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.
Cell Rep ; 42(4): 112276, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36965484

RESUMEN

Although the skeleton is essential for locomotion, endocrine functions, and hematopoiesis, the molecular mechanisms of human skeletal development remain to be elucidated. Here, we introduce an integrative method to model human skeletal development by combining in vitro sclerotome induction from human pluripotent stem cells and in vivo endochondral bone formation by implanting the sclerotome beneath the renal capsules of immunodeficient mice. Histological and scRNA-seq analyses reveal that the induced bones recapitulate endochondral ossification and are composed of human skeletal cells and mouse circulatory cells. The skeletal cell types and their trajectories are similar to those of human embryos. Single-cell multiome analysis reveals dynamic changes in chromatin accessibility associated with multiple transcription factors constituting cell-type-specific gene-regulatory networks (GRNs). We further identify ZEB2, which may regulate the GRNs in human osteogenesis. Collectively, these results identify components of GRNs in human skeletal development and provide a valuable model for its investigation.


Asunto(s)
Multiómica , Células Madre Pluripotentes , Humanos , Ratones , Animales , Diferenciación Celular , Factores de Transcripción/metabolismo , Regulación de la Expresión Génica , Células Madre Pluripotentes/metabolismo
2.
J Biomed Mater Res A ; 111(8): 1200-1215, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36728346

RESUMEN

Cell therapy is emerging as an effective treatment strategy for many diseases. Here we describe a novel approach to bone tissue repair that combines hydrogel-based cell therapy with low intensity pulsed ultrasound (LIPUS), an FDA approved treatment for fracture repair. Bone marrow-derived stromal cells (BMSCs) have been encapsulated in type I collagen hydrogels and mechanically stimulated using LIPUS-derived acoustic radiation force (ARF). We observed the expression and upward trend of load-sensitive, osteoblast-specific markers and determined that the extent of cell response is dependent on an optimal combination of both hydrogel stiffness and ARF intensity. Specifically, cells encapsulated in hydrogels of optimal stiffness respond at the onset of ultrasound by upregulating early bone-sensitive markers such as calcium, cyclooxygenase-2, and prostaglandin E2 , and later by supporting mineralized tissue formation after 21 days of culture. In vivo evaluation of a critical size calvarial defect in NOD scid gamma (NSG) mice indicated that the implantation of BMSC-laden hydrogels of optimal stiffness improved healing of calvarial defects after daily administration of ARF over 4 weeks. Collectively, these findings validate the efficacy of our system of localized cell delivery for treating bone defects where undifferentiated BMSCs are induced to the osteoblastic lineage. Further, in vivo healing may be enhanced via non-invasive transdermal mechanical stimulation of implanted cells using ARF.


Asunto(s)
Hidrogeles , Células Madre Mesenquimatosas , Ratones , Animales , Hidrogeles/farmacología , Ultrasonografía , Colágeno/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos
3.
NPJ Regen Med ; 6(1): 34, 2021 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-34117259

RESUMEN

The osteogenic potential of bone marrow mesenchymal stem cells (BMSCs) declines dramatically with aging. By using a calvarial defect model, we showed that a senolytic cocktail (dasatinib+quercetin; D + Q) improved osteogenic capacity of aged BMSC both in vitro and in vivo. The study presented a model to assess strategies to improve bone-forming potential on aged BMSCs. D + Q might hold promise for improving BMSC function in aged populations.

4.
Saudi J Biol Sci ; 27(9): 2491-2497, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32884431

RESUMEN

OBJECTIVE: To study the therapeutic effect and mechanism of levotinib on choroidal neovascularization (CNV) in mice. METHODS: 45 healthy C57BL/6 mice were selected and randomly divided into three groups: control group (group A), model group (group B) and levotinib group (group C). The model of CNV in mice was established. The fluorescence leakage of choroidal lesions in mice was observed by fundus fluorescein angiography. The morphological changes of retinal vessels in mice were observed by retinal slice preparation, the pathological changes of eyeball tissues in mice were observed by hematoxylin-eosin (HE) staining, the expression of vascular endothelial growth factor (VEGF) in mice retina was detected by real-time quantitative fluorescence PCR, and the protein expression of VEGF in mice retina was detected by Western blotting. RESULT: On the 7th, 14th and 21st day after modeling, compared with group B, the fluorescence leakage area of group C mice was significantly reduced, and the difference was statistically significant (P < 0.05). The morphology of retinal vessels in group A was normal. In group B, the retinal vessels showed large areas of ischemia without perfusion and abundant neovascularization clusters and capillaries. Compared with group B, the morphology of retinal vessels in group C was significantly improved. Group A mice had normal eyeball structure, group B mice had visible spindle-like damage to the inner and outer retina, while group C mice had significantly less spindle-like damage than group B. Compared with group A, group B mice had significantly higher expression of retinal VEGF and the difference was statistically significant (P < 0.05), but compared with group B mice, the expression of VEGF in the retina of mice in group C was significantly decreased, and the difference was statistically significant (P < 0.05). Compared with group A, the expression of VEGF in retina of group B mice was significantly increased, and the difference was statistically significant (P < 0.05). Compared with group B, the expression of VEGF in retina of group C mice was significantly decreased, and the difference was statistically significant (P < 0.05). CONCLUSION: Levatinib has obvious therapeutic effect on CNV, which may be achieved by inhibiting the high expression of VEGF in CNV.

5.
ACS Appl Mater Interfaces ; 12(16): 18235-18249, 2020 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-32212615

RESUMEN

As one of the major challenges in the field of tissue engineering, large skeletal defects have attracted wide attention from researchers. Collagen (Col) and hydroxyapatite (HA), the most abundant protein and the main component in natural bone, respectively, are usually used as a biomimetic composite material in tissue engineering due to their excellent biocompatibility and biodegradability. In this study, novel intrafibrillar mineralized Col-HA-based scaffolds, constructed in either cellular or lamellar microstructures, were established through a biomimetic method to enhance the new bone-regenerating capability of tissue engineering scaffolds. Moreover, iron (Fe) and manganese (Mn), two of the essential trace elements in the body, were successfully incorporated into the lamellar scaffold to further improve the osteoinductivity of these biomaterials. It was found that the lamellar scaffolds demonstrated better osteogenic abilities compared to both in-house and commercial Col-HA-based cellular scaffolds in vitro and in vivo. Meanwhile, Fe/Mn incorporation further amplified the osteogenic promotion of the lamellar scaffolds. More importantly, a synergistic effect was observed in the Fe and Mn dual-element-incorporated lamellar scaffolds for both in vitro osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and in vivo bone regeneration loaded with fresh bone marrow cells. This study provides a simple but practical strategy for the creation of functional scaffolds for bone regeneration.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Colágeno , Durapatita , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Células 3T3 , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/química , Colágeno/farmacología , Durapatita/química , Durapatita/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Cráneo/efectos de los fármacos , Cráneo/patología
6.
Regen Ther ; 14: 19-31, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31988991

RESUMEN

Clinically relevant human induced pluripotent stem cell (hiPSC) derivatives require efficient protocols to differentiate hiPSCs into specific lineages. Here we developed a fully defined xeno-free strategy to direct hiPSCs toward osteoblasts within 21 days. The strategy successfully achieved the osteogenic induction of four independently derived hiPSC lines by a sequential use of combinations of small-molecule inducers. The induction first generated mesodermal cells, which subsequently recapitulated the developmental expression pattern of major osteoblast genes and proteins. Importantly, Col2.3-Cherry hiPSCs subjected to this strategy strongly expressed the cherry fluorescence that has been observed in bone-forming osteoblasts in vivo. Moreover, the protocol combined with a three-dimensional (3D) scaffold was suitable for the generation of a xeno-free 3D osteogenic system. Thus, our strategy offers a platform with significant advantages for bone biology studies and it will also contribute to clinical applications of hiPSCs to skeletal regenerative medicine.

7.
Nano Energy ; 762020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38074984

RESUMEN

Electrical stimulation (ES) has been shown to induce and enhance bone regeneration. By combining this treatment with tissue-engineering approaches (which rely on biomaterial scaffolds to construct artificial tissues), a replacement bone-graft with strong regenerative properties can be achieved while avoiding the use of potentially toxic levels of growth factors. Unfortunately, there is currently a lack of safe and effective methods to induce electrical cues directly on cells/tissues grown on the biomaterial scaffolds. Here, we present a novel bone regeneration method which hybridizes ES and tissue-engineering approaches by employing a biodegradable piezoelectric PLLA (Poly(L-lactic acid)) nanofiber scaffold which, together with externally-controlled ultrasound (US), can generate surface-charges to drive bone regeneration. We demonstrate that the approach of using the piezoelectric scaffold and US can enhance osteogenic differentiation of different stem cells in vitro, and induce bone growth in a critical-sized calvarial defect in vivo. The biodegradable piezoelectric scaffold with applied US could significantly impact the field of tissue engineering by offering a novel biodegradable, battery-free and remotely-controlled electrical stimulator.

8.
Ann Biomed Eng ; 48(3): 992-1005, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31037444

RESUMEN

Despite its regenerative ability, long and segmental bone defect repair remains a significant orthopedic challenge. Conventional tissue engineering efforts induce bone formation through intramembranous ossification (IO) which limits vascular formation and leads to poor bone regeneration. To overcome this challenge, a novel hybrid matrix comprised of a load-bearing polymer template and a gel phase is designed and assessed for bone regeneration. Our previous studies developed a synthetic ECM, hyaluronan (HA)-fibrin (FB), that is able to mimic cartilage-mediated bone formation in vitro. In this study, the well-characterized HA-FB hydrogel is combined with a biodegradable polymer template to form a hybrid matrix. In vitro evaluation of the matrix showed cartilage template formation, cell recruitment and recruited cell osteogenesis, essential stages in endochondral ossification. A transgenic reporter-mouse critical-defect model was used to evaluate the bone healing potential of the hybrid matrix in vivo. The results demonstrated host cell recruitment into the hybrid matrix that led to new bone formation and subsequent remodeling of the mineralization. Overall, the study developed and evaluated a novel load-bearing graft system for bone regeneration via endochondral ossification.


Asunto(s)
Regeneración Ósea , Células Madre Mesenquimatosas/fisiología , Osteogénesis , Cráneo/fisiología , Ingeniería de Tejidos/métodos , Animales , Células Cultivadas , Matriz Extracelular , Fibrina , Humanos , Ácido Hialurónico , Hidrogeles , Ratones SCID , Porosidad
9.
Proc Natl Acad Sci U S A ; 117(1): 214-220, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31871178

RESUMEN

Piezoelectric materials, a type of "smart" material that generates electricity while deforming and vice versa, have been used extensively for many important implantable medical devices such as sensors, transducers, and actuators. However, commonly utilized piezoelectric materials are either toxic or nondegradable. Thus, implanted devices employing these materials raise a significant concern in terms of safety issues and often require an invasive removal surgery, which can damage directly interfaced tissues/organs. Here, we present a strategy for materials processing, device assembly, and electronic integration to 1) create biodegradable and biocompatible piezoelectric PLLA [poly(l-lactic acid)] nanofibers with a highly controllable, efficient, and stable piezoelectric performance, and 2) demonstrate device applications of this nanomaterial, including a highly sensitive biodegradable pressure sensor for monitoring vital physiological pressures and a biodegradable ultrasonic transducer for blood-brain barrier opening that can be used to facilitate the delivery of drugs into the brain. These significant applications, which have not been achieved so far by conventional piezoelectric materials and bulk piezoelectric PLLA, demonstrate the PLLA nanofibers as a powerful material platform that offers a profound impact on various medical fields including drug delivery, tissue engineering, and implanted medical devices.


Asunto(s)
Implantes Absorbibles , Sistemas Microelectromecánicos/instrumentación , Nanofibras/química , Transductores , Sistemas de Liberación de Medicamentos , Electricidad , Electrónica , Diseño de Equipo , Monitoreo Fisiológico/instrumentación , Presión , Prótesis e Implantes , Ingeniería de Tejidos , Ultrasonido
10.
J Histochem Cytochem ; 67(6): 401-417, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30848692

RESUMEN

The effectiveness of autologous cell-based skeletal repair continues to be controversial in part because in vitro predictors of in vivo human bone formation by cultured human progenitor cells are not reliable. To assist in the development of in vivo assays of human osteoprogenitor potential, a fluorescence-based histology of nondecalcified mineralized tissue is presented that provides multiple criteria to distinguish human and host osteoblasts, osteocytes, and accumulated bone matrix in a mouse calvarial defect model. These include detection of an ubiquitously expressed red fluorescent protein reporter by the implanted human cells, antibodies specific to human bone sialoprotein and a human nuclear antigen, and expression of a bone/fibroblast restricted green fluorescent protein reporter in the host tissue. Using low passage bone marrow-derived stromal cells, robust human bone matrix formation was obtained. However, a striking feature is the lack of mouse bone marrow investment and osteoclasts within the human bone matrix. This deficiency may account for the accumulation of a disorganized human bone matrix that has not undergone extensive remodeling. These features, which would not be appreciated by traditional decalcified paraffin histology, indicate the human bone matrix is not undergoing active remodeling and thus the full differentiation potential of the implanted human cells within currently used mouse models is not being realized.


Asunto(s)
Osteogénesis , Animales , Humanos , Ratones , Imagen Óptica , Osteoblastos/citología , Cráneo/citología , Cráneo/diagnóstico por imagen , Cráneo/fisiología
11.
Tissue Eng Part A ; 25(5-6): 468-486, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30136616

RESUMEN

IMPACT STATEMENT: This study represents significant advancement in the use of biomimetic scaffolds to direct zonal osteochondral tissue formation. We describe the use of a novel fluorescent reporter system that enables the real-time evaluation of cellular differentiation in a nondestructive manner. In this study, we use this tool to confirm the osteogenic and chondrogenic capabilities of our scaffold alongside control scaffolds, and use cryohistological methods to probe zone-specific differences in cell and tissue quality. We believe this approach can be widely adopted by others for a variety of biomaterial and cell systems in the development of tissue engineered therapeutics.


Asunto(s)
Materiales Biocompatibles/farmacología , Diferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Animales , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Colágeno Tipo I/metabolismo , Colágeno Tipo II/metabolismo , Fluorescencia , Regulación de la Expresión Génica/efectos de los fármacos , Implantes Experimentales , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones Transgénicos , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Tejido Subcutáneo/efectos de los fármacos , Tejido Subcutáneo/fisiología , Andamios del Tejido/química
12.
Methods Mol Biol ; 1723: 385-396, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29344873

RESUMEN

Laser-capture microdissection (LCM) coupled to downstream RNA analysis poses unique difficulties for the evaluation of mineralized tissues. A rapid protocol was thus developed to enable sufficient integrity of bone and cartilage tissue for reliable sectioning, while minimizing RNA loss associated with prolonged decalcification and purification steps. Specifically, the protocol involves pump-assisted, cardiac perfusion-fixation with paraformaldehyde, and moderate digestion of LCM-acquired tissue with proteinase K followed by DNase treatment and separation of RNA using magnetic beads. Reverse transcription and cDNA synthesis are performed immediately after RNA purification, without need for further protein removal.


Asunto(s)
Huesos/metabolismo , Cartílago/metabolismo , Modelos Animales de Enfermedad , Células Madre Pluripotentes Inducidas/metabolismo , Captura por Microdisección con Láser/métodos , Células Madre Mesenquimatosas/metabolismo , ARN/análisis , Cráneo/metabolismo , Animales , Huesos/patología , Cartílago/patología , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/patología , Células Madre Mesenquimatosas/patología , Ratones , Perfusión , ARN/genética , ARN/aislamiento & purificación , Cráneo/patología
13.
Stem Cells Transl Med ; 3(10): 1125-37, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25122686

RESUMEN

The use of human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) for study and treatment of bone diseases or traumatic bone injuries requires efficient protocols to differentiate hESCs/iPSCs into cells with osteogenic potential and the ability to isolate differentiated osteoblasts for analysis. We have used zinc finger nuclease technology to deliver a construct containing the Col2.3 promoter driving GFPemerald to the AAVS1 site (referred to as a "safe harbor" site), in human embryonic stem cells (H9Zn2.3GFP), with the goal of marking the cells that have become differentiated osteoblasts. In teratomas formed using these cells, we identified green fluorescent protein (GFP)-positive cells specifically associated with in vivo bone formation. We also differentiated the cells into a mesenchymal stem cell population with osteogenic potential and implanted them into a mouse calvarial defect model. We observed GFP-positive cells associated with alizarin complexone-labeled newly formed bone surfaces. The cells were alkaline phosphatase-positive, and immunohistochemistry with human specific bone sialoprotein (BSP) antibody indicates that the GFP-positive cells are also associated with the human BSP-containing matrix, demonstrating that the Col2.3GFP construct marks cells in the osteoblast lineage. Single-cell cloning generated a 100% Col2.3GFP-positive cell population, as demonstrated by fluorescence in situ hybridization using a GFP probe. The karyotype was normal, and pluripotency was demonstrated by Tra1-60 immunostaining, pluripotent low density reverse transcription-polymerase chain reaction array and embryoid body formation. These cells will be useful to develop optimal osteogenic differentiation protocols and to isolate osteoblasts from normal and diseased iPSCs for analysis.


Asunto(s)
Células Madre Embrionarias/citología , Proteínas Fluorescentes Verdes , Osteoblastos/citología , Animales , Matriz Ósea/citología , Diferenciación Celular/fisiología , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Genes Reporteros , Xenoinjertos , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Ratones , Células Madre Pluripotentes/citología , Ratas
14.
Artículo en Inglés | MEDLINE | ID: mdl-25570850

RESUMEN

Bone defect repair is a significant clinical challenge in orthopedic surgery. Despite tremendous efforts, the majority of the current bone tissue engineering strategies depend on bone formation via intramembranous ossification (IO), which often results in poor vascularization and limited-area bone regeneration. Recently, there has been increasing interest in exploring bone regeneration through a cartilage-mediated process similar to endochondral ossification (EO). This method is advantageous because long bones are originally developed through EO and moreover, vascularization is an inherent step of this process. Therefore, it may be possible to effectively employ the EO method for the repair and regeneration of large and segmental bone defects. Although a number of studies have demonstrated engineered bone formation through EO, there are no approaches aiming for their clinical translation. In this study, we propose a strategy modeled after the U.S. Food and Drug Administration (FDA) approved autologus chondrocyte implantation (ACI) procedure. In its implementation, we concentrated human bone marrow aspirate via a minimally manipulated process and demonstrated the potential of human bone marrow derived cells for in vitro pre-cartilage template formation and bone regeneration in vivo.


Asunto(s)
Huesos/fisiología , Ingeniería de Tejidos , Adolescente , Adulto , Animales , Células de la Médula Ósea/citología , Regeneración Ósea , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Femenino , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ratones , Ratones Transgénicos , Osteogénesis , Radiografía , Cráneo/diagnóstico por imagen , Cráneo/patología , Trasplante Heterólogo , Cicatrización de Heridas , Adulto Joven
15.
Tissue Eng Part A ; 20(1-2): 365-77, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23952622

RESUMEN

The in vivo osteogenesis potential of mesenchymal-like cells derived from human embryonic stem cells (hESC-MCs) was evaluated in vivo by implantation on collagen/hydroxyapatite scaffolds into calvarial defects in immunodeficient mice. This study is novel because no osteogenic or chondrogenic differentiation protocols were applied to the cells prior to implantation. After 6 weeks, X-ray, microCT, and histological analysis showed that the hESC-MCs had consistently formed a highly vascularized new bone that bridged the bone defect and seamlessly integrated with host bone. The implanted hESC-MCs differentiated in situ to functional hypertrophic chondrocytes, osteoblasts, and osteocytes forming new bone tissue via an endochondral ossification pathway. Evidence for the direct participation of the human cells in bone morphogenesis was verified by two separate assays: with Alu and by human mitochondrial antigen positive staining in conjunction with co-localized expression of human bone sialoprotein in histologically verified regions of new bone. The large volume of new bone in a calvarial defect and the direct participation of the hESC-MCs far exceeds that of previous studies and that of the control adult hMSCs. This study represents a key step forward for bone tissue engineering because of the large volume, vascularity, and reproducibility of new bone formation and the discovery that it is advantageous to not over-commit these progenitor cells to a particular lineage prior to implantation. The hESC-MCs were able to recapitulate the mesenchymal developmental pathway and were able to repair the bone defect semi-autonomously without preimplantation differentiation to osteo- or chondroprogenitors.


Asunto(s)
Regeneración Ósea/fisiología , Células Madre Embrionarias/citología , Células Madre Mesenquimatosas/citología , Adulto , Animales , Regeneración Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Colágeno/farmacología , Durapatita/farmacología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Morfogénesis/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Fenotipo , Implantación de Prótesis , Regeneración/efectos de los fármacos , Cráneo/efectos de los fármacos , Cráneo/patología , Trasplante de Células Madre , Andamios del Tejido/química
16.
Mol Pharmacol ; 82(4): 738-45, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22828798

RESUMEN

Kalirin-7 (Kal7), the major kalirin isoform in adult brain, plays a key role in the formation of dendritic spines in hippocampal/cortical neurons. Its role in the GABAergic medium spiny neurons (MSNs) of the nucleus accumbens (NAc) and striatum, the areas known to play a key role in the common reward pathway, is not as well understood. Although Kal7 expression in mouse NAc increased in response to cocaine, MSN dendritic spine density did not differ from that for the wild type in Kal7-null mice. Unlike wild-type mice, Kal7-null mice did not respond to cocaine with an increase in MSN dendritic spine density. To explore further the role of Kal7 in cocaine-induced alterations in MSN morphology, we turned to the rat. Based on immunostaining, both Kal7 and Kal12 are expressed at moderate levels in the MSNs of the NAc and striatum. Expression of Kal7 and Kal12 in MSNs of both areas increases after repeated cocaine treatments. Overexpression of Kal7 in cultured MSN neurons increases dendritic spine density, as observed in rats after long-term cocaine administration. Reducing endogenous expression of all major kalirin isoforms in cultured MSN neurons causes a decrease in total dendritic length and dendritic spine density. These data suggest that kalirin is essential for maintaining spine density in NAc MSNs under normal conditions and that Kal7 is an obligatory intermediate in the response of MSNs to repeated exposure to cocaine.


Asunto(s)
Cocaína/farmacología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Neuronas/efectos de los fármacos , Animales , Células Cultivadas , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/ultraestructura , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Masculino , Neuronas/metabolismo , Neuronas/ultraestructura , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Núcleo Accumbens/ultraestructura , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
17.
Biol Psychiatry ; 68(3): 249-55, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20452575

RESUMEN

BACKGROUND: Long-lasting increases in dendritic spine density and gene expression in the nucleus accumbens and in the ambulatory response to cocaine occur following chronic cocaine treatment. Despite numerous reports of these findings, the molecular mechanisms leading to these morphological, biochemical, and behavioral changes remain unclear. METHODS: We used mice genetically lacking Kalirin7 (Kal7(KO)), a Rho guanine nucleotide exchange factor that regulates dendritic spine formation and function. Both wild-type (Wt) and Kal7(KO) mice were given high-dose cocaine (20 mg/kg) for 4 or 8 consecutive days. Locomotor sensitization and conditioned place preference elicited by cocaine were evaluated. The nucleus accumbens core was diolistically labeled and spine density and morphology were quantified using confocal microscopy. RESULTS: Cocaine increased Kalirin7 messenger RNA and protein expression in the nucleus accumbens of Wt mice. The Kal7(KO) animals showed greater locomotor sensitization to cocaine than Wt mice. In contrast, Kal7(KO) mice exhibited decreased place preference for cocaine, despite displaying a normal place preference for food. While Wt mice showed a robust increase in dendritic spine density after 4 and 8 days of cocaine treatment, dendritic spine density failed to increase in cocaine-exposed Kal7(KO) mice. Wild-type mice treated with cocaine for 8 days exhibited larger dendritic spines than cocaine-treated Kal7(KO) mice. CONCLUSIONS: Kalirin7 is an essential determinant of dendritic spine formation following cocaine treatment. The absence of this single isoform of one of the many Rho guanine nucleotide exchange factors expressed in the nucleus accumbens results in enhanced locomotor sensitization and diminished place preference in response to cocaine.


Asunto(s)
Conducta Animal/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Cocaína/farmacología , Espinas Dendríticas/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Núcleo Accumbens/efectos de los fármacos , Animales , Aprendizaje por Asociación/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Espinas Dendríticas/metabolismo , Factores de Intercambio de Guanina Nucleótido/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Núcleo Accumbens/citología , Núcleo Accumbens/metabolismo , ARN Mensajero/análisis
18.
BMC Neurosci ; 10: 61, 2009 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-19534784

RESUMEN

BACKGROUND: Guanine nucleotide exchange factors (GEFs) and their target Rho GTPases regulate cytoskeletal changes and membrane trafficking. Dynamin, a large force-generating GTPase, plays an essential role in membrane tubulation and fission in cells. Kalirin12, a neuronal RhoGEF, is found in growth cones early in development and in dendritic spines later in development. RESULTS: The IgFn domain of Kalirin12, not present in other Kalirin isoforms, binds dynamin1 and dynamin2. An inactivating mutation in the GTPase domain of dynamin diminishes this interaction and the isolated GTPase domain of dynamin retains the ability to bind Kalirin12. Co-immunoprecipitation demonstrates an interaction of Kalirin12 and dynamin2 in embryonic brain. Purified recombinant Kalirin-IgFn domain inhibits the ability of purified rat brain dynamin to oligomerize in response to the presence of liposomes containing phosphatidylinositol-4,5-bisphosphate. Consistent with this, expression of exogenous Kalirin12 or its IgFn domain in PC12 cells disrupts clathrin-mediated transferrin endocytosis. Similarly, expression of exogenous Kalirin12 disrupts transferrin endocytosis in cortical neurons. Expression of Kalirin7, a shorter isoform which lacks the IgFn domain, was previously shown to inhibit clathrin-mediated endocytosis; the GTPase domain of dynamin does not interact with Kalirin7. CONCLUSION: Kalirin12 may play a role in coordinating Rho GTPase-mediated changes in the actin cytoskeleton with dynamin-mediated changes in membrane trafficking.


Asunto(s)
Dinaminas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Neuronas/fisiología , Animales , Células Cultivadas , Corteza Cerebral/citología , Clatrina/farmacología , Dinaminas/clasificación , Embrión de Mamíferos , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , GTP Fosfohidrolasas/metabolismo , Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/genética , Conos de Crecimiento/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Inmunoprecipitación/métodos , Liposomas/metabolismo , Neuronas/citología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Ratas , Transfección/métodos , Transferrina/metabolismo
19.
J Cell Sci ; 121(Pt 15): 2601-11, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18628310

RESUMEN

Kalirin, one of the few Rho guanine nucleotide exchange factors (GEFs) that contains spectrin-like repeats, plays a critical role in axon extension and maintenance of dendritic spines. PC12 cells were used to determine whether Cdk5, a critical participant in both processes, regulates the action of Kalirin. Expression of Kalirin-7 in nondifferentiated PC12 cells caused GEF-activity-dependent extension of broad cytoplasmic protrusions; coexpression of dominant-negative Cdk5 largely eliminated this response. The spectrin-like repeat region of Kalirin plays an essential role in this response, which is not mimicked by the GEF domain alone. Thr1590, which follows the first GEF domain of Kalirin, is the only Cdk5 phosphorylation site in Kalirin-7. Although mutant Kalirin-7 with Ala1590 retains GEF activity, it is unable to cause extension of protrusions. Kalirin-7 with an Asp1590 mutation has slightly increased GEF activity and dominant-negative Cdk5 fails to block its ability to cause extension of protrusions. Phosphorylation of Thr1590 causes a slight increase in GEF activity and Kalirin-7 solubility. Dendritic spines formed by cortical neurons in response to the expression of Kalirin-7 with Ala1590 differ in shape from those formed in response to wild-type Kalirin-7 or Kalirin-7 containing Asp1590. The presence of Thr1590 in each major Kalirin isoform would allow Cdk5 to regulate Kalirin function throughout development.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Animales , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Células Cultivadas , Espinas Dendríticas/metabolismo , Modelos Biológicos , Células PC12 , Fosforilación , Proteína Fosfatasa 1/metabolismo , Ratas , Treonina/genética , Treonina/metabolismo
20.
J Biol Chem ; 279(46): 48159-67, 2004 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-15337741

RESUMEN

Based on sequence comparisons, MOX (monooxygenase X), is a member of the copper monooxygenase family that includes dopamine beta-monooxygenase (DBM) and peptidylglycine alpha-hydroxylating monooxygenase (PHM). MOX has all of the residues expected to be critical for copper binding, and its cysteine residues can yield the intramolecular disulfide bond pattern observed in DBM. Although DBM and PHM function within the lumen of the secretory pathway, the published sequence for human MOX lacks a signal sequence, suggesting that it does not enter this compartment. We identified an upstream exon that encodes the signal sequence of human MOX. A retained intron yields minor amounts of transcript encoding MOX without a signal sequence. MOX transcripts are widely expressed, with the highest levels in the salivary gland and ovary and moderate levels in brain, pituitary, and heart. Despite the presence of a signal sequence, exogenous MOX is not secreted, and it localizes throughout the endoplasmic reticulum in both endocrine or nonendocrine cells. Neither appending green fluorescent protein to its C terminus nor deleting the hydrophobic domain near its C terminus facilitates secretion of MOX. MOX is N-glycosylated, is tightly membrane-associated, and forms oligomers that are not disulfide-linked. Based on its sequence and localization, MOX is predicted to hydroxylate a hydrophobic substrate in the endoplasmic reticulum.


Asunto(s)
Cobre/metabolismo , Retículo Endoplásmico/enzimología , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Secuencia de Aminoácidos , Animales , Fraccionamiento Celular , Línea Celular , Exones , Humanos , Intrones , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Datos de Secuencia Molecular , Familia de Multigenes , Señales de Clasificación de Proteína , Estructura Cuaternaria de Proteína , Alineación de Secuencia , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...