Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell Tissue Res ; 372(3): 507-522, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29318389

RESUMEN

Advances in stem cell biology and materials science have provided a basis for developing tissue engineering methods to repair muscle injury. Among stem cell populations with potential to aid muscle repair, adipose-derived mesenchymal stem cells (ASC) hold great promise. To evaluate the possibility of using porcine ASC for muscle regeneration studies, we co-cultured porcine ASC with murine C2C12 myoblasts. These experiments demonstrated that porcine ASC display significant myogenic potential. Co-culture of ASC expressing green fluorescent protein (GFP) with C2C12 cells resulted in GFP+ myotube formation, indicating fusion of ASC with myoblasts to form myotubes. The presence of porcine lamin A/C positive nuclei in myotubes and RTqPCR analysis of porcine myogenin and desmin expression confirmed that myotube nuclei derived from ASC contribute to muscle gene expression. Co-culturing GFP+ASC with porcine satellite cells demonstrated enhanced myogenic capability of ASC, as the percentage of labeled myotubes increased compared to mouse co-cultures. Enhancing myogenic potential of ASC through soluble factor treatment or expansion of ASC with innate myogenic capacity should allow for their therapeutic use to regenerate muscle tissue lost to disease or injury.


Asunto(s)
Tejido Adiposo/citología , Separación Celular , Células Madre Mesenquimatosas/citología , Desarrollo de Músculos , Animales , Diferenciación Celular , Linaje de la Célula , Núcleo Celular/metabolismo , Técnicas de Cocultivo , Medios de Cultivo , Regulación de la Expresión Génica , Ratones , Fibras Musculares Esqueléticas/metabolismo , Especificidad de Órganos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Células Madre/citología , Porcinos
2.
Anim Biotechnol ; 28(4): 275-287, 2017 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-28267421

RESUMEN

Bone is a plastic tissue with a large healing capability. However, extensive bone loss due to disease or trauma requires extreme therapy such as bone grafting or tissue-engineering applications. Presently, bone grafting is the gold standard for bone repair, but presents serious limitations including donor site morbidity, rejection, and limited tissue regeneration. The use of stem cells appears to be a means to overcome such limitations. Bone marrow mesenchymal stem cells (BMSC) have been the choice thus far for stem cell therapy for bone regeneration. However, adipose-derived stem cells (ASC) have similar immunophenotype, morphology, multilineage potential, and transcriptome compared to BMSC, and both types have demonstrated extensive osteogenic capacity both in vitro and in vivo in several species. The use of scaffolds in combination with stem cells and growth factors provides a valuable tool for guided bone regeneration, especially for complex anatomic defects. Before translation to human medicine, regenerative strategies must be developed in animal models to improve effectiveness and efficiency. The pig presents as a useful model due to similar macro- and microanatomy and favorable logistics of use. This review examines data that provides strong support for the clinical translation of the pig model for bone regeneration.


Asunto(s)
Regeneración Ósea , Trasplante de Células Madre Mesenquimatosas , Porcinos , Animales , Modelos Animales de Enfermedad , Humanos , Ingeniería de Tejidos , Andamios del Tejido
3.
Homeopathy ; 106(2): 87-92, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28552178

RESUMEN

OBJECTIVE: This study investigated the effect of two different follicle stimulating hormone (FSH) preparations (diluted/dynamised and diluted) on the in vitro development and steroid production (estradiol, progesterone and testosterone) of isolated porcine preantral follicle after in vitro culture. METHODS: Secondary follicles were cultured in Alpha Minimum Essential Medium (α-MEM+) supplemented with grain ethanol (AL - 0.2%, v/v), diluted/dynamised FSH (rFSH 6cH - 0.05 fg/mL) or diluted-only FSH (1.5 ng/mL) for 4 days. Follicle development was evaluated on the basis of follicular growth, morphology and hormone production. RESULTS: The percentage of follicular integrity and extrusion were not affected by the treatments after culture. For all treatments, follicular diameter increased significantly from Day 0 to Day 4. On Day 2 of culture, the estradiol production was significantly higher in AL and diluted-only FSH treatments compared with diluted/dynamised FSH. However, diluted/dynamised FSH showed a significantly higher progesterone production on Day 2. Only on Day 4, the testosterone production was higher in the AL than diluted-only FSH treatments, but similar to diluted/dynamised FSH treatment. Except for diluted/dynamised FSH treatment, progesterone production increased (P < 0.05) from Day 2 to Day 4; only for AL treatment, a significant increase of testosterone production was observed during culture. CONCLUSION: Compared to control the diluted/dynamised FSH addition increased progesterone production but decreased the estradiol production after in vitro culture of isolated porcine preantral follicles. Taken together the results suggest that at least for progesterone production the mechanism of action of diluted/dynamised FSH differs from its vehicle.


Asunto(s)
Estradiol/metabolismo , Hormona Folículo Estimulante/farmacología , Homeopatía , Folículo Ovárico/efectos de los fármacos , Animales , Femenino , Modelos Animales , Folículo Ovárico/metabolismo , Porcinos
4.
BMC Dev Biol ; 14: 32, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-25063185

RESUMEN

BACKGROUND: To gain insight into what differences might restrict the capacity for limb regeneration in Xenopus froglets, we used High Performance Liquid Chromatography (HPLC)/double mass spectrometry to characterize protein expression during fibroblastema formation in the amputated froglet hindlimb, and compared the results to those obtained previously for blastema formation in the axolotl limb. RESULTS: Comparison of the Xenopus fibroblastema and axolotl blastema revealed several similarities and significant differences in proteomic profiles. The most significant similarity was the strong parallel down regulation of muscle proteins and enzymes involved in carbohydrate metabolism. Regenerating Xenopus limbs differed significantly from axolotl regenerating limbs in several ways: deficiency in the inositol phosphate/diacylglycerol signaling pathway, down regulation of Wnt signaling, up regulation of extracellular matrix (ECM) proteins and proteins involved in chondrocyte differentiation, lack of expression of a key cell cycle protein, ecotropic viral integration site 5 (EVI5), that blocks mitosis in the axolotl, and the expression of several patterning proteins not seen in the axolotl that may dorsalize the fibroblastema. CONCLUSIONS: We have characterized global protein expression during fibroblastema formation after amputation of the Xenopus froglet hindlimb and identified several differences that lead to signaling deficiency, failure to retard mitosis, premature chondrocyte differentiation, and failure of dorsoventral axial asymmetry. These differences point to possible interventions to improve blastema formation and pattern formation in the froglet limb.


Asunto(s)
Ambystoma/metabolismo , Miembro Posterior/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Ambystoma/crecimiento & desarrollo , Animales , Regeneración Ósea/fisiología , Cromatografía Líquida de Alta Presión , Regulación del Desarrollo de la Expresión Génica , Espectrometría de Masas , Proteómica , Transducción de Señal , Proteínas de Xenopus/genética , Xenopus laevis/crecimiento & desarrollo
5.
Hum Mol Genet ; 21(7): 1592-603, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22180459

RESUMEN

The dystrophin-glycoprotein complex connects myofibers with extracellular matrix laminin. In Duchenne muscular dystrophy, this linkage system is absent and the integrity of muscle fibers is compromised. One potential therapy for addressing muscular dystrophy is to augment the amount of α7ß1 integrin, the major laminin-binding integrin in skeletal muscle. Whereas transgenic over-expression of α7 chain may alleviate development of muscular dystrophy and extend the lifespan of severely dystrophic mdx/utrn(-/-) mice, further enhancing levels of α7 chain provided little additional membrane integrin and negligible additional improvement in mdx mice. We demonstrate here that normal levels of ß1 chain limit formation of integrin heterodimer and that increasing ß1D chain in mdx mice results in more functional integrin at the sarcolemma, more matrix laminin and decreased damage of muscle fibers. Moreover, increasing the amount of ß1D chain in vitro enhances transcription of α7 integrin and α2 laminin genes and the amounts of these proteins. Thus manipulation of ß1D integrin expression offers a novel approach to enhance integrin-mediated therapy for muscular dystrophy.


Asunto(s)
Integrina beta1/genética , Integrinas/metabolismo , Laminina/metabolismo , Distrofias Musculares/metabolismo , Sarcolema/metabolismo , Animales , Antígenos CD/biosíntesis , Antígenos CD/genética , Antígenos CD/metabolismo , Línea Celular , Retículo Endoplásmico/metabolismo , Cadenas alfa de Integrinas/biosíntesis , Cadenas alfa de Integrinas/genética , Cadenas alfa de Integrinas/metabolismo , Integrina beta1/metabolismo , Laminina/biosíntesis , Laminina/genética , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Ratones Transgénicos , Fibras Musculares Esqueléticas/metabolismo , Distrofias Musculares/prevención & control , Sarcolema/ultraestructura
6.
Curr Top Microbiol Immunol ; 367: 133-59, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23224711

RESUMEN

Skeletal muscle possesses a robust innate capability for repair of tissue damage. Natural repair of muscle damage is a stepwise process that requires the coordinated activity of a number of cell types, including infiltrating macrophages, resident myogenic and non-myogenic stem cells, and connective tissue fibroblasts. Despite the proficiency of this intrinsic repair capability, severe injuries that result in significant loss of muscle tissue overwhelm the innate repair process and require intervention if muscle function is to be restored. Recent advances in stem cell biology, regenerative medicine, and materials science have led to attempts at developing tissue engineering-based methods for repairing severe muscle defects. Muscle tissue also plays a role in the ability of tailed amphibians to regenerate amputated limbs through epimorphic regeneration. Muscle contributes adult stem cells to the amphibian regeneration blastema, but it can also contribute blastemal cells through the dedifferentiation of multinucleate myofibers into mononuclear precursors. This fascinating plasticity and its contributions to limb regeneration have prompted researchers to investigate the potential for mammalian muscle to undergo dedifferentiation. Several works have shown that mammalian myotubes can be fragmented into mononuclear cells and induced to re-enter the cell cycle, but mature myofibers are resistant to fragmentation. However, recent works suggest that there may be a path to inducing fragmentation of mature myofibers into proliferative multipotent cells with the potential for use in muscle tissue engineering and regenerative therapies.


Asunto(s)
Anfibios/fisiología , Extremidades/fisiología , Músculo Esquelético/fisiología , Regeneración/fisiología , Animales , Desdiferenciación Celular , Macrófagos/fisiología , Fibras Musculares Esqueléticas/citología , Ingeniería de Tejidos
7.
Curr Top Microbiol Immunol ; 367: 113-32, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23239235

RESUMEN

Bone has the natural ability to remodel and repair. Fractures and small noncritical size bone defects undergo regenerative healing via coordinated concurrent development of skeletal and vascular elements in a soft cartilage callus environment. Within this environment bone regeneration recapitulates many of the same cellular and molecular mechanisms that form embryonic bone. Angiogenesis is intimately involved with embryonic bone formation and with both endochondral and intramembranous bone formation in differentiated bone. During bone regeneration osteogenic cells are first associated with vascular tissue in the adjacent periosteal space or the adjacent injured marrow cavity that houses endosteal blood vessels. Critical size bone defects cannot heal without the assistance of therapeutic aids or materials designed to encourage bone regeneration. We discuss the prospects for using synthetic hydrogels in a bioengineering approach to repair critical size bone defects. Hydrogel scaffolds can be designed and fabricated to potentially trigger the same bone morphogenetic cascade that heals bone fractures and noncritical size defects naturally. Lastly, we introduce adult Xenopus laevis hind limb as a novel small animal model system for bone regeneration research. Xenopus hind limbs have been used successfully to screen promising scaffolds designed to heal critical size bone defects.


Asunto(s)
Regeneración Ósea , Curación de Fractura , Animales , Miembro Posterior/fisiología , Humanos , Osteogénesis , Xenopus laevis/fisiología
8.
bioRxiv ; 2024 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-38328159

RESUMEN

Optimal imaging strategies remain underdeveloped to maximize information for fluorescence microscopy while minimizing the harm to fragile living systems. Taking hint from the supercontinuum generation in ultrafast laser physics, we generated supercontinuum fluorescence from untreated unlabeled live samples before nonlinear photodamage onset. Our imaging achieved high-content cell phenotyping and tissue histology, identified bovine embryo polarization, quantified aging-related stress across cell types and species, demystified embryogenesis before and after implantation, sensed drug cytotoxicity in real-time, scanned brain area for targeted patching, optimized machine learning to track small moving organisms, induced two-photon phototropism of leaf chloroplasts under two-photon photosynthesis, unraveled microscopic origin of autumn colors, and interrogated intestinal microbiome. The results enable a facility-type microscope to freely explore vital molecular biology across life sciences.

9.
Animals (Basel) ; 13(13)2023 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-37443900

RESUMEN

Approximately 80% of the ~1.5 million bovine embryos transferred in 2021 were in vitro produced. However, only ~27% of the transferred IVP embryos will result in live births. The ~73% pregnancy failures are partly due to transferring poor-quality embryos, a result of erroneous stereomicroscopy-based morphological evaluation, the current method of choice for pre-transfer embryo evaluation. Numerous microscopic (e.g., differential interference contrast, electron, fluorescent, time-lapse, and artificial-intelligence-based microscopy) and non-microscopic (e.g., genomics, transcriptomics, epigenomics, proteomics, metabolomics, and nuclear magnetic resonance) methodologies have been tested to find an embryo evaluation technique that is superior to morphologic evaluation. Many of these research tools can accurately determine embryo quality/viability; however, most are invasive, expensive, laborious, technically sophisticated, and/or time-consuming, making them futile in the context of in-field embryo evaluation. However accurate they may be, using complex methods, such as RNA sequencing, SNP chips, mass spectrometry, and multiphoton microscopy, at thousands of embryo production/collection facilities is impractical. Therefore, future research is warranted to innovate field-friendly, simple benchtop tests using findings already available, particularly from omics-based research methodologies. Time-lapse monitoring and artificial-intelligence-based automated image analysis also have the potential for accurate embryo evaluation; however, further research is warranted to innovate economically feasible options for in-field applications.

10.
BMC Bioinformatics ; 12: 80, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21418574

RESUMEN

BACKGROUND: Studies on amphibian limb regeneration began in the early 1700's but we still do not completely understand the cellular and molecular events of this unique process. Understanding a complex biological process such as limb regeneration is more complicated than the knowledge of the individual genes or proteins involved. Here we followed a systems biology approach in an effort to construct the networks and pathways of protein interactions involved in formation of the accumulation blastema in regenerating axolotl limbs. RESULTS: We used the human orthologs of proteins previously identified by our research team as bait to identify the transcription factor (TF) pathways and networks that regulate blastema formation in amputated axolotl limbs. The five most connected factors, c-Myc, SP1, HNF4A, ESR1 and p53 regulate ~50% of the proteins in our data. Among these, c-Myc and SP1 regulate 36.2% of the proteins. c-Myc was the most highly connected TF (71 targets). Network analysis showed that TGF-ß1 and fibronectin (FN) lead to the activation of these TFs. We found that other TFs known to be involved in epigenetic reprogramming, such as Klf4, Oct4, and Lin28 are also connected to c-Myc and SP1. CONCLUSIONS: Our study provides a systems biology approach to how different molecular entities inter-connect with each other during the formation of an accumulation blastema in regenerating axolotl limbs. This approach provides an in silico methodology to identify proteins that are not detected by experimental methods such as proteomics but are potentially important to blastema formation. We found that the TFs, c-Myc and SP1 and their target genes could potentially play a central role in limb regeneration. Systems biology has the potential to map out numerous other pathways that are crucial to blastema formation in regeneration-competent limbs, to compare these to the pathways that characterize regeneration-deficient limbs and finally, to identify stem cell markers in regeneration.


Asunto(s)
Extremidades/fisiología , Proteómica , Regeneración/genética , Factores de Transcripción/genética , Ambystoma mexicanum/genética , Ambystoma mexicanum/fisiología , Animales , ADN Complementario/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Factor 4 Similar a Kruppel , Factor de Crecimiento Transformador beta1/genética
11.
Biofabrication ; 14(1)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34663761

RESUMEN

Regenerative medicine approaches for massive craniomaxillofacial (CMF) bone defects face challenges associated with the scale of missing bone, the need for rapid graft-defect integration, and challenges related to inflammation and infection. Mineralized collagen scaffolds have been shown to promote mesenchymal stem cell osteogenesis due to their porous nature and material properties, but are mechanically weak, limiting surgical practicality. Previously, these scaffolds were combined with 3D-printed polycaprolactone (PCL) mesh to form a scaffold-mesh composite to increase strength and promote bone formation in sub-critical sized porcine ramus defects. Here, we compare the performance of mineralized collagen-PCL composites to the PCL mesh in a critical-sized porcine ramus defect model. While there were no differences in overall healing response between groups, our data demonstrated broadly variable metrics of healing regarding new bone infiltration and fibrous tissue formation. Abscesses were present surrounding some implants and PCL polymer was still present after 9-10 months of implantation. Overall, while there was limited successful healing, with 2 of 22 implants showed substantial levels of bone regeneration, and others demonstrating some form of new bone formation, the results suggest targeted improvements to improve repair of large animal models to more accurately represent CMF bone healing. Notably, strategies to increase osteogenesis throughout the implant, modulate the immune system to support repair, and employ shape-fitting tactics to avoid implant micromotion and resultant fibrosis. Improvements to the mineralized collagen scaffolds involve changes in pore size and shape to increase cell migration and osteogenesis and inclusion or delivery of factors to aid vascular ingrowth and bone regeneration.


Asunto(s)
Materiales Biocompatibles , Andamios del Tejido , Animales , Materiales Biocompatibles/farmacología , Regeneración Ósea , Colágeno/farmacología , Osteogénesis , Poliésteres , Porcinos
12.
BMC Biol ; 7: 83, 2009 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-19948009

RESUMEN

BACKGROUND: Following amputation, urodele salamander limbs reprogram somatic cells to form a blastema that self-organizes into the missing limb parts to restore the structure and function of the limb. To help understand the molecular basis of blastema formation, we used quantitative label-free liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS)-based methods to analyze changes in the proteome that occurred 1, 4 and 7 days post amputation (dpa) through the mid-tibia/fibula of axolotl hind limbs. RESULTS: We identified 309 unique proteins with significant fold change relative to controls (0 dpa), representing 10 biological process categories: (1) signaling, (2) Ca2+ binding and translocation, (3) transcription, (4) translation, (5) cytoskeleton, (6) extracellular matrix (ECM), (7) metabolism, (8) cell protection, (9) degradation, and (10) cell cycle. In all, 43 proteins exhibited exceptionally high fold changes. Of these, the ecotropic viral integrative factor 5 (EVI5), a cell cycle-related oncoprotein that prevents cells from entering the mitotic phase of the cell cycle prematurely, was of special interest because its fold change was exceptionally high throughout blastema formation. CONCLUSION: Our data were consistent with previous studies indicating the importance of inositol triphosphate and Ca2+ signaling in initiating the ECM and cytoskeletal remodeling characteristic of histolysis and cell dedifferentiation. In addition, the data suggested that blastema formation requires several mechanisms to avoid apoptosis, including reduced metabolism, differential regulation of proapoptotic and antiapoptotic proteins, and initiation of an unfolded protein response (UPR). Since there is virtually no mitosis during blastema formation, we propose that high levels of EVI5 function to arrest dedifferentiated cells somewhere in the G1/S/G2 phases of the cell cycle until they have accumulated under the wound epidermis and enter mitosis in response to neural and epidermal factors. Our findings indicate the general value of quantitative proteomic analysis in understanding the regeneration of complex structures.


Asunto(s)
Ambystoma/fisiología , Extremidades/fisiología , Proteómica , Regeneración/fisiología , Amputación Quirúrgica , Animales , Señalización del Calcio/genética , Cromatografía Líquida de Alta Presión , Matriz Extracelular/metabolismo , Extremidades/cirugía , Inositol 1,4,5-Trifosfato/metabolismo , Mapeo Peptídico , Espectrometría de Masas en Tándem , Cicatrización de Heridas
13.
Regen Biomater ; 7(3): 247-258, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32523727

RESUMEN

Defects in craniofacial bones occur congenitally, after high-energy impacts, and during the course of treatment for stroke and cancer. These injuries are difficult to heal due to the overwhelming size of the injury area and the inflammatory environment surrounding the injury. Significant inflammatory response after injury may greatly inhibit regenerative healing. We have developed mineralized collagen scaffolds that can induce osteogenic differentiation and matrix biosynthesis in the absence of osteogenic media or supplemental proteins. The amniotic membrane is derived from placentas and has been recently investigated as an extracellular matrix to prevent chronic inflammation. Herein, we hypothesized that a mineralized collagen-amnion composite scaffold could increase osteogenic activity in the presence of inflammatory cytokines. We report mechanical properties of a mineralized collagen-amnion scaffold and investigated osteogenic differentiation and mineral deposition of porcine adipose-derived stem cells within these scaffolds as a function of inflammatory challenge. Incorporation of amniotic membrane matrix promotes osteogenesis similarly to un-modified mineralized collagen scaffolds, and increases in mineralized collagen-amnion scaffolds under inflammatory challenge. Together, these findings suggest that a mineralized collagen-amnion scaffold may provide a beneficial environment to aid craniomaxillofacial bone repair, especially in the course of defects presenting significant inflammatory complications.

14.
Laryngoscope ; 128(7): E251-E257, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29668079

RESUMEN

OBJECTIVES/HYPOTHESIS: Reconstruction of craniofacial cartilagenous defects are among the most challenging surgical procedures in facial plastic surgery. Bioengineered craniofacial cartilage holds immense potential to surpass current reconstructive options, but limitations to clinical translation exist. We endeavored to determine the viability of utilizing adipose-derived stem cell-chondrocyte co-culture and three-dimensional (3D) printing to produce 3D bioscaffolds for cartilage tissue engineering. We describe a feasibility study revealing a novel approach for cartilage tissue engineering with in vitro and in vivo animal data. METHODS: Porcine adipose-derived stem cells and chondrocytes were isolated and co-seeded at 1:1, 2:1, 5:1, 10:1, and 0:1 experimental ratios in a hyaluronic acid/collagen hydrogel in the pores of 3D-printed polycaprolactone scaffolds to form 3D bioscaffolds for cartilage tissue engineering. Bioscaffolds were cultured in vitro without growth factors for 4 weeks and then implanted into the subcutaneous tissue of athymic rats for an additional 4 weeks before sacrifice. Bioscaffolds were subjected to histologic, immunohistochemical, and biochemical analysis. RESULTS: Successful production of cartilage was achieved using a co-culture model of adipose-derived stem cells and chondrocytes without the use of exogenous growth factors. Histology demonstrated cartilage growth for all experimental ratios at the post-in vivo time point confirmed with type II collagen immunohistochemistry. There was no difference in sulfated-glycosaminoglycan production between experimental groups. CONCLUSION: Tissue-engineered cartilage was successfully produced on 3D-printed bioresorbable scaffolds using an adipose-derived stem cell and chondrocyte co-culture technique. This potentiates co-culture as a solution for several key barriers to a clinically translatable cartilage tissue engineering process. LEVEL OF EVIDENCE: NA. Laryngoscope, 128:E251-E257, 2018.


Asunto(s)
Cartílago/citología , Condrocitos/citología , Técnicas de Cocultivo/métodos , Células Madre Mesenquimatosas/citología , Ingeniería de Tejidos/métodos , Animales , Anomalías Craneofaciales , Estudios de Factibilidad , Inmunohistoquímica , Impresión Tridimensional , Ratas , Porcinos , Andamios del Tejido
15.
Tissue Eng Part A ; 24(11-12): 943-954, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29264958

RESUMEN

A tissue engineering approach to address craniofacial defects requires a biomaterial that balances macro-scale mechanical stiffness and strength with the micron-scale features that promote cell expansion and tissue biosynthesis. Such criteria are often in opposition, leading to suboptimal mechanical competence or bioactivity. We report the use of a multiscale composite biomaterial that integrates a polycaprolactone (PCL) reinforcement structure with a mineralized collagen-glycosaminoglycan scaffold to circumvent conventional tradeoffs between mechanics and bioactivity. The composite promotes activation of the canonical bone morphogenetic protein 2 (BMP-2) pathway and subsequent mineralization of adipose-derived stem cells in the absence of supplemental BMP-2 or osteogenic media. We subsequently examined new bone infill in the acellular composite, scaffold alone, or PCL support in 10 mm dia. ramus mandibular defects in Yorkshire pigs. We report an analytical approach to quantify radial, angular, and depth bone infill from micro-computed tomography data. The collagen-PCL composite showed improved overall infill, and significantly increased radial and angular bone infill versus the PCL cage alone. Bone infill was further enhanced in the composite for defects that penetrated the medullary cavity, suggesting recruitment of marrow-derived cells. These results indicate a multiscale mineralized collagen-PCL composite offers strategic advantages for regenerative repair of craniofacial bone defects.


Asunto(s)
Colágeno/química , Enfermedades Mandibulares/tratamiento farmacológico , Poliésteres/química , Animales , Huesos/patología , Enfermedades Mandibulares/metabolismo , Porcinos , Cicatrización de Heridas/efectos de los fármacos
16.
Mol Biol Cell ; 13(7): 2347-59, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12134074

RESUMEN

The sarcolemma of fast-twitch muscle is organized into "costameres," structures that are oriented transversely, over the Z and M lines of nearby myofibrils, and longitudinally, to form a rectilinear lattice. Here we examine the role of desmin, the major intermediate filament protein of muscle in organizing costameres. In control mouse muscle, desmin is enriched at the sarcolemmal domains that lie over nearby Z lines and that also contain beta-spectrin. In tibialis anterior muscle from mice lacking desmin due to homologous recombination, most costameres are lost. In myofibers from desmin -/- quadriceps, by contrast, most costameric structures are stable. Alternatively, Z line domains may be lost, whereas domains oriented longitudinally or lying over M lines are retained. Experiments with pan-specific antibodies to intermediate filament proteins and to cytokeratins suggest that control and desmin -/- muscles express similar levels of cytokeratins. Cytokeratins concentrate at the sarcolemma at all three domains of costameres when the latter are retained in desmin -/- muscle and redistribute with beta-spectrin at the sarcolemma when costameres are lost. Our results suggest that desmin associates with and selectively stabilizes the Z line domains of costameres, but that cytokeratins associate with all three domains of costameres, even in the absence of desmin.


Asunto(s)
Citoesqueleto/metabolismo , Desmina/metabolismo , Queratinas/metabolismo , Músculo Esquelético/fisiología , Músculo Esquelético/ultraestructura , Sarcolema/metabolismo , Animales , Desmina/genética , Inmunohistoquímica , Filamentos Intermedios/metabolismo , Ratones , Ratones Endogámicos , Ratones Noqueados , Modelos Anatómicos , Fibras Musculares de Contracción Rápida/citología , Fibras Musculares de Contracción Rápida/metabolismo , Proteínas Musculares/metabolismo , Espectrina/metabolismo
17.
Free Radic Biol Med ; 110: 206-218, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28629836

RESUMEN

Oxidative stress has been linked strongly to cell death and cardiac remodeling processes, all hallmarks of heart failure. Mice deficient for desmin (des-/-), the major muscle specific intermediate filament protein, develop dilated cardiomyopathy and heart failure characterized by mitochondrial defects and cardiomyocyte death. The cellular and biochemical alterations in the hearts of these mice strongly suggest that oxidative stress is one of the mechanisms contributing to the pathogenesis of the phenotype. Recently, we showed that indeed the desmin deficient cardiomyocytes are under increased oxidative stress. In order to verify these findings in vivo, we generated transgenic animals overexpressing SOD2 (MnSOD) and/or catalase in the heart and crossed them with des-/- mice, thus allowing us to evaluate the contribution of oxidative injury in inherited cardiomyopathies, as well as the therapeutic potential of antioxidant strategies. Moderate MnSOD and/or catalase overexpression in des-/- hearts leads to a marked decrease in intracellular reactive oxygen species (ROS), ameliorates mitochondrial and other ultrastructural defects, minimizes myocardial degeneration and leads to a significant improvement of cardiac function. Importantly, catalase overexpression increased the 50% survival rate of des-/- mice in an obligatory exercise to 100%. In contrast, MnSOD overexpression enhanced the lethality of des-/- mice, underscoring the importance of a fine balanced cellular redox status. Overall, the present study supports the contribution of oxidative stress in the development of des-/- cardiomyopathy and points to a well-considered antioxidant treatment as therapeutic for cardiomyopathies.


Asunto(s)
Cardiomiopatía Dilatada/genética , Catalasa/genética , Desmina/genética , Insuficiencia Cardíaca/genética , Miocitos Cardíacos/enzimología , Superóxido Dismutasa/genética , Animales , Cardiomiopatía Dilatada/enzimología , Cardiomiopatía Dilatada/mortalidad , Cardiomiopatía Dilatada/patología , Catalasa/metabolismo , Citosol/enzimología , Desmina/deficiencia , Regulación de la Expresión Génica , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/mortalidad , Insuficiencia Cardíaca/patología , Ratones , Ratones Noqueados , Mitocondrias/enzimología , Miocitos Cardíacos/patología , Estrés Oxidativo , Condicionamiento Físico Animal , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Superóxido Dismutasa/metabolismo , Análisis de Supervivencia
18.
J Clin Invest ; 123(10): 4294-308, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24091324

RESUMEN

Ischemic damage is recognized to cause cardiomyocyte (CM) death and myocardial dysfunction, but the role of cell-matrix interactions and integrins in this process has not been extensively studied. Expression of α7ß1D integrin, the dominant integrin in normal adult CMs, increases during ischemia/reperfusion (I/R), while deficiency of ß1 integrins increases ischemic damage. We hypothesized that the forced overexpression of integrins on the CM would offer protection from I/R injury. Tg mice with CM-specific overexpression of integrin α7ß1D exposed to I/R had a substantial reduction in infarct size compared with that of α5ß1D-overexpressing mice and WT littermate controls. Using isolated CMs, we found that α7ß1D preserved mitochondrial membrane potential during hypoxia/reoxygenation (H/R) injury via inhibition of mitochondrial Ca2+ overload but did not alter H/R effects on oxidative stress. Therefore, we assessed Ca2+ handling proteins in the CM and found that ß1D integrin colocalized with ryanodine receptor 2 (RyR2) in CM T-tubules, complexed with RyR2 in human and rat heart, and specifically bound to RyR2 amino acids 165-175. Integrins stabilized the RyR2 interdomain interaction, and this stabilization required integrin receptor binding to its ECM ligand. These data suggest that α7ß1D integrin modifies Ca2+ regulatory pathways and offers a means to protect the myocardium from ischemic injury.


Asunto(s)
Integrinas/metabolismo , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Hipoxia de la Célula , Células Cultivadas , Humanos , Integrinas/química , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/patología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley , Canal Liberador de Calcio Receptor de Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
19.
Tissue Eng Part A ; 17(5-6): 691-701, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20929280

RESUMEN

We used the tarsus of an adult Xenopus laevis frog as an in vivo load-bearing model to study the regeneration of critical-size defects (CSD) in long bones. We found the CSD for this bone to be about 35% of the tarsus length. To promote regeneration, we implanted biocompatible 1,6 hexanediol diacrylate scaffolds soaked with bone morphogenetic proteins-4 and vascular endothelial growth factors. In contrast to studies that use scaffolds as templates for bone formation, we used scaffolds as a growth factor delivery vehicle to promote cartilage-to-bone regeneration. Defects in control frogs were filled with scaffolds lacking growth factors. The limbs were harvested at a series of time points ranging from 3 weeks to 6 months after implantation and evaluated using micro-computed tomography and histology. In frogs treated with growth factor-loaded scaffolds, we observed a cartilage-to-bone regeneration in the skeletal defect. Five out of eight defects were completely filled with cartilage by 6 weeks. Blood vessels had invaded the cartilage, and bone was beginning to form in ossifying centers. By 3 months, these processes were well advanced, and extensive ossification was observed in 6-month samples. In contrast, the defects in control frogs showed only formation of fibrous scar tissue. This study demonstrates the utility of a Xenopus model system for tissue engineering research and that the normal in vivo mechanism of endochondral bone development and fracture repair can be mimicked in the repair of CSD with scaffolds used as growth factor delivery mechanisms.


Asunto(s)
Proteína Morfogenética Ósea 4/farmacología , Regeneración Ósea/efectos de los fármacos , Huesos/efectos de los fármacos , Huesos/patología , Modelos Animales , Factor A de Crecimiento Endotelial Vascular/farmacología , Cicatrización de Heridas/efectos de los fármacos , Animales , Huesos/cirugía , Humanos , Implantes Experimentales , Microscopía Electrónica de Rastreo , Porosidad/efectos de los fármacos , Andamios del Tejido/química , Xenopus laevis
20.
J Cell Biol ; 181(5): 761-75, 2008 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-18519735

RESUMEN

We explored the involvement of the muscle-specific intermediate filament protein desmin in the model of tumor necrosis factor alpha (TNF-alpha)-induced cardiomyopathy. We demonstrate that in mice overexpressing TNF-alpha in the heart (alpha-myosin heavy chain promoter-driven secretable TNF-alpha [MHCsTNF]), desmin is modified, loses its intercalated disk (ID) localization, and forms aggregates that colocalize with heat shock protein 25 and ubiquitin. Additionally, other ID proteins such as desmoplakin and beta-catenin show similar localization changes in a desmin-dependent fashion. To address underlying mechanisms, we examined whether desmin is a substrate for caspase-6 in vivo as well as the implications of desmin cleavage in MHCsTNF mice. We generated transgenic mice with cardiac-restricted expression of a desmin mutant (D263E) and proved that it is resistant to caspase cleavage in the MHCsTNF myocardium. The aggregates are diminished in these mice, and D263E desmin, desmoplakin, and beta-catenin largely retain their proper ID localization. Importantly, D263E desmin expression attenuated cardiomyocyte apoptosis, prevented left ventricular wall thinning, and improved the function of MHCsTNF hearts.


Asunto(s)
Desmina/fisiología , Insuficiencia Cardíaca/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis , Cardiomiopatías/patología , Caspasa 6/metabolismo , Desmina/metabolismo , Desmoplaquinas/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Mutación , Miocardio/metabolismo , Ubiquitina/química , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA