Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell ; 159(2): 428-39, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25303535

RESUMEN

The generation of insulin-producing pancreatic ß cells from stem cells in vitro would provide an unprecedented cell source for drug discovery and cell transplantation therapy in diabetes. However, insulin-producing cells previously generated from human pluripotent stem cells (hPSC) lack many functional characteristics of bona fide ß cells. Here, we report a scalable differentiation protocol that can generate hundreds of millions of glucose-responsive ß cells from hPSC in vitro. These stem-cell-derived ß cells (SC-ß) express markers found in mature ß cells, flux Ca(2+) in response to glucose, package insulin into secretory granules, and secrete quantities of insulin comparable to adult ß cells in response to multiple sequential glucose challenges in vitro. Furthermore, these cells secrete human insulin into the serum of mice shortly after transplantation in a glucose-regulated manner, and transplantation of these cells ameliorates hyperglycemia in diabetic mice.


Asunto(s)
Técnicas de Cultivo de Célula , Células Secretoras de Insulina/citología , Animales , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Insulina/genética , Insulina/metabolismo , Islotes Pancreáticos , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
2.
Nature ; 569(7756): 368-373, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31068696

RESUMEN

In vitro differentiation of human stem cells can produce pancreatic ß-cells; the loss of this insulin-secreting cell type underlies type 1 diabetes. Here, as a step towards understanding this differentiation process, we report the transcriptional profiling of more than 100,000 human cells undergoing in vitro ß-cell differentiation, and describe the cells that emerged. We resolve populations that correspond to ß-cells, α-like poly-hormonal cells, non-endocrine cells that resemble pancreatic exocrine cells and a previously unreported population that resembles enterochromaffin cells. We show that endocrine cells maintain their identity in culture in the absence of exogenous growth factors, and that changes in gene expression associated with in vivo ß-cell maturation are recapitulated in vitro. We implement a scalable re-aggregation technique to deplete non-endocrine cells and identify CD49a (also known as ITGA1) as a surface marker of the ß-cell population, which allows magnetic sorting to a purity of 80%. Finally, we use a high-resolution sequencing time course to characterize gene-expression dynamics during the induction of human pancreatic endocrine cells, from which we develop a lineage model of in vitro ß-cell differentiation. This study provides a perspective on human stem-cell differentiation, and will guide future endeavours that focus on the differentiation of pancreatic islet cells, and their applications in regenerative medicine.


Asunto(s)
Diferenciación Celular , Células Secretoras de Insulina/citología , Células Madre/citología , Animales , Biomarcadores/metabolismo , Linaje de la Célula , Separación Celular , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/clasificación , Células Secretoras de Insulina/metabolismo , Integrina alfa1/metabolismo , Masculino , Ratones , RNA-Seq , Análisis de la Célula Individual , Células Madre/metabolismo
3.
Langmuir ; 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38318799

RESUMEN

There is a critical need for sorting complex materials, such as pancreatic islets of Langerhans, exocrine acinar tissues, and embryoid bodies. These materials are cell clusters, which have highly heterogeneous physical properties (such as size, shape, morphology, and deformability). Selecting such materials on the basis of specific properties can improve clinical outcomes and help advance biomedical research. In this work, we focused on sorting one such complex material, human stem cell-derived ß cell clusters (SC-ß cell clusters), by size. For this purpose, we developed a microfluidic device in which an image detection system was coupled to an actuation mechanism based on traveling surface acoustic waves (TSAWs). SC-ß cell clusters of varying size (∼100-500 µm in diameter) were passed through the sorting device. Inside the device, the size of each cluster was estimated from their bright-field images. After size identification, larger clusters, relative to the cutoff size for separation, were selectively actuated using TSAW pulses. As a result of this selective actuation, smaller and larger clusters exited the device from different outlets. At the current sample dilutions, the experimental sorting efficiency ranged between 78% and 90% for a separation cutoff size of 250 µm, yielding sorting throughputs of up to 0.2 SC-ß cell clusters/s using our proof-of-concept design. The biocompatibility of this sorting technique was also established, as no difference in SC-ß cell cluster viability due to TSAW pulse usage was found. We conclude the proof-of-concept sorting work by discussing a few ways to optimize sorting of SC-ß cell clusters for potentially higher sorting efficiency and throughput. This sorting technique can potentially help in achieving a better distribution of islets for clinical islet transplantation (a potential cure for type 1 diabetes). Additionally, the use of this technique for sorting islets can help in characterizing islet biophysical properties by size and selecting suitable islets for improved islet cryopreservation.

4.
Proc Natl Acad Sci U S A ; 112(50): 15498-503, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26621734

RESUMEN

Type 2 diabetes is characterized by a reduction in insulin function and an increase in glucagon activity that together result in hyperglycemia. Glucagon receptor antagonists have been developed as drugs for diabetes; however, they often increase glucagon plasma levels and induce the proliferation of glucagon-secreting α-cells. We find that the secreted protein Angiopoietin-like 4 (Angptl4) is up-regulated via Pparγ activation in white adipose tissue and plasma following an acute treatment with a glucagon receptor antagonist. Induction of adipose angptl4 and Angptl4 supplementation promote α-cell proliferation specifically. Finally, glucagon receptor antagonist improves glycemia in diet-induced obese angptl4 knockout mice without increasing glucagon levels or α-cell proliferation, underscoring the importance of this protein. Overall, we demonstrate that triglyceride metabolism in adipose tissue regulates α-cells in the endocrine pancreas.


Asunto(s)
Tejido Adiposo/metabolismo , Angiopoyetinas/metabolismo , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Receptores de Glucagón/antagonistas & inhibidores , Triglicéridos/metabolismo , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/sangre , Animales , Restricción Calórica , Proliferación Celular , Regulación de la Expresión Génica , Glucagón/sangre , Ratones Endogámicos C57BL , Ratones SCID , PPAR gamma/agonistas , PPAR gamma/metabolismo , Receptores de Glucagón/metabolismo
5.
Mol Aspects Med ; 91: 101138, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36050142

RESUMEN

Regenerative medicine as a field has emerged as a new component of modern medicine and medical research that encompasses a wide range of products including cellular and acellular therapies. As this new field emerged, regulatory agencies like the Food and Drug Administration (FDA) rapidly adapted existing regulatory frameworks to address the transplantation, gene therapy, cell-based therapeutics, and acellular biologics that fall under the broader regenerative medicine umbrella. Where it has not been possible to modify existing regulation and processes, entirely new frameworks have been generated with the intention of providing flexible, forward-facing systems to regulate this rapidly growing field. This review discusses the current state of FDA regulatory affairs in the context of stem cells and extracellular vesicles by highlighting gaps in the current regulatory system and then discussing where regulatory science in regenerative medicine may be headed based on these gaps and the FDA's historical ability to deal with emerging fields. Lastly, we utilize case studies in stem cell and acellular based treatments to demonstrate how regulatory science has evolved in regenerative medicine and highlight the ongoing clinical efforts and challenges of these therapies.


Asunto(s)
Investigación Biomédica , Vesículas Extracelulares , Estados Unidos , Humanos , Medicina Regenerativa , United States Food and Drug Administration , Células Madre
6.
Cells ; 12(15)2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37566061

RESUMEN

Human pluripotent stem cells (hPSCs) are capable of unlimited proliferation and can undergo differentiation to give rise to cells and tissues of the three primary germ layers. While directing lineage selection of hPSCs has been an active area of research, improving the efficiency of differentiation remains an important objective. In this study, we describe a two-compartment microfluidic device for co-cultivation of adult human hepatocytes and stem cells. Both cell types were cultured in a 3D or spheroid format. Adult hepatocytes remained highly functional in the microfluidic device over the course of 4 weeks and served as a source of instructive paracrine cues to drive hepatic differentiation of stem cells cultured in the neighboring compartment. The differentiation of stem cells was more pronounced in microfluidic co-cultures compared to a standard hepatic differentiation protocol. In addition to improving stem cell differentiation outcomes, the microfluidic co-culture system described here may be used for parsing signals and mechanisms controlling hepatic cell fate.


Asunto(s)
Microfluídica , Células Madre Pluripotentes , Humanos , Técnicas de Cocultivo , Microfluídica/métodos , Hepatocitos/metabolismo , Diferenciación Celular
7.
Mol Pharm ; 9(5): 1425-34, 2012 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-22486564

RESUMEN

The evasion of apoptosis is a key characteristic of cancer, and thus strategies to selectively induce apoptosis in cancer cells hold considerable promise in personalized anticancer therapy. Structurally similar procaspase activating compounds PAC-1 and S-PAC-1 restore procaspase-3 activity through the chelation of inhibitory zinc ions in vitro, induce apoptotic death of cancer cells in culture, and reduce tumor burden in vivo. Ip or iv administrations of high doses of PAC-1 are transiently neurotoxic in vivo, while S-PAC-1 is safe even at very high doses and has been evaluated in a phase I clinical trial of pet dogs with spontaneously occurring lymphoma. Here we show that PAC-1 and S-PAC-1 have similar mechanisms of cell death induction at low concentrations (less than 50 µM), but at high concentrations PAC-1 displays unique cell death induction features. Cells treated with a high concentration of PAC-1 have a distinctive gene expression profile, unusual cellular and mitochondrial morphology, and an altered intracellular Ca(2+) concentration, indicative of endoplasmic reticulum (ER) stress-induced apoptosis. These studies suggest strategies for anticancer clinical development, specifically bolus dosing for PAC-1 and continuous rate infusion for S-PAC-1.


Asunto(s)
Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Calcio/metabolismo , Línea Celular Tumoral , Perros , Femenino , Células HL-60 , Células HeLa , Humanos , Hidrazonas/farmacología , Ratones , Microscopía Confocal , Microscopía Electrónica de Transmisión , Piperazinas/farmacología , Zinc/metabolismo
8.
Nat Med ; 28(4): 798-808, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35288694

RESUMEN

Pancreatic islet transplantation can cure diabetes but requires accessible, high-quality islets in sufficient quantities. Cryopreservation could solve islet supply chain challenges by enabling quality-controlled banking and pooling of donor islets. Unfortunately, cryopreservation has not succeeded in this objective, as it must simultaneously provide high recovery, viability, function and scalability. Here, we achieve this goal in mouse, porcine, human and human stem cell (SC)-derived beta cell (SC-beta) islets by comprehensive optimization of cryoprotectant agent (CPA) composition, CPA loading and unloading conditions and methods for vitrification and rewarming (VR). Post-VR islet viability, relative to control, was 90.5% for mouse, 92.1% for SC-beta, 87.2% for porcine and 87.4% for human islets, and it remained unchanged for at least 9 months of cryogenic storage. VR islets had normal macroscopic, microscopic, and ultrastructural morphology. Mitochondrial membrane potential and adenosine triphosphate (ATP) levels were slightly reduced, but all other measures of cellular respiration, including oxygen consumption rate (OCR) to produce ATP, were unchanged. VR islets had normal glucose-stimulated insulin secretion (GSIS) function in vitro and in vivo. Porcine and SC-beta islets made insulin in xenotransplant models, and mouse islets tested in a marginal mass syngeneic transplant model cured diabetes in 92% of recipients within 24-48 h after transplant. Excellent glycemic control was seen for 150 days. Finally, our approach processed 2,500 islets with >95% islets recovery at >89% post-thaw viability and can readily be scaled up for higher throughput. These results suggest that cryopreservation can now be used to supply needed islets for improved transplantation outcomes that cure diabetes.


Asunto(s)
Diabetes Mellitus , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Adenosina Trifosfato/metabolismo , Animales , Criopreservación/métodos , Crioprotectores/metabolismo , Crioprotectores/farmacología , Diabetes Mellitus/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Porcinos , Vitrificación
9.
Bioact Mater ; 15: 1-14, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35386345

RESUMEN

Human pluripotent stem cells (hPSC) hold considerable promise as a source of adult cells for treatment of diseases ranging from diabetes to liver failure. Some of the challenges that limit the clinical/translational impact of hPSCs are high cost and difficulty in scaling-up of existing differentiation protocols. In this paper, we sought to address these challenges through the development of bioactive microcapsules. A co-axial flow focusing microfluidic device was used to encapsulate hPSCs in microcapsules comprised of an aqueous core and a hydrogel shell. Importantly, the shell contained heparin moieties for growth factor (GF) binding and release. The aqueous core enabled rapid aggregation of hPSCs into 3D spheroids while the bioactive hydrogel shell was used to load inductive cues driving pluripotency maintenance and endodermal differentiation. Specifically, we demonstrated that one-time, 1 h long loading of pluripotency signals, fibroblast growth factor (FGF)-2 and transforming growth factor (TGF)-ß1, into bioactive microcapsules was sufficient to induce and maintain pluripotency of hPSCs over the course of 5 days at levels similar to or better than a standard protocol with soluble GFs. Furthermore, stem cell-carrying microcapsules that previously contained pluripotency signals could be reloaded with an endodermal cue, Nodal, resulting in higher levels of endodermal markers compared to stem cells differentiated in a standard protocol. Overall, bioactive heparin-containing core-shell microcapsules decreased GF usage five-fold while improving stem cell phenotype and are well suited for 3D cultivation of hPSCs.

10.
Invest New Drugs ; 29(5): 901-11, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20499133

RESUMEN

PAC-1 is a preferential small molecule activator of procaspase-3 and has potential to become a novel and effective anticancer agent. The rational development of PAC-1 for translational oncologic applications would be advanced by coupling relevant in vitro cytotoxicity studies with pharmacokinetic investigations conducted in large mammalian models possessing similar metabolism and physiology as people. In the present study, we investigated whether concentrations and exposure durations of PAC-1 that induce cytotoxicity in lymphoma cell lines in vitro can be achievable in healthy dogs through a constant rate infusion (CRI) intravenous delivery strategy. Time- and dose-dependent procaspase-3 activation by PAC-1 with subsequent cytotoxicity was determined in a panel of B-cell lymphoma cells in vitro. The pharmacokinetics of PAC-1 administered orally or intravenously was studied in 6 healthy dogs using a crossover design. The feasibility of maintaining steady state plasma concentration of PAC-1 for 24 or 48 h that paralleled in vitro cytotoxic concentrations was investigated in 4 healthy dogs. In vitro, PAC-1 induced apoptosis in lymphoma cell lines in a time- and dose-dependent manner. The oral bioavailability of PAC-1 was relatively low and highly variable (17.8 ± 9.5%). The achievement and maintenance of predicted PAC-1 cytotoxic concentrations in normal dogs was safely attained via intravenous CRI lasting for 24 or 48 h in duration. Using the dog as a large mammalian model, PAC-1 can be safely administered as an intravenous CRI while achieving predicted in vitro cytotoxic concentrations.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Caspasa 3/metabolismo , Activadores de Enzimas/farmacocinética , Salud , Hidrazonas/farmacocinética , Piperazinas/farmacocinética , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/farmacocinética , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Extractos Celulares , Línea Celular Tumoral , Perros , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Activadores de Enzimas/administración & dosificación , Activadores de Enzimas/efectos adversos , Activadores de Enzimas/farmacología , Humanos , Hidrazonas/administración & dosificación , Hidrazonas/efectos adversos , Hidrazonas/farmacología , Piperazinas/administración & dosificación , Piperazinas/efectos adversos , Piperazinas/farmacología , Bibliotecas de Moléculas Pequeñas/efectos adversos , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Tiempo
11.
Sci Rep ; 11(1): 7177, 2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33785778

RESUMEN

Cellular therapies based on human pluripotent stem cells (hPSCs) offer considerable promise for treating numerous diseases including diabetes and end stage liver failure. Stem cell spheroids may be cultured in stirred bioreactors to scale up cell production to cell numbers relevant for use in humans. Despite significant progress in bioreactor culture of stem cells, areas for improvement remain. In this study, we demonstrate that microfluidic encapsulation of hPSCs and formation of spheroids. A co-axial droplet microfluidic device was used to fabricate 400 µm diameter capsules with a poly(ethylene glycol) hydrogel shell and an aqueous core. Spheroid formation was demonstrated for three hPSC lines to highlight broad utility of this encapsulation technology. In-capsule differentiation of stem cell spheroids into pancreatic ß-cells in suspension culture was also demonstrated.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes/fisiología , Esferoides Celulares/fisiología , Reactores Biológicos , Cápsulas/química , Técnicas de Cultivo de Célula/instrumentación , Diferenciación Celular , Línea Celular , Supervivencia Celular , Trasplante de Células/métodos , Diabetes Mellitus/terapia , Enfermedad Hepática en Estado Terminal/terapia , Humanos , Hidrogeles/química , Células Secretoras de Insulina/fisiología , Técnicas Analíticas Microfluídicas/instrumentación , Células Madre Pluripotentes/trasplante , Polietilenglicoles/química
12.
Sci Rep ; 10(1): 19374, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33168920

RESUMEN

Small-molecule inhibitors of non-canonical IκB kinases TANK-binding kinase 1 (TBK1) and IκB kinase ε (IKKε) have shown to stimulate ß-cell regeneration in multiple species. Here we demonstrate that TBK1 is predominantly expressed in ß-cells in mammalian islets. Proteomic and transcriptome analyses revealed that genetic silencing of TBK1 increased expression of proteins and genes essential for cell proliferation in INS-1 832/13 rat ß-cells. Conversely, TBK1 overexpression decreased sensitivity of ß-cells to the elevation of cyclic AMP (cAMP) levels and reduced proliferation of ß-cells in a manner dependent on the activity of cAMP-hydrolyzing phosphodiesterase 3 (PDE3). While the mitogenic effect of (E)3-(3-phenylbenzo[c]isoxazol-5-yl)acrylic acid (PIAA) is derived from inhibition of TBK1, PIAA augmented glucose-stimulated insulin secretion (GSIS) and expression of ß-cell differentiation and proliferation markers in human embryonic stem cell (hESC)-derived ß-cells and human islets. TBK1 expression was increased in ß-cells upon diabetogenic insults, including in human type 2 diabetic islets. PIAA enhanced expression of cell cycle control molecules and ß-cell differentiation markers upon diabetogenic challenges, and accelerated restoration of functional ß-cells in streptozotocin (STZ)-induced diabetic mice. Altogether, these data suggest the critical function of TBK1 as a ß-cell autonomous replication barrier and present PIAA as a valid therapeutic strategy augmenting functional ß-cells.


Asunto(s)
Proliferación Celular , Regulación Enzimológica de la Expresión Génica , Células Secretoras de Insulina/enzimología , Proteínas Serina-Treonina Quinasas/biosíntesis , Regeneración , Animales , Línea Celular Tumoral , Silenciador del Gen , Células Madre Embrionarias Humanas/enzimología , Humanos , Insulina/genética , Insulina/metabolismo , Secreción de Insulina , Proteínas Serina-Treonina Quinasas/genética , Ratas
13.
Nat Commun ; 11(1): 2241, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32382023

RESUMEN

The generation of pancreatic cell types from renewable cell sources holds promise for cell replacement therapies for diabetes. Although most effort has focused on generating pancreatic beta cells, considerable evidence indicates that glucagon secreting alpha cells are critically involved in disease progression and proper glucose control. Here we report on the generation of stem cell-derived human pancreatic alpha (SC-alpha) cells from pluripotent stem cells via a transient pre-alpha cell intermediate. These pre-alpha cells exhibit a transcriptional profile similar to mature alpha cells and although they produce proinsulin protein, they do not secrete significant amounts of processed insulin. Compound screening identified a protein kinase c activator that promotes maturation of pre-alpha cells into SC-alpha cells. The resulting SC-alpha cells do not express insulin, share an ultrastructure similar to cadaveric alpha cells, express and secrete glucagon in response to glucose and some glucagon secretagogues, and elevate blood glucose upon transplantation in mice.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Secretoras de Glucagón/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Madre Pluripotentes/citología , Western Blotting , Diferenciación Celular/fisiología , Línea Celular , Electrofisiología , Técnica del Anticuerpo Fluorescente , Humanos , Páncreas/citología
14.
Curr Biol ; 16(13): 1366-72, 2006 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-16824925

RESUMEN

Angioblasts are multipotent progenitor cells that give rise to arteries or veins . Genetic disruption of the gridlock gene perturbs the artery/vein balance, resulting in generation of insufficient numbers of arterial cells . However, within angioblasts the precise biochemical signals that determine the artery/vein cell-fate decision are poorly understood. We have identified by chemical screening two classes of compounds that compensate for a mutation in the gridlock gene . Both target the VEGF signaling pathway and reveal two downstream branches emanating from the VEGF receptor with opposing effects on arterial specification. We show that activation of ERK (p42/44 MAP kinase) is a specific marker of early arterial progenitors and is among the earliest known determinants of arterial specification. In embryos, cells fated to contribute to arteries express high levels of activated ERK, whereas cells fated to contribute to veins do not. Inhibiting the phosphatidylinositol-3 kinase (PI3K) branch with GS4898 or known PI3K inhibitors, or by expression of a dominant-negative form of AKT promotes arterial specification. Conversely, inhibition of the ERK branch blocks arterial specification, and expression of constitutively active AKT promotes venous specification. In summary, chemical genetic analysis has uncovered unanticipated opposing roles of PI3K and ERK in artery/vein specification.


Asunto(s)
Arterias/embriología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Venas/embriología , Pez Cebra/embriología , Animales , Arterias/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Células Endoteliales/citología , Endotelio Vascular/citología , Mutación , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Venas/citología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
15.
J Chromatogr A ; 1118(1): 100-5, 2006 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-16516902

RESUMEN

Macrocycle-based ion chromatography provides a convenient, reliable method for the determination of perchlorate ion, which is currently of great interest to the environmental community. This study shows that effective perchlorate determinations can be made using standard conductimetric detection by combining an 18-crown-6-based mobile phase with an underivatized reversed-phase mobile phase ion chromatography (MPIC) column. One unique feature of this method is the flexibility in column capacity that is achieved through simple variations in eluent concentrations of 18-crown-6 and KOH, facilitating the separation of target analyte anions such as perchlorate. Using a standard anion exchange column as concentrator makes possible the determination of perchlorate as low as 0.2 ug/L in low ionic strength matrices. Determination of perchlorate at the sub-ug/L level in pure water and in spiked local city hard water samples with high background ion concentrations can be achieved this way. However, like other IC techniques, this method is challenged to achieve analyses at the ug/L level in the demanding high ionic strength matrix described by the United States Environmental Protection Agency (EPA) (1,000 mg/L chloride, sulfate and carbonate). We approached this challenge by use of the Cryptand C1 concentrator column, provided by Dionex Corporation, to effectively preconcentrate perchlorate while reducing background ion concentrations in the high ionic strength matrix. The retention characteristics of the concentrator column were studied in order to maximize its effectiveness for perchlorate determinations. The method makes possible the determination of perchlorate at the 5 ug/L level in the highest ionic strength matrix described by the EPA.


Asunto(s)
Cromatografía por Intercambio Iónico/métodos , Percloratos/análisis , Abastecimiento de Agua/análisis , Cromatografía por Intercambio Iónico/instrumentación , Cromatografía por Intercambio Iónico/normas , Éteres Corona/química , Éteres Cíclicos/química , Percloratos/normas , Reproducibilidad de los Resultados , Bases de Schiff/química , Compuestos de Sodio/análisis , Estados Unidos , United States Environmental Protection Agency
16.
Stem Cell Reports ; 6(6): 993-1008, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27304920

RESUMEN

Human pluripotent stem cells (hPSCs) offer a renewable source of cells that can be expanded indefinitely and differentiated into virtually any type of cell in the human body, including neurons. This opens up unprecedented possibilities to study neuronal cell and developmental biology and cellular pathology of the nervous system, provides a platform for the screening of chemical libraries that affect these processes, and offers a potential source of transplantable cells for regenerative approaches to neurological disease. However, defining protocols that permit a large number and high yield of neurons has proved difficult. We present differentiation protocols for the generation of distinct subtypes of neurons in a highly reproducible manner, with minimal experiment-to-experiment variation. These neurons form synapses with neighboring cells, exhibit spontaneous electrical activity, and respond appropriately to depolarization. hPSC-derived neurons exhibit a high degree of maturation and survive in culture for up to 4-5 months, even without astrocyte feeder layers.


Asunto(s)
Técnicas de Cultivo de Célula , Red Nerviosa/citología , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Biomarcadores/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Diferenciación Celular/efectos de los fármacos , Factor Neurotrófico Ciliar/farmacología , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Red Nerviosa/fisiología , Neurogénesis/genética , Neuronas/clasificación , Neuronas/citología , Neuronas/metabolismo , Variaciones Dependientes del Observador , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reproducibilidad de los Resultados , Proteínas Smad/antagonistas & inhibidores , Proteínas Smad/genética , Proteínas Smad/metabolismo , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
17.
Elife ; 3: e02809, 2014 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25233132

RESUMEN

Dysfunction or death of pancreatic ß cells underlies both types of diabetes. This functional decline begins with ß cell stress and de-differentiation. Current drugs for type 2 diabetes (T2D) lower blood glucose levels but they do not directly alleviate ß cell stress nor prevent, let alone reverse, ß cell de-differentiation. We show here that Urocortin 3 (Ucn3), a marker for mature ß cells, is down-regulated in the early stages of T2D in mice and when ß cells are stressed in vitro. Using an insulin expression-coupled lineage tracer, with Ucn3 as a reporter for the mature ß cell state, we screen for factors that reverse ß cell de-differentiation. We find that a small molecule inhibitor of TGFß receptor I (Alk5) protects cells from the loss of key ß cell transcription factors and restores a mature ß cell identity even after exposure to prolonged and severe diabetes.


Asunto(s)
Desdiferenciación Celular/efectos de los fármacos , Células Secretoras de Insulina/patología , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Biomarcadores/metabolismo , Citocinas/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Humanos , Resistencia a la Insulina , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Estrés Fisiológico/efectos de los fármacos , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Urocortinas/metabolismo
18.
Nat Protoc ; 5(2): 294-302, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20134429

RESUMEN

This protocol describes the gram-scale solution-phase synthesis of the colorimetric caspase-3/7 substrate Ac-DEVD-pNA. The caspase enzymes are integral to cellular inflammation and apoptotic cascades, and are commonly studied by cell biologists, medicinal chemists and chemical biologists. In particular, the assessment of caspase enzymatic activity is a standard method to evaluate cell death pathways and new apoptosis-modulating agents. Caspase enzymatic activity can be conveniently monitored with peptidic chromogenic or fluorogenic substrates, with certain peptide sequences imparting selectivity for certain caspases. The synthesis of these peptide substrates is typically carried out by solid-phase synthesis, a method that is not ideal for production of the gram quantities needed for high-throughput screening. Described herein is a facile method for the synthesis of the Ac-DEVD-pNA caspase-3/7 substrate using solution-phase peptide synthesis. This protocol, involving iterative PyBOP-mediated couplings and Fmoc deprotections, is rapid (about 5 d), operationally simple and can be used to generate over 1 g of product at a fraction of the cost of the commercial substrate.


Asunto(s)
Anilidas/metabolismo , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Oligopéptidos/metabolismo , Anilidas/síntesis química , Compuestos de Anilina/metabolismo , Apoptosis/efectos de los fármacos , Activación Enzimática , Humanos , Hidrólisis , Inflamación/enzimología , Inflamación/patología , Oligopéptidos/síntesis química , Péptidos/síntesis química , Soluciones , Solventes , Especificidad por Sustrato
19.
Cancer Res ; 70(18): 7232-41, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20823163

RESUMEN

A critical event in the apoptotic cascade is the proteolytic activation of procaspases to active caspases. The caspase autoactivating compound PAC-1 induces cancer cell apoptosis and exhibits antitumor activity in murine xenograft models when administered orally as a lipid-based formulation or implanted s.c. as a cholesterol pellet. However, high doses of PAC-1 were found to induce neurotoxicity, prompting us to design and assess a novel PAC-1 derivative called S-PAC-1. Similar to PAC-1, S-PAC-1 activated procaspase-3 and induced cancer cell apoptosis. However, S-PAC-1 did not induce neurotoxicity in mice or dogs. Continuous i.v. infusion of S-PAC-1 in dogs led to a steady-state plasma concentration of ∼10 µmol/L for 24 to 72 hours. In a small efficacy trial of S-PAC-1, evaluation of six pet dogs with lymphoma revealed that S-PAC-1 was well tolerated and that the treatments induced partial tumor regression or stable disease in four of six subjects. Our results support this canine setting for further evaluation of small-molecule procaspase-3 activators, including S-PAC-1, a compound that is an excellent candidate for further clinical evaluation as a novel cancer chemotherapeutic.


Asunto(s)
Enfermedades de los Perros/tratamiento farmacológico , Hidrazonas/farmacología , Linfoma de Células B/veterinaria , Piperazinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Enfermedades de los Perros/enzimología , Enfermedades de los Perros/patología , Perros , Activación Enzimática/efectos de los fármacos , Células HeLa , Humanos , Hidrazonas/farmacocinética , Hidrazonas/toxicidad , Células Jurkat , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/enzimología , Linfoma de Células B/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Piperazinas/farmacocinética , Piperazinas/toxicidad , Células U937
20.
J Mol Biol ; 388(1): 144-58, 2009 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-19281821

RESUMEN

The direct induction of apoptosis has emerged as a powerful anticancer strategy, and small molecules that either inhibit or activate certain proteins in the apoptotic pathway have great potential as novel chemotherapeutic agents. Central to apoptosis is the activation of the zymogen procaspase-3 to caspase-3. Caspase-3 is the key "executioner" caspase, catalyzing the hydrolysis of a multitude of protein substrates within the cell. Interestingly, procaspase-3 levels are often elevated in cancer cells, suggesting a compound that directly stimulates the activation of procaspase-3 to caspase-3 could selectively induce apoptosis in cancer cells. We recently reported the discovery of a compound, PAC-1, which enhances procaspase-3 activity in vitro and induces apoptotic death in cancer cells in culture and in mouse xenograft models. Described herein is the mechanism by which PAC-1 activates procaspase-3 in vitro. We show that zinc inhibits the enzymatic activity of procaspase-3 and that PAC-1 strongly activates procaspase-3 in buffers that contain zinc. PAC-1 and zinc form a tight complex with one another, with a dissociation constant of approximately 42 nM. The combined data indicate that PAC-1 activates procaspase-3 in vitro by sequestering inhibitory zinc ions, thus allowing procaspase-3 to autoactivate itself to caspase-3. The small-molecule-mediated activation of procaspases has great therapeutic potential and thus this discovery of the in vitro mechanism of action of PAC-1 is critical to the development and optimization of other procaspase-activating compounds.


Asunto(s)
Caspasa 3/metabolismo , Fosfatasa 2 de Especificidad Dual/metabolismo , Precursores Enzimáticos/metabolismo , Zinc/metabolismo , Caspasa 7/metabolismo , Inhibidores de Caspasas , Línea Celular Tumoral , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA