Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Clin Immunol ; 41(2): 414-426, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33159275

RESUMEN

PURPOSE: Allogeneic bone marrow transplantation (alloBMT) is the only cure for many primary immune deficiency disorders (PIDD), primary immune regulatory disorders (PIRD), and inherited bone marrow failure syndromes (IBMFS). METHODS: We report the results of 25 patients who underwent alloBMT using reduced intensity conditioning (RIC), alternative donors, and post-transplantation cyclophosphamide (PTCy). In an attempt to reduce regimen-related toxicities, we removed low-dose TBI from the prep and added mycophenolate mofetil and tacrolimus for graft-versus-host disease (GVHD) prophylaxis for all donor types in the latter 14 patients. Donors were haploidentical related (n = 14), matched unrelated (n = 9), or mismatched unrelated (n = 2). The median age was 9 years (range 5 months-21 years). RESULTS: With a median follow-up of 26 months (range 7 months-9 years), the 2-year overall survival is 92%. There were two deaths, one from infection, and one from complications after a second myeloablative BMT. Three patients developed secondary graft failure, one at 2 years and two at >3 years, successfully treated with CD34 cell boost in one or second BMT in two. The remaining 20 patients have full or stable mixed donor chimerism and are disease-free. The incidence of mixed chimerism is increased since removing TBI from the prep. The 6-month cumulative incidence of grade II acute GVHD is 17%, with no grade III-IV. The 1-year cumulative incidence of chronic GVHD is 14%, with severe of 5%. CONCLUSION: This alloBMT platform using alternative donors, RIC, and PTCy is associated with excellent rates of engraftment and low rates of GVHD and non-relapse mortality, and offers a curative option for patients with PIDD, PIRD, and IBMFS. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT04232085.


Asunto(s)
Trastornos de Fallo de la Médula Ósea/tratamiento farmacológico , Trasplante de Médula Ósea/efectos adversos , Ciclofosfamida/uso terapéutico , Síndromes de Inmunodeficiencia/tratamiento farmacológico , Adolescente , Adulto , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Lactante , Recién Nacido , Masculino , Ácido Micofenólico/farmacología , Tacrolimus/uso terapéutico , Donantes de Tejidos , Acondicionamiento Pretrasplante/métodos , Adulto Joven
2.
Exp Cell Res ; 390(1): 111935, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32151493

RESUMEN

Tankyrase 1 (TNKS1; PARP-5a) and Tankyrase 2 (TNKS2; PARP-5b) are poly-ADP-ribosyl-polymerase (PARP)-domain-containing proteins that regulate the activities of a wide repertoire of target proteins via post-translational addition of poly-ADP-ribose polymers (PARylation). Although tankyrases were first identified as regulators of human telomere elongation, important and expansive roles of tankyrase activity have recently emerged in the development and maintenance of stem cell states. Herein, we summarize the current state of knowledge of the various tankyrase-mediated activities that may promote human naïve and 'extended' pluripotency'. We review the putative role of tankyrase and PARP inhibition in trophectoderm specification, telomere elongation, DNA repair and chromosomal segregation, metabolism, and PTEN-mediated apoptosis. Importantly, tankyrases possess PARP-independent activities that include regulation of MDC1-associated DNA repair by homologous recombination (HR) and autophagy/pexophagy, which is an essential mechanism of protein synthesis in the preimplantation embryo. Additionally, tankyrases auto-regulate themselves via auto-PARylation which augments their cellular protein levels and potentiates their non-PARP tankyrase functions. We propose that these non-PARP-related activities of tankyrase proteins may further independently affect both naïve and extended pluripotency via mechanisms that remain undetermined. We broadly outline a hypothetical framework for how inclusion of a tankyrase/PARP inhibitor in small molecule cocktails may stabilize and potentiate naïve and extended pluripotency via pleiotropic routes and mechanisms.


Asunto(s)
Células Madre Pluripotentes/metabolismo , Tanquirasas/metabolismo , Apoptosis , Diferenciación Celular , Reparación del ADN , Humanos , Células Madre Pluripotentes/citología , Tanquirasas/genética , Homeostasis del Telómero
4.
Development ; 143(23): 4368-4380, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27660325

RESUMEN

The derivation and maintenance of human pluripotent stem cells (hPSCs) in stable naïve pluripotent states has a wide impact in human developmental biology. However, hPSCs are unstable in classical naïve mouse embryonic stem cell (ESC) WNT and MEK/ERK signal inhibition (2i) culture. We show that a broad repertoire of conventional hESC and transgene-independent human induced pluripotent stem cell (hiPSC) lines could be reverted to stable human preimplantation inner cell mass (ICM)-like naïve states with only WNT, MEK/ERK, and tankyrase inhibition (LIF-3i). LIF-3i-reverted hPSCs retained normal karyotypes and genomic imprints, and attained defining mouse ESC-like functional features, including high clonal self-renewal, independence from MEK/ERK signaling, dependence on JAK/STAT3 and BMP4 signaling, and naïve-specific transcriptional and epigenetic configurations. Tankyrase inhibition promoted a stable acquisition of a human preimplantation ICM-like ground state via modulation of WNT signaling, and was most efficacious in efficiently reprogrammed conventional hiPSCs. Importantly, naïve reversion of a broad repertoire of conventional hiPSCs reduced lineage-primed gene expression and significantly improved their multilineage differentiation capacities. Stable naïve hPSCs with reduced genetic variability and improved functional pluripotency will have great utility in regenerative medicine and human disease modeling.


Asunto(s)
Diferenciación Celular/fisiología , Autorrenovación de las Células/fisiología , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Tanquirasas/antagonistas & inhibidores , Vía de Señalización Wnt/fisiología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Células Cultivadas , Reprogramación Celular/fisiología , Estratos Germinativos/embriología , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Humanos , Quinasas Janus/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Ratones , Factor de Transcripción STAT3/metabolismo
5.
Biol Blood Marrow Transplant ; 23(12): 2127-2136, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28807769

RESUMEN

High-risk, recurrent, or refractory solid tumors in pediatric, adolescent, and young adult (AYA) patients have an extremely poor prognosis despite current intensive treatment regimens. We piloted an allogeneic bone marrow transplant platform using reduced-intensity conditioning (RIC) and partially HLA-mismatched (haploidentical) related donors for this population of pediatric and AYA solid tumor patients. Sixteen patients received fludarabine, cyclophosphamide, melphalan, and low-dose total body irradiation RIC haploidentical BMT (haploBMT) followed by post-transplantation cyclophosphamide (PTCy), mycophenolate mofetil, and sirolimus. All assessable patients were full donor chimeras on day 30 with a median neutrophil recovery of 19 days and platelet recovery of 21 days. One patient (7%) exhibited secondary graft failure associated with concomitant infection. The median follow-up time was 15 months. Overall survival was 88%, 56%, and 21% at 6, 12, and 24 months, respectively. Median survival from transplant date was 14 months with a median progression-free survival 7 months. We observed limited graft-versus-host disease in 3 patients and nonrelapse mortality in 1 patient. We demonstrated that RIC haploBMT with PTCy is feasible and has acceptable toxicities in patients with incurable pediatric and AYA solid tumors; thus, this approach serves as a platform for post-transplant strategies to prevent relapse and optimize progression-free survival.


Asunto(s)
Trasplante de Médula Ósea/métodos , Ciclofosfamida/uso terapéutico , Neoplasias/terapia , Adolescente , Adulto , Trasplante de Médula Ósea/mortalidad , Niño , Preescolar , Supervivencia de Injerto , Humanos , Neoplasias/mortalidad , Trasplante Haploidéntico/mortalidad , Resultado del Tratamiento , Adulto Joven
6.
Biol Blood Marrow Transplant ; 23(2): 325-332, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27888014

RESUMEN

Lower-intensity conditioning regimens for haploidentical blood or marrow transplantation (BMT) are safe and efficacious for adult patients with hematologic malignancies. We report data for pediatric/young adult patients with high-risk hematologic malignancies (n = 40) treated with nonmyeloablative haploidentical BMT with post-transplantation cyclophosphamide from 2003 to 2015. Patients received a preparative regimen of fludarabine, cyclophosphamide, and total body irradiation. Post-transplantation immunosuppression consisted of cyclophosphamide, mycophenolate mofetil, and tacrolimus. Donor engraftment occurred in 29 of 32 (91%), with median time to engraftment of neutrophils >500/µL of 16 days (range, 13 to 22) and for platelets >20,000/µL without transfusion of 18 days (range, 12 to 62). Cumulative incidences of acute graft-versus-host disease (GVHD) grades II to IV and grades III and IV at day 100 were 33% and 5%, respectively. The cumulative incidence of chronic GVHD was 23%, with 7% moderate-to-severe chronic GVHD, according to National Institutes of Health consensus criteria. Transplantation-related mortality (TRM) at 1 year was 13%. The cumulative incidence of relapse at 2 years was 52%. With a median follow-up of 20 months (range, 3 to 148), 1-year actuarial overall and event-free survival were 56% and 43%, respectively. Thus, we demonstrate excellent rates of engraftment, GVHD, and TRM in pediatric/young adult patients treated with this regimen. This approach is a widely available, safe, and feasible option for pediatric and young adult patients with high-risk hematologic malignancies, including those with a prior history of myeloablative BMT and/or those with comorbidities or organ dysfunction that preclude eligibility for myeloablative BMT.


Asunto(s)
Trasplante de Médula Ósea , Ciclofosfamida/uso terapéutico , Neoplasias Hematológicas/terapia , Inmunosupresores/uso terapéutico , Acondicionamiento Pretrasplante/métodos , Adolescente , Aloinjertos , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Enfermedad Injerto contra Huésped/etiología , Histocompatibilidad , Humanos , Lactante , Recién Nacido , Masculino , Síndromes Mielodisplásicos/terapia , Estudios Retrospectivos , Riesgo , Resultado del Tratamiento , Adulto Joven
7.
Biol Blood Marrow Transplant ; 22(1): 112-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26343947

RESUMEN

High-dose cyclophosphamide given after HLA-matched related and unrelated allogeneic bone marrow transplantation (BMT) for patients with hematologic malignancies is effective single-agent graft-versus-host disease (GVHD) prophylaxis in adults. Data describing outcomes for pediatric and young adult patients have not been reported. Between the years 2007 and 2013, 29 pediatric and young adult patients ages ≤21 years of age treated at our institution for high-risk hematologic malignancies underwent myeloablative HLA-matched related T cell-replete BMT. Eleven patients received post-transplantation cyclophosphamide (PTCy) as single-agent GVHD prophylaxis and were followed prospectively. Eighteen patients received calcineurin inhibitor (CNI)-based standard GVHD prophylaxis and were studied retrospectively as a control group. No acute GVHD (aGVHD) developed in patients receiving PTCy, whereas patients receiving CNI-based GVHD prophylaxis had cumulative incidences of grades II to IV and grades III and IV aGVHD of 27% and 5%, respectively. No patients receiving PTCy developed chronic GHVD, compared to 1 in the control group. Two-year overall survival was similar between the 2 groups (54% PTCy versus 58% CNI-based prophylaxis), as was event-free survival (42% PTCy versus 47% CNI-based). The 5-year cumulative incidence of relapse was 58% for PTCy and 42% for CNI-based GVHD prophylaxis (P = .45). These results suggest that PTCy is a safe and efficacious method of GVHD prophylaxis after an HLA-matched related BMT in the pediatric and young adult population that affords patients to be off all post-transplantation immunosuppression on day +5.


Asunto(s)
Trasplante de Médula Ósea , Ciclofosfamida/administración & dosificación , Enfermedad Injerto contra Huésped/prevención & control , Neoplasias Hematológicas/terapia , Donantes de Tejidos , Adolescente , Adulto , Aloinjertos , Niño , Preescolar , Ciclofosfamida/efectos adversos , Femenino , Enfermedad Injerto contra Huésped/etiología , Humanos , Lactante , Masculino , Adulto Joven
8.
Biol Blood Marrow Transplant ; 22(5): 895-901, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26860634

RESUMEN

Allogeneic hematopoietic stem cell transplantation (HSCT) is curative for many nonmalignant pediatric disorders, including hemoglobinopathies, bone marrow failure syndromes, and immunodeficiencies. There is great success using HLA-matched related donors for these patients; however, the use of alternative donors has been associated with increased graft failure, graft-versus-host disease (GVHD), and transplant-related mortality (TRM). HSCT using alternative donors with post-transplantation cyclophosphamide (PT/Cy) for GVHD prophylaxis has been performed for hematologic malignancies with engraftment, GVHD, and TRM comparable with that seen with HLA-matched related donors. There are limited reports of HSCT in nonmalignant pediatric disorders other than hemoglobinopathies using alternative donors and PT/Cy. We transplanted 11 pediatric patients with life-threatening nonmalignant conditions using reduced-intensity conditioning, alternative donors, and PT/Cy alone or in combination with tacrolimus and mycophenolate mofetil. We observed limited GVHD, no TRM, and successful engraftment sufficient to eliminate manifestations of disease in all patients. Allogeneic HSCT using alternative donors and PT/Cy shows promise for curing nonmalignant disorders; development of prospective clinical trials to confirm these observations is warranted.


Asunto(s)
Ciclofosfamida/administración & dosificación , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas , Acondicionamiento Pretrasplante , Donante no Emparentado , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Ácido Micofenólico/administración & dosificación , Tacrolimus/administración & dosificación
9.
Circulation ; 129(3): 359-72, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24163065

RESUMEN

BACKGROUND: The generation of vascular progenitors (VPs) from human induced pluripotent stem cells (hiPSCs) has great potential for treating vascular disorders such as ischemic retinopathies. However, long-term in vivo engraftment of hiPSC-derived VPs into the retina has not yet been reported. This goal may be limited by the low differentiation yield, greater senescence, and poor proliferation of hiPSC-derived vascular cells. To evaluate the potential of hiPSCs for treating ischemic retinopathies, we generated VPs from a repertoire of viral-integrated and nonintegrated fibroblast and cord blood (CB)-derived hiPSC lines and tested their capacity for homing and engrafting into murine retina in an ischemia-reperfusion model. METHODS AND RESULTS: VPs from human embryonic stem cells and hiPSCs were generated with an optimized vascular differentiation system. Fluorescence-activated cell sorting purification of human embryoid body cells differentially expressing endothelial/pericytic markers identified a CD31(+)CD146(+) VP population with high vascular potency. Episomal CB-induced pluripotent stem cells (iPSCs) generated these VPs with higher efficiencies than fibroblast-iPSC. Moreover, in contrast to fibroblast-iPSC-VPs, CB-iPSC-VPs maintained expression signatures more comparable to human embryonic stem cell VPs, expressed higher levels of immature vascular markers, demonstrated less culture senescence and sensitivity to DNA damage, and possessed fewer transmitted reprogramming errors. Luciferase transgene-marked VPs from human embryonic stem cells, CB-iPSCs, and fibroblast-iPSCs were injected systemically or directly into the vitreous of retinal ischemia-reperfusion-injured adult nonobese diabetic-severe combined immunodeficient mice. Only human embryonic stem cell- and CB-iPSC-derived VPs reliably homed and engrafted into injured retinal capillaries, with incorporation into damaged vessels for up to 45 days. CONCLUSIONS: VPs generated from CB-iPSCs possessed augmented capacity to home, integrate into, and repair damaged retinal vasculature.


Asunto(s)
Células Madre Embrionarias/citología , Sangre Fetal/citología , Células Madre Pluripotentes/citología , Daño por Reperfusión/terapia , Enfermedades de la Retina/terapia , Trasplante de Células Madre/métodos , Animales , Capilares/citología , Senescencia Celular , Daño del ADN , Modelos Animales de Enfermedad , Fibroblastos/citología , Supervivencia de Injerto , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Regeneración , Daño por Reperfusión/patología , Enfermedades de la Retina/patología , Transcriptoma
10.
Stem Cells ; 32(9): 2338-49, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24801745

RESUMEN

Gaucher disease (GD) is an autosomal recessive disorder caused by mutations in the acid ß-glucocerebrosidase (GCase; GBA) gene. The hallmark of GD is the presence of lipid-laden Gaucher macrophages, which infiltrate bone marrow and other organs. These pathological macrophages are believed to be the sources of elevated levels of inflammatory mediators present in the serum of GD patients. The alteration in the immune environment caused by GD is believed to play a role in the increased risk of developing multiple myeloma and other malignancies in GD patients. To determine directly whether Gaucher macrophages are abnormally activated and whether their functional defects can be reversed by pharmacological intervention, we generated GD macrophages by directed differentiation of human induced pluripotent stem cells (hiPSC) derived from patients with types 1, 2, and 3 GD. GD hiPSC-derived macrophages expressed higher levels of tumor necrosis factor α, IL-6, and IL-1ß than control cells, and this phenotype was exacerbated by treatment with lipopolysaccharide. In addition, GD hiPSC macrophages exhibited a striking delay in clearance of phagocytosed red blood cells, recapitulating the presence of red blood cell remnants in Gaucher macrophages from bone marrow aspirates. Incubation of GD hiPSC macrophages with recombinant GCase, or with the chaperones isofagomine and ambroxol, corrected the abnormal phenotypes of GD macrophages to an extent that reflected their known clinical efficacies. We conclude that Gaucher macrophages are the likely source of the elevated levels of inflammatory mediators in the serum of GD patients and that GD hiPSC are valuable new tools for studying disease mechanisms and drug discovery.


Asunto(s)
Citocinas/biosíntesis , Enfermedad de Gaucher/metabolismo , Enfermedad de Gaucher/patología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Femenino , Enfermedad de Gaucher/genética , Humanos , Células Madre Pluripotentes Inducidas/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID
11.
Proc Natl Acad Sci U S A ; 109(44): 18054-9, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23071332

RESUMEN

Gaucher disease (GD) is an autosomal recessive disorder caused by mutations in the acid ß-glucocerebrosidase gene. To model GD, we generated human induced pluripotent stem cells (hiPSC), by reprogramming skin fibroblasts from patients with type 1 (N370S/N370S), type 2 (L444P/RecNciI), and type 3 (L444P/L444P) GD. Pluripotency was demonstrated by the ability of GD hiPSC to differentiate to all three germ layers and to form teratomas in vivo. GD hiPSC differentiated efficiently to the cell types most affected in GD, i.e., macrophages and neuronal cells. GD hiPSC-macrophages expressed macrophage-specific markers, were phagocytic, and were capable of releasing inflammatory mediators in response to LPS. Moreover, GD hiPSC-macrophages recapitulated the phenotypic hallmarks of the disease. They exhibited low glucocerebrosidase (GC) enzymatic activity and accumulated sphingolipids, and their lysosomal functions were severely compromised. GD hiPSC-macrophages had a defect in their ability to clear phagocytosed RBC, a phenotype of tissue-infiltrating GD macrophages. The kinetics of RBC clearance by types 1, 2, and 3 GD hiPSC-macrophages correlated with the severity of the mutations. Incubation with recombinant GC completely reversed the delay in RBC clearance from all three types of GD hiPSC-macrophages, indicating that their functional defects were indeed caused by GC deficiency. However, treatment of induced macrophages with the chaperone isofagomine restored phagocytosed RBC clearance only partially, regardless of genotype. These findings are consistent with the known clinical efficacies of recombinant GC and isofagomine. We conclude that cell types derived from GD hiPSC can effectively recapitulate pathologic hallmarks of the disease.


Asunto(s)
Enfermedad de Gaucher/patología , Células Madre Pluripotentes/citología , Diferenciación Celular , Linaje de la Célula , Humanos , Activación de Macrófagos , Modelos Biológicos
12.
Biochim Biophys Acta ; 1830(2): 2385-94, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23104383

RESUMEN

BACKGROUND: Induced pluripotent stem cells (iPSC) derived from reprogrammed patient somatic cells possess enormous therapeutic potential. However, unlocking the full capabilities of iPSC will require an improved understanding of the molecular mechanisms which govern the induction and maintenance of pluripotency, as well as directed differentiation to clinically relevant lineages. Induced pluripotency of a differentiated cell is mediated by sequential cascades of genetic and epigenetic reprogramming of somatic histone and DNA CpG methylation marks. These genome-wide changes are mediated by a coordinated activity of transcription factors and epigenetic modifying enzymes. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), are now recognized as an important third class of regulators of the pluripotent state. SCOPE OF REVIEW: This review surveys the currently known roles and mechanisms of ncRNAs in regulating the embryonic and induced pluripotent states. MAJOR CONCLUSIONS: Through a variety of mechanisms, ncRNAs regulate constellations of key pluripotency genes and epigenetic regulators, and thus critically determine induction and maintenance of the pluripotent state. GENERAL SIGNIFICANCE: A further understanding of the roles of ncRNAs in regulating pluripotency may help assess the quality of human iPSC reprogramming. Additionally, ncRNA biology may help decipher potential transcriptional and epigenetic commonalities between the self renewal processes that govern both ESC and tumor initiating cancer stem cells (CSC). This article is part of a Special Issue entitled Biochemistry of Stem Cells.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Pluripotentes/citología , ARN no Traducido/fisiología , Islas de CpG , Metilación de ADN , Humanos
13.
bioRxiv ; 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38915486

RESUMEN

PARP1 (ARTD1) and Tankyrases (TNKS1/TNKS2; PARP5a/5b) are poly-ADP-ribose polymerases (PARPs) with catalytic and non-catalytic functions that regulate both the genome and proteome during zygotic genome activation (ZGA), totipotent, and pluripotent embryonic stages. Here, we show that primed, conventional human pluripotent stem cells (hPSC) cultured continuously under non-specific TNKS1/TNKS2/PARP1-inhibited chemical naive reversion conditions underwent epigenetic reprogramming to clonal blastomere-like stem cells. TIRN stem cells concurrently expressed hundreds of gene targets of the ZGA-priming pioneer factor DUX4, as well as a panoply of four-cell (4C)-specific (e.g., TPRXL, HOX clusters), eight-cell (8C)-specific (e.g., DUXA, GSC, GATA6), primitive endoderm-specific (e.g., GATA4, SOX17), trophectoderm-specific (e.g., CDX2, TFAP2C), and naive epiblast-specific (e.g., DNMT3L, NANOG, POU5F1(OCT4)) factors; all in a hybrid, combinatorial single-cell manner. Mapping of proteomic and single-cell expressions of TIRN cells against human preimplantation embryo references identified them as relatively homogenous 4C-8C stage populations. Injection of TIRN cells into murine 8C-16C-staged embryos resulted in efficient totipotent-like single cell contributions of human cells to both extra-embryonic (trophectoderm, placenta) and embryonic (neural, fetal liver, hematopoietic) lineages in human-murine blastocyst and fetal chimeras. Pairing of proteome with ubiquitinome analyses of TIRN cells revealed a global shutdown of ADP-ribosylation, and a perturbed TNKS/PARP1 equilibrium which not only impacted the protein levels of hundreds of TNKS/PARP1 substrates via a rewiring of the ubiquitin-proteosome system (UPS), but also de-repressed expression of hundreds of developmental genes associated with PARP1 suppression. ChIP-Seq analysis of core NANOG-SOX2-OCT4 (NSO) pluripotency factors in TIRN cells identified reprogrammed DUX4-accessible distal and cis-regulatory enhancer regions that were co-bound by PARP1 (NSOP). These NSOP enhancer regions possessed co-binding motifs for hundreds of the same ZGA-associated, embryonic, and extraembryonic lineage-specifying pioneer factors (e.g., HOX, FOX, GATA, SOX, TBX, CDX families) that were concurrently co-expressed in TIRN cells; suggesting that PARP1 and DUX4 cooperate with NSO pluripotency core factors to regulate the epigenetic plasticity of a human totipotency program. These findings provide the first demonstration that global, proteome-wide perturbations of post-translational modifications (i.e., ADP-ribosylation, ubiquitination) can regulate epigenetic reprogramming during human embryogenesis. Totipotent TIRN stem cells will provide a valuable cell culture model for studying the proteogenomic regulation of lineage specification from human blastomere stages and may facilitate the efficient generation of human organs in interspecies chimeras.

14.
Nat Commun ; 15(1): 3258, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637498

RESUMEN

Viral infections remain a major risk in immunocompromised pediatric patients, and virus-specific T cell (VST) therapy has been successful for treatment of refractory viral infections in prior studies. We performed a phase II multicenter study (NCT03475212) for the treatment of pediatric patients with inborn errors of immunity and/or post allogeneic hematopoietic stem cell transplant with refractory viral infections using partially-HLA matched VSTs targeting cytomegalovirus, Epstein-Barr virus, or adenovirus. Primary endpoints were feasibility, safety, and clinical responses (>1 log reduction in viremia at 28 days). Secondary endpoints were reconstitution of antiviral immunity and persistence of the infused VSTs. Suitable VST products were identified for 75 of 77 clinical queries. Clinical responses were achieved in 29 of 47 (62%) of patients post-HSCT including 73% of patients evaluable at 1-month post-infusion, meeting the primary efficacy endpoint (>52%). Secondary graft rejection occurred in one child following VST infusion as described in a companion article. Corticosteroids, graft-versus-host disease, transplant-associated thrombotic microangiopathy, and eculizumab treatment correlated with poor response, while uptrending absolute lymphocyte and CD8 T cell counts correlated with good response. This study highlights key clinical factors that impact response to VSTs and demonstrates the feasibility and efficacy of this therapy in pediatric HSCT.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Trasplante de Células Madre Hematopoyéticas , Virosis , Humanos , Niño , Herpesvirus Humano 4 , Factores de Riesgo , Trasplante de Células Madre Hematopoyéticas/efectos adversos
15.
Cytometry A ; 83(1): 114-26, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22736485

RESUMEN

The hematopoietic and vascular lineages are intimately entwined as they arise together from bipotent hemangioblasts and hemogenic endothelial precursors during human embryonic development. In vitro differentiation of human pluripotent stem cells toward these lineages provides opportunities for elucidating the mechanisms of hematopoietic genesis. We previously demonstrated the stepwise in vitro differentiation of human embryonic stem cells (hESC) to definitive erythromyelopoiesis through clonogenic bipotent primitive hemangioblasts. This system recapitulates an orderly hematopoiesis similar to human yolk sac development via the generation of mesodermal-hematoendothelial progenitor cells that give rise to endothelium followed by embryonic primitive and definitive hematopoietic cells. Here, we report that under modified feeder-free endothelial culture conditions, multipotent CD34⁺ CD45⁺ hematopoietic progenitors arise in mass quantities from differentiated hESC and human induced pluripotent stem cells (hiPSC). These hematopoietic progenitors arose directly from adherent endothelial/stromal cell layers in a manner resembling in vivo hematopoiesis from embryonic hemogenic endothelium. Although fibroblast-derived hiPSC lines were previously found inefficient in hemato-endothelial differentiation capacity, our culture system also supported robust hiPSC hemato-vascular differentiation at levels comparable to hESC. We present comparative differentiation results for simultaneously generating hematopoietic and vascular progenitors from both hESC and fibroblast-hiPSC. This defined, optimized, and low-density differentiation system will be ideal for direct single-cell time course studies of the earliest hematopoietic events using time-lapse videography, or bulk kinetics using flow cytometry analyses on emerging hematopoietic progenitors.


Asunto(s)
Vasos Sanguíneos/citología , Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes Inducidas/citología , Células Madre/citología , Antígenos CD34/metabolismo , Vasos Sanguíneos/fisiología , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Citometría de Flujo , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/fisiología , Humanos , Técnicas In Vitro , Células Madre Pluripotentes Inducidas/fisiología , Antígenos Comunes de Leucocito/metabolismo , Células Madre/inmunología , Células Madre/fisiología , Imagen de Lapso de Tiempo
16.
NPJ Regen Med ; 8(1): 66, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38086850

RESUMEN

Granulocytes and macrophages are the frontline defenders of the innate immune system. These myeloid cells play a crucial role in not only eliminating pathogens and tumor cells, but also regulating adaptive immune responses. In neonatal sepsis and post-chemotherapy agranulocytosis, the absence of these cells leaves the host highly vulnerable to infections. Beyond replacement to prevent or control neutropenic sepsis, engineered myeloid cells may offer distinct opportunities for cell therapies. For example, the mobility and specific homing capacities of neutrophils to sites of inflammation could be exploited to deliver biocidal agents, or anti-inflammatory healing signals during sepsis, autoimmunity, and organ transplantation. Additionally, myeloid cells can be engineered to express chimeric antigen receptors (CAR), carry chemotherapeutics, or enhance lymphoid tumor killing. However, traditional methods of cell isolation are incapable of providing sufficient cell numbers of these short-lived cells; their propensity for premature activation further complicates their cell engineering. Here, we review current and future biotherapeutic innovations that employ engineered multipotent myeloid progenitors derived from either self-renewing human induced pluripotent stem cells (hiPSC) or primary CD34+ hematopoietic stem-progenitors. We provide a roadmap for solving the challenges of sourcing, cost, and production of engineered myeloid cell therapies.

17.
J Mol Cell Cardiol ; 53(1): 15-23, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22713758

RESUMEN

In this study, we characterized the electrophysiological benefits of engrafting human embryonic stem cell-derived cardiomyocytes (hESC-CMs) in a model of arrhythmogenic cardiac tissue. Using transforming growth factor-ß treated monolayers of neonatal rat ventricular cells (NRVCs), which retain several key aspects of the healing infarct such as an excess of contractile myofibroblasts and slowed, heterogeneous conduction, we assessed the ability of hESC-CMs to improve conduction and prevent arrhythmias. Cells from beating embryoid bodies (hESC-CMs) can form functional monolayers which beat spontaneously and can be electrically stimulated, with mean action potential duration of 275 ± 36 ms and conduction velocity (CV) of 10.6 ± 4.2 cm/s (n = 3). These cells, or cells from non-beating embryoid bodies (hEBCs) were added to anisotropic, NRVC monolayers. Immunostaining demonstrated hESC-CM survival and engraftment, and dye transfer assays confirmed functional coupling between hESC-CMs and NRVCs. Conduction velocities significantly increased in anisotropic NRVC monolayers after engraftment of hESC-CMs (13.4 ± 0.9 cm/s, n = 35 vs. 30.1 ± 3.2 cm/s, n = 20 in the longitudinal direction and 4.3 ± 0.3 cm/s vs. 9.3 ± 0.9 cm/s in the transverse direction), but decreased to even lower values after engraftment of non-cardiac hEBCs (to 10.6 ± 1.3 cm/s and 3.1 ± 0.5 cm/s, n = 11, respectively). Furthermore, reentrant wave vulnerability in NRVC monolayers decreased by 20% after engraftment of hESC-CMs, but did not change with engraftment of hEBCs. Finally, the culture of hESC-CMs in transwell inserts, which prevents juxtacrine interactions, or engraftment with connexin43-silenced hESC-CMs provided no functional improvement to NRVC monolayers. These results demonstrate that hESC-CMs can reverse the slowing of conduction velocity, reduce the incidence of reentry, and augment impaired electrical propagation via gap junction coupling to host cardiomyocytes in this arrhythmogenic in vitro model.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Fenómenos Electrofisiológicos , Células Madre Embrionarias/citología , Miocitos Cardíacos/trasplante , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/terapia , Comunicación Celular , Diferenciación Celular , Línea Celular , Silenciador del Gen , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas
18.
Methods Mol Biol ; 2416: 133-156, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34870835

RESUMEN

Tankyrase/PARP inhibitor-regulated naïve human pluripotent stem cells (TIRN-hPSC) represent a new class of human stem cells for regenerative medicine that can differentiate into multi-lineage progenitors with improved in vivo functionality. Chemical reversion of conventional, primed hPSC to a TIRN-hPSC state alleviates dysfunctional epigenetic donor cell memory, lineage-primed gene expression, and potentially disease-associated aberrations in their differentiated progeny. Here, we provide methods for the reversion of normal or diseased patient-specific primed hPSC to TIRN-hPSC and describe their subsequent differentiation into embryonic-like pericytic-endothelial "naïve" vascular progenitors (N-VP). N-VP possess improved vascular functionality, high epigenetic plasticity, maintain greater genomic stability, and are more efficient in migrating to and re-vascularizing ischemic tissues than those generated from primed isogenic hPSC. We also describe detailed methods for the ocular transplantation and quantitation of vascular engraftment of N-VP into the ischemia-damaged neural retina of a humanized mouse model of ischemic retinopathy. The application of TIRN-hPSC-derived N-VP will advance vascular cell therapies of ischemic retinopathy, myocardial infarction, and cerebral vascular stroke.


Asunto(s)
Células Madre Pluripotentes , Animales , Diferenciación Celular/efectos de los fármacos , Humanos , Isquemia , Ratones , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Enfermedades de la Retina , Tanquirasas
19.
Artículo en Inglés | MEDLINE | ID: mdl-36683654

RESUMEN

We present a patient case of a 73-year-old man with new-onset substernal chest pain and B symptoms, found on computed tomography imaging to have massive mediastinal lymphadenopathy of more than 6 cm. Positron emission tomography imaging revealed fluorodeoxyglucose-avid nodes further extending to the axillary, abdominal, and inguinal regions. After a broad patient work-up for infectious, malignant, and rheumatic causes, he was ultimately diagnosed with Rosai-Dorfman disease, a rare histiocytic neoplasm, by excisional lymph node biopsy.

20.
NPJ Regen Med ; 6(1): 25, 2021 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-34001907

RESUMEN

Human pluripotent stem cells (hPSCs) can generate specialized cell lineages that have great potential for regenerative therapies and disease modeling. However, the developmental stage of the lineages generated from conventional hPSC cultures in vitro are embryonic in phenotype, and may not possess the cellular maturity necessary for corrective regenerative function in vivo in adult recipients. Here, we present the scientific evidence for how adult human tissues could generate human-animal interspecific chimeras to solve this problem. First, we review the phenotypes of the embryonic lineages differentiated from conventional hPSC in vitro and through organoid technologies and compare their functional relevance to the tissues generated during normal human in utero fetal and adult development. We hypothesize that the developmental incongruence of embryo-stage hPSC-differentiated cells transplanted into a recipient adult host niche is an important mechanism ultimately limiting their utility in cell therapies and adult disease modeling. We propose that this developmental obstacle can be overcome with optimized interspecies chimeras that permit the generation of adult-staged, patient-specific whole organs within animal hosts with human-compatible gestational time-frames. We suggest that achieving this goal may ultimately have to await the derivation of alternative, primitive totipotent-like stem cells with improved embryonic chimera capacities. We review the scientific challenges of deriving alternative human stem cell states with expanded embryonic potential, outline a path forward for conducting this emerging research with appropriate ethical and regulatory oversight, and defend the case of why current federal funding restrictions on this important category of biomedical research should be liberalized.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA