Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 207
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 171(7): 1495-1507.e15, 2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29224783

RESUMEN

Current genome-editing systems generally rely on inducing DNA double-strand breaks (DSBs). This may limit their utility in clinical therapies, as unwanted mutations caused by DSBs can have deleterious effects. CRISPR/Cas9 system has recently been repurposed to enable target gene activation, allowing regulation of endogenous gene expression without creating DSBs. However, in vivo implementation of this gain-of-function system has proven difficult. Here, we report a robust system for in vivo activation of endogenous target genes through trans-epigenetic remodeling. The system relies on recruitment of Cas9 and transcriptional activation complexes to target loci by modified single guide RNAs. As proof-of-concept, we used this technology to treat mouse models of diabetes, muscular dystrophy, and acute kidney disease. Results demonstrate that CRISPR/Cas9-mediated target gene activation can be achieved in vivo, leading to measurable phenotypes and amelioration of disease symptoms. This establishes new avenues for developing targeted epigenetic therapies against human diseases. VIDEO ABSTRACT.


Asunto(s)
Sistemas CRISPR-Cas , Epigénesis Genética , Marcación de Gen/métodos , Terapia Genética/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Utrofina/genética , Animales , Secuencia de Bases , Modelos Animales de Enfermedad , Distrofina/genética , Interleucina-10/genética , Proteínas Klotho , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Activación Transcripcional
2.
Cell ; 168(3): 473-486.e15, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28129541

RESUMEN

Interspecies blastocyst complementation enables organ-specific enrichment of xenogenic pluripotent stem cell (PSC) derivatives. Here, we establish a versatile blastocyst complementation platform based on CRISPR-Cas9-mediated zygote genome editing and show enrichment of rat PSC-derivatives in several tissues of gene-edited organogenesis-disabled mice. Besides gaining insights into species evolution, embryogenesis, and human disease, interspecies blastocyst complementation might allow human organ generation in animals whose organ size, anatomy, and physiology are closer to humans. To date, however, whether human PSCs (hPSCs) can contribute to chimera formation in non-rodent species remains unknown. We systematically evaluate the chimeric competency of several types of hPSCs using a more diversified clade of mammals, the ungulates. We find that naïve hPSCs robustly engraft in both pig and cattle pre-implantation blastocysts but show limited contribution to post-implantation pig embryos. Instead, an intermediate hPSC type exhibits higher degree of chimerism and is able to generate differentiated progenies in post-implantation pig embryos.


Asunto(s)
Quimerismo , Edición Génica , Mamíferos/embriología , Animales , Blastocisto , Sistemas CRISPR-Cas , Bovinos , Embrión de Mamíferos/citología , Femenino , Humanos , Masculino , Mamíferos/clasificación , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Células Madre Pluripotentes , Ratas , Ratas Sprague-Dawley , Sus scrofa
3.
Cell ; 167(7): 1719-1733.e12, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27984723

RESUMEN

Aging is the major risk factor for many human diseases. In vitro studies have demonstrated that cellular reprogramming to pluripotency reverses cellular age, but alteration of the aging process through reprogramming has not been directly demonstrated in vivo. Here, we report that partial reprogramming by short-term cyclic expression of Oct4, Sox2, Klf4, and c-Myc (OSKM) ameliorates cellular and physiological hallmarks of aging and prolongs lifespan in a mouse model of premature aging. Similarly, expression of OSKM in vivo improves recovery from metabolic disease and muscle injury in older wild-type mice. The amelioration of age-associated phenotypes by epigenetic remodeling during cellular reprogramming highlights the role of epigenetic dysregulation as a driver of mammalian aging. Establishing in vivo platforms to modulate age-associated epigenetic marks may provide further insights into the biology of aging.


Asunto(s)
Envejecimiento/genética , Reprogramación Celular , Epigénesis Genética , Enfermedades Metabólicas/genética , Factores de Transcripción/metabolismo , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Animales , Diabetes Mellitus Tipo 2/inducido químicamente , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Lamina Tipo A/genética , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/prevención & control , Ratones , Modelos Animales , Páncreas/metabolismo , Sarcopenia/metabolismo
4.
Cell ; 161(3): 459-469, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25910206

RESUMEN

Mitochondrial diseases include a group of maternally inherited genetic disorders caused by mutations in mtDNA. In most of these patients, mutated mtDNA coexists with wild-type mtDNA, a situation known as mtDNA heteroplasmy. Here, we report on a strategy toward preventing germline transmission of mitochondrial diseases by inducing mtDNA heteroplasmy shift through the selective elimination of mutated mtDNA. As a proof of concept, we took advantage of NZB/BALB heteroplasmic mice, which contain two mtDNA haplotypes, BALB and NZB, and selectively prevented their germline transmission using either mitochondria-targeted restriction endonucleases or TALENs. In addition, we successfully reduced human mutated mtDNA levels responsible for Leber's hereditary optic neuropathy (LHOND), and neurogenic muscle weakness, ataxia, and retinitis pigmentosa (NARP), in mammalian oocytes using mitochondria-targeted TALEN (mito-TALENs). Our approaches represent a potential therapeutic avenue for preventing the transgenerational transmission of human mitochondrial diseases caused by mutations in mtDNA. PAPERCLIP.


Asunto(s)
Marcación de Gen , Enfermedades Mitocondriales/genética , Animales , Fusión Celular , ADN Mitocondrial , Embrión de Mamíferos/metabolismo , Endonucleasas/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NZB , Enfermedades Mitocondriales/prevención & control , Mutación , Oocitos/metabolismo
5.
Nephrol Dial Transplant ; 39(1): 114-121, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-37715343

RESUMEN

BACKGROUND: Ischemia-reperfusion injury (IRI) upon transplantation is one of the most impactful events that the kidney graft suffers during its life. Its clinical manifestation in the recipient, delayed graft function (DGF), has serious prognostic consequences. However, the different definitions of DGF are subject to physicians' choices and centers' policies, and a more objective tool to quantify IRI is needed. Here, we propose the use of donor-derived cell-free DNA (ddcfDNA) for this scope. METHODS: ddcfDNA was assessed in 61 kidney transplant recipients of either living or deceased donors at 24 h, and 7, 14 and 30 days after transplantation using the AlloSeq cfDNA Kit (CareDx, San Francisco, CA, USA). Patients were followed-up for 6 months and 7-year graft survival was estimated through the complete and functional iBox tool. RESULTS: Twenty-four-hour ddcfDNA was associated with functional DGF [7.20% (2.35%-15.50%) in patients with functional DGF versus 2.70% (1.55%-4.05%) in patients without it, P = .023] and 6-month estimated glomerular filtration rate (r = -0.311, P = .023). At Day 7 after transplantation, ddcfDNA was associated with dialysis duration in DGF patients (r = 0.612, P = .005) and worse 7-year iBox-estimated graft survival probability (ß -0.42, P = .001) at multivariable analysis. Patients with early normalization of ddcfDNA (<0.5% at 1 week) had improved functional iBox-estimated probability of graft survival (79.5 ± 16.8%) in comparison with patients with 7-day ddcfDNA ≥0.5% (67.7 ± 24.1%) (P = .047). CONCLUSIONS: ddcfDNA early kinetics after transplantation reflect recovery from IRI and are associated with short-, medium- and long-term graft outcome. This may provide a more objective estimate of IRI severity in comparison with the clinical-based definitions of DGF.


Asunto(s)
Ácidos Nucleicos Libres de Células , Trasplante de Riñón , Humanos , Funcionamiento Retardado del Injerto , Diálisis Renal , Trasplante de Riñón/efectos adversos , Donantes de Tejidos , Supervivencia de Injerto , Rechazo de Injerto/diagnóstico , Factores de Riesgo
6.
Transpl Int ; 36: 10840, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36713113

RESUMEN

Extracorporeal photopheresis (ECP) is an immunomodulatory therapy based on the infusion of autologous cellular products exposed to ultraviolet light (UV) in the presence of a photosensitizer. The study evaluates the ECP efficacy as induction therapy in a full-mismatch kidney transplant rat model. Dark Agouti to Lewis (DA-L) kidney transplant model has been established. ECP product was obtained from Lewis rat recipients after DA kidney graft transplantation (LewDA). Leukocytes of those LewDA rats were exposed to 8-methoxy psoralen, and illuminated with UV-A. The ECP doses assessed were 10 × 106 and 100 × 106 cells/time point. Lewis recipients received seven ECP infusions. DA-L model was characterized by the appearance of donor-specific antibodies (DSA) and kidney function deterioration from day three after kidney transplant. The dysfunction progressed rapidly until graft loss (6.1 ± 0.5 days). Tacrolimus at 0.25 mg/kg prolonged rat survival until 11.4 ± 0.7 days (p = 0.0004). In this context, the application of leukocytes from LewDA sensitized rats accelerated the rejection (8.7 ± 0.45, p = 0.0012), whereas ECP product at high dose extended kidney graft survival until 26.3 ± 7.3 days, reducing class I and II DSA in surviving rats. ECP treatment increases kidney graft survival in full-mismatch rat model of acute rejection and is a suitable immunomodulatory therapy to be explored in kidney transplantation.


Asunto(s)
Trasplante de Riñón , Fotoféresis , Ratas , Animales , Supervivencia de Injerto , Ratas Endogámicas Lew , Rechazo de Injerto/prevención & control , Anticuerpos
7.
Kidney Int ; 102(2): 421-430, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35644282

RESUMEN

Several organ allocation protocols give priority to wait-listed simultaneous kidney-pancreas (SPK) transplant recipients to mitigate the higher cardiovascular risk of patients with diabetes mellitus on dialysis. The available information regarding the impact of preemptive simultaneous kidney-pancreas transplantation on recipient and graft outcomes is nonetheless controversial. To help resolve this, we explored the influence of preemptive simultaneous kidney-pancreas transplants on patient and graft survival through a retrospective analysis of the OPTN/UNOS database, encompassing 9690 simultaneous transplant recipients between 2000 and 2017. Statistical analysis was performed applying a propensity score analysis to minimize bias. Of these patients, 1796 (19%) were transplanted preemptively. At ten years, recipient survival was significantly superior in the preemptive group when compared to the non-preemptive group (78.9% vs 71.8%). Dialysis at simultaneous kidney-pancreas transplantation was an independent significant risk for patient survival (hazard ratio 1.66 [95% confidence interval 1.32-2.09]), especially if the dialysis duration was 12 months or longer. Preemptive transplantation was also associated with significant superior kidney graft survival compared to those on dialysis (death-censored: 84.3% vs 75.4%, respectively; estimated half-life of 38.57 [38.33 -38.81] vs 22.35 [22.17 - 22.53] years, respectively). No differences were observed between both groups neither for pancreas graft survival nor for post-transplant surgical complications. Thus, our results sustain the relevance of early referral for pancreas transplantation and the importance of pancreas allocation priority in reducing patient mortality after simultaneous kidney-pancreas transplantation.


Asunto(s)
Diabetes Mellitus Tipo 1 , Trasplante de Riñón , Trasplante de Páncreas , Diabetes Mellitus Tipo 1/cirugía , Supervivencia de Injerto , Humanos , Trasplante de Riñón/métodos , Páncreas , Trasplante de Páncreas/efectos adversos , Estudios Retrospectivos
8.
Transpl Int ; 35: 10639, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36466442

RESUMEN

Profiling of circulating immune cells provides valuable insight to the pathophysiology of acute rejection in organ transplantation. Herein we characterized the peripheral blood mononuclear cells in simultaneous kidney-pancreas transplant recipients. We conducted a retrospective analysis in a biopsy-matched cohort (n = 67) and compared patients with biopsy proven acute rejection (BPAR; 41%) to those without rejection (No-AR). We observed that CD3+ T cells, both CD8+ and CD4+, as well as CD19+ B cells were increased in patients with BPAR, particularly in biopsies performed in the early post-transplant period (<3 months). During this period immune subsets presented a good discriminative ability (CD4+ AUC 0.79; CD8+ AUC 0.80; B cells AUC 0.86; p < 0.05) and outperformed lipase (AUC 0.62; p = 0.12) for the diagnosis of acute rejection. We further evaluated whether this could be explained by differences in frequencies prior to transplantation. Patients presenting with early post-transplant rejection (<3 months) had a significant increase in T-cell frequencies pre-transplant, both CD4+ T cells and CD8+ T cells (p < 0.01), which were associated with a significant inferior rejection-free graft survival. T cell frequencies in peripheral blood correlated with pancreas acute rejection episodes, and variations prior to transplantation were associated with pancreas early acute rejection.


Asunto(s)
Leucocitos Mononucleares , Trasplante de Páncreas , Humanos , Estudios Retrospectivos , Páncreas , Riñón
9.
Nature ; 540(7631): 144-149, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27851729

RESUMEN

Targeted genome editing via engineered nucleases is an exciting area of biomedical research and holds potential for clinical applications. Despite rapid advances in the field, in vivo targeted transgene integration is still infeasible because current tools are inefficient, especially for non-dividing cells, which compose most adult tissues. This poses a barrier for uncovering fundamental biological principles and developing treatments for a broad range of genetic disorders. Based on clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9) technology, here we devise a homology-independent targeted integration (HITI) strategy, which allows for robust DNA knock-in in both dividing and non-dividing cells in vitro and, more importantly, in vivo (for example, in neurons of postnatal mammals). As a proof of concept of its therapeutic potential, we demonstrate the efficacy of HITI in improving visual function using a rat model of the retinal degeneration condition retinitis pigmentosa. The HITI method presented here establishes new avenues for basic research and targeted gene therapies.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Marcación de Gen/métodos , Genoma/genética , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Animales , División Celular , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Terapia Genética/métodos , Neuronas/citología , Neuronas/metabolismo , Ratas , Homología de Secuencia
10.
Br J Haematol ; 193(1): 181-187, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32469083

RESUMEN

Carfilzomib has been associated with the development of thrombotic microangiopathy (TMA) in relapsed/refractory multiple myeloma patients, a severe disease with no currently available aetiological treatment. We evaluated the potential role of terminal complement pathway in four patients with carfilzomib-induced TMA. Membrane attack complex (C5b-9) deposition on endothelial cells in culture exposed to plasma from patients during the acute phase of the disease suggests complement overactivation as a mechanism of potential endothelial damage in three out of four patients. If confirmed in larger cohorts, C5b-9 evaluation will allow early identification of patients who could benefit from complement blockade and treatment monitoring.


Asunto(s)
Proteínas del Sistema Complemento/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Oligopéptidos/efectos adversos , Microangiopatías Trombóticas/inducido químicamente , Ubiquitina/antagonistas & inhibidores , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Complejo de Ataque a Membrana del Sistema Complemento/efectos adversos , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/metabolismo , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/complicaciones , Oligopéptidos/uso terapéutico , Estudios Prospectivos , Inhibidores de Proteasoma/efectos adversos , Inhibidores de Proteasoma/uso terapéutico , Microangiopatías Trombóticas/tratamiento farmacológico , Microangiopatías Trombóticas/etiología , Microangiopatías Trombóticas/metabolismo , Ubiquitina/metabolismo
11.
Ann Surg ; 273(6): e230-e238, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30829695

RESUMEN

OBJECTIVE: The aim of the study was to evaluate the effect of cardiac arrest time (CAT) in donors after brain death (DBD) donors on pancreas transplant outcome. SUMMARY OF BACKGROUND DATA: Results from donors after circulatory death report good outcomes despite warm ischemia times up to 57 minutes. Previous cardiac arrest in DBD has been addressed as a potential risk factor, but duration of the CAT has never been evaluated. METHODS: We conducted a retrospective analysis including 342 pancreas transplants performed at our center from 2000 to 2016, and evaluated the effect of previous cardiac arrest in DBD (caDBD) on pancreas transplant outcomes. RESULTS: A total of 49 (14.3%) caDBD were accepted for transplantation [median CAT of 5.0 min (IQR 2.5-15.0)]. Anoxic encephalopathy was most frequent and P-PASS higher (16.9 vs 15.6) in caDBD group when compared with other DBD. No differences were found in all other characteristics evaluated.Graft survival was similar between both groups, as was the incidence of early graft failure (EGF). CAT increased the risk for EGF [OR 1.09 (95% CI, 1.01-1.17)], and the duration of CPR discriminated for EGF [AUC of 0.86 (95% CI, 0.74-0.98)], with a sensitivity and specificity of 100% and 75% at a cutoff of 15 minutes. When evaluated separately, caDBD >15 min increased over 5 times the risk for EGF [HR 5.80 (95% CI, 1.82-18.56); P = 0.003], and these presented fewer days on the ICU (1.0 vs 3.0 d). CONCLUSION: CaDBD donors are suitable for routine pancreas transplantation without increasing EGF risk, and in those with longer CAT it may be prudent to postpone donation a few days to allow a thorough evaluation of organ damage following cardiac arrest.


Asunto(s)
Paro Cardíaco , Trasplante de Páncreas , Donantes de Tejidos , Obtención de Tejidos y Órganos , Adulto , Muerte Encefálica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
12.
N Engl J Med ; 379(1): 11-21, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29972753

RESUMEN

BACKGROUND: Patisiran, an investigational RNA interference therapeutic agent, specifically inhibits hepatic synthesis of transthyretin. METHODS: In this phase 3 trial, we randomly assigned patients with hereditary transthyretin amyloidosis with polyneuropathy, in a 2:1 ratio, to receive intravenous patisiran (0.3 mg per kilogram of body weight) or placebo once every 3 weeks. The primary end point was the change from baseline in the modified Neuropathy Impairment Score+7 (mNIS+7; range, 0 to 304, with higher scores indicating more impairment) at 18 months. Other assessments included the Norfolk Quality of Life-Diabetic Neuropathy (Norfolk QOL-DN) questionnaire (range, -4 to 136, with higher scores indicating worse quality of life), 10-m walk test (with gait speed measured in meters per second), and modified body-mass index (modified BMI, defined as [weight in kilograms divided by square of height in meters]×albumin level in grams per liter; lower values indicated worse nutritional status). RESULTS: A total of 225 patients underwent randomization (148 to the patisiran group and 77 to the placebo group). The mean (±SD) mNIS+7 at baseline was 80.9±41.5 in the patisiran group and 74.6±37.0 in the placebo group; the least-squares mean (±SE) change from baseline was -6.0±1.7 versus 28.0±2.6 (difference, -34.0 points; P<0.001) at 18 months. The mean (±SD) baseline Norfolk QOL-DN score was 59.6±28.2 in the patisiran group and 55.5±24.3 in the placebo group; the least-squares mean (±SE) change from baseline was -6.7±1.8 versus 14.4±2.7 (difference, -21.1 points; P<0.001) at 18 months. Patisiran also showed an effect on gait speed and modified BMI. At 18 months, the least-squares mean change from baseline in gait speed was 0.08±0.02 m per second with patisiran versus -0.24±0.04 m per second with placebo (difference, 0.31 m per second; P<0.001), and the least-squares mean change from baseline in the modified BMI was -3.7±9.6 versus -119.4±14.5 (difference, 115.7; P<0.001). Approximately 20% of the patients who received patisiran and 10% of those who received placebo had mild or moderate infusion-related reactions; the overall incidence and types of adverse events were similar in the two groups. CONCLUSIONS: In this trial, patisiran improved multiple clinical manifestations of hereditary transthyretin amyloidosis. (Funded by Alnylam Pharmaceuticals; APOLLO ClinicalTrials.gov number, NCT01960348 .).


Asunto(s)
Neuropatías Amiloides Familiares/terapia , ARN Interferente Pequeño/uso terapéutico , Tratamiento con ARN de Interferencia , Administración Intravenosa , Adulto , Anciano , Anciano de 80 o más Años , Neuropatías Amiloides Familiares/sangre , Neuropatías Amiloides Familiares/complicaciones , Progresión de la Enfermedad , Método Doble Ciego , Edema/inducido químicamente , Femenino , Trastornos Neurológicos de la Marcha/etiología , Humanos , Infusiones Intravenosas/efectos adversos , Análisis de los Mínimos Cuadrados , Masculino , Persona de Mediana Edad , Polineuropatías/etiología , Polineuropatías/terapia , Prealbúmina/análisis , Prealbúmina/genética , Calidad de Vida , ARN Interferente Pequeño/efectos adversos , Índice de Severidad de la Enfermedad , Prueba de Paso
13.
N Engl J Med ; 379(1): 22-31, 2018 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-29972757

RESUMEN

BACKGROUND: Hereditary transthyretin amyloidosis is caused by pathogenic single-nucleotide variants in the gene encoding transthyretin ( TTR) that induce transthyretin misfolding and systemic deposition of amyloid. Progressive amyloid accumulation leads to multiorgan dysfunction and death. Inotersen, a 2'- O-methoxyethyl-modified antisense oligonucleotide, inhibits hepatic production of transthyretin. METHODS: We conducted an international, randomized, double-blind, placebo-controlled, 15-month, phase 3 trial of inotersen in adults with stage 1 (patient is ambulatory) or stage 2 (patient is ambulatory with assistance) hereditary transthyretin amyloidosis with polyneuropathy. Patients were randomly assigned, in a 2:1 ratio, to receive weekly subcutaneous injections of inotersen (300 mg) or placebo. The primary end points were the change in the modified Neuropathy Impairment Score+7 (mNIS+7; range, -22.3 to 346.3, with higher scores indicating poorer function; minimal clinically meaningful change, 2 points) and the change in the score on the patient-reported Norfolk Quality of Life-Diabetic Neuropathy (QOL-DN) questionnaire (range, -4 to 136, with higher scores indicating poorer quality of life). A decrease in scores indicated improvement. RESULTS: A total of 172 patients (112 in the inotersen group and 60 in the placebo group) received at least one dose of a trial regimen, and 139 (81%) completed the intervention period. Both primary efficacy assessments favored inotersen: the difference in the least-squares mean change from baseline to week 66 between the two groups (inotersen minus placebo) was -19.7 points (95% confidence interval [CI], -26.4 to -13.0; P<0.001) for the mNIS+7 and -11.7 points (95% CI, -18.3 to -5.1; P<0.001) for the Norfolk QOL-DN score. These improvements were independent of disease stage, mutation type, or the presence of cardiomyopathy. There were five deaths in the inotersen group and none in the placebo group. The most frequent serious adverse events in the inotersen group were glomerulonephritis (in 3 patients [3%]) and thrombocytopenia (in 3 patients [3%]), with one death associated with one of the cases of grade 4 thrombocytopenia. Thereafter, all patients received enhanced monitoring. CONCLUSIONS: Inotersen improved the course of neurologic disease and quality of life in patients with hereditary transthyretin amyloidosis. Thrombocytopenia and glomerulonephritis were managed with enhanced monitoring. (Funded by Ionis Pharmaceuticals; NEURO-TTR ClinicalTrials.gov number, NCT01737398 .).


Asunto(s)
Neuropatías Amiloides Familiares/terapia , Oligonucleótidos Antisentido/uso terapéutico , Prealbúmina/antagonistas & inhibidores , Tratamiento con ARN de Interferencia , Adulto , Anciano , Anciano de 80 o más Años , Neuropatías Amiloides Familiares/sangre , Neuropatías Amiloides Familiares/complicaciones , Progresión de la Enfermedad , Método Doble Ciego , Femenino , Glomerulonefritis/inducido químicamente , Humanos , Inyecciones Subcutáneas , Análisis de los Mínimos Cuadrados , Masculino , Persona de Mediana Edad , Oligonucleótidos Antisentido/efectos adversos , Polineuropatías/etiología , Polineuropatías/terapia , Prealbúmina/análisis , Prealbúmina/genética , Calidad de Vida , Índice de Severidad de la Enfermedad , Trombocitopenia/inducido químicamente
14.
Transpl Int ; 34(1): 175-184, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33131120

RESUMEN

The dialysis-based definition of Delayed Graft Function (dDGF) is not necessarily objective as it depends on the individual physician's decision. The functional definition of DGF (fDGF, the failure of serum creatinine to decrease by at least 10% daily on 3 consecutive days during the first week post-transplant), may be more sensitive to reflect recovery after the ischemia-reperfusion injury. We retrospectively analyzed both definitions in 253 deceased donor kidney transplant recipients for predicting death-censored graft failure as primary outcome, using eGFR < 25 ml/min/1.73 m2 as a surrogate end-point for graft failure. Secondary outcome was a composite outcome that included graft failure as above and also patient's death. Median follow-up was 3.22 [2.38-4.21] years. Seventy-nine patients developed dDGF (31.2%) and 127 developed fDGF (50.2%). Sixty-three patients fulfilled criteria for both definitions (24.9%). At multivariable analysis, the two definitions were significantly associated with the primary [HR (95%CI) 2.07 (1.09-3.94), P = 0.026 for fDGF and HR (95%CI) 2.41 (1.33-4.37), P = 0.004 for dDGF] and the secondary composite outcome [HR (95%CI) 1.58 (1.01-2.51), P = 0.047 for fDGF and HR (95%CI) 1.67 (1.05-2.66), P = 0.028 for dDGF]. Patients who met criteria for both definitions had the worst prognosis, with a three-year estimates (95%CI) of survival from the primary and secondary outcomes of 2.31 (2.02-2.59) and 2.20 (1.91-2.49) years for fDGF+/dDGF+, in comparison with the other groups (P < 0.01 for trend). fDGF provides supplementary information about graft outcomes on top of the dDGF definition in a modern series of kidney transplantation.


Asunto(s)
Trasplante de Riñón , Funcionamiento Retardado del Injerto , Rechazo de Injerto , Supervivencia de Injerto , Humanos , Estudios Retrospectivos , Factores de Riesgo , Factores de Tiempo , Donantes de Tejidos
15.
Nature ; 521(7552): 316-21, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25945737

RESUMEN

Pluripotency, the ability to generate any cell type of the body, is an evanescent attribute of embryonic cells. Transitory pluripotent cells can be captured at different time points during embryogenesis and maintained as embryonic stem cells or epiblast stem cells in culture. Since ontogenesis is a dynamic process in both space and time, it seems counterintuitive that these two temporal states represent the full spectrum of organismal pluripotency. Here we show that by modulating culture parameters, a stem-cell type with unique spatial characteristics and distinct molecular and functional features, designated as region-selective pluripotent stem cells (rsPSCs), can be efficiently obtained from mouse embryos and primate pluripotent stem cells, including humans. The ease of culturing and editing the genome of human rsPSCs offers advantages for regenerative medicine applications. The unique ability of human rsPSCs to generate post-implantation interspecies chimaeric embryos may facilitate our understanding of early human development and evolution.


Asunto(s)
Quimera , Células Madre Pluripotentes/citología , Animales , Técnicas de Cultivo de Célula/métodos , Línea Celular , Células Madre Embrionarias/citología , Femenino , Estratos Germinativos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino , Ratones , Pan troglodytes , Células Madre Pluripotentes/metabolismo , Medicina Regenerativa , Especificidad de la Especie
16.
Health Care Manag Sci ; 24(1): 55-71, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32946046

RESUMEN

The main applications of Data Envelopment Analysis (DEA) to medicine focus on evaluating the efficiency of different health structures, hospitals and departments within them. The evolution of patients after undergoing a medical procedure or their response to a given treatment are not generally studied through this programming technique. In addition to the difficulty inherent to the collection of this type of data, the use of a technique that is mainly applied to evaluate the efficiency of decision making units representing industrial and production structures to analyze the evolution of human patients may seem inappropriate. In the current paper, we illustrate how this is not actually the case and implement a decision engineering approach to model kidney transplantation patients as decision making units. As such, patients undergo three different phases, each composed by specific as well as interrelated variables, determining the potential success of the transplantation process. DEA is applied to a set of 12 input and 6 output variables - retrieved over a 10-year period - describing the evolution of 485 patients undergoing kidney transplantation from living donors. The resulting analysis allows us to classify the set of patients in terms of the efficiency of the transplantation process and identify the specific characteristics across which potential improvements could be defined on a per patient basis.


Asunto(s)
Toma de Decisiones Clínicas , Trasplante de Riñón/estadística & datos numéricos , Modelos Estadísticos , Factores de Edad , Diabetes Mellitus , Eficiencia Organizacional , Rechazo de Injerto/epidemiología , Histocompatibilidad , Humanos , Hipertensión , Trasplante de Riñón/efectos adversos , Donadores Vivos , Diálisis Renal/estadística & datos numéricos , España
17.
Am J Transplant ; 20(7): 1875-1878, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32198834

RESUMEN

COVID-19 is novel infectious disease with an evolving understanding of its epidemiology and clinical manifestations. Immunocompromised patients often present atypical presentations of viral diseases. Herein we report a case of a COVID-19 infection in a solid organ transplant recipient, in which the first clinical symptoms were of gastrointestinal viral disease and fever, which further progressed to respiratory symptoms in 48 hours. In these high risk populations, protocols for screening for SARS-Cov2 may be needed to be re-evaluated.


Asunto(s)
Infecciones por Coronavirus/terapia , Glomerulonefritis por IGA/cirugía , Huésped Inmunocomprometido , Terapia de Inmunosupresión , Fallo Renal Crónico/cirugía , Trasplante de Riñón , Neumonía Viral/terapia , Betacoronavirus , COVID-19 , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/diagnóstico , Glomerulonefritis por IGA/complicaciones , Humanos , Tolerancia Inmunológica , Fallo Renal Crónico/complicaciones , Masculino , Persona de Mediana Edad , Pandemias , Neumonía Viral/complicaciones , Neumonía Viral/diagnóstico , Respiración Artificial , Factores de Riesgo , SARS-CoV-2
18.
Nat Mater ; 18(4): 397-405, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30778227

RESUMEN

The generation of organoids is one of the biggest scientific advances in regenerative medicine. Here, by lengthening the time that human pluripotent stem cells (hPSCs) were exposed to a three-dimensional microenvironment, and by applying defined renal inductive signals, we generated kidney organoids that transcriptomically matched second-trimester human fetal kidneys. We validated these results using ex vivo and in vitro assays that model renal development. Furthermore, we developed a transplantation method that utilizes the chick chorioallantoic membrane. This approach created a soft in vivo microenvironment that promoted the growth and differentiation of implanted kidney organoids, as well as providing a vascular component. The stiffness of the in ovo chorioallantoic membrane microenvironment was recapitulated in vitro by fabricating compliant hydrogels. These biomaterials promoted the efficient generation of renal vesicles and nephron structures, demonstrating that a soft environment accelerates the differentiation of hPSC-derived kidney organoids.


Asunto(s)
Espacio Extracelular/metabolismo , Riñón/citología , Organoides/citología , Células Madre Pluripotentes/citología , Técnicas de Cultivo de Tejidos/métodos , Diferenciación Celular , Microambiente Celular , Femenino , Humanos , Cinética , Células Madre Pluripotentes/metabolismo , Embarazo , Tercer Trimestre del Embarazo , Transcriptoma
19.
Am J Nephrol ; 51(5): 357-365, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32187607

RESUMEN

BACKGROUND: Recurrence of immunoglobulin (Ig)A nephropathy (rIgAN) is a growing cause of kidney allograft dysfunction. This study was aimed at investigating factors associated with rIgAN and the subsequent progression to end-stage renal disease (ESRD). METHODS: Retrospective study including consecutive patients with IgA nephropathy (IgAN) who received a kidney transplant in our center between 1992 and 2016 and had a renal biopsy by clinical indication. The date of detection of chronic kidney disease (CKD) 5 was used as renal outcome. RESULTS: Eighty-six kidney transplants were performed in patients with IgAN, 38 (44%) were from living donors (related n = 26). rIgAN was diagnosed in 23 allografts (27%). Renal function and proteinuria at the end of the follow-up period were worst in the rIgAN patients compared to those without rIgAN (2.2 vs. 1.4 mg/dL, p = 0.014, and 1.16 vs. 0.49 g/day, p = 0.005, respectively). Risk of rIgAN and progression to CKD 5 decreased with patient's age (hazard ratio [HR] 0.95, 95% CI 0.92-0.98, p = 0.002, and HR 0.97, 95% CI 0.83-0.97, p = 0.008 per year, respectively). Patients with rIgAN had a higher risk of progression to CKD 5 (HR 6.7, 95% CI 1.3-35.7, p = 0.025). Full donor-recipient mismatch in the human leukocyte antigen (HLA)-B loci decreased the risk of rIgAN (HR 0.22, 95% CI 0.06-0.76, p = 0.017). CONCLUSIONS: rIgAN was an independent risk factor for ESRD after renal allograft. Younger age increased the risk of rIgAN and CKD 5. Conversely, HLA-B mismatching was a potential protective factor for rIgAN of this glomerular disease.


Asunto(s)
Glomerulonefritis por IGA/diagnóstico , Antígenos HLA-B/inmunología , Fallo Renal Crónico/cirugía , Trasplante de Riñón/efectos adversos , Adulto , Factores de Edad , Aloinjertos/inmunología , Aloinjertos/patología , Biopsia , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Glomerulonefritis por IGA/complicaciones , Glomerulonefritis por IGA/inmunología , Glomerulonefritis por IGA/cirugía , Antígenos HLA-B/análisis , Prueba de Histocompatibilidad , Humanos , Riñón/inmunología , Riñón/patología , Fallo Renal Crónico/inmunología , Masculino , Persona de Mediana Edad , Factores Protectores , Recurrencia , Estudios Retrospectivos , Factores de Riesgo , Trasplante Homólogo
20.
J Clin Apher ; 35(3): 188-199, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32219886

RESUMEN

INTRODUCTION: Nonantigen-specific immunoadsorption (IA) has proven to be effective in acute antibody-mediated rejection (aAMR). However, there is a lack of solid studies evaluating the safety and efficacy of IA with antihuman Ig-columns in aAMR. For chronic-active AMR (cAMR), no studies have evaluated the efficacy of nonantingen-specific IA with antihuman Ig-columns. The purpose of this study was to evaluate the role of nonantigen-specific IA with antihuman Ig-columns in the treatment of both aAMR and cAMR in kidney transplantation. MATERIAL AND METHODS: In retrospective and observational study, kidney graft and recipient survival rates were assessed after treatment of aAMR and cAMR with nonantigen-specific IA with Ig-Flex columns (Therasorb) between January 2012 and May 2018. Protocols included nonantigen-specific IA, rituximab, intravenous immunoglobulin, and rescue plasma exchange, if necessary. RESULTS: The study included 14 patients with AMR (acute in 9, chronic active in 5). For aAMR, mean follow-up was 13 ± 6 months, and patient and graft survival were, respectively, of 100% and 83%, with a mean increase in estimated glomerular filtration rate (eGFR) of 7.98 ± 12.96, 10.18 ± 16.71, and 11.43 ± 13.85 mL/min/1.72 m2 (P > .05) at 3, 12 months after treatment, and at the end of follow-up, respectively. For cAMR, mean follow-up was 14 ± 8 months, and patient and graft survival were, respectively, of 100% and 60%, with an average increase in eGFR of 4.30 ± 7.86, 5.64 ± 10.47, and 14.5 ± 7.86 mL/min/m2 (P > .05) at 3, 12 months after IA treatment, and at the end of the follow-up, respectively, although 40% did not respond and required chronic hemodialysis. CONCLUSION: Nonantigen-specific IA with Ig-Flex columns was safe and effective for aAMR treatment in kidney transplantation. In cAMR, IA with Ig-Flex columns was associated with a satisfactory kidney graft survival, suggesting that IA could potentially offer some benefits supporting its indication in cAMR.


Asunto(s)
Rechazo de Injerto , Sistema Inmunológico/inmunología , Inmunoglobulina G/inmunología , Trasplante de Riñón/métodos , Adulto , Anciano , Anticuerpos , Femenino , Estudios de Seguimiento , Tasa de Filtración Glomerular , Supervivencia de Injerto , Humanos , Inmunoglobulinas Intravenosas , Masculino , Persona de Mediana Edad , Plasmaféresis , Regeneración , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA