Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 150(16)2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37497580

RESUMEN

Earlier data on liver development demonstrated that morphogenesis of the bile duct, portal mesenchyme and hepatic artery is interdependent, yet how this interdependency is orchestrated remains unknown. Here, using 2D and 3D imaging, we first describe how portal mesenchymal cells become organised to form hepatic arteries. Next, we examined intercellular signalling active during portal area development and found that axon guidance genes are dynamically expressed in developing bile ducts and portal mesenchyme. Using tissue-specific gene inactivation in mice, we show that the repulsive guidance molecule BMP co-receptor A (RGMA)/neogenin (NEO1) receptor/ligand pair is dispensable for portal area development, but that deficient roundabout 2 (ROBO2)/SLIT2 signalling in the portal mesenchyme causes reduced maturation of the vascular smooth muscle cells that form the tunica media of the hepatic artery. This arterial anomaly does not impact liver function in homeostatic conditions, but is associated with significant tissular damage following partial hepatectomy. In conclusion, our work identifies new players in development of the liver vasculature in health and liver regeneration.


Asunto(s)
Orientación del Axón , Arteria Hepática , Animales , Ratones , Conductos Biliares , Morfogénesis , Silenciador del Gen
2.
Physiol Rev ; 98(3): 1143-1167, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29717931

RESUMEN

Diabetes mellitus results from disturbed glucose homeostasis due to an absolute (type 1) or relative (type 2) deficiency of insulin, a peptide hormone almost exclusively produced by the beta cells of the endocrine pancreas in a tightly regulated manner. Current therapy only delays disease progression through insulin injection and/or oral medications that increase insulin secretion or sensitivity, decrease hepatic glucose production, or promote glucosuria. These drugs have turned diabetes into a chronic disease as they do not solve the underlying beta cell defects or entirely prevent the long-term complications of hyperglycemia. Beta cell replacement through islet transplantation is a more physiological therapeutic alternative but is severely hampered by donor shortage and immune rejection. A curative strategy should combine newer approaches to immunomodulation with beta cell replacement. Success of this approach depends on the development of practical methods for generating beta cells, either in vitro or in situ through beta cell replication or beta cell differentiation. This review provides an overview of human beta cell generation.


Asunto(s)
Técnicas de Cultivo de Célula , Células Secretoras de Insulina/fisiología , Regeneración , Animales , Homeostasis , Humanos , Células Secretoras de Insulina/trasplante
3.
Gut ; 2021 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-34330784

RESUMEN

OBJECTIVE: The aggressive basal-like molecular subtype of pancreatic ductal adenocarcinoma (PDAC) harbours a ΔNp63 (p40) gene expression signature reminiscent of a basal cell type. Distinct from other epithelia with basal tumours, ΔNp63+ basal cells reportedly do not exist in the normal pancreas. DESIGN: We evaluated ΔNp63 expression in human pancreas, chronic pancreatitis (CP) and PDAC. We further studied in depth the non-cancerous tissue and developed a three-dimensional (3D) imaging protocol (FLIP-IT, Fluorescence Light sheet microscopic Imaging of Paraffin-embedded or Intact Tissue) to study formalin-fixed paraffin-embedded samples at single cell resolution. Pertinent mouse models and HPDE cells were analysed. RESULTS: In normal human pancreas, rare ΔNp63+ cells exist in ducts while their prevalence increases in CP and in a subset of PDAC. In non-cancer tissue, ΔNp63+ cells are atypical KRT19+ duct cells that overall lack SOX9 expression while they do express canonical basal markers and pertain to a niche of cells expressing gastrointestinal stem cell markers. 3D views show that the basal cells anchor on the basal membrane of normal medium to large ducts while in CP they exist in multilayer dome-like structures. In mice, ΔNp63 is not found in adult pancreas nor in selected models of CP or PDAC, but it is induced in organoids from larger Sox9low ducts. In HPDE, ΔNp63 supports a basal cell phenotype at the expense of a classical duct cell differentiation programme. CONCLUSION: In larger human pancreatic ducts, basal cells exist. ΔNp63 suppresses duct cell identity. These cells may play an important role in pancreatic disease, including PDAC ontogeny, but are not present in mouse models.

4.
Diabetologia ; 62(11): 1961-1968, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31414144

RESUMEN

Reciprocal signalling between the endothelium and the pancreatic epithelium is crucial for coordinated differentiation of the embryonic endocrine and exocrine pancreas. In the adult pancreas, islets depend on their dense capillary network to adequately respond to changes in plasma glucose levels. Vascular changes contribute to the onset and progression of both type 1 and type 2 diabetes. Impaired revascularisation of islets transplanted in individuals with type 1 diabetes is linked to islet graft failure and graft loss. This review summarises our understanding of the role of vascular endothelial growth factor-A (VEGF-A) and endothelial cells in beta cell development, physiology and disease. In addition, the therapeutic potential of modulating VEGF-A levels in beta and beta-like cells for transplantation is discussed.


Asunto(s)
Vasos Sanguíneos/metabolismo , Células Endoteliales/citología , Células Secretoras de Insulina/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Glucemia/análisis , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Progresión de la Enfermedad , Homeostasis , Humanos , Células Secretoras de Insulina/trasplante , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos NOD , Neovascularización Fisiológica , Páncreas/metabolismo , Ratas , Regeneración , Transducción de Señal
5.
Diabetologia ; 61(8): 1804-1810, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29789879

RESUMEN

AIMS/HYPOTHESIS: The initial avascular period following islet transplantation seriously compromises graft function and survival. Enhancing graft revascularisation to improve engraftment has been attempted through virus-based delivery of angiogenic triggers, but risks associated with viral vectors have hampered clinical translation. In vitro transcribed mRNA transfection circumvents these risks and may be used for improving islet engraftment. METHODS: Mouse and human pancreatic islet cells were transfected with mRNA encoding the angiogenic growth factor vascular endothelial growth factor A (VEGF-A) before transplantation under the kidney capsule in mice. RESULTS: At day 7 post transplantation, revascularisation of grafts transfected with Vegf-A (also known as Vegfa) mRNA was significantly higher compared with non-transfected or Gfp mRNA-transfected controls in mouse islet grafts (2.11- and 1.87-fold, respectively) (vessel area/graft area, mean ± SEM: 0.118 ± 0.01 [n = 3] in Vegf-A mRNA transfected group (VEGF) vs 0.056 ± 0.01 [n = 3] in no RNA [p < 0.05] vs 0.063 ± 0.02 [n = 4] in Gfp mRNA transfected group (GFP) [p < 0.05]); EndoC-bH3 grafts (2.85- and 2.48-fold. respectively) (0.085 ± 0.02 [n = 4] in VEGF vs 0.030 ± 0.004 [n = 4] in no RNA [p < 0.05] vs 0.034 ± 0.01 [n = 5] in GFP [p < 0.05]); and human islet grafts (3.17- and 3.80-fold, respectively) (0.048 ± 0.013 [n = 3] in VEGF vs 0.015 ± 0.0051 [n = 4] in no RNA [p < 0.01] vs 0.013 ± 0.0046 [n = 4] in GFP [p < 0.01]). At day 30 post transplantation, human islet grafts maintained a vascularisation benefit (1.70- and 1.82-fold, respectively) (0.049 ± 0.0042 [n = 8] in VEGF vs 0.029 ± 0.0052 [n = 5] in no RNA [p < 0.05] vs 0.027 ± 0.0056 [n = 4] in GFP [p < 0.05]) and a higher beta cell volume (1.64- and 2.26-fold, respectively) (0.0292 ± 0.0032 µl [n = 7] in VEGF vs 0.0178 ± 0.0021 µl [n = 5] in no RNA [p < 0.01] vs 0.0129 ± 0.0012 µl [n = 4] in GFP [p < 0.001]). CONCLUSIONS/INTERPRETATION: Vegf-A mRNA transfection before transplantation provides a promising and safe strategy to improve engraftment of islets and other cell-based implants.


Asunto(s)
Células Secretoras de Insulina/citología , Islotes Pancreáticos/citología , Neovascularización Fisiológica , ARN Mensajero/genética , Transfección , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Supervivencia Celular , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/trasplante , Trasplante de Islotes Pancreáticos , Ratones
6.
Diabetologia ; 60(6): 1051-1056, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28299380

RESUMEN

AIMS/HYPOTHESIS: Endothelial-endocrine cell interactions and vascular endothelial growth factor (VEGF)-A signalling are deemed essential for maternal islet vascularisation, glucose control and beta cell expansion during mouse pregnancy. The aim of this study was to assess whether pregnancy-associated beta cell expansion was affected under conditions of islet hypovascularisation. METHODS: Soluble fms-like tyrosine kinase 1 (sFLT1), a VEGF-A decoy receptor, was conditionally overexpressed in maternal mouse beta cells from 1.5 to 14.5 days post coitum. Islet vascularisation, glycaemic control, beta cell proliferation, individual beta cell size and total beta cell volume were assessed in both pregnant mice and non-pregnant littermates. RESULTS: Conditional overexpression of sFLT1 in beta cells resulted in islet hypovascularisation and glucose intolerance in both pregnant and non-pregnant mice. In contrast to non-pregnant littermates, glucose intolerance in pregnant mice was transient. sFLT1 overexpression did not affect pregnancy-associated changes in beta cell proliferation, individual beta cell size or total beta cell volume. CONCLUSIONS/INTERPRETATION: Reduced intra-islet VEGF-A signalling results in maternal islet hypovascularisation and impaired glycaemic control but does not preclude beta cell expansion during mouse pregnancy.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Animales , Proliferación Celular/genética , Proliferación Celular/fisiología , Tamaño de la Célula , Femenino , Islotes Pancreáticos/metabolismo , Ratones , Embarazo , Ratas , Transducción de Señal/genética , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
7.
Diabetologia ; 59(7): 1474-1479, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27130279

RESUMEN

AIMS/HYPOTHESIS: Pw1 or paternally-expressed gene 3 (Peg3) encodes a zinc finger transcription factor that is widely expressed during mouse embryonic development and later restricted to multiple somatic stem cell lineages in the adult. The aim of the present study was to define Pw1 expression in the embryonic and adult pancreas and investigate its role in the beta cell cycle in Pw1 wild-type and mutant mice. METHODS: We analysed PW1 expression by immunohistochemistry in pancreas of nonpregant and pregnant mice and following injury by partial duct ligation. Its role in the beta cell cycle was studied in vivo using a novel conditional knockout mouse and in vitro by lentivirus-mediated gene knockdown. RESULTS: We showed that PW1 is expressed in early pancreatic progenitors at E9.5 but becomes progressively restricted to fully differentiated beta cells as they become established after birth and withdraw from the cell cycle. Notably, PW1 expression declines when beta cells are induced to proliferate and loss of PW1 function activates the beta cell cycle. CONCLUSIONS/INTERPRETATION: These results indicate that PW1 is a co-regulator of the beta cell cycle and can thus be considered a novel therapeutic target in diabetes.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Páncreas/metabolismo , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Inmunohistoquímica , Células Secretoras de Insulina/citología , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Páncreas/embriología
8.
Diabetologia ; 59(9): 1834-7, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27053238

RESUMEN

The generation of beta(-like) cells to compensate for their absolute or relative shortage in type 1 and type 2 diabetes is an obvious therapeutic strategy. Patients first received grafts of donor islet cells over 25 years ago, but this procedure has not become routine in clinical practice because of a donor cell shortage and (auto)immune problems. Transplantation of differentiated embryonic and induced pluripotent stem cells may overcome some but not all the current limitations. Reprogramming exocrine cells towards functional beta(-like) cells would offer an alternative abundant and autologous source of beta(-like) cells. This review focuses on work by our research group towards achieving such a source of cells. It summarises a presentation given at the 'Can we make a better beta cell?' symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Amin Ardestani and Kathrin Maedler, DOI: 10.1007/s00125-016-3892-9 , and by Heiko Lickert and colleagues, DOI: 10.1007/s00125-016-3949-9 ) and a commentary by the Session Chair, Shanta Persaud (DOI: 10.1007/s00125-016-3870-2 ).


Asunto(s)
Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Páncreas/citología , Animales , Diferenciación Celular/fisiología , Humanos , Macrófagos/metabolismo , Factores de Transcripción/metabolismo
9.
Eur J Immunol ; 45(5): 1482-93, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25645754

RESUMEN

Pancreas injury by partial duct ligation (PDL) activates a healing response, encompassing ß-cell neogenesis and proliferation. Macrophages (MΦs) were recently shown to promote ß-cell proliferation after PDL, but they remain poorly characterized. We assessed myeloid cell diversity and the factors driving myeloid cell dynamics following acute pancreas injury by PDL. In naive and sham-operated pancreas, the myeloid cell compartment consisted mainly of two distinct tissue-resident MΦ types, designated MHC-II(lo) and MHC-II(hi) MΦs, the latter being predominant. MHC-II(lo) and MHC-II(hi) pancreas MΦs differed at the molecular level, with MHC-II(lo) MΦs being more M2-activated. After PDL, there was an early surge of Ly6C(hi) monocyte infiltration in the pancreas, followed by a transient MHC-II(lo) MΦ peak and ultimately a restoration of the MHC-II(hi) MΦ-dominated steady-state equilibrium. These intricate MΦ dynamics in PDL pancreas depended on monocyte recruitment by C-C chemokine receptor 2 and macrophage-colony stimulating factor receptor as well as on macrophage-colony stimulating factor receptor-dependent local MΦ proliferation. Functionally, MHC-II(lo) MΦs were more angiogenic. We further demonstrated that, at least in C-C chemokine receptor 2-KO mice, tissue MΦs, rather than Ly6C(hi) monocyte-derived MΦs, contributed to ß-cell proliferation. Together, our study fully characterizes the MΦ subsets in the pancreas and clarifies the complex dynamics of MΦs after PDL injury.


Asunto(s)
Macrófagos/inmunología , Macrófagos/patología , Monocitos/inmunología , Monocitos/patología , Páncreas/inmunología , Páncreas/lesiones , Animales , Antígenos Ly/metabolismo , Movimiento Celular/inmunología , Proliferación Celular , Microambiente Celular/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Ligadura , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/clasificación , Células Mieloides/inmunología , Células Mieloides/patología , Páncreas/patología , Conductos Pancreáticos/lesiones , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Regeneración/inmunología
10.
Diabetologia ; 57(1): 140-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24121626

RESUMEN

AIMS/HYPOTHESIS: Vascular endothelial growth factor (VEGF) has been recognised by loss-of-function experiments as a pleiotropic factor with importance in embryonic pancreas development and postnatal beta cell function. Chronic, nonconditional overexpression of VEGF-A has a deleterious effect on beta cell development and function. We report, for the first time, a conditional gain-of-function study to evaluate the effect of transient VEGF-A overexpression by adult pancreatic beta cells on islet vasculature and beta cell proliferation and survival, under both normal physiological and injury conditions. METHODS: In a transgenicmouse strain, overexpressing VEGF-A in a doxycycline-inducible and beta cell-specific manner, we evaluated the ability of VEGF-A to affect islet vessel density, beta cell proliferation and protection of the adult beta cell mass from toxin-induced injury. RESULTS: Short-term VEGF-A overexpression resulted in islet hypervascularisation, increased beta cell proliferation and protection from toxin-mediated beta cell death, and thereby prevented the development of hyperglycaemia. Extended overexpression of VEGF-A led to impaired glucose tolerance, elevated fasting glycaemia and a decreased beta cell mass. CONCLUSIONS/INTERPRETATION: Overexpression of VEGF-A in beta cells time-dependently affects glycometabolic control and beta cell protection and proliferation. These data nourish further studies to examine the role of controlled VEGF delivery in (pre)clinical applications aimed at protecting and/or restoring the injured beta cell mass.


Asunto(s)
Diabetes Mellitus/prevención & control , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Proliferación Celular , Supervivencia Celular/fisiología , Diabetes Mellitus/metabolismo , Islotes Pancreáticos/irrigación sanguínea , Islotes Pancreáticos/metabolismo , Ratones , Ratones Transgénicos , Ratas , Factor A de Crecimiento Endotelial Vascular/genética
11.
Artículo en Inglés | MEDLINE | ID: mdl-38644094

RESUMEN

The pandemic scale of diabetes mellitus is alarming, its complications remain devastating, and current treatments still pose a major burden on those affected and on the healthcare system as a whole. As the disease emanates from the destruction or dysfunction of insulin-producing pancreatic ß-cells, a real cure requires their restoration and protection. An attractive strategy is to regenerate ß-cells directly within the pancreas; however, while several approaches for ß-cell regeneration have been proposed in the past, clinical translation has proven challenging. This review scrutinizes recent findings in ß-cell regeneration and discusses their potential clinical implementation. Hereby, we aim to delineate a path for innovative, targeted therapies to help shift from 'caring for' to 'curing' diabetes.

12.
J Exp Med ; 204(1): 129-39, 2007 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-17227908

RESUMEN

For decades, in vitro expansion of transplantable hematopoietic stem cells (HSCs) has been an elusive goal. Here, we demonstrate that multipotent adult progenitor cells (MAPCs), isolated from green fluorescent protein (GFP)-transgenic mice and expanded in vitro for >40-80 population doublings, are capable of multilineage hematopoietic engraftment of immunodeficient mice. Among MAPC-derived GFP+CD45.2+ cells in the bone marrow of engrafted mice, HSCs were present that could radioprotect and reconstitute multilineage hematopoiesis in secondary and tertiary recipients, as well as myeloid and lymphoid hematopoietic progenitor subsets and functional GFP+ MAPC-derived lymphocytes that were functional. Although hematopoietic contribution by MAPCs was comparable to control KTLS HSCs, approximately 10(3)-fold more MAPCs were required for efficient engraftment. Because GFP+ host-derived CD45.1+ cells were not observed, fusion is not likely to account for the generation of HSCs by MAPCs.


Asunto(s)
Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Multipotentes/trasplante , Animales , Linfocitos B/inmunología , Supervivencia de Injerto , Proteínas Fluorescentes Verdes/genética , Hematopoyesis/inmunología , Sistema Hematopoyético/citología , Técnicas In Vitro , Tejido Linfoide/citología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Células Madre Multipotentes/inmunología , Especificidad de Órganos , Proteínas Recombinantes/genética , Linfocitos T/inmunología
13.
Theranostics ; 13(15): 5483-5500, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37908728

RESUMEN

Rationale: Although promising responses are obtained in patients treated with immune checkpoint inhibitors targeting programmed death ligand 1 (PD-L1) and its receptor programmed death-1 (PD-1), only a fraction of patients benefits from this immunotherapy. Cancer vaccination may be an effective approach to improve the response to immune checkpoint inhibitors anti-PD-L1/PD-1 therapy. However, there is a lack of research on the dynamics of PD-L1 expression in response to cancer vaccination. Methods: We performed non-invasive whole-body imaging to visualize PD-L1 expression at different timepoints after vaccination of melanoma-bearing mice. Mice bearing ovalbumin (OVA) expressing B16 tumors were i.v. injected with the Galsome mRNA vaccine: OVA encoding mRNA lipoplexes co-encapsulating a low or a high dose of the atypical adjuvant α-galactosylceramide (αGC) to activate invariant natural killer T (iNKT) cells. Serial non-invasive whole-body immune imaging was performed using a technetium-99m (99mTc)-labeled anti-PD-L1 nanobody, single-photon emission computerized tomography (SPECT) and X-ray computed tomography (CT) images were quantified. Additionally, cellular expression of PD-L1 was evaluated with flow cytometry. Results: SPECT/CT-imaging showed a rapid and systemic upregulation of PD-L1 after vaccination. PD-L1 expression could not be correlated to the αGC-dose, although we observed a dose-dependent iNKT cell activation. Dynamics of PD-L1 expression were organ-dependent and most pronounced in lungs and liver, organs to which the vaccine was distributed. PD-L1 expression in lungs increased immediately after vaccination and gradually decreased over time, whereas in liver, vaccination-induced PD-L1 upregulation was short-lived. Flow cytometric analysis of these organs further showed myeloid cells as well as non-immune cells with elevated PD-L1 expression in response to vaccination. SPECT/CT imaging of the tumor demonstrated that the expression of PD-L1 remained stable over time and was overall not affected by vaccination although flow cytometric analysis at the cellular level demonstrated changes in PD-L1 expression in various immune cell populations following vaccination. Conclusion: Repeated non-invasive whole-body imaging using 99mTc-labeled anti-PD-L1 nanobodies allows to document the dynamic nature of PD-L1 expression upon vaccination. Galsome vaccination rapidly induced systemic upregulation of PD-L1 expression with the most pronounced upregulation in lungs and liver while flow cytometry analysis showed upregulation of PD-L1 in the tumor microenvironment. This study shows that imaging using nanobodies may be useful for monitoring vaccine-mediated PD-L1 modulation in patients and could provide a rationale for combination therapy. To the best of our knowledge, this is the first report that visualizes PD-L1 expression upon cancer vaccination.


Asunto(s)
Melanoma , Células T Asesinas Naturales , Anticuerpos de Dominio Único , Humanos , Ratones , Animales , Antígeno B7-H1 , Células T Asesinas Naturales/metabolismo , Anticuerpos de Dominio Único/metabolismo , Inhibidores de Puntos de Control Inmunológico/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T CD8-positivos , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X , Vacunas Sintéticas , Melanoma/diagnóstico por imagen , Melanoma/terapia , Microambiente Tumoral , Vacunas de ARNm
14.
Cells ; 10(1)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33477961

RESUMEN

Diabetes mellitus is a pandemic metabolic disorder that results from either the autoimmune destruction or the dysfunction of insulin-producing pancreatic beta cells. A promising cure is beta cell replacement through the transplantation of islets of Langerhans. However, donor shortage hinders the widespread implementation of this therapy. Human pluripotent stem cells, including embryonic stem cells and induced pluripotent stem cells, represent an attractive alternative beta cell source for transplantation. Although major advances over the past two decades have led to the generation of stem cell-derived beta-like cells that share many features with genuine beta cells, producing fully mature beta cells remains challenging. Here, we review the current status of beta cell differentiation protocols and highlight specific challenges that are associated with producing mature beta cells. We address the challenges and opportunities that are offered by monogenic forms of diabetes. Finally, we discuss the remaining hurdles for clinical application of stem cell-derived beta cells and the status of ongoing clinical trials.


Asunto(s)
Diferenciación Celular , Diabetes Mellitus Tipo 1 , Células Madre Embrionarias Humanas , Células Madre Pluripotentes Inducidas , Células Secretoras de Insulina , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/terapia , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/patología , Células Madre Embrionarias Humanas/trasplante , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/trasplante , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/trasplante
15.
Cell Death Differ ; 28(9): 2601-2615, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33762742

RESUMEN

Maintenance of the pancreatic acinar cell phenotype suppresses tumor formation. Hence, repetitive acute or chronic pancreatitis, stress conditions in which the acinar cells dedifferentiate, predispose for cancer formation in the pancreas. Dedifferentiated acinar cells acquire a large panel of duct cell-specific markers. However, it remains unclear to what extent dedifferentiated acini differ from native duct cells and which genes are uniquely regulating acinar cell dedifferentiation. Moreover, most studies have been performed on mice since the availability of human cells is scarce. Here, we applied a non-genetic lineage tracing method of human pancreatic exocrine acinar and duct cells that allowed cell-type-specific gene expression profiling by RNA sequencing. Subsequent to this discovery analysis, one transcription factor that was unique for dedifferentiated acinar cells was functionally characterized. RNA sequencing analysis showed that human dedifferentiated acinar cells expressed genes in "Pathways of cancer" with a prominence of MECOM (EVI-1), a transcription factor that was not expressed by duct cells. During mouse embryonic development, pre-acinar cells also transiently expressed MECOM and in the adult mouse pancreas, MECOM was re-expressed when mice were subjected to acute and chronic pancreatitis, conditions in which acinar cells dedifferentiate. In human cells and in mice, MECOM expression correlated with and was directly regulated by SOX9. Mouse acinar cells that, by genetic manipulation, lose the ability to upregulate MECOM showed impaired cell adhesion, more prominent acinar cell death, and suppressed acinar cell dedifferentiation by limited ERK signaling. In conclusion, we transcriptionally profiled the two major human pancreatic exocrine cell types, acinar and duct cells, during experimental stress conditions. We provide insights that in dedifferentiated acinar cells, cancer pathways are upregulated in which MECOM is a critical regulator that suppresses acinar cell death by permitting cellular dedifferentiation.


Asunto(s)
Células Acinares/metabolismo , Muerte Celular/genética , Proteína del Locus del Complejo MDS1 y EV11/metabolismo , Oncogenes/genética , Animales , Desdiferenciación Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Transducción de Señal
16.
J Cell Biol ; 159(2): 303-12, 2002 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-12403815

RESUMEN

Regulatory proteins have been identified in embryonic development of the endocrine pancreas. It is unknown whether these factors can also play a role in the formation of pancreatic endocrine cells from postnatal nonendocrine cells. The present study demonstrates that adult human pancreatic duct cells can be converted into insulin-expressing cells after ectopic, adenovirus-mediated expression of the class B basic helix-loop-helix factor neurogenin 3 (ngn3), which is a critical factor in embryogenesis of the mouse endocrine pancreas. Infection with adenovirus ngn3 (Adngn3) induced gene and/or protein expression of NeuroD/beta2, Pax4, Nkx2.2, Pax6, and Nkx6.1, all known to be essential for beta-cell differentiation in mouse embryos. Expression of ngn3 in adult human duct cells induced Notch ligands Dll1 and Dll4 and neuroendocrine- and beta-cell-specific markers: it increased the percentage of synaptophysin- and insulin-positive cells 15-fold in ngn3-infected versus control cells. Infection with NeuroD/beta2 (a downstream target of ngn3) induced similar effects. These data indicate that the Delta-Notch pathway, which controls embryonic development of the mouse endocrine pancreas, can also operate in adult human duct cells driving them to a neuroendocrine phenotype with the formation of insulin-expressing cells.


Asunto(s)
Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Proteínas del Tejido Nervioso/genética , Conductos Pancreáticos/citología , Conductos Pancreáticos/fisiología , Adulto , Factores de Edad , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Biomarcadores , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio , Humanos , Insulina/fisiología , Péptidos y Proteínas de Señalización Intracelular , Islotes Pancreáticos/embriología , Proteínas de la Membrana/genética , Proteínas Nucleares , Conductos Pancreáticos/embriología , Fenotipo , Receptores Notch , Factores de Transcripción , Transcripción Genética/fisiología
17.
Liver Transpl ; 14(3): 333-45, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18306374

RESUMEN

Widespread use of liver transplantation in the treatment of hepatic diseases is restricted by the limited availability of donated organs. One potential solution to this problem would be isolation and propagation of liver progenitor cells or stem cells. Here, we report on the isolation of a novel progenitor cell population from unmanipulated (that is, no prior exposure to chemicals and no injury) adult rat liver. Rat liver cells were cultured following a protocol developed in our laboratory to generate a unique progenitor cell population called liver-derived progenitor cells (LDPCs). LDPCs were analyzed by fluorescence-activated cell sorting, real-time polymerase chain reaction (RT-PCR), immunostaining and microarray gene expression. LDPCs were also differentiated into hepatocytes and biliary epithelium in vitro and examined for mature hepatic markers and urea and albumin production. These analyses showed that, LDPCs expressed stem cell markers such as cluster domain (CD)45, CD34, c-kit, and Thy 1, similar to hematopoietic stem cells, as well as endodermal/hepatic markers such as hepatocyte nuclear factor (HNF)3beta, hematopoietically-expressed homeobox gene-1, c-met, and transthyretin. LDPCs were negative for OV-6, cytokeratins (CKs), albumin, and HNF1alpha. The microarray gene expression profile demonstrated that they showed some similarities to known liver progenitor/stem cells such as oval cells. In addition, LDPCs differentiated into functional hepatocytes in vitro as shown by albumin expression and urea production. In conclusion, LDPCs are a population of unique liver progenitors that can be generated from unmanipulated adult liver, which makes them potentially useful for clinical applications, especially for cell transplantation in the treatment of liver diseases.


Asunto(s)
Separación Celular/métodos , Hígado/citología , Células Madre/citología , Células Madre/metabolismo , Albúminas/metabolismo , Animales , Conductos Biliares/citología , Conductos Biliares/metabolismo , Diferenciación Celular , Células Cultivadas , Citometría de Flujo , Hepatocitos/citología , Hepatocitos/metabolismo , Cariotipificación , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/metabolismo , Hígado/metabolismo , Análisis por Micromatrices , Prealbúmina/genética , Prealbúmina/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Urea/metabolismo
18.
PLoS One ; 13(2): e0191249, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29408875

RESUMEN

Pancreas injury by partial duct ligation (PDL) activates beta cell differentiation and proliferation in adult mouse pancreas but remains controversial regarding the anticipated increase in beta cell volume. Several reports unable to show beta cell volume augmentation in PDL pancreas used automated digital image analysis software. We hypothesized that fully automatic beta cell morphometry without manual micrograph artifact remediation introduces bias and therefore might be responsible for reported discrepancies and controversy. However, our present results prove that standard digital image processing with automatic thresholding is sufficiently robust albeit less sensitive and less adequate to demonstrate a significant increase in beta cell volume in PDL versus Sham-operated pancreas. We therefore conclude that other confounding factors such as quality of surgery, selection of samples based on relative abundance of the transcription factor Neurogenin 3 (Ngn3) and tissue processing give rise to inter-laboratory inconsistencies in beta cell volume quantification in PDL pancreas.


Asunto(s)
Automatización , Islotes Pancreáticos/patología , Animales
19.
PLoS One ; 11(5): e0156204, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27243814

RESUMEN

Transcription factor mediated lineage reprogramming of human pancreatic exocrine tissue could conceivably provide an unlimited supply of islets for transplantation in the treatment of diabetes. Exocrine tissue can be efficiently reprogrammed to islet-like cells using a cocktail of transcription factors: Pdx1, Ngn3, MafA and Pax4 in combination with growth factors. We show here that overexpression of exogenous Pax4 in combination with suppression of the endogenous transcription factor ARX considerably enhances the production of functional insulin-secreting ß-like cells with concomitant suppression of α-cells. The efficiency was further increased by culture on laminin-coated plates in media containing low glucose concentrations. Immunocytochemistry revealed that reprogrammed cultures were composed of ~45% islet-like clusters comprising >80% monohormonal insulin+ cells. The resultant ß-like cells expressed insulin protein levels at ~15-30% of that in adult human islets, efficiently processed proinsulin and packaged insulin into secretory granules, exhibited glucose responsive insulin secretion, and had an immediate and prolonged effect in normalising blood glucose levels upon transplantation into diabetic mice. We estimate that approximately 3 billion of these cells would have an immediate therapeutic effect following engraftment in type 1 diabetes patients and that one pancreas would provide sufficient tissue for numerous transplants.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Técnicas de Reprogramación Celular/métodos , Diabetes Mellitus Experimental/terapia , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/trasplante , Factores de Transcripción Paired Box/metabolismo , Páncreas Exocrino/citología , Factores de Transcripción/genética , Adulto , Animales , Células Cultivadas , Femenino , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones SCID , Páncreas Exocrino/metabolismo , Proinsulina/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Estreptozocina , Adulto Joven
20.
Best Pract Res Clin Endocrinol Metab ; 29(6): 849-57, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26696514

RESUMEN

One of the key promises of regenerative medicine is providing a cure for diabetes. Cell-based therapies are proving their safety and efficiency, but donor beta cell shortages and immunological issues remain major hurdles. Reprogramming of human pancreatic exocrine cells towards beta cells would offer a major advantage by providing an abundant and autologous source of beta cells. Over the past decade our understanding of transdifferentiation processes greatly increased allowing us to design reprogramming protocols that fairly aim for clinical trials.


Asunto(s)
Transdiferenciación Celular , Técnicas de Reprogramación Celular/métodos , Células Secretoras de Insulina/citología , Páncreas Exocrino/citología , Animales , Humanos , Células Secretoras de Insulina/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Páncreas Exocrino/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA