Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Mol Ther ; 32(8): 2535-2548, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38867450

RESUMEN

Stem and progenitor cells hold great promise for regenerative medicine and gene therapy approaches. However, transplantation of living cells entails a fundamental risk of unwanted growth, potentially exacerbated by CRISPR-Cas9 or other genetic manipulations. Here, we describe a safety system to control cell proliferation while allowing robust and efficient cell manufacture, without any added genetic elements. Inactivating TYMS, a key nucleotide metabolism enzyme, in several cell lines resulted in cells that proliferate only when supplemented with exogenous thymidine. Under supplementation, TYMS-/--pluripotent stem cells proliferate, produce teratomas, and successfully differentiate into potentially therapeutic cell types such as pancreatic ß cells. Our results suggest that supplementation with exogenous thymidine affects stem cell proliferation, but not the function of stem cell-derived cells. After differentiation, postmitotic cells do not require thymidine in vitro or in vivo, as shown by the production of functional human insulin in mice up to 5 months after implantation of stem cell-derived pancreatic tissue.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Timidina , Timidilato Sintasa , Humanos , Animales , Ratones , Timidina/metabolismo , Timidilato Sintasa/genética , Timidilato Sintasa/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Línea Celular , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Sistemas CRISPR-Cas
2.
Diabetologia ; 67(9): 1912-1929, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38871836

RESUMEN

AIMS/HYPOTHESIS: Stem cell-derived islets (SC-islets) are being used as cell replacement therapy for insulin-dependent diabetes. Non-invasive long-term monitoring methods for SC-islet grafts, which are needed to detect misguided differentiation in vivo and to optimise their therapeutic effectiveness, are lacking. Positron emission tomography (PET) has been used to monitor transplanted primary islets. We therefore aimed to apply PET as a non-invasive monitoring method for SC-islet grafts. METHODS: We implanted different doses of human SC-islets, SC-islets derived using an older protocol or a state-of-the-art protocol and SC-islets genetically rendered hyper- or hypoactive into mouse calf muscle to yield different kinds of grafts. We followed the grafts with PET using two tracers, glucagon-like peptide 1 receptor-binding [18F]F-dibenzocyclooctyne-exendin-4 ([18F]exendin) and the dopamine precursor 6-[18F]fluoro-L-3,4-dihydroxyphenylalanine ([18F]FDOPA), for 5 months, followed by histological assessment of graft size and composition. Additionally, we implanted a kidney subcapsular cohort with different SC-islet doses to assess the connection between C-peptide and stem cell-derived beta cell (SC-beta cell) mass. RESULTS: Small but pure and large but impure grafts were derived from SC-islets. PET imaging allowed detection of SC-islet grafts even <1 mm3 in size, [18F]exendin having a better detection rate than [18F]FDOPA (69% vs 44%, <1 mm3; 96% vs 85%, >1 mm3). Graft volume quantified with [18F]exendin (r2=0.91) and [18F]FDOPA (r2=0.86) strongly correlated with actual graft volume. [18F]exendin PET delineated large cystic structures and its uptake correlated with graft SC-beta cell proportion (r2=0.68). The performance of neither tracer was affected by SC-islet graft hyper- or hypoactivity. C-peptide measurements under fasted or glucose-stimulated conditions did not correlate with SC-islet graft volume or SC-beta cell mass, with C-peptide under hypoglycaemia having a weak correlation with SC-beta cell mass (r2=0.52). CONCLUSIONS/INTERPRETATION: [18F]exendin and [18F]FDOPA PET enable non-invasive assessment of SC-islet graft size and aspects of graft composition. These methods could be leveraged for optimising SC-islet cell replacement therapy in diabetes.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Tomografía de Emisión de Positrones , Trasplante de Islotes Pancreáticos/métodos , Animales , Ratones , Humanos , Tomografía de Emisión de Positrones/métodos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/citología , Células Madre/citología , Células Madre/metabolismo , Masculino , Diabetes Mellitus Tipo 1/cirugía , Diabetes Mellitus Tipo 1/metabolismo , Femenino
3.
Diabetes ; 73(7): 1127-1139, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38603470

RESUMEN

Pluripotent stem cell-derived islets (SC-islets) have emerged as a new source for ß-cell replacement therapy. The function of human islet transplants is hampered by excessive cell death posttransplantation; contributing factors include inflammatory reactions, insufficient revascularization, and islet amyloid formation. However, there is a gap in knowledge of the engraftment process of SC-islets. In this experimental study, we investigated the engraftment capability of SC-islets at 3 months posttransplantation and observed that cell apoptosis rates were lower but vascular density was similar in SC-islets compared with human islets. Whereas the human islet transplant vascular structures were a mixture of remnant donor endothelium and ingrowing blood vessels, the SC-islets contained ingrowing blood vessels only. Oxygenation in the SC-islet grafts was twice as high as that in the corresponding grafts of human islets, suggesting better vascular functionality. Similar to the blood vessel ingrowth, reinnervation of the SC-islets was four- to fivefold higher than that of the human islets. Both SC-islets and human islets contained amyloid at 1 and 3 months posttransplantation. We conclude that the vascular and neural engraftment of SC-islets are superior to those of human islets, but grafts of both origins develop amyloid, with potential long-term consequences.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Humanos , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/irrigación sanguínea , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Animales , Ratones , Apoptosis/fisiología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Supervivencia de Injerto/fisiología , Masculino
4.
Am J Hum Genet ; 111(4): 714-728, 2024 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-38579669

RESUMEN

Argininosuccinate lyase deficiency (ASLD) is a recessive metabolic disorder caused by variants in ASL. In an essential step in urea synthesis, ASL breaks down argininosuccinate (ASA), a pathognomonic ASLD biomarker. The severe disease forms lead to hyperammonemia, neurological injury, and even early death. The current treatments are unsatisfactory, involving a strict low-protein diet, arginine supplementation, nitrogen scavenging, and in some cases, liver transplantation. An unmet need exists for improved, efficient therapies. Here, we show the potential of a lipid nanoparticle-mediated CRISPR approach using adenine base editors (ABEs) for ASLD treatment. To model ASLD, we first generated human-induced pluripotent stem cells (hiPSCs) from biopsies of individuals homozygous for the Finnish founder variant (c.1153C>T [p.Arg385Cys]) and edited this variant using the ABE. We then differentiated the hiPSCs into hepatocyte-like cells that showed a 1,000-fold decrease in ASA levels compared to those of isogenic non-edited cells. Lastly, we tested three different FDA-approved lipid nanoparticle formulations to deliver the ABE-encoding RNA and the sgRNA targeting the ASL variant. This approach efficiently edited the ASL variant in fibroblasts with no apparent cell toxicity and minimal off-target effects. Further, the treatment resulted in a significant decrease in ASA, to levels of healthy donors, indicating restoration of the urea cycle. Our work describes a highly efficient approach to editing the disease-causing ASL variant and restoring the function of the urea cycle. This method relies on RNA delivered by lipid nanoparticles, which is compatible with clinical applications, improves its safety profile, and allows for scalable production.


Asunto(s)
Argininosuccinatoliasa , Aciduria Argininosuccínica , Humanos , Argininosuccinatoliasa/genética , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/terapia , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , ARN Guía de Sistemas CRISPR-Cas , Urea , Edición Génica/métodos
5.
Nat Commun ; 13(1): 6363, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36289205

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease that results in the destruction of insulin producing pancreatic ß-cells. One of the genes associated with T1D is TYK2, which encodes a Janus kinase with critical roles in type-Ι interferon (IFN-Ι) mediated intracellular signalling. To study the role of TYK2 in ß-cell development and response to IFNα, we generated TYK2 knockout human iPSCs and directed them into the pancreatic endocrine lineage. Here we show that loss of TYK2 compromises the emergence of endocrine precursors by regulating KRAS expression, while mature stem cell-islets (SC-islets) function is not affected. In the SC-islets, the loss or inhibition of TYK2 prevents IFNα-induced antigen processing and presentation, including MHC Class Ι and Class ΙΙ expression, enhancing their survival against CD8+ T-cell cytotoxicity. These results identify an unsuspected role for TYK2 in ß-cell development and support TYK2 inhibition in adult ß-cells as a potent therapeutic target to halt T1D progression.


Asunto(s)
Diabetes Mellitus Tipo 1 , Insulinas , Humanos , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Insulinas/metabolismo , Interferón-alfa/farmacología , Interferón-alfa/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , TYK2 Quinasa/genética , TYK2 Quinasa/metabolismo , Células Secretoras de Insulina
6.
Nat Biotechnol ; 40(7): 1042-1055, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35241836

RESUMEN

Transplantation of pancreatic islet cells derived from human pluripotent stem cells is a promising treatment for diabetes. Despite progress in the generation of stem-cell-derived islets (SC-islets), no detailed characterization of their functional properties has been conducted. Here, we generated functionally mature SC-islets using an optimized protocol and benchmarked them comprehensively against primary adult islets. Biphasic glucose-stimulated insulin secretion developed during in vitro maturation, associated with cytoarchitectural reorganization and the increasing presence of alpha cells. Electrophysiology, signaling and exocytosis of SC-islets were similar to those of adult islets. Glucose-responsive insulin secretion was achieved despite differences in glycolytic and mitochondrial glucose metabolism. Single-cell transcriptomics of SC-islets in vitro and throughout 6 months of engraftment in mice revealed a continuous maturation trajectory culminating in a transcriptional landscape closely resembling that of primary islets. Our thorough evaluation of SC-islet maturation highlights their advanced degree of functionality and supports their use in further efforts to understand and combat diabetes.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Células Madre Pluripotentes , Animales , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Ratones , Células Madre Pluripotentes/metabolismo
7.
Front Endocrinol (Lausanne) ; 13: 837450, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35250887

RESUMEN

Congenital hyperinsulinism (CHI) is a severe inherited neonatal disorder characterized by inappropriate insulin secretion caused by genetic defects of the pancreatic beta cells. Several open questions remain in CHI research, such as the optimal treatment for the most common type of CHI, caused by mutations in the genes encoding ATP-sensitive potassium channels, and the molecular mechanisms of newly identified CHI genes. Answering these questions requires robust preclinical models, particularly since primary patient material is extremely scarce and accurate animal models are not available. In this short review, we explain why pluripotent stem cell derived islets present an attractive solution to these issues and outline the current progress in stem-cell based modeling of CHI. Stem cell derived islets enable the study of molecular mechanisms of CHI and the discovery of novel antihypoglycemic drugs, while also providing a valuable model to study the biology of variable functional states of beta cells.


Asunto(s)
Hiperinsulinismo Congénito , Islotes Pancreáticos , Animales , Hiperinsulinismo Congénito/tratamiento farmacológico , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Células Madre/metabolismo
8.
Nat Cell Biol ; 24(2): 148-154, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35165416

RESUMEN

Metabolic characteristics of adult stem cells are distinct from their differentiated progeny, and cellular metabolism is emerging as a potential driver of cell fate conversions1-4. How these metabolic features are established remains unclear. Here we identified inherited metabolism imposed by functionally distinct mitochondrial age-classes as a fate determinant in asymmetric division of epithelial stem-like cells. While chronologically old mitochondria support oxidative respiration, the electron transport chain of new organelles is proteomically immature and they respire less. After cell division, selectively segregated mitochondrial age-classes elicit a metabolic bias in progeny cells, with oxidative energy metabolism promoting differentiation in cells that inherit old mitochondria. Cells that inherit newly synthesized mitochondria with low levels of Rieske iron-sulfur polypeptide 1 have a higher pentose phosphate pathway activity, which promotes de novo purine biosynthesis and redox balance, and is required to maintain stemness during early fate determination after division. Our results demonstrate that fate decisions are susceptible to intrinsic metabolic bias imposed by selectively inherited mitochondria.


Asunto(s)
Células Madre Adultas/metabolismo , Diferenciación Celular , Linaje de la Célula , ADN Mitocondrial/genética , Metabolismo Energético , Genes Mitocondriales , Glándulas Mamarias Humanas/metabolismo , Mitocondrias/metabolismo , Animales , Línea Celular , Proliferación Celular , Senescencia Celular , Femenino , Humanos , Glándulas Mamarias Humanas/citología , Metaboloma , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/genética , Fenotipo , Proteoma
9.
Diabetes ; 71(4): 653-668, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35044456

RESUMEN

Type 1 diabetes (T1D) results from autoimmune destruction of ß-cells in the pancreas. Protein tyrosine phosphatases (PTPs) are candidate genes for T1D and play a key role in autoimmune disease development and ß-cell dysfunction. Here, we assessed the global protein and individual PTP profiles in the pancreas from nonobese mice with early-onset diabetes (NOD) mice treated with an anti-CD3 monoclonal antibody and interleukin-1 receptor antagonist. The treatment reversed hyperglycemia, and we observed enhanced expression of PTPN2, a PTP family member and T1D candidate gene, and endoplasmic reticulum (ER) chaperones in the pancreatic islets. To address the functional role of PTPN2 in ß-cells, we generated PTPN2-deficient human stem cell-derived ß-like and EndoC-ßH1 cells. Mechanistically, we demonstrated that PTPN2 inactivation in ß-cells exacerbates type I and type II interferon signaling networks and the potential progression toward autoimmunity. Moreover, we established the capacity of PTPN2 to positively modulate the Ca2+-dependent unfolded protein response and ER stress outcome in ß-cells. Adenovirus-induced overexpression of PTPN2 partially protected from ER stress-induced ß-cell death. Our results postulate PTPN2 as a key protective factor in ß-cells during inflammation and ER stress in autoimmune diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Animales , Apoptosis/genética , Diabetes Mellitus Tipo 1/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Humanos , Células Secretoras de Insulina/metabolismo , Interferón gamma/farmacología , Ratones , Ratones Endogámicos NOD , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética
10.
J Nucl Med ; 63(2): 310-315, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34215672

RESUMEN

Surgery with curative intent can be offered to congenital hyperinsulinism (CHI) patients, provided that the lesion is focal. Radiolabeled exendin-4 specifically binds the glucagonlike peptide 1 receptor on pancreatic ß-cells. In this study, we compared the performance of 18F-DOPA PET/CT, the current standard imaging method for CHI, and PET/CT with the new tracer 68Ga-NODAGA-exendin-4 in the preoperative detection of focal CHI. Methods: Nineteen CHI patients underwent both 18F-DOPA PET/CT and 68Ga-NODAGA-exendin-4 PET/CT before surgery. The images were evaluated in 3 settings: a standard clinical reading, a masked expert reading, and a joint reading. The target (lesion)-to-nontarget (normal pancreas) ratio was determined using SUVmax Image quality was rated by pediatric surgeons in a questionnaire. Results: Fourteen of 19 patients having focal lesions underwent surgery. On the basis of clinical readings, the sensitivity of 68Ga-NODAGA-exendin-4 PET/CT (100%; 95% CI, 77%-100%) was higher than that of 18F-DOPA PET/CT (71%; 95% CI, 42%-92%). Interobserver agreement between readings was higher for 68Ga-NODAGA-exendin-4 than for 18F-DOPA PET/CT (Fleiss κ = 0.91 vs. 0.56). 68Ga-NODAGA-exendin-4 PET/CT provided significantly (P = 0.021) higher target-to-nontarget ratios (2.02 ± 0.65) than did 18F-DOPA PET/CT (1.40 ± 0.40). On a 5-point scale, pediatric surgeons rated 68Ga-NODAGA-exendin-4 PET/CT as superior to 18F-DOPA PET/CT. Conclusion: For the detection of focal CHI, 68Ga-NODAGA-exendin-4 PET/CT has higher clinical sensitivity and better interobserver correlation than 18F-DOPA PET/CT. Better contrast and image quality make 68Ga-NODAGA-exendin-4 PET/CT superior to 18F-DOPA PET/CT in surgeons' intraoperative quest for lesion localization.


Asunto(s)
Hiperinsulinismo Congénito , Tomografía Computarizada por Tomografía de Emisión de Positrones , Acetatos , Niño , Hiperinsulinismo Congénito/diagnóstico por imagen , Exenatida , Radioisótopos de Galio , Compuestos Heterocíclicos con 1 Anillo , Humanos , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos
11.
Diabetologia ; 64(3): 630-640, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33404684

RESUMEN

AIMS/HYPOTHESIS: Congenital hyperinsulinism caused by mutations in the KATP-channel-encoding genes (KATPHI) is a potentially life-threatening disorder of the pancreatic beta cells. No optimal medical treatment is available for patients with diazoxide-unresponsive diffuse KATPHI. Therefore, we aimed to create a model of KATPHI using patient induced pluripotent stem cell (iPSC)-derived islets. METHODS: We derived iPSCs from a patient carrying a homozygous ABCC8V187D mutation, which inactivates the sulfonylurea receptor 1 (SUR1) subunit of the KATP-channel. CRISPR-Cas9 mutation-corrected iPSCs were used as controls. Both were differentiated to stem cell-derived islet-like clusters (SC-islets) and implanted into NOD-SCID gamma mice. RESULTS: SUR1-mutant and -corrected iPSC lines both differentiated towards the endocrine lineage, but SUR1-mutant stem cells generated 32% more beta-like cells (SC-beta cells) (64.6% vs 49.0%, p = 0.02) and 26% fewer alpha-like cells (16.1% vs 21.8% p = 0.01). SUR1-mutant SC-beta cells were 61% more proliferative (1.23% vs 0.76%, p = 0.006), and this phenotype could be induced in SUR1-corrected cells with pharmacological KATP-channel inactivation. The SUR1-mutant SC-islets secreted 3.2-fold more insulin in low glucose conditions (0.0174% vs 0.0054%/min, p = 0.0021) and did not respond to KATP-channel-acting drugs in vitro. Mice carrying grafts of SUR1-mutant SC-islets presented with 38% lower fasting blood glucose (4.8 vs 7.7 mmol/l, p = 0.009) and their grafts failed to efficiently shut down insulin secretion during induced hypoglycaemia. Explanted SUR1-mutant grafts displayed an increase in SC-beta cell proportion and SC-beta cell nucleomegaly, which was independent of proliferation. CONCLUSIONS/INTERPRETATION: We have created a model recapitulating the known pathophysiology of KATPHI both in vitro and in vivo. We have also identified a novel role for KATP-channel activity during human islet development. This model will enable further studies for the improved understanding and clinical management of KATPHI without the need for primary patient tissue.


Asunto(s)
Hiperinsulinismo Congénito/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Islotes Pancreáticos/metabolismo , Receptores de Sulfonilureas/metabolismo , Animales , Diferenciación Celular , Línea Celular , Proliferación Celular , Hiperinsulinismo Congénito/genética , Hiperinsulinismo Congénito/patología , Hiperinsulinismo Congénito/fisiopatología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/trasplante , Secreción de Insulina , Islotes Pancreáticos/patología , Islotes Pancreáticos/fisiopatología , Trasplante de Islotes Pancreáticos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Mutación , Fenotipo , Receptores de Sulfonilureas/genética
12.
J Clin Endocrinol Metab ; 106(4): e1542-e1551, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33475139

RESUMEN

CONTEXT: The management of congenital hyperinsulinism (CHI) has improved. OBJECTIVE: To examine the treatment and long-term outcome of Finnish patients with persistent and transient CHI (P-CHI and T-CHI). DESIGN: A population-based retrospective study of CHI patients treated from 1972 to 2015. PATIENTS: 106 patients with P-CHI and 132 patients with T-CHI (in total, 42 diagnosed before and 196 after year 2000) with median follow-up durations of 12.5 and 6.2 years, respectively. MAIN OUTCOME MEASURES: Recovery, diabetes, pancreatic exocrine dysfunction, neurodevelopment. RESULTS: The overall incidence of CHI (n = 238) was 1:11 300 live births (1972-2015). From 2000 to 2015, the incidence of P-CHI (n = 69) was 1:13 500 and of T-CHI (n = 127) 1:7400 live births. In the 21st century P-CHI group, hyperinsulinemic medication was initiated and normoglycemia achieved faster relative to earlier. Of the 74 medically treated P-CHI patients, 68% had discontinued medication. Thirteen (12%) P-CHI patients had partial pancreatic resection and 19 (18%) underwent near-total pancreatectomy. Of these, 0% and 84% developed diabetes and 23% and 58% had clinical pancreatic exocrine dysfunction, respectively. Mild neurological difficulties (21% vs 16%, respectively) and intellectual disability (9% vs 5%, respectively) were as common in the P-CHI and T-CHI groups. However, the 21st century P-CHI patients had significantly more frequent normal neurodevelopment and significantly more infrequent diabetes and pancreatic exocrine dysfunction compared with those diagnosed earlier. CONCLUSIONS: Our results demonstrated improved treatment and long-term outcome in the 21st century P-CHI patients relative to earlier.


Asunto(s)
Hiperinsulinismo Congénito/epidemiología , Hiperinsulinismo Congénito/terapia , Adolescente , Adulto , Niño , Preescolar , Femenino , Finlandia , Edad Gestacional , Humanos , Lactante , Masculino , Estudios Retrospectivos , Resultado del Tratamiento , Adulto Joven
13.
Viruses ; 12(9)2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32867368

RESUMEN

CRISPR activation (CRISPRa) has revealed great potential as a tool to modulate the expression of targeted cellular genes. Here, we successfully applied the CRISPRa system to trigger the Kaposi's sarcoma-associated herpesvirus (KSHV) reactivation in latently infected cells by selectively activating ORF50 gene directly from the virus genome. We found that a nuclease-deficient Cas9 (dCas9) fused to a destabilization domain (DD) and 12 copies of the VP16 activation domain (VP192) triggered a more efficient KSHV lytic cycle and virus production when guided to two different sites on the ORF50 promoter, instead of only a single site. To our surprise, the virus reactivation induced by binding of the stable DD-dCas9-VP192 on the ORF50 promoter was even more efficient than reactivation induced by ectopic expression of ORF50. This suggests that recruitment of additional transcriptional activators to the ORF50 promoter, in addition to ORF50 itself, are needed for the efficient virus production. Further, we show that CRISPRa can be applied to selectively express the early lytic gene, ORF57, without disturbing the viral latency. Therefore, CRISPRa-based systems can be utilized to facilitate virus-host interaction studies by controlling the expression of not only cellular but also of specific KSHV genes.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Herpesvirus Humano 8/fisiología , Proteínas Inmediatas-Precoces/genética , Sarcoma de Kaposi/virología , Transactivadores/genética , Proteína 9 Asociada a CRISPR/química , Proteína 9 Asociada a CRISPR/genética , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/genética , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Regiones Promotoras Genéticas , Dominios Proteicos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transactivadores/metabolismo , Activación Transcripcional , Activación Viral , Latencia del Virus
14.
J Clin Endocrinol Metab ; 105(4)2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32170320

RESUMEN

CONTEXT: Major advances have been made in the genetics and classification of congenital hyperinsulinism (CHI). OBJECTIVE: To examine the genetics and clinical characteristics of patients with persistent and transient CHI. DESIGN: A cross-sectional study with the register data and targeted sequencing of 104 genes affecting glucose metabolism. PATIENTS: Genetic and phenotypic data were collected from 153 patients with persistent (n = 95) and transient (n = 58) CHI diagnosed between 1972 and 2015. Of these, 86 patients with persistent and 58 with transient CHI participated in the analysis of the selected 104 genes affecting glucose metabolism, including 10 CHI-associated genes, and 9 patients with persistent CHI were included because of their previously confirmed genetic diagnosis. MAIN OUTCOME MEASURES: Targeted next-generation sequencing results and genotype-phenotype associations. RESULTS: Five novel and 21 previously reported pathogenic or likely pathogenic variants in ABCC8, KCNJ11, GLUD1, GCK, HNF4A, and SLC16A1 genes were found in 68% (n = 65) and 0% of the patients with persistent and transient CHI, respectively. KATP channel mutations explained 82% of the mutation positive cases. CONCLUSIONS: The genetic variants found in this nationwide CHI cohort are in agreement with previous studies, mutations in the KATP channel genes being the major causes of the disease. Pathogenic CHI-associated variants were not identified in patients who were both diazoxide responsive and able to discontinue medication within the first 4 months. Therefore, our results support the notion that genetic testing should be focused on patients with inadequate response or prolonged need for medication.


Asunto(s)
Biomarcadores/análisis , Hiperinsulinismo Congénito/patología , Estudios de Asociación Genética , Mutación , Niño , Preescolar , Hiperinsulinismo Congénito/epidemiología , Hiperinsulinismo Congénito/genética , Estudios Transversales , Femenino , Finlandia , Estudios de Seguimiento , Quinasas del Centro Germinal/genética , Glutamato Deshidrogenasa/genética , Factor Nuclear 4 del Hepatocito/genética , Humanos , Lactante , Recién Nacido , Masculino , Transportadores de Ácidos Monocarboxílicos/genética , Canales de Potasio de Rectificación Interna/genética , Pronóstico , Estudios Retrospectivos , Receptores de Sulfonilureas/genética , Simportadores/genética
15.
Ups J Med Sci ; 124(4): 228-237, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31623497

RESUMEN

Background: Murine boundary cap-derived neural crest stem cells (NCSCs) are capable of enhancing islet function by stimulating beta cell proliferation as well as increasing the neural and vascular density in the islets both in vitro and in vivo. This study aimed to isolate NCSC-like cells from human bone marrow.Methods: CD271 magnetic cell separation and culture techniques were used to purify a NCSC-enriched population of human bone marrow. Analyses of the CD271+ and CD271- fractions in terms of protein expression were performed, and the capacity of the CD271+ bone marrow cells to form 3-dimensional spheres when grown under non-adherent conditions was also investigated. Moreover, the NCSC characteristics of the CD271+ cells were evaluated by their ability to migrate toward human islets as well as human islet-like cell clusters (ICC) derived from pluripotent stem cells.Results: The CD271+ bone marrow population fulfilled the criterion of being multipotent stem cells, having the potential to differentiate into glial cells, neurons as well as myofibroblasts in vitro. They had the capacity to form 3-dimensional spheres as well as an ability to migrate toward human islets, further supporting their NCSC identity. Additionally, we demonstrated similar migration features toward stem cell-derived ICC.Conclusion: The results support the NCSC identity of the CD271-enriched human bone marrow population. It remains to investigate whether the human bone marrow-derived NCSCs have the ability to improve transplantation efficacy of not only human islets but stem cell-derived ICC as well.


Asunto(s)
Técnicas de Cultivo de Célula , Separación Celular/métodos , Islotes Pancreáticos/citología , Cresta Neural/citología , Células Madre Pluripotentes/citología , Adapaleno/metabolismo , Adulto , Anciano , Células de la Médula Ósea/citología , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Humanos , Trasplante de Islotes Pancreáticos , Masculino , Persona de Mediana Edad , Adulto Joven
16.
Nat Commun ; 10(1): 620, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30728358

RESUMEN

Elevated MYC expression sensitizes tumor cells to apoptosis but the therapeutic potential of this mechanism remains unclear. We find, in a model of MYC-driven breast cancer, that pharmacological activation of AMPK strongly synergizes with BCL-2/BCL-XL inhibitors to activate apoptosis. We demonstrate the translational potential of an AMPK and BCL-2/BCL-XL co-targeting strategy in ex vivo and in vivo models of MYC-high breast cancer. Metformin combined with navitoclax or venetoclax efficiently inhibited tumor growth, conferred survival benefits and induced tumor infiltration by immune cells. However, withdrawal of the drugs allowed tumor re-growth with presentation of PD-1+/CD8+ T cell infiltrates, suggesting immune escape. A two-step treatment regimen, beginning with neoadjuvant metformin+venetoclax to induce apoptosis and followed by adjuvant metformin+venetoclax+anti-PD-1 treatment to overcome immune escape, led to durable antitumor responses even after drug withdrawal. We demonstrate that pharmacological reactivation of MYC-dependent apoptosis is a powerful antitumor strategy involving both tumor cell depletion and immunosurveillance.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Genes myc/efectos de los fármacos , Inmunoterapia , Compuestos de Anilina/farmacología , Animales , Anticuerpos Monoclonales Humanizados , Apoptosis/inmunología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Linfocitos T CD8-positivos , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Combinación de Medicamentos , Femenino , Células HEK293 , Xenoinjertos , Humanos , Metformina/farmacología , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-bcl-2 , Sulfonamidas/farmacología , Proteína bcl-X
17.
Elife ; 72018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30412052

RESUMEN

Insulin gene mutations are a leading cause of neonatal diabetes. They can lead to proinsulin misfolding and its retention in endoplasmic reticulum (ER). This results in increased ER-stress suggested to trigger beta-cell apoptosis. In humans, the mechanisms underlying beta-cell failure remain unclear. Here we show that misfolded proinsulin impairs developing beta-cell proliferation without increasing apoptosis. We generated induced pluripotent stem cells (iPSCs) from people carrying insulin (INS) mutations, engineered isogenic CRISPR-Cas9 mutation-corrected lines and differentiated them to beta-like cells. Single-cell RNA-sequencing analysis showed increased ER-stress and reduced proliferation in INS-mutant beta-like cells compared with corrected controls. Upon transplantation into mice, INS-mutant grafts presented reduced insulin secretion and aggravated ER-stress. Cell size, mTORC1 signaling, and respiratory chain subunits expression were all reduced in INS-mutant beta-like cells, yet apoptosis was not increased at any stage. Our results demonstrate that neonatal diabetes-associated INS-mutations lead to defective beta-cell mass expansion, contributing to diabetes development.


Asunto(s)
Diabetes Mellitus/genética , Estrés del Retículo Endoplásmico/genética , Células Madre Pluripotentes Inducidas/química , Proinsulina/genética , Animales , Apoptosis/genética , Sistemas CRISPR-Cas/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Diabetes Mellitus/patología , Retículo Endoplásmico/genética , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Recién Nacido , Células Secretoras de Insulina/química , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Mutación , Proinsulina/química , Pliegue de Proteína , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de la Célula Individual
18.
Cell Death Dis ; 8(9): e3034, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28880267

RESUMEN

The generation of induced pluripotent stem cells (iPSCs) by somatic cell reprogramming holds great potential for modeling human diseases. However, the reprogramming process remains very inefficient and a better understanding of its basic biology is required. The mesenchymal-to-epithelial transition (MET) has been recognized as a crucial step for the successful reprogramming of fibroblasts into iPSCs. It has been reported that the p53 tumor suppressor gene acts as a barrier of this process, while its homolog p63 acts as an enabling factor. In this regard, the information concerning the role of the third homolog, p73, during cell reprogramming is limited. Here, we derive total Trp73 knockout mouse embryonic fibroblasts, with or without Trp53, and examine their reprogramming capacity. We show that p73 is required for effective reprogramming by the Yamanaka factors, even in the absence of p53. Lack of p73 affects the early stages of reprogramming, impairing the MET and resulting in altered maturation and stabilization phases. Accordingly, the obtained p73-deficient iPSCs have a defective epithelial phenotype and alterations in the expression of pluripotency markers. We demonstrate that p73 deficiency impairs the MET, at least in part, by hindering BMP pathway activation. We report that p73 is a positive modulator of the BMP circuit, enhancing its activation by DNp73 repression of the Smad6 promoter. Collectively, these findings provide mechanistic insight into the MET process, proposing p73 as an enhancer of MET during cellular reprogramming.


Asunto(s)
Proteína Morfogenética Ósea 4/farmacología , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Fosfoproteínas/genética , Transactivadores/genética , Proteína Tumoral p73/genética , Proteína p53 Supresora de Tumor/genética , Animales , Línea Celular , Reprogramación Celular , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fosfoproteínas/deficiencia , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Proteína smad6/genética , Proteína smad6/metabolismo , Transactivadores/deficiencia , Proteína Tumoral p73/deficiencia , Proteína p53 Supresora de Tumor/deficiencia
19.
Transplantation ; 100(7): e25-31, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27082827

RESUMEN

The International Pancreas and Islet Transplant Association (IPITA), in conjunction with the Transplantation Society (TTS), convened a workshop to consider the future of pancreas and islet transplantation in the context of potential competing technologies that are under development, including the artificial pancreas, transplantation tolerance, xenotransplantation, encapsulation, stem cell derived beta cells, beta cell proliferation, and endogenous regeneration. Separate workgroups for each topic and then the collective group reviewed the state of the art, hurdles to application, and proposed research agenda for each therapy that would allow widespread application. Herein we present the executive summary of this workshop that focuses on obstacles to application and the research agenda to overcome them; the full length article with detailed background for each topic is published as an online supplement to Transplantation.


Asunto(s)
Células Secretoras de Insulina/citología , Trasplante de Islotes Pancreáticos/métodos , Trasplante de Páncreas/métodos , Animales , Proliferación Celular , Congresos como Asunto , Diabetes Mellitus Tipo 1/terapia , Humanos , Tolerancia Inmunológica , Insulina/administración & dosificación , Islotes Pancreáticos/metabolismo , Páncreas/metabolismo , Regeneración , Sociedades Médicas , Porcinos , Trasplante Heterólogo/métodos , Trasplante Homólogo , Estados Unidos
20.
Ups J Med Sci ; 121(2): 146-50, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27007444

RESUMEN

Isolated human islets are a rare and precious material for diabetes research. However, their availability is limited, and it is impossible to obtain them from patients with specific genotypes. Human pluripotent stem cells provide an alternative. Induced pluripotent stem cells can be generated from any individual's somatic cells and differentiated into pancreatic cells. Currently, this approach is limited by the immaturity of the islet-like cells derived from stem cells. However, this approach can already be used to model developmental defects, and the possibilities for studying insulin secretion are continuously improving. In addition, genome editing using the CRISPR/Cas9 technology provides powerful possibilities to study the impact of specific genotypes. The same technology can also be used for transcriptional regulation in order to improve the functional maturation of stem cell-derived islets. These tools are today becoming available for tomorrow's translational diabetes research.


Asunto(s)
Reprogramación Celular , Diabetes Mellitus Tipo 1/genética , Edición Génica , Islotes Pancreáticos/metabolismo , Páncreas/metabolismo , Animales , Investigación Biomédica , Sistemas CRISPR-Cas , Diferenciación Celular , Diabetes Mellitus Experimental/genética , Genotipo , Humanos , Insulina/metabolismo , Células Madre Pluripotentes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA