chemokine activities, selectively induce apoptosis in endothelial cells and are up-regulated in the lungs of cigarette smoke-exposed humans. Previously, we showed that EMAP II is an essential mediator of cigarette smoke-induced lung emphysema in mice linking endothelial cell apoptosis with inflammation. Here we addressed the role of the CXCR3 receptor in EMAP II-induced and IP-10-induced apoptosis in endothelial cells and its regulation by cigarette smoke. We found that both neutralizing antibodies and small inhibitory RNA to CXCR3 abrogated EMAP II-induced and IP-10-induced endothelial caspase-3 activation and DNA fragmentation. CXCR3 receptor surface expression in human lung microvascular endothelial cells and in lung tissue endothelium was up-regulated by exposure to cigarette smoke. In tissue culture conditions, EMAP II-induced and IP-10-induced apoptosis was enhanced by preincubation with cigarette smoke extract. Interestingly, serum starvation also induced CXCR3 up-regulation and enhanced EMAP II-induced endothelial apoptosis. Signal transduction via p38 mitogen-activated protein kinase activation was essential for CXCR3-induced cell death, but not for CXCR3 receptor up-regulation by cigarette smoke. In turn, protein nitration was required for CXCR3 receptor up-regulation by cigarette smoke and consequently for subsequent CXCR3-induced cell death. In conclusion, the concerted up-regulation of proinflammatory EMAP II, IP-10, and CXCR3 by cigarette smoke could sustain a cascade of cell death that may promote the alveolar tissue loss noted in human emphysema.


Asunto(s)
Apoptosis , Células Endoteliales/metabolismo , Nicotiana/química , Extractos Vegetales/farmacología , Receptores CXCR3/metabolismo , Humo , Regulación hacia Arriba/efectos de los fármacos , Animales , Células Cultivadas , Quimiocina CXCL10/farmacología , Medio de Cultivo Libre de Suero , Citocinas/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Endotelio Vascular/citología , Femenino , Humanos , Pulmón/citología , Pulmón/efectos de los fármacos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Microvasos/citología , Proteínas de Neoplasias/farmacología , Proteínas de Unión al ARN/farmacología , Receptores CXCR3/genética , Tirosina/análogos & derivados , Tirosina/metabolismo
19.
AIDS Res Hum Retroviruses ; 28(10): 1207-15, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22463742

RESUMEN

Untreated HIV infection is associated with endothelial dysfunction and subsequent cardiovascular disease, likely due to both direct effects of the virus and to indirect effects of systemic inflammation on the vasculature. We have recently shown that treatment with the antiinflammatory agent pentoxifylline (PTX) improved in vivo endothelial function and reduced circulating levels of the inflammatory markers vascular cell adhesion molecule-1 (VCAM-1) and interferon-gamma-induced protein (IP-10) in HIV-infected patients. To delineate the mechanisms underlying this therapeutic effect, we tested whether clinically relevant concentrations of PTX suppress VCAM-1 or IP-10 release in cultivated human lung microvascular endothelial cells. Indeed, we found that tumor necrosis factor (TNF)-α-induced VCAM-1 was reduced with concentrations of PTX in the low nanomolar range, comparable to plasma levels in PTX-treated groups. We also investigated the effect of HIV proteins and found that HIV transactivator of transcription (HIV-Tat) and HIV-envelope-derived recombinant gp120 enhanced TNF-α-induced VCAM-1 gene expression in lung microvascular and coronary macrovascular endothelial cells, respectively. In addition, PTX and a NF-κB-specific inhibitor reduced this enhanced VCAM-1 gene induction in microvascular and macrovascular endothelial cells. These results provide novel insights in how the antiinflammatory agent PTX can directly reduce HIV-associated proinflammatory endothelial activation, which may underlie vascular dysfunction and coronary vascular diseases.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/inmunología , Antiinflamatorios/farmacología , Quimiocina CXCL10/inmunología , VIH-1/inmunología , Pentoxifilina/farmacología , Factor de Necrosis Tumoral alfa/inmunología , Molécula 1 de Adhesión Celular Vascular/inmunología , Síndrome de Inmunodeficiencia Adquirida/tratamiento farmacológico , Células Cultivadas , Quimiocina CXCL10/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , VIH-1/efectos de los fármacos , Humanos , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa , Activación Transcripcional/efectos de los fármacos , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/efectos de los fármacos , Vasodilatadores/farmacología , Replicación Viral/efectos de los fármacos
20.
Antivir Ther ; 16(8): 1335-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22155915

RESUMEN

BACKGROUND: The use of abacavir and didanosine in HAART has been associated with an increased risk of myocardial infarction in HIV-infected patients. The aim of this study was to address the development of endothelial dysfunction in cultivated coronary artery endothelial cells (HCAECs) in response to abacavir, didanosine and tenofovir. We examined the impact of these drugs on the expression levels of the proinflammatory, oxidative stress and apoptosis regulating genes in HCAECs. METHODS: We tested gene and protein expression changes in HCAECs in response to abacavir, didanosine and tenofovir using quantitative real-time reverse transciptase PCR, FACS and ELISA. The assessed genes/proteins included the proinflammatory molecules VCAM-1, ICAM-1, MCP-1, RANTES and IL-6. In addition, we assessed the gene expression of the intracellular reactive oxygen producing NADPH oxidase subunit gp91(PHOX) and the apoptosis regulating molecules Bcl-2 and BAD. RESULTS: Exposure of HCAECs to abacavir, didanosine and tenofovir resulted in no statistically significant changes in any of the tested genes/proteins at any time point or at any concentration. CONCLUSIONS: We found no evidence that abacavir, didanosine or tenofovir had direct in vitro effects on coronary endothelial cell gene transcription and protein expression of the selected mediators. If abacavir or didanosine increase cardiovascular risk, it is likely not through the direct endothelial activation pathways tested in these experiments. However, further studies are needed to completely exclude the toxicity of abacavir or didanosine on endothelial cells.


Asunto(s)
Fármacos Anti-VIH/administración & dosificación , Moléculas de Adhesión Celular/metabolismo , Vasos Coronarios/efectos de los fármacos , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Expresión Génica , NADPH Oxidasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Adenina/administración & dosificación , Adenina/efectos adversos , Adenina/análogos & derivados , Adenina/uso terapéutico , Fármacos Anti-VIH/efectos adversos , Fármacos Anti-VIH/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Cardiovasculares/etiología , Moléculas de Adhesión Celular/genética , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Citocinas/genética , Didanosina/administración & dosificación , Didanosina/efectos adversos , Didanosina/uso terapéutico , Didesoxinucleósidos/administración & dosificación , Didesoxinucleósidos/efectos adversos , Didesoxinucleósidos/uso terapéutico , Células Endoteliales/citología , Células Endoteliales/metabolismo , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/fisiología , Humanos , Inflamación/genética , Inflamación/metabolismo , NADPH Oxidasas/genética , Organofosfonatos/administración & dosificación , Organofosfonatos/efectos adversos , Organofosfonatos/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Cultivo Primario de Células , Proteínas Proto-Oncogénicas/genética , Tenofovir
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...