Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Blood Adv ; 8(9): 2290-2299, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38447116

RESUMEN

ABSTRACT: Patients treated with antineoplastic therapy often develop thrombocytopenia requiring platelet transfusion, which has potential to exacerbate pulmonary injury. This study tested the hypothesis that amotosalen-UVA pathogen-reduced platelet components (PRPCs) do not potentiate pulmonary dysfunction compared with conventional platelet components (CPCs). A prospective, multicenter, open-label, sequential cohort study evaluated the incidence of treatment-emergent assisted mechanical ventilation initiated for pulmonary dysfunction (TEAMV-PD). The first cohort received CPC. After the CPC cohort, each site enrolled a second cohort transfused with PRPC. Other outcomes included clinically significant pulmonary adverse events (CSPAE) and the incidence of treatment-emergent acute respiratory distress syndrome (TEARDS) diagnosed by blinded expert adjudication. The incidence of TEAMV-PD in all patients (1068 PRPC and 1223 CPC) was less for PRPC (1.7 %) than CPC (3.1%) with a treatment difference of -1.5% (95% confidence interval [CI], -2.7 to -0.2). In patients requiring ≥2 PCs, the incidence of TEAMV-PD was reduced for PRPC recipients compared with CPC recipients (treatment difference, -2.4%; 95% CI, -4.2 to -0.6). CSPAE increased with increasing PC exposure but were not significantly different between the cohorts. For patients receiving ≥2 platelet transfusions, TEARDS occurred in 1.3% PRPC and 2.6% CPC recipients (P = .086). Bayesian analysis demonstrated PRPC may be superior in reducing TEAMV-PD and TEARDS for platelet transfusion recipients compared with CPC recipients, with 99.2% and 88.8% probability, respectively. In this study, PRPC compared with CPC demonstrated high probability of reduced severe pulmonary injury requiring assisted mechanical ventilation in patients with hematology disorders dependent on platelet transfusion. This trial was registered at www.ClinicalTrials.gov as #NCT02549222.


Asunto(s)
Transfusión de Plaquetas , Humanos , Transfusión de Plaquetas/efectos adversos , Femenino , Persona de Mediana Edad , Masculino , Anciano , Lesión Pulmonar Aguda/etiología , Plaquetas , Estudios Prospectivos , Adulto , Trombocitopenia/etiología , Enfermedades Hematológicas/terapia
2.
Transfusion ; 62(7): 1365-1376, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35748490

RESUMEN

BACKGROUND: Platelet transfusion carries risk of transfusion-transmitted infection (TTI). Pathogen reduction of platelet components (PRPC) is designed to reduce TTI. Pulmonary adverse events (AEs), including transfusion-related acute lung injury and acute respiratory distress syndrome (ARDS) occur with platelet transfusion. STUDY DESIGN: An open label, sequential cohort study of transfusion-dependent hematology-oncology patients was conducted to compare pulmonary safety of PRPC with conventional PC (CPC). The primary outcome was the incidence of treatment-emergent assisted mechanical ventilation (TEAMV) by non-inferiority. Secondary outcomes included: time to TEAMV, ARDS, pulmonary AEs, peri-transfusion AE, hemorrhagic AE, transfusion reactions (TRs), PC and red blood cell (RBC) use, and mortality. RESULTS: By modified intent-to-treat (mITT), 1068 patients received 5277 PRPC and 1223 patients received 5487 CPC. The cohorts had similar demographics, primary disease, and primary therapy. PRPC were non-inferior to CPC for TEAMV (treatment difference -1.7%, 95% CI: (-3.3% to -0.1%); odds ratio = 0.53, 95% CI: (0.30, 0.94). The cumulative incidence of TEAMV for PRPC (2.9%) was significantly less than CPC (4.6%, p = .039). The incidence of ARDS was less, but not significantly different, for PRPC (1.0% vs. 1.8%, p = .151; odds ratio = 0.57, 95% CI: (0.27, 1.18). AE, pulmonary AE, and mortality were not different between cohorts. TRs were similar for PRPC and CPC (8.3% vs. 9.7%, p = .256); and allergic TR were significantly less with PRPC (p = .006). PC and RBC use were not increased with PRPC. DISCUSSION: PRPC demonstrated reduced TEAMV with no excess treatment-related pulmonary morbidity.


Asunto(s)
Síndrome de Dificultad Respiratoria , Reacción a la Transfusión , Plaquetas , Transfusión Sanguínea , Estudios de Cohortes , Humanos , Fármacos Fotosensibilizantes , Transfusión de Plaquetas/efectos adversos , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/terapia , Reacción a la Transfusión/epidemiología , Reacción a la Transfusión/etiología
3.
Blood Transfus ; 19(6): 456-466, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34369863

RESUMEN

BACKGROUND: Leukoreduction to eliminate mononuclear cells within blood products is necessary to prevent graft-versus-host disease after transfusion. Published reports document low concentrations of mononuclear cells leftover in fresh-frozen plasma products, however the phenotype and the proliferative potential of these cells has not been tested. MATERIALS AND METHODS: We investigated residual cellular components contained within fresh and fresh-frozen plasma products and characterised their proliferative potential in co-cultures with unrelated allogeneic cells. We designed a flow-based assay to phenotype cells and quantify cell division by measuring the dilution of fluorescently labeled protein as cells divide. Leukocytes from consenting donors were purified from fresh liquid or fresh-frozen plasma units and cultured for three to seven days with unrelated irradiated allogeneic targets. RESULTS: We discovered a median of 1.6×107 viable lymphocytes were detectable in fresh plasma units after collection (n=8), comprised of a mixture of CD3+ CD8+ and CD3+ CD4+ cells. Furthermore, we identified a median of 8.4% of live CD3+ plasma lymphocytes divided as early as Day 4 when co-cultured with unrelated allogeneic cells, expanding to a median 88.8% by Day 7 (n=3). Although freezing the plasma product reduced the total number of viable leukocyte cells down to 2.3×105 (n=10), residual naive CD3+ cells were viable and demonstrated division through Day 7 of co-culture. DISCUSSION: The evidence of viable proliferative lymphocytes in fresh and fresh-frozen plasma products derived from centrifugation suggests that additional leukoreduction measures should be investigated to fully eradicate reactive lymphocytes from centrifuged plasma products.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , División Celular , Humanos , Leucocitos , Prueba de Cultivo Mixto de Linfocitos , Linfocitos
4.
Transfusion ; 61(6): 1780-1788, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33665853

RESUMEN

BACKGROUND: Severe blood donor adverse events are rare, but due to their rarity studying them can be difficult. To get an accurate estimate of their frequency and rate in the donor population it may be necessary to combine donation data across countries. STUDY DESIGN AND METHODS: International blood collection organizations (BCOs) provided data on rare/severe donor reactions as well as denominator information for their donor populations from 2015 to 2017. Donor reactions were classified using standardized definitions. RESULTS: BCOs from six countries provided reaction data for more than 22 million donations. A total of 480 rare reactions were reported of which 76.7% were imputed as definite and 11% probable. Rates of rare reactions were higher in females and first-time donors. Systemic rare reactions were the most common reaction type, accounting for over three quarters of reactions reported. Of systemic reactions, vasovagal reactions with loss of consciousness and injury or off-site (n = 350) made up the majority and occurred 1.53 per 100,000 donations. For the 22.3% that were localized reactions, the majority of these were cellulitis (n = 71, 0.31 per 100,000 donations) followed by deep venous thrombosis (n = 21, 0.09 per 100,000 donations). CONCLUSION: Pulling together data from multiple BCOs across countries allows for a better understanding of rare reactions, such as vasovagal reaction with injury or cellulitis, and for generating a reliable incidence rate for air embolism or compartment syndrome. However, gaps remain due to missing elements such as unknown donor status or location of reaction.


Asunto(s)
Donantes de Sangre , Celulitis (Flemón)/etiología , Síncope Vasovagal/etiología , Trombosis de la Vena/etiología , Adolescente , Adulto , Anciano , Eliminación de Componentes Sanguíneos/efectos adversos , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Factores de Riesgo , Adulto Joven
5.
Transfusion ; 61(2): 641-648, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33616945

RESUMEN

BACKGROUND: Pathogen reduction technology and enhanced bacterial culture screening promise to significantly reduce the risk of transfusion-associated septic reactions due to contaminated platelets. Recent reports suggest that these interventions lack efficacy for post-collection and processing contamination with environmental organisms if the storage bag integrity is compromised. CASE REPORT: We report a fatal septic transfusion reaction in a 63-year-old patient with chronic kidney and liver disease who received a pathogen reduced platelet transfusion in anticipation of surgery. METHODS: The residual platelet concentrate was cultured, with the detected microorganisms undergoing 16S genotype sequencing. Separate pathogen reduction studies were performed on the recovered bacteria, including assessment for amotosalen photoproducts. The storage container was subjected to pressure testing and microscopic examination. Environmental culture screening was performed at the hospital. RESULTS: Gram negative rods were detected in the platelet unit and cultures of both platelet component and the patient's blood grew Acinetobacter baumannii complex, Leclercia adecarboxylata and Staphylococcus saprophyticus. These strains were effectively inactivated with >7.2, 7.7, and >7.1 log10 kill, respectively. The platelet storage container revealed a leak visible only on pressure testing. Hospital environmental cultures were negative and the contamination source is unknown. A. baumannii complex and S. saprophyticus 16S genotyping sequences were identical to those implicated in a previously reported septic reaction. CONCLUSION: Findings are compatible with post-processing environmental contamination of a pathogen reduced platelet concentrate via a non-visible, acquired storage container leak. Efforts are warranted to actively prevent damage to, and detect defects in, platelet storage containers, and to store and transport components in clean environments.


Asunto(s)
Infecciones por Acinetobacter/etiología , Coinfección/etiología , Infección Hospitalaria/etiología , Infecciones por Enterobacteriaceae/etiología , Contaminación de Equipos , Falla de Equipo , Transfusión de Plaquetas/efectos adversos , Transfusión de Plaquetas/instrumentación , Sepsis/etiología , Infecciones Estafilocócicas/etiología , Reacción a la Transfusión/etiología , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/aislamiento & purificación , Plaquetas/microbiología , Patógenos Transmitidos por la Sangre/efectos de los fármacos , Patógenos Transmitidos por la Sangre/efectos de la radiación , Coinfección/microbiología , Infección Hospitalaria/microbiología , Enterobacteriaceae/aislamiento & purificación , Infecciones por Enterobacteriaceae/microbiología , Resultado Fatal , Furocumarinas , Fracturas de Cadera/complicaciones , Humanos , Masculino , Persona de Mediana Edad , Sepsis/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus saprophyticus/aislamiento & purificación , Trombocitopenia/complicaciones , Trombocitopenia/terapia , Reacción a la Transfusión/microbiología , Rayos Ultravioleta
6.
Ann Surg ; 273(3): 395-401, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33065652

RESUMEN

OBJECTIVE: To address the clinical and regulatory challenges of optimal primary endpoints for bleeding patients by developing consensus-based recommendations for primary clinical outcomes for pivotal trials in patients within 6 categories of significant bleeding, (1) traumatic injury, (2) intracranial hemorrhage, (3) cardiac surgery, (4) gastrointestinal hemorrhage, (5) inherited bleeding disorders, and (6) hypoproliferative thrombocytopenia. BACKGROUND: A standardized primary outcome in clinical trials evaluating hemostatic products and strategies for the treatment of clinically significant bleeding will facilitate the conduct, interpretation, and translation into clinical practice of hemostasis research and support alignment among funders, investigators, clinicians, and regulators. METHODS: An international panel of experts was convened by the National Heart Lung and Blood Institute and the United States Department of Defense on September 23 and 24, 2019. For patients suffering hemorrhagic shock, the 26 trauma working-group members met for almost a year, utilizing biweekly phone conferences and then an in-person meeting, evaluating the strengths and weaknesses of previous high quality studies. The selection of the recommended primary outcome was guided by goals of patient-centeredness, expected or demonstrated sensitivity to beneficial treatment effects, biologic plausibility, clinical and logistical feasibility, and broad applicability. CONCLUSIONS: For patients suffering hemorrhagic shock, and especially from truncal hemorrhage, the recommended primary outcome was 3 to 6-hour all-cause mortality, chosen to coincide with the physiology of hemorrhagic death and to avoid bias from competing risks. Particular attention was recommended to injury and treatment time, as well as robust assessments of multiple safety related outcomes.


Asunto(s)
Ensayos Clínicos como Asunto , Hemostasis Quirúrgica/métodos , Evaluación de Resultado en la Atención de Salud , Choque Hemorrágico/etiología , Choque Hemorrágico/prevención & control , Consenso , Medicina Basada en la Evidencia , Hemostáticos/uso terapéutico , Humanos , Atención Dirigida al Paciente , Choque Hemorrágico/mortalidad
7.
Pediatr Blood Cancer ; 67(11): e28693, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32885904

RESUMEN

There are no proven safe and effective therapies for children who develop life-threatening complications of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Convalescent plasma (CP) has demonstrated potential benefit in adults with SARS-CoV-2, but has theoretical risks.We present the first report of CP in children with life-threatening coronavirus disease 2019 (COVID-19), providing data on four pediatric patients with acute respiratory distress syndrome. We measured donor antibody levels and recipient antibody response prior to and following CP infusion. Infusion of CP was not associated with antibody-dependent enhancement (ADE) and did not suppress endogenous antibody response. We found CP was safe and possibly efficacious. Randomized pediatric trials are needed.


Asunto(s)
COVID-19/terapia , Síndrome de Dificultad Respiratoria/terapia , Adolescente , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/uso terapéutico , COVID-19/complicaciones , Humanos , Inmunización Pasiva/métodos , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Síndrome de Dificultad Respiratoria/etiología , SARS-CoV-2/inmunología , Índice de Severidad de la Enfermedad , Sueroterapia para COVID-19
8.
Transfusion ; 60(8): 1811-1820, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32654200

RESUMEN

BACKGROUND: Growth in size and complexity of clinical hematopoietic progenitor cell (HPC) transplant programs necessitates parallel increases in cellular therapy laboratory (CTL) workload. Typically individually developed, HPC product processing is labor and time intensive. Variation in procedure type and numbers across CTLs complicates direct comparisons, and benchmark data are not readily available. STUDY DESIGN AND METHODS: Studies were undertaken at seven CTLs. Transplant volume and staff numbers were determined. Staff recorded time performing tasks broken down into steps: paperwork, product acceptance, transport/infusion, processing, and cryopreservation. Times were added to obtain total times for 15 common CTL procedures. RESULTS: Annual transplant volume ranged from 53.4 to 463.2, with products processed by a range of 2 to 10 dedicated CTL staff. Paperwork time constituted 23.7% to 62.3% total time; product processing time accounted for 1.8 (for National Marrow Donor Program product receipt) to 62.6% (for red blood cell reduction of allogeneic HPC products from bone marrow) of total processing time. Mean time for 15 procedures ranged from 1.27 to 8.28 hours (standard deviation range, 0.35-2.71 hr). Mean time for products from bone marrow versus peripheral blood was 6.6 ± 2.0 versus 5.5 ± 1.1 hours (p = 0.02). Cryopreservation (6.5 ± 1.6 vs. 4.4 ± 0.85 hr; p < 0.01) and manipulation (6.4 ± 1.5 vs. 4.4 ± 0.85 hr; p < 0.01) added time. CONCLUSION: CTL procedures are time intensive, with wide intra- and inter-CTL variation. Paperwork accounted for substantial portion of total time across procedures. Bone marrow source, cryopreservation, and manipulation contributed to longer times. These findings provide concrete data on which to build regarding CTL workload capacity.


Asunto(s)
Trasplante de Médula Ósea , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Laboratorios de Hospital , Carga de Trabajo , Aloinjertos , Humanos
9.
Nat Commun ; 10(1): 3027, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31289275

RESUMEN

Fibrosis accompanying wound healing can drive the failure of many different organs. Activated fibroblasts are the principal determinants of post-injury pathological fibrosis along with physiological repair, making them a difficult therapeutic target. Although activated fibroblasts are phenotypically heterogeneous, they are not recognized as distinct functional entities. Using mice that express GFP under the FSP1 or αSMA promoter, we characterized two non-overlapping fibroblast subtypes from mouse hearts after myocardial infarction. Here, we report the identification of FSP1-GFP+ cells as a non-pericyte, non-hematopoietic fibroblast subpopulation with a predominant pro-angiogenic role, characterized by in vitro phenotypic/cellular/ultrastructural studies and in vivo granulation tissue formation assays combined with transcriptomics and proteomics. This work identifies a fibroblast subtype that is functionally distinct from the pro-fibrotic αSMA-expressing myofibroblast subtype. Our study has the potential to shift our focus towards viewing fibroblasts as molecularly and functionally heterogeneous and provides a paradigm to approach treatment for organ fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Infarto del Miocardio/patología , Miocardio/patología , Neovascularización Fisiológica/fisiología , Cicatrización de Heridas/fisiología , Actinas/genética , Actinas/metabolismo , Animales , Trasplante de Médula Ósea , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Fibroblastos/patología , Fibrosis/etiología , Fibrosis/patología , Proteínas Fluorescentes Verdes/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/etiología , Infarto del Miocardio/fisiopatología , Miocardio/citología , Proteína de Unión al Calcio S100A4/genética , Proteína de Unión al Calcio S100A4/metabolismo , Quimera por Trasplante
10.
Transfusion ; 58(9): 2184-2191, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30204955

RESUMEN

BACKGROUND: Viability testing is a common practice in laboratories. The goal of this study was to ascertain current laboratory practices internationally for performing viability testing for cryopreserved cord blood (CB) products and glean information about how to standardize the method to improve interlaboratory reproducibility. STUDY DESIGN AND METHODS: A survey to evaluate current laboratory practices for viability testing was designed and distributed internationally. The question topics included sampling and testing methods, responses to unexpected results, and the rating of the reliability of the CB quality tests, together with expectations for standardization. RESULTS: There were 32 respondents to the survey, of whom 28 responded to the more detailed questionnaire about viability methods. Overall, responses indicated that various stains were used among the laboratories, and when multiple sites used the same viability stain the methods differed. The majority of the respondents were in favor of standardizing the viability testing methods. A wide variety of preferences were communicated about how to standardize the method, but a majority did advocate the use of 7-aminoactinomycin D (7-AAD) with flow cytometry. CONCLUSIONS: The survey results revealed a variety of tests and inconsistent interlaboratory practices for performing the viability assay. Flow cytometry with a 7-AAD dye was suggested as a first step toward standardization.


Asunto(s)
Conservación de la Sangre/métodos , Seguridad de la Sangre , Trasplante de Células Madre de Sangre del Cordón Umbilical , Criopreservación/métodos , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Núcleo Celular/ultraestructura , Separación Celular/métodos , Supervivencia Celular , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Trasplante de Células Madre de Sangre del Cordón Umbilical/normas , Dactinomicina/análogos & derivados , Citometría de Flujo/métodos , Colorantes Fluorescentes , Encuestas de Atención de la Salud , Células Madre Hematopoyéticas/ultraestructura , Humanos , Recién Nacido , Cooperación Internacional , Internet , Laboratorios/normas , Utilización de Procedimientos y Técnicas , Reproducibilidad de los Resultados
11.
JCI Insight ; 3(3)2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29415892

RESUMEN

Osteoarthritis (OA) is a degenerative joint disease involving both cartilage and synovium. The canonical Wnt/ß-catenin pathway, which is activated in OA, is emerging as an important regulator of tissue repair and fibrosis. This study seeks to examine Wnt pathway effects on synovial fibroblasts and articular chondrocytes as well as the therapeutic effects of Wnt inhibition on OA disease severity. Mice underwent destabilization of the medial meniscus surgery and were treated by intra-articular injection with XAV-939, a small-molecule inhibitor of Wnt/ß-catenin signaling. Wnt/ß-catenin signaling was highly activated in murine synovial fibroblasts as well as in OA-derived human synovial fibroblasts. XAV-939 ameliorated OA severity associated with reduced cartilage degeneration and synovitis in vivo. Wnt inhibition using mechanistically distinct small-molecule inhibitors, XAV-939 and C113, attenuated the proliferation and type I collagen synthesis in synovial fibroblasts in vitro but did not affect human OA-derived chondrocyte proliferation. However, Wnt modulation increased COL2A1 and PRG4 transcripts, which are downregulated in chondrocytes in OA. In conclusion, therapeutic Wnt inhibition reduced disease severity in a model of traumatic OA via promoting anticatabolic effects on chondrocytes and antifibrotic effects on synovial fibroblasts and may be a promising class of drugs for the treatment of OA.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Osteoartritis/tratamiento farmacológico , Vía de Señalización Wnt/efectos de los fármacos , Animales , Cartílago Articular/citología , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Colágeno Tipo II/metabolismo , Modelos Animales de Enfermedad , Humanos , Inyecciones Intraarticulares , Masculino , Ratones , Células 3T3 NIH , Osteoartritis/patología , Cultivo Primario de Células , Proteoglicanos/metabolismo , Membrana Sinovial/citología , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/patología , beta Catenina/metabolismo
12.
PLoS One ; 12(9): e0184620, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28886156

RESUMEN

Vascular smooth muscle cells (VSMCs) represent important modulators of plaque stability in advanced lesions. We previously reported that loss of small proline-rich repeat protein 3 (Sprr3), leads to VSMC apoptosis in a PI3K/Akt-dependent manner and accelerates lesion progression. Here, we investigated the role of Sprr3 in modulating plaque stability in hyperlipidemic ApoE-/- mice. We show that loss of Sprr3 increased necrotic core size and reduced cap collagen content of atheromas in brachiocephalic arteries with evidence of plaque rupture and development of intraluminal thrombi. Moreover, Sprr3-/-ApoE-/- mice developed advanced coronary artery lesions accompanied by intraplaque hemorrhage and left ventricle microinfarcts. SPRR3 is known to reduce VSMC survival in lesions by promoting their apoptosis. In addition, we demonstrated that Sprr3-/- VSMCs displayed reduced expression of procollagen in a PI3K/Akt dependent manner. SPRR3 loss also increased MMP gelatinase activity in lesions, and increased MMP2 expression, migration and contraction of VSMCs independently of PI3K/Akt. Consequently, Sprr3 represents the first described VSMC modulator of each of the critical features of cap stability, including VSMC numbers, collagen type I synthesis, and protease activity through Akt dependent and independent pathways.


Asunto(s)
Apolipoproteínas E/metabolismo , Proteínas Ricas en Prolina del Estrato Córneo/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Apolipoproteínas E/genética , Proteínas Ricas en Prolina del Estrato Córneo/genética , Femenino , Fibronectinas/metabolismo , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Transducción de Señal/fisiología
13.
N Engl J Med ; 372(15): 1419-29, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25853746

RESUMEN

BACKGROUND: Some observational studies have reported that transfusion of red-cell units that have been stored for more than 2 to 3 weeks is associated with serious, even fatal, adverse events. Patients undergoing cardiac surgery may be especially vulnerable to the adverse effects of transfusion. METHODS: We conducted a randomized trial at multiple sites from 2010 to 2014. Participants 12 years of age or older who were undergoing complex cardiac surgery and were likely to undergo transfusion of red cells were randomly assigned to receive leukocyte-reduced red cells stored for 10 days or less (shorter-term storage group) or for 21 days or more (longer-term storage group) for all intraoperative and postoperative transfusions. The primary outcome was the change in Multiple Organ Dysfunction Score (MODS; range, 0 to 24, with higher scores indicating more severe organ dysfunction) from the preoperative score to the highest composite score through day 7 or the time of death or discharge. RESULTS: The median storage time of red-cell units provided to the 1098 participants who received red-cell transfusion was 7 days in the shorter-term storage group and 28 days in the longer-term storage group. The mean change in MODS was an increase of 8.5 and 8.7 points, respectively (95% confidence interval for the difference, -0.6 to 0.3; P=0.44). The 7-day mortality was 2.8% in the shorter-term storage group and 2.0% in the longer-term storage group (P=0.43); 28-day mortality was 4.4% and 5.3%, respectively (P=0.57). Adverse events did not differ significantly between groups except that hyperbilirubinemia was more common in the longer-term storage group. CONCLUSIONS: The duration of red-cell storage was not associated with significant differences in the change in MODS. We did not find that the transfusion of red cells stored for 10 days or less was superior to the transfusion of red cells stored for 21 days or more among patients 12 years of age or older who were undergoing complex cardiac surgery. (Funded by the National Heart, Lung, and Blood Institute; RECESS ClinicalTrials.gov number, NCT00991341.).


Asunto(s)
Conservación de la Sangre , Procedimientos Quirúrgicos Cardíacos , Transfusión de Eritrocitos , Adulto , Anciano , Tipificación y Pruebas Cruzadas Sanguíneas , Transfusión de Eritrocitos/efectos adversos , Femenino , Humanos , Análisis de Intención de Tratar , Tiempo de Internación , Masculino , Persona de Mediana Edad , Mortalidad , Insuficiencia Multiorgánica/clasificación , Modelos de Riesgos Proporcionales , Índice de Severidad de la Enfermedad , Factores de Tiempo
14.
Transfusion ; 55(2): 441-51; quiz 440, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25145464

RESUMEN

Cell-based therapy is an exciting, promising, and a developing new treatment for cardiac diseases. Stem cell-based therapies have the potential to fundamentally transform the treatment of ischemic cardiac injury and heart failure by achieving what would have been unthinkable only a few years ago-the Holy Grail of myocardial regeneration. Recent therapeutic approaches involve bone marrow (BM)-derived mononuclear cells and their subsets such as mesenchymal stem/stromal cells (MSCs), endothelial progenitor cells as well as adipose tissue-derived MSCs, cardiac tissue-derived stem cells, and cell combinations. Clinical trials employing these cells have demonstrated that cellular therapy is feasible and safe. Regarding delivery methods, the safety of catheter-based, transendocardial and -epicardial stem cell injection has been established. However, the results, while variable, suggest rather modest clinical efficacy overall in both heart failure and ischemic heart disease, such as in acute myocardial infarction. Future studies will focus on determining the most efficacious cell type(s) and/or cell combinations and the most reasonable indications and optimal timing of transplantation, as well as the mechanisms underlying their therapeutic effects. We will review and summarize the clinical trial results to date. In addition, we discuss challenges and operational issues in cell processing for cardiac applications.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Cardiopatías/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Miocardio/metabolismo , Ensayos Clínicos como Asunto , Cardiopatías/metabolismo , Cardiopatías/patología , Humanos , Miocardio/patología
15.
Stem Cells ; 33(4): 1333-44, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25537659

RESUMEN

MSCs encounter extended hypoxia in the wound microenvironment yet little is known about their adaptability to this prolonged hypoxic milieu. In this study, we evaluated the cellular and molecular response of MSCs in extended hypoxia (1% O2 ) versus normoxia (20% O2 ) culture. Prolonged hypoxia induced a switch toward anaerobic glycolysis transcriptome and a dramatic increase in the transcript and protein levels of monocarboxylate transporter-4 (MCT4) in MSCs. To clarify the impact of MCT4 upregulation on MSC biology, we generated MSCs which stably overexpressed MCT4 (MCT4-MSCs) at levels similar to wild-type MSCs following prolonged hypoxic culture. Consistent with its role to efflux lactate to maintain intracellular pH, MCT4-MSCs demonstrated reduced intracellular lactate. To explore the in vivo significance of MCT4 upregulation in MSC therapy, mice were injected intramuscularly following MI with control (GFP)-MSCs, MCT4-MSCs, or MSCs in which MCT4 expression was stably silenced (KDMCT4-MSCs). Overexpression of MCT4 worsened cardiac remodeling and cardiac function whereas silencing of MCT4 significantly improved cardiac function. MCT4-overexpressing MSC secretome induced reactive oxygen species-mediated cardiomyocyte but not fibroblast apoptosis in vitro and in vivo; lactate alone recapitulated the effects of the MCT4-MSC secretome. Our findings suggest that lactate extruded by MCT4-overexpressing MSCs preferentially induced cell death in cardiomyocytes but not in fibroblasts, leading ultimately to a decline in cardiac function and increased scar size. A better understanding of stem cells response to prolonged hypoxic stress and the resultant stem cell-myocyte/fibroblast cross-talk is necessary to optimize MSC-based therapy for cardiac regeneration.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Transportadores de Ácidos Monocarboxílicos/biosíntesis , Proteínas Musculares/biosíntesis , Isquemia Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Transcriptoma/fisiología , Animales , Hipoxia de la Célula/fisiología , Células Cultivadas , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Isquemia Miocárdica/patología , Células 3T3 NIH , Factores de Tiempo
16.
Arterioscler Thromb Vasc Biol ; 34(12): 2527-36, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25278290

RESUMEN

OBJECTIVE: Atherosclerosis is the primary driver of cardiovascular disease, the leading cause of death worldwide. Identification of naturally occurring atheroprotective genes has become a major goal for the development of interventions that will limit atheroma progression and associated adverse events. To this end, we have identified small proline-rich repeat protein (SPRR3) as selectively upregulated in vascular smooth muscle cells (VSMCs) of atheroma-bearing arterial tissue versus healthy arterial tissue. In this study, we sought to determine the role of SPRR3 in atheroma pathophysiology. APPROACH AND RESULTS: We found that atheroprone apolipoprotein E-null mice lacking SPRR3 developed significantly greater atheroma burden. To determine the cellular driver(s) of this increase, we evaluated SPRR3-dependent changes in bone marrow-derived cells, endothelial cells, and VSMCs. Bone marrow transplant of SPRR3-expressing cells into SPRR3(-/-)apolipoprotein E-deficient recipients failed to rescue atheroma burden. Similarly, endothelial cells did not exhibit a response to SPRR3 loss. However, atheromas from SPRR3-deficient mice exhibited increased TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling)-positive VSMCs compared with control. Cell death in SPRR3-deficient VSMCs was significantly increased in vitro. Conversely, SPRR3-overexpressing VSMCs exhibited reduced apoptosis compared with control. We also observed a PI3K (phosphatidylinositol 3-kinase)/Akt-dependent positive association between SPRR3 expression and levels of active Akt in VSMCs. The survival advantage seen in SPRR3-overexpressing VSMCs was abrogated after the addition of a PI3K/Akt pathway inhibitor. CONCLUSIONS: These results indicate that SPRR3 protects the lesion from VSMC loss by promoting survival signaling in plaque VSMCs, thereby significantly decreasing atherosclerosis progression. As the first identified atheroma-specific VSMC prosurvival factor, SPRR3 represents a potential target for lesion-specific modulation of VSMC survival.


Asunto(s)
Proteínas Ricas en Prolina del Estrato Córneo/metabolismo , Miocitos del Músculo Liso/metabolismo , Placa Aterosclerótica/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Adaptación Fisiológica , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apoptosis , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proliferación Celular , Supervivencia Celular , Proteínas Ricas en Prolina del Estrato Córneo/deficiencia , Proteínas Ricas en Prolina del Estrato Córneo/genética , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/patología , Fosforilación , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología , Transducción de Señal
18.
J Mol Signal ; 8(1): 12, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24180579

RESUMEN

BACKGROUND: Programmed necrosis is a form of caspase-independent cell death whose molecular regulation is poorly understood. While tumor necrosis factor-alpha (TNF-α) has been identified as an activator of programmed necrosis, the specific context under which this can happen is unclear. Recently we reported that TNF-α can be expressed by human tumor cells as both a membrane tethered (mTNF-α) and a soluble (sTNF-α) form. Whereas low level, tumor-derived sTNF-α acts as a tumor promoter, tumor cell expression of mTNF-α significantly delays tumor growth in mice, in large part by induction of programmed necrosis of tumor associated myeloid cells. In this study we sought to determine the molecular mechanism involved in mTNF-α oxidative stress-induced cell death by evaluating the known pathways involved in TNF receptor-induced programmed necrosis. METHODS: The source of Reactive Oxygen Species (ROS) in mTNF-α treated cells was determined by coculturing RAW 264.7 monocytic and L929 fibroblasts cells with fixed B16F10 control or mTNF-α expressing-melanoma cells in the presence of inhibitors of NADPH and mitochondria ROS. To identify the down-stream effector of TNF-a receptors (TNFR), level of phospho-RIP-1 and ceramide activity were evaluated.To determine whether mTNF-mediated cell death was dependent on a specific TNFR, cell death was measured in primary CD11b myeloid cells isolated from wild-type or TNFR-1, TNFR-2, TNFR-1 and TNFR-2 double knockout mice, cocultured with various TNF-α isoform. RESULTS: Tumor derived-mTNF-α increased ROS-mediated cytotoxicity, independent of caspase-3 activity. Although TNFR on target cells were required for this effect, we observed that mTNF-induced cell death could be mediated through both TNFR-1 and the death domain-lacking TNFR-2. ROS generation and cytotoxicity were inhibited by a mitochondrial respiratory chain inhibitor but not by an inhibitor of NADPH oxidase. mTNF-α mediated cytotoxicity was independent of RIP-1, a serine/threonine kinase that serves as a main adaptor protein of sTNF-α induced programmed necrosis. Instead, mTNF-α-induced ROS and cell death was prohibited by the ceramide-activated protein kinase (CAPK) inhibitor. CONCLUSION: These findings demonstrate that the mTNF-α isoform is an effective inducer of programmed necrosis through a caspase independent, ceramide-related pathway. Interestingly, unlike sTNFα, mTNF-induced programmed necrosis is not dependent on the presence of TNFR1.

19.
Cancer Res ; 73(13): 3938-50, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23704210

RESUMEN

TNF-α, produced by most malignant cells, orchestrates the interplay between malignant cells and myeloid cells, which have been linked to tumor growth and metastasis. Although TNF-α can exist as one of two isoforms, a 26-kDa membrane tethered form (mTNF-α) or a soluble 17-kDa cytokine (sTNF-α), the vast majority of published studies have only investigated the biologic effects of the soluble form. We show for the first time that membrane and soluble isoforms have diametrically opposing effects on both tumor growth and myeloid content. Mouse lung and melanoma tumor lines expressing mTNF-α generated small tumors devoid of monocytes versus respective control lines or lines expressing sTNF-α. The lack of myeloid cells was due to a direct effect of mTNF-α on myeloid survival via induction of cell necrosis by increasing reactive oxygen species. Human non-small cell lung carcinoma expressed varying levels of both soluble and membrane TNF-α, and gene expression patterns favoring mTNF-α were predictive of improved lung cancer survival. These data suggest that there are significant differences in the role of different TNF-α isoforms in tumor progression and the bioavailability of each isoform may distinctly regulate tumor progression. This insight is critical for effective intervention in cancer therapy with the available TNF-α inhibitors, which can block both TNF-α isoforms.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Supervivencia Celular , Neoplasias Pulmonares/metabolismo , Células Mieloides/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Antígeno CD11b/metabolismo , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/patología , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Muerte Celular , Línea Celular Tumoral , Movimiento Celular , Expresión Génica , Humanos , Neoplasias Pulmonares/mortalidad , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Isoformas de Proteínas/fisiología , Especies Reactivas de Oxígeno/metabolismo , Solubilidad , Análisis de Matrices Tisulares
20.
Stem Cells Transl Med ; 2(2): 151-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23362238

RESUMEN

Multipotent mesenchymal stromal cells (MSCs) have the potential to repair and regenerate damaged tissues, making them attractive candidates for cell-based therapies. To maximize efficacy of MSCs, prediction of their therapeutic abilities must be made so that only the best cells will be used. Our goal was to identify feasible and reproducible in vitro assays to predict MSC potency. We generated cell lines from 10 normal human bone marrow samples and used the International Society for Cellular Therapy's minimal criteria to define them as MSCs: plastic adherence, appropriate surface marker expression, and trilineage differentiation. Each MSC line was further characterized by its growth, proliferation, and viability as determined by cell count, bromodeoxyuridine incorporation, and cellular ATP levels, respectively. To determine whether these tests reliably predict the therapeutic aptitude of the MSCs, several lines were implanted in vivo to examine their capacity to engraft and form granulation tissue in a well-established murine wound model using polyvinyl alcohol sponges. Long-term engraftment of MSCs in the sponges was quantified through the presence of the human-specific Alu gene in sponge sections. Sections were also stained for proliferating cells, vascularity, and granulation tissue formation to determine successful engraftment and repair. We found that high performance in a combination of the in vitro tests accurately predicted which lines functioned well in vivo. These findings suggest that reliable and reproducible in vitro assays may be used to measure the functional potential of MSCs for therapeutic use.


Asunto(s)
Citometría de Flujo/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo/normas , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas/normas , Células Madre Mesenquimatosas/metabolismo , Ratones , Persona de Mediana Edad , Células Madre Multipotentes/metabolismo , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Piel/lesiones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA