Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Med (Lausanne) ; 8: 660877, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33937296

RESUMEN

Islet transplantation can restore glycemic control in patients with type 1 diabetes. Using this procedure, the early stages of engraftment are often crucial to long-term islet function, and outcomes are not always successful. Numerous studies have shown that mesenchymal stem cells (MSCs) facilitate islet graft function. However, experimental data can be inconsistent due to variables associated with MSC generation (including donor characteristics and tissue source), thus, demonstrating the need for a well-characterized and uniform cell product before translation to the clinic. Unlike bone marrow- or adipose tissue-derived MSCs, human embryonic stem cell-derived-MSCs (hESC-MSCs) offer an unlimited source of stable and highly-characterized cells that are easily scalable. Here, we studied the effects of human hemangioblast-derived mesenchymal cells (HMCs), (i.e., MSCs differentiated from hESCs using a hemangioblast intermediate), on islet cell transplantation using a minimal islet mass model. The co-transplantation of the HMCs allowed a mass of islets that was insufficient to correct diabetes on its own to restore glycemic control in all recipients. Our in vitro studies help to elucidate the mechanisms including reduction of cytokine stress by which the HMCs support islet graft protection in vivo. Derivation, stability, and scalability of the HMC source may offer unique advantages for clinical applications, including fewer islets needed for successful islet transplantation.

3.
Nat Rev Drug Discov ; 19(7): 463-479, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32612263

RESUMEN

Naturally occurring stem cells isolated from humans have been used therapeutically for decades. This has primarily involved the transplantation of primary cells such as haematopoietic and mesenchymal stem cells and, more recently, derivatives of pluripotent stem cells. However, the advent of cell-engineering approaches is ushering in a new generation of stem cell-based therapies, greatly expanding their therapeutic utility. These next-generation stem cells are being used as 'Trojan horses' to improve the delivery of drugs and oncolytic viruses to intractable tumours and are also being engineered with angiogenic, neurotrophic and anti-inflammatory molecules to accelerate the repair of injured or diseased tissues. Moreover, gene therapy and gene editing technologies are being used to create stem cell derivatives with improved functionality, specificity and responsiveness compared with their natural counterparts. Here, we review these engineering approaches and areas in which they will help broaden the utility and clinical applicability of stem cells.


Asunto(s)
Ingeniería Celular/métodos , Trasplante de Células Madre/métodos , Células Madre/citología , Animales , Sistemas de Liberación de Medicamentos , Edición Génica , Terapia Genética/métodos , Humanos , Viroterapia Oncolítica/métodos
4.
Ocul Immunol Inflamm ; 26(8): 1228-1236, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28914568

RESUMEN

PURPOSE: We investigated the effect of exogenously administered human embryonic stem cell-derived mesenchymal stromal cells (hESC-MSCs) in experimental autoimmune uveitis (EAU) in B10.RIII mice, a murine model of severe uveitis. METHODS: B10.RIII mice were immunized with an uveitogenic peptide, and intraperitoneal injections of 5 million hESC-MSCs per animal were given on the same day. Behavioral light sensitivity assays, histological evaluation, cytokine production, and regulatory T cells were analyzed at the peak of the disease. RESULTS: Histological and behavioral evidence demonstrated that early systemic treatment with hESC-MSCs decreases the development of severe EAU in B10.RIII mice. hESC-MSCs suppress Th17 and upregulate Th1 and Th2 responses as well as IL-2 and GM-CSF in splenocytes from hESC-MSC-treated mice. CONCLUSIONS: MSCs that originate from hESC decrease the development of severe EAU in B10.RIII mice, likely through systemic immune modulation. Further investigation is needed to determine any potential effect on active EAU.


Asunto(s)
Enfermedades Autoinmunes/prevención & control , Células Madre Embrionarias Humanas/citología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Células del Estroma/fisiología , Uveítis/prevención & control , Animales , Enfermedades Autoinmunes/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Humanos , Ratones , Células TH1/inmunología , Células Th17/inmunología , Células Th2/inmunología , Uveítis/inmunología
5.
Regen Med ; 11(8): 831-847, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27908220

RESUMEN

Pluripotent stem cells (PSCs) can differentiate into virtually any cell type in the body, making them attractive for both regenerative medicine and drug discovery. Over the past 10 years, technological advances and innovative platforms have yielded first-in-man PSC-based clinical trials and opened up new approaches for disease modeling and drug development. Induced PSCs have become the foremost alternative to embryonic stem cells and accelerated the development of disease-in-a-dish models. Over the years and with each new discovery, PSCs have proven to be extremely versatile. This review article highlights key advancements in PSC research, from 2006 to 2016, and how they will guide the direction of the field over the next decade.


Asunto(s)
Células Madre Pluripotentes/citología , Medicina Regenerativa , Trasplante de Células Madre , Ingeniería de Tejidos/métodos , Animales , Humanos
6.
Regen Med ; 11(1): 33-43, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26387424

RESUMEN

AIM: To evaluate the safety and efficacy of intralesional injection of human embryonic stem cell (hESC)-derived mesenchymal stem/stromal cells (MSCs) in canine anal furunculosis dogs. MATERIALS & METHODS: Dogs naturally develop an immune-mediated disease called canine anal furunculosis, which shares many features with human fistulizing Crohn's disease. RESULTS: The hESC-MSCs were well tolerated and 1 month postinjection, accompanied by reduced serum levels of IL-2 and IL-6, two inflammatory cytokines associated with Crohn's disease. All six dogs were found to be completely free of fistulas at 3 months postinjection. However, at 6 months, two dogs had some fistula relapse. CONCLUSION: Results of this study provide the first evidence of the safety and therapeutic potential of hESC-MSCs in a large animal model.


Asunto(s)
Enfermedad de Crohn/terapia , Células Madre Embrionarias Humanas/citología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Fístula Rectal/terapia , Animales , Enfermedad de Crohn/tratamiento farmacológico , Ciclosporina/farmacología , Ciclosporina/uso terapéutico , Citocinas/sangre , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Estudios de Seguimiento , Xenoinjertos , Humanos , Inmunomodulación , Células del Estroma/citología
7.
Sci Rep ; 5: 17685, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26628350

RESUMEN

Adult tissue-derived mesenchymal stromal cells (MSCs) are showing promise in clinical trials for systemic lupus erythematosus (SLE). However, the inability to manufacture large quantities of functional cells from a single donor as well as donor-dependent variability in quality limits their clinical utility. Human embryonic stem cell (hESC)-derived MSCs are an alternative to adult MSCs that can circumvent issues regarding scalability and consistent quality due to their derivation from a renewable starting material. Here, we show that hESC-MSCs prevent the progression of fatal lupus nephritis (LN) in NZB/W F1 (BWF1) mice. Treatment led to statistically significant reductions in proteinuria and serum creatinine and preserved renal architecture. Specifically, hESC-MSC treatment prevented disease-associated interstitial inflammation, protein cast deposition, and infiltration of CD3(+) lymphocytes in the kidneys. This therapy also led to significant reductions in serum levels of tumor necrosis factor alpha (TNFα) and interleukin 6 (IL-6), two inflammatory cytokines associated with SLE. Mechanistically, in vitro data support these findings, as co-culture of hESC-MSCs with lipopolysaccharide (LPS)-stimulated BWF1 lymphocytes decreased lymphocyte secretion of TNFα and IL-6, and enhanced the percentage of putative regulatory T cells. This study represents an important step in the development of a commercially scalable and efficacious cell therapy for SLE/LN.


Asunto(s)
Células Madre Embrionarias Humanas/metabolismo , Riñón , Nefritis Lúpica , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Células Madre Adultas/metabolismo , Animales , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Nefritis Lúpica/metabolismo , Nefritis Lúpica/patología , Nefritis Lúpica/fisiopatología , Nefritis Lúpica/terapia , Ratones
8.
Nat Rev Drug Discov ; 14(10): 681-92, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26391880

RESUMEN

Pluripotent stem cells (PSCs) hold great promise for drug discovery and regenerative medicine owing to their ability to differentiate into any cell type in the body. After more than three decades of research, including delays due to the potential tumorigenicity of PSCs and inefficiencies in differentiation methods, the field is at a turning point, with a number of clinical trials across the globe now testing PSC-derived products in humans. Ocular diseases dominate these first-in-man trials, and Phase l/ll results are showing promising safety data as well as possible efficacy. In addition, the advent of induced PSC (iPSC) technology is enabling the development of a wide range of cell-based disease models from genetically predisposed patients, thereby facilitating drug discovery. In this Review, we discuss the recent progress and remaining challenges for the use of PSCs in regenerative medicine and drug development.


Asunto(s)
Descubrimiento de Drogas , Células Madre Pluripotentes/trasplante , Medicina Regenerativa , Ensayos Clínicos como Asunto , Cardiopatías/terapia , Humanos , Enfermedades Pulmonares/terapia , Degeneración Macular/terapia , Enfermedades Neurodegenerativas/terapia , Epitelio Pigmentado de la Retina/trasplante
10.
Stem Cell Reports ; 3(5): 817-31, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25418726

RESUMEN

Human induced pluripotent stem cells (iPSCs) provide a potentially replenishable source for the production of transfusable platelets. Here, we describe a method to generate megakaryocytes (MKs) and functional platelets from iPSCs in a scalable manner under serum/feeder-free conditions. The method also permits the cryopreservation of MK progenitors, enabling a rapid "surge" capacity when large numbers of platelets are needed. Ultrastructural/morphological analyses show no major differences between iPSC platelets and human blood platelets. iPSC platelets form aggregates, lamellipodia, and filopodia after activation and circulate in macrophage-depleted animals and incorporate into developing mouse thrombi in a manner identical to human platelets. By knocking out the ß2-microglobulin gene, we have generated platelets that are negative for the major histocompatibility antigens. The scalable generation of HLA-ABC-negative platelets from a renewable cell source represents an important step toward generating universal platelets for transfusion as well as a potential strategy for the management of platelet refractoriness.


Asunto(s)
Plaquetas/citología , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Megacariocitos/citología , Animales , Antígenos CD34/metabolismo , Plaquetas/metabolismo , Plaquetas/ultraestructura , Técnicas de Cultivo de Célula/métodos , Proliferación Celular , Células Cultivadas , Técnicas de Inactivación de Genes , Antígenos HLA/genética , Antígenos HLA/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/ultraestructura , Leucosialina/metabolismo , Masculino , Megacariocitos/metabolismo , Megacariocitos/ultraestructura , Ratones Endogámicos NOD , Ratones SCID , Microscopía Electrónica , Microscopía Fluorescente , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Transfusión de Plaquetas/métodos , Reproducibilidad de los Resultados , Trasplante Heterólogo , Microglobulina beta-2/genética , Microglobulina beta-2/metabolismo
11.
Stem Cell Reports ; 3(1): 115-30, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-25068126

RESUMEN

Current therapies for multiple sclerosis (MS) are largely palliative, not curative. Mesenchymal stem cells (MSCs) harbor regenerative and immunosuppressive functions, indicating a potential therapy for MS, yet the variability and low potency of MSCs from adult sources hinder their therapeutic potential. MSCs derived from human embryonic stem cells (hES-MSCs) may be better suited for clinical treatment of MS because of their unlimited and stable supply. Here, we show that hES-MSCs significantly reduce clinical symptoms and prevent neuronal demyelination in a mouse experimental autoimmune encephalitis (EAE) model of MS, and that the EAE disease-modifying effect of hES-MSCs is significantly greater than that of human bone-marrow-derived MSCs (BM-MSCs). Our evidence also suggests that increased IL-6 expression by BM-MSCs contributes to the reduced anti-EAE therapeutic activity of these cells. A distinct ability to extravasate and migrate into inflamed CNS tissues may also be associated with the robust therapeutic effects of hES-MSCs on EAE.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Embrionarias/citología , Encefalomielitis Autoinmune Experimental/terapia , Células Madre Mesenquimatosas/citología , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Animales , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Humanos , Trasplante de Células Madre Mesenquimatosas , Ratones
12.
Cell Stem Cell ; 14(6): 777-80, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24746675

RESUMEN

Derivation of patient-specific human pluripotent stem cells via somatic cell nuclear transfer (SCNT) has the potential for applications in a range of therapeutic contexts. However, successful SCNT with human cells has proved challenging to achieve, and thus far has only been reported with fetal or infant somatic cells. In this study, we describe the application of a recently developed methodology for the generation of human ESCs via SCNT using dermal fibroblasts from 35- and 75-year-old males. Our study therefore demonstrates the applicability of SCNT for adult human cells and supports further investigation of SCNT as a strategy for regenerative medicine.


Asunto(s)
Células Madre Adultas/citología , Técnicas de Transferencia Nuclear , Células Madre Pluripotentes/citología , Adulto , Anciano , Dermis/citología , Fibroblastos/citología , Humanos , Masculino
13.
Stem Cells Dev ; 23(14): 1611-24, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24650034

RESUMEN

Mesenchymal stem cells (MSCs) are being tested in a wide range of human diseases; however, loss of potency and inconsistent quality severely limit their use. To overcome these issues, we have utilized a developmental precursor called the hemangioblast as an intermediate cell type in the derivation of a highly potent and replenishable population of MSCs from human embryonic stem cells (hESCs). This method circumvents the need for labor-intensive hand-picking, scraping, and sorting that other hESC-MSC derivation methods require. Moreover, unlike previous reports on hESC-MSCs, we have systematically evaluated their immunomodulatory properties and in vivo potency. As expected, they dynamically secrete a range of bioactive factors, display enzymatic activity, and suppress T-cell proliferation that is induced by either allogeneic cells or mitogenic stimuli. However, they also display unique immunophenotypic properties, as well as a smaller size and >30,000-fold proliferative capacity than bone marrow-derived MSCs. In addition, this is the first report which demonstrates that hESC-MSCs can inhibit CD83 up-regulation and IL-12p70 secretion from dendritic cells and enhance regulatory T-cell populations induced by interleukin 2 (IL-2). This is also the first report which shows that hESC-MSCs have therapeutic efficacy in two different autoimmune disorder models, including a marked increase in survival of lupus-prone mice and a reduction of symptoms in an autoimmune model of uveitis. Our data suggest that this novel and therapeutically active population of MSCs could overcome many of the obstacles that plague the use of MSCs in regenerative medicine and serve as a scalable alternative to current MSC sources.


Asunto(s)
Diferenciación Celular/genética , Inmunomodulación , Células Madre Mesenquimatosas/citología , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular/inmunología , Linaje de la Célula , Proliferación Celular/genética , Células Dendríticas/citología , Células Dendríticas/inmunología , Humanos , Activación de Linfocitos/inmunología , Células Madre Mesenquimatosas/inmunología , Ratones , Células Madre Pluripotentes/inmunología , Linfocitos T Reguladores/inmunología
15.
Stem Cells Int ; 2011: 273076, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21437192

RESUMEN

The ability of human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) to divide indefinitely without losing pluripotency and to theoretically differentiate into any cell type in the body makes them highly attractive cell sources for large scale regenerative medicine purposes. The current use of adult stem cell-derived products in hematologic intervention sets an important precedent and provides a guide for developing hESC/iPSC based therapies for the blood system. In this review, we highlight biological functions of mature cells of the blood, clinical conditions requiring the transfusion or stimulation of these cells, and the potential for hESC/iPSC-derivatives to serve as functional replacements. Many researchers have already been able to differentiate hESCs and/or iPSCs into specific mature blood cell types. For example, hESC-derived red blood cells and platelets are functional in tasks such as oxygen delivery and blood clotting, respectively and may be able to serve as substitutes for their donor-derived counterparts in emergencies. hESC-derived dendritic cells are functional in antigen-presentation and may be used as off-the-shelf vaccine therapies to stimulate antigen-specific immune responses against cancer cells. However, in vitro differentiation systems used to generate these cells will need further optimization before hESC/iPSC-derived blood components can be used clinically.

16.
Cell Res ; 21(3): 530-45, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21221130

RESUMEN

Platelets play an essential role in hemostasis and atherothrombosis. Owing to their short storage time, there is constant demand for this life-saving blood component. In this study, we report that it is feasible to generate functional megakaryocytes and platelets from human embryonic stem cells (hESCs) on a large scale. Differential-interference contrast and electron microscopy analyses showed that ultrastructural and morphological features of hESC-derived platelets were indistinguishable from those of normal blood platelets. In functional assays, hESC-derived platelets responded to thrombin stimulation, formed microaggregates, and facilitated clot formation/retraction in vitro. Live cell microscopy demonstrated that hESC-platelets formed lamellipodia and filopodia in response to thrombin activation, and tethered to each other as observed in normal blood. Using real-time intravital imaging with high-speed video microscopy, we have also shown that hESC-derived platelets contribute to developing thrombi at sites of laser-induced vascular injury in mice, providing the first evidence for in vivo functionality of hESC-derived platelets. These results represent an important step toward generating an unlimited supply of platelets for transfusion. Since platelets contain no genetic material, they are ideal candidates for early clinical translation involving human pluripotent stem cells.


Asunto(s)
Plaquetas/citología , Células Madre Embrionarias/citología , Animales , Plaquetas/fisiología , Plaquetas/ultraestructura , Diferenciación Celular , Citometría de Flujo , Humanos , Masculino , Megacariocitos/citología , Megacariocitos/fisiología , Megacariocitos/ultraestructura , Ratones , Ratones Endogámicos C57BL , Microcirculación , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Transfusión de Plaquetas , Seudópodos/fisiología
18.
Mol Endocrinol ; 24(12): 2292-302, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20980435

RESUMEN

Both pro- and antimitogenic activities have been ascribed to progesterone receptor (PR) agonists and antagonists in breast cancer cells; however, the transcriptional responses that underlie these paradoxical functions are not apparent. Using nontransformed, normal human mammary epithelial cells engineered to express PR and standard microarray technology, we defined 2370 genes that were significantly regulated by the PR agonist R5020. Gene ontology (GO) analysis revealed that GO terms involved in inflammation and nuclear factor-κB (NF-κB) signaling were among the most significantly regulated. Interestingly, on those NF-κB responsive genes that were inhibited by agonist-activated PR, antagonists either 1) mimicked the actions of agonists or 2) reversed the inhibitory actions of agonists. This difference in pharmacological response could be attributed to the fact that although agonist- and antagonist-activated PR is recruited to NF-κB-responsive promoters, the physical presence of PR tethered to the promoter of some genes is sufficient for transcriptional inhibition, whereas on others, an agonist-activated PR conformation is required for inhibition of NF-κB signaling. Importantly, the actions of PR on the latter class of genes were reversed by an activation function-2-inhibiting, LXXLL-containing peptide. Consideration of the relative activities of these distinct antiinflammatory pathways in breast cancer may be instructive with respect to the likely therapeutic activity of PR agonists or antagonists in the treatment of breast cancer.


Asunto(s)
Neoplasias Inflamatorias de la Mama/patología , Receptores de Progesterona/fisiología , Secuencia de Aminoácidos , Línea Celular , Línea Celular Tumoral , Elementos de Facilitación Genéticos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Gonanos/farmacología , Humanos , Neoplasias Inflamatorias de la Mama/genética , Neoplasias Inflamatorias de la Mama/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Mifepristona/farmacología , Datos de Secuencia Molecular , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Progesterona/genética , Progesterona/metabolismo , Progestinas/metabolismo , Regiones Promotoras Genéticas , Isoformas de Proteínas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Progesterona/agonistas , Receptores de Progesterona/antagonistas & inhibidores , Transducción de Señal , Activación Transcripcional , Translocación Genética
19.
Blood ; 114(23): 4804-12, 2009 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-19822904

RESUMEN

Cyclic adenosine monophosphate response element binding (CREB)-binding protein (CBP) and p300 are multidomain transcriptional coactivators that help assemble large regulatory complexes at sites of active transcription. Nullizygosity of CBP or p300 results in pervasive defects in hematopoiesis. To systematically assess the structural domains of p300 required for normal hematopoiesis, we used recombinase-mediated cassette exchange to create an allelic series of coisogenic embryonic stem cells, each expressing a different mutant of p300 from the endogenous locus. We found that deletion of either the KIX or CH1 domain caused profound and pervasive defects in hematopoiesis, whereas the loss of most other domains had only lineage-restricted effects. When expressed from the p300 locus, an extra copy of CBP largely compensated for a lack of p300. Surprisingly, mutation of the p300 histone acetyltransferase (HAT) domain had minimal effects on hematopoiesis, and actually increased progenitor and stem cell numbers and proliferative potential. Our results suggest that, in distinct contrast to other organ systems, HAT activity does not provide a critical function for hematopoietic development and emphasizes the importance of enzyme-independent functions of p300.


Asunto(s)
Proteína p300 Asociada a E1A/fisiología , Hematopoyesis/fisiología , Animales , Trasplante de Médula Ósea , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Linaje de la Célula , Quimera , Proteína p300 Asociada a E1A/química , Proteína p300 Asociada a E1A/genética , Transferencia de Embrión , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Femenino , Dosificación de Gen , Técnicas de Inactivación de Genes , Genes Sintéticos , Hematopoyesis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Mutación , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Estructura Terciaria de Proteína , Empalme del ARN , Quimera por Radiación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/fisiología , Eliminación de Secuencia , Relación Estructura-Actividad
20.
Mol Imaging ; 8(3): 140-7, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19723471

RESUMEN

Inhibiting the proteolytic activity of the 26S proteasome has been shown to have selective apoptotic effects on cancer cells and to be clinically efficacious in certain malignancies. There is an unmet medical need for additional proteasome inhibitors, and their development will be facilitated by surrogate markers of proteasome function. Toward this end, ectopic fusion of the destruction domain from ornithine decarboxylase (ODC) to reporter proteins is often used for assessing proteasome function. For luciferase-based reporters, we hypothesized that the oxygen-dependent destruction domain (ODD) from hypoxia-inducible factor 1 alpha (HIF-1 alpha) may provide improved sensitivity over luciferase-ODC, owing to its extremely rapid turnover by the proteasome (HIF-1 alpha has a half-life of less than 5 minutes). In the current study, we show that ODD-luciferase affords a greater dynamic range and faster kinetics than luciferase-ODC in sensing proteasome inhibition in vitro. Importantly, ODD-luciferase also serves as an effective in vivo marker of proteasome function in xenograft tumor models, with inhibition being detected by noninvasive imaging within 3 hours of bortezomib administration. These data establish ODD-luciferase as a surrogate marker of proteasome function that can be used both in vitro and in vivo for the development of novel proteasome inhibitors.


Asunto(s)
Mediciones Luminiscentes/métodos , Proteínas Luminiscentes/metabolismo , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Imagen de Cuerpo Entero/métodos , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Animales , Genes Reporteros , Vectores Genéticos , Células HCT116 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Luciferasas/química , Luciferasas/genética , Luciferasas/metabolismo , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Ratones , Ratones Desnudos , Ornitina Descarboxilasa/química , Ornitina Descarboxilasa/genética , Ornitina Descarboxilasa/metabolismo , Inhibidores de Proteasas/química , Complejo de la Endopetidasa Proteasomal/química , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...