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.
Nat Microbiol ; 7(12): 1951-1955, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36344621

RESUMEN

The ongoing monkeypox virus (MPXV) outbreak is the largest ever recorded outside of Africa. We isolated and sequenced a virus from the first clinical MPXV case diagnosed in France (May 2022). We report that tecovirimat (ST-246), a US Food and Drug Administration approved drug, is efficacious against this isolate in vitro at nanomolar concentrations, whereas cidofovir is only effective at micromolar concentrations. Our results support the use of tecovirimat in ongoing human clinical trials.


Asunto(s)
Monkeypox virus , Mpox , Estados Unidos , Humanos , Mpox/tratamiento farmacológico , Isoindoles/farmacología , Isoindoles/uso terapéutico , Benzamidas/farmacología , Benzamidas/uso terapéutico
2.
Biomedicines ; 10(6)2022 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-35740407

RESUMEN

Biomaterial use is a promising approach to facilitate wound healing of the bone tissue. Biomaterials induce the formation of membrane capsules and the recruitment of different types of macrophages. Macrophages are immune cells that produce diverse combinations of cytokines playing an important role in bone healing and regeneration, but the exact mechanism remains to be studied. Our work aimed to identify in vivo macrophages in the Masquelet induced membrane in a rat model. Most of the macrophages in the damaged area were M2-like, with smaller numbers of M1-like macrophages. In addition, high expression of IL-1ß and IL-6 cytokines were detected in the membrane region by RT-qPCR. Using an innovative combination of two hybridization techniques (in situ hybridization and in situ hybridization chain reaction (in situ HCR)), M2b-like macrophages were identified for the first time in cryosections of non-decalcified bone. Our work has also demonstrated that microspectroscopical analysis is essential for macrophage characterization, as it allows the discrimination of fluorescence and autofluorescence. Finally, this work has revealed the limitations of immunolabelling and the potential of in situ HCR to provide valuable information for in vivo characterization of macrophages.

3.
Biomedicines ; 10(2)2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35203694

RESUMEN

Bone is a very complex tissue that is constantly changing throughout the lifespan. The precise mechanism of bone regeneration remains poorly understood. Large bone defects can be caused by gunshot injury, trauma, accidents, congenital anomalies and tissue resection due to cancer. Therefore, understanding bone homeostasis and regeneration has considerable clinical and scientific importance in the development of bone therapy. Macrophages are well known innate immune cells secreting different combinations of cytokines and their role in bone regeneration during bone healing is essential. Here, we present a method to identify mRNA transcripts in cryosections of non-decalcified rat bone using in situ hybridization and hybridization chain reaction to explore gene expression in situ for better understanding the gene expression of the bone tissues.

4.
Hypoxia (Auckl) ; 7: 41-52, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31440522

RESUMEN

PURPOSE: Bone marrow response to an organismal stress is made by orchestrating the interplay between hematopoietic stem/progenitor cells (HSPCs) and mesenchymal stromal cells (MSCs). Neither the cellular nor the molecular factors that regulate this process are fully understood, especially since this mechanism probably varies depending on the type of stress. Herein, we explored the differentiation and fate of MSCs and HSPCs in mice challenged with a hematopoietic stress or a mechanical stress applied separately or in combination. METHODS: Mice were subjected to 4 days of hypobaric hypoxia (hematopoietic challenge) and/or 7 days of hindlimb suspension (stromal challenge) and then sacrificed for blood and bone collection. Using hematological measurements, colony-forming unit assays, bone histomorphometry and array-based multiplex ELISA analysis, we evaluated challenge influences on both MSC and HSPC mobilization, differentiation (osteoblasts, osteoclasts, and mature blood cells) and fate. RESULTS: We found that hypoxia leads to HSPC mobilization and that an imbalance between bone formation and bone resorption accounts for this mobilization. Whilst suspension is also associated with an imbalance between bone formation and bone resorption, it does not induce HSPC mobilization. Then, we revealed cellular interactions by combining hematopoietic and stromal challenges together in mice. We showed that the hypoxia-driven HSPC mobilization is moderated by suspension. Moreover, when applied in a hypoxic environment, suspension offsets bone imbalance. We identified stroma cell-derived factors MIP-1α, HGF and SDF-1 as potent molecular key players sustaining interactions between hindlimb suspension and hypobaric hypoxia. CONCLUSION: Taken together, our data highlight the benefit of combining different types of stress to better understand the interplay between MSCs and HSPCs.

5.
PLoS One ; 12(8): e0182454, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28854256

RESUMEN

We previously reported the development of a new acquired neurogenic HO (NHO) mouse model, combining spinal cord transection (SCI) and chemical muscle injury. Pathological mechanisms responsible for ectopic osteogenesis after central neurological damage are still to be elucidated. In this study, we first hypothesized that peripheral nervous system (PNS) might convey pathological signals from injured spinal cord to muscles in NHO mouse model. Secondly, we sought to determine whether SCI could lead to intramuscular modifications of BMP2 signaling pathways. Twenty one C57Bl6 mice were included in this protocol. Bilateral cardiotoxin (CTX) injection in hamstring muscles was associated with a two-stage surgical procedure, combining thoracic SCI with unilateral peripheral denervation. Volumes of HO (Bone Volume, BV) were measured 28 days after surgery using micro-computed tomography imaging techniques and histological analyses were made to confirm intramuscular osteogenesis. Volume comparisons were conducted between right and left hind limb of each animal, using a Wilcoxon signed rank test. Quantitative polymerase chain reaction (qPCR) was performed to explore intra muscular expression of BMP2, Alk3 and Id1. Nineteen mice survive the complete SCI and peripheral denervation procedure. When CTX injections were done right after surgery (n = 7), bilateral HO were detected in all animals after 28 days. Micro-CT measurements showed significantly increased BV in denervated paws (1.47 mm3 +/- 0.5) compared to contralateral sides (0.56 mm3 +/-0.4), p = 0.03. When peripheral denervation and CTX injections were performed after sham SCI surgery (n = 6), bilateral HO were present in three mice at day 28. Quantitative PCR analyses showed no changes in intra muscular BMP2 expression after SCI as compared to control mice (shamSCI). Peripheral denervation can be reliably added to spinal cord transection in NHO mouse model. This new experimental design confirms that neuro inflammatory mechanisms induced by central or peripheral nervous system injury plays a key role in triggering ectopic osteogenesis.


Asunto(s)
Músculos/patología , Osificación Heterotópica/patología , Traumatismos de la Médula Espinal/patología , Médula Espinal/patología , Animales , Proteína Morfogenética Ósea 2/análisis , Proteínas Cardiotóxicas de Elápidos , Desnervación , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos C57BL , Músculos/efectos de los fármacos , Músculos/inervación , Osificación Heterotópica/inducido químicamente , Osificación Heterotópica/diagnóstico por imagen , Osificación Heterotópica/etiología , Médula Espinal/diagnóstico por imagen , Médula Espinal/efectos de los fármacos , Traumatismos de la Médula Espinal/inducido químicamente , Traumatismos de la Médula Espinal/diagnóstico por imagen , Traumatismos de la Médula Espinal/etiología , Microtomografía por Rayos X
6.
J Mater Sci Mater Med ; 28(2): 35, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28110459

RESUMEN

Research in bone tissue engineering is focused on the development of alternatives to autologous bone grafts for bone reconstruction. Although multiple stem cell-based products and biomaterials are currently being investigated, comparative studies are rarely achieved to evaluate the most appropriate approach in this context. Here, we aimed to compare different clinically relevant bone tissue engineering methods and evaluated the kinetic repair and the bone healing efficiency supported by mesenchymal stem cells and two different biomaterials, a new hydrogel scaffold and a commercial hydroxyapatite/tricalcium phosphate ceramic, alone or in combination.Syngeneic mesenchymal stem cells (5 × 105) and macroporous biphasic calcium phosphate ceramic granules (Calciresorb C35®, Ceraver) or porous pullulan/dextran-based hydrogel scaffold were implanted alone or combined in a drilled-hole bone defect in rats. Using quantitative microtomography measurements and qualitative histological examinations, their osteogenic properties were evaluated 7, 30, and 90 days after implantation. Three months after surgery, only minimal repair was evidenced in control rats while newly mineralized bone was massively observed in animals treated with either hydrogels (bone volume/tissue volume = 20%) or ceramics (bone volume/tissue volume = 26%). Repair mechanism and resorption kinetics were strikingly different: rapidly-resorbed hydrogels induced a dense bone mineralization from the edges of the defect while ceramics triggered newly woven bone formation in close contact with the ceramic surface that remained unresorbed. Delivery of mesenchymal stem cells in combination with these biomaterials enhanced both bone healing (>20%) and neovascularization after 1 month, mainly in hydrogel.Osteogenic and angiogenic properties combined with rapid resorption make hydrogels a promising alternative to ceramics for bone repair by cell therapy.


Asunto(s)
Regeneración Ósea , Fosfatos de Calcio/química , Hidrogeles/química , Células Madre Mesenquimatosas/citología , Polisacáridos/química , Andamios del Tejido/química , Animales , Materiales Biocompatibles/química , Células de la Médula Ósea/citología , Resorción Ósea , Trasplante Óseo/métodos , Cerámica/química , Fémur/patología , Masculino , Neovascularización Patológica , Ratas , Ratas Endogámicas Lew , Ingeniería de Tejidos/métodos , Microtomografía por Rayos X
7.
Physiol Rep ; 3(8)2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26265754

RESUMEN

Within the framework of earlier publications, we have consistently dedicated our investigations to eliciting the effects of both seasonal vitamin D deficiency and submarine-induced hypercapnia on serum parameters for acid-base balance and bone metabolism in submariners over a 2-month winter (WP) or summer (SP) patrols. The latest findings reported herein, contribute further evidence with regard to overall physiological regulations in the same submariner populations that underwent past scrutiny. Hence, urine and blood samples were collected in WP and SP submariners at control prepatrol time as well as on submarine patrol days 20, 41, and 58. Several urine and serum metabolic markers were quantified, namely, deoxypyridinoline (DPD), lactate, albumin, creatinine, nonesterified fatty acids (NEFA), and ionized sodium (Na(+)) or potassium (K(+)), with a view to assessing bone, muscle, liver, or kidney metabolisms. We evidenced bone metabolism alteration (urine DPD, calcium, and phosphorus) previously recorded in submarine crewmembers under prolonged patrols. We also highlighted transitory modifications in liver metabolism (serum albumin) occurring within the first 20 days of submersion. We further evidenced changes in submariners' renal physiology (serum creatinine) throughout the entire patrol time span. Measurements of ionic homeostasis (serum Na(+) and K(+)) displayed potential seasonal impact over active ionic pumps in submariners. Finally, there is some evidence that submersion provides beneficial conditions prone to fend off seasonal lactic acidosis (serum lactate) detected in WP submariners.

8.
Stem Cells Transl Med ; 3(8): 958-68, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24944208

RESUMEN

We investigated the effects of respiratory hypobaric hypoxia on femoral bone-defect repair in mice because hypoxia is believed to influence both mesenchymal stromal cell (MSC) and hematopoietic stem cell mobilization, a process involved in the bone-healing mechanism. To mimic conditions of non-weight-bearing limb immobilization in patients suffering from bone trauma, our hypoxic mouse model was further subjected to hind-limb unloading. A hole was drilled in the right femur of adult male C57/BL6J mice. Four days after surgery, mice were subjected to hind-limb unloading for 1 week. Seven days after surgery, mice were either housed for 4 days in a hypobaric room (FiO2 at 10%) or kept under normoxic conditions. Unsuspended control mice were housed in either hypobaric or normoxic conditions. Animals were sacrificed on postsurgery day 11 to allow for collection of both contralateral and lesioned femurs, blood, and spleen. As assessed by microtomography, delayed hypoxia enhanced bone-healing efficiency by increasing the closing of the cortical defect and the newly synthesized bone volume in the cavity by +55% and +35%, respectively. Proteome analysis and histomorphometric data suggested that bone-repair improvement likely results from the acceleration of the natural bone-healing process rather than from extended mobilization of MSC-derived osteoprogenitors. Hind-limb unloading had hardly any effect beyond delayed hypoxia-enhanced bone-healing efficiency.


Asunto(s)
Remodelación Ósea , Fracturas del Fémur/complicaciones , Fémur/fisiopatología , Curación de Fractura , Hipoxia/complicaciones , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Fracturas del Fémur/diagnóstico por imagen , Fracturas del Fémur/metabolismo , Fracturas del Fémur/fisiopatología , Fémur/diagnóstico por imagen , Fémur/metabolismo , Células Madre Hematopoyéticas/metabolismo , Suspensión Trasera , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteómica , Factores de Tiempo , Microtomografía por Rayos X
9.
Stem Cell Res Ther ; 4(1): 1, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23290259

RESUMEN

INTRODUCTION: This study investigated the promising effect of a new Platelet Glue obtained from Cryoprecipitation of Apheresis Platelet products (PGCAP) used in combination with Mesenchymal Stromal Cells (MSC) loaded on ceramic biomaterials to provide novel strategies enhancing bone repair. METHODS: PGCAP growth factor content was analyzed by ELISA and compared to other platelet and plasma-derived products. MSC loaded on biomaterials (65% hydroxyapatite/35% beta-TCP or 100% beta-TCP) were embedded in PGCAP and grown in presence or not of osteogenic induction medium for 21 days. Biomaterials were then implanted subcutaneously in immunodeficient mice for 28 days. Effect of PGCAP on MSC was evaluated in vitro by proliferation and osteoblastic gene expression analysis and in vivo by histology and immunohistochemistry. RESULTS: We showed that PGCAP, compared to other platelet-derived products, allowed concentrating large amount of growth factors and cytokines which promoted MSC and osteoprogenitor proliferation. Next, we found that PGCAP improves the proliferation of MSC and osteogenic-induced MSC. Furthermore, we demonstrated that PGCAP up-regulates the mRNA expression of osteogenic markers (Collagen type I, Osteonectin, Osteopontin and Runx2). In vivo, type I collagen expressed in ectopic bone-like tissue was highly enhanced in biomaterials embedded in PGCAP in the absence of osteogenic pre-induction. Better results were obtained with 65% hydroxyapatite/35% beta-TCP biomaterials as compared to 100% beta-TCP. CONCLUSIONS: We have demonstrated that PGCAP is able to enhance in vitro MSC proliferation, osteoblastic differentiation and in vivo bone formation in the absence of osteogenic pre-induction. This clinically adaptable platelet glue could be of interest for improving bone repair.


Asunto(s)
Materiales Biocompatibles/farmacología , Plaquetas/efectos de los fármacos , Huesos/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Adhesivos/farmacología , Animales , Biomarcadores/metabolismo , Plaquetas/metabolismo , Huesos/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , ARN Mensajero/metabolismo , Ingeniería de Tejidos/métodos
10.
Biomed Mater Eng ; 22(4): 219-26, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22785365

RESUMEN

Skeletal unloading provokes bone loss. These bone alterations have been shown to be associated with impairment of osteoblastic activity. In the present study, we evaluated the effect of skeletal unloading on bone marrow progenitor cells, for exploration of the underlying mechanism. Wistar rats were randomized to be either hindlimb unloaded for 9 days or to act as controls. Micro-CT was used to detect tibial trabecular architecture changes in response to skeletal unloading. Microgravity conditions for 9 days resulted in a decreased number and an increased spacing of the bone trabeculae in the proximal tibia. The proliferative capacity of the femoral bone marrow samples was assessed (fibroblast-colony-forming assay). By using qPCR, the expression of selected markers of vascularization (Vegfa; Hif1a; Angpt1), energy metabolism (Prkaa2; Mtor), bone formation (Runx2; Alp; Bglap; Bmp2; Bmp4; Bmp7) and bone resorption (Acp5; Tnfsf11; Tnfrsf11b) in these bone marrow suspensions was measured. We demonstrated a striking decrease in the number of fibroblastic progenitors in response to hindlimb unloading. This deficit in proliferation was shown to be accompanied by altered hindlimb perfusion and cellular energy homeostasis. Ex vivo culture assays of the bone marrow-derived progenitor cells screened for osteogenic (Runx2; Alp; Bglap) and adipogenic (Pparg; Fabp4) differentiation alterations in response to microgravity. Induced progenitor cells from unloaded rats showed a delay in osteogenic differentiation and impaired adipogenic differentiation compared to control. The data of this multi-level approach demonstrate that skeletal unloading significantly affects the bone tissue and its metabolism at the progenitor stage. The molecular expressions of the bone marrow population support a role of cellular metabolic stresses in skeletal alterations induced by inactivity.


Asunto(s)
Células de la Médula Ósea/citología , Suspensión Trasera , Huesos de la Pierna/fisiología , Osteogénesis , Células Madre/citología , Animales , Médula Ósea/irrigación sanguínea , Médula Ósea/fisiología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Metabolismo Energético , Femenino , Regulación de la Expresión Génica , Miembro Posterior/irrigación sanguínea , Miembro Posterior/fisiología , Huesos de la Pierna/irrigación sanguínea , Neovascularización Fisiológica , Osteoblastos/citología , Ratas , Ratas Wistar , Células Madre/metabolismo
11.
PLoS One ; 7(1): e30788, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22303456

RESUMEN

The mitogen-activated protein kinases (MAPK) ERK1 and ERK2 are among the major signal transduction molecules but little is known about their specific functions in vivo. ERK activity is provided by two isoforms, ERK1 and ERK2, which are ubiquitously expressed and share activators and substrates. However, there are not in vivo studies which have reported a role for ERK1 or ERK2 in HSCs and the bone marrow microenvironment. The present study shows that the ERK1-deficient mice present a mild osteopetrosis phenotype. The lodging and the homing abilities of the ERK1(-/-) HSC are impaired, suggesting that the ERK1(-/-)-defective environment may affect the engrafment of HSCs. Serial transplantations demonstrate that ERK1 is involved in the maintenance of an appropriate medullar microenvironment, but that the intrinsic properties of HSCs are not altered by the ERK1(-/-) defective microenvironment. Deletion of ERK1 impaired in vitro and in vivo osteoclastogenesis while osteoblasts were unaffected. As osteoclasts derive from precursors of the monocyte/macrophage lineage, investigation of the monocytic compartment was performed. In vivo analysis of the myeloid lineage progenitors revealed that the frequency of CMPs increased by approximately 1.3-fold, while the frequency of GMPs significantly decreased by almost 2-fold, compared with the respective WT compartments. The overall mononuclear-phagocyte lineage development was compromised in these mice due to a reduced expression of the M-CSF receptor on myeloid progenitors. These results show that the cellular targets of ERK1 are M-CSFR-responsive cells, upstream to osteoclasts. While ERK1 is well known to be activated by M-CSF, the present results are the first to point out an ERK1-dependent M-CSFR regulation on hematopoietic progenitors. This study reinforces the hypothesis of an active cross-talk between HSCs, their progeny and bone cells in the maintenance of the homeostasis of these compartments.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/enzimología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Nicho de Células Madre , Animales , Densidad Ósea , Médula Ósea/patología , Huesos/enzimología , Huesos/patología , Compartimento Celular , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Microambiente Celular , Eliminación de Gen , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 3 Activada por Mitógenos/deficiencia , Monocitos , Osteoblastos/enzimología , Osteoblastos/patología , Osteoclastos/enzimología , Osteoclastos/patología , Osteogénesis
12.
J Appl Physiol (1985) ; 112(4): 587-96, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22134698

RESUMEN

The aim of the study was to determine the seasonal influence of vitamin D status on bone metabolism in French submariners over a 2-mo patrol. Blood samples were collected as follows: prepatrol and patrol days 20, 41, and 58 on crewmembers from both a winter (WP; n = 20) and a summer patrol (SP; n = 20), respectively. Vitamin D status was evaluated for WP and SP. Moreover, extended parameters for acid-base balance (Pco(2), pH, and bicarbonate), bone metabolism (bone alkaline phosphatase and COOH-terminal telopeptide of type I collagen), and mineral homeostasis (parathyroid hormone, ionized calcium and phosphorus) were scrutinized. As expected, SP vitamin D status was higher than WP vitamin D status, regardless of the considered experimental time. A mild chronic respiratory acidosis (CRA) was identified in both SP and WP submariners, up to patrol day 41. Such an occurrence paired up with an altered bone remodeling coupling (decreased bone alkaline phosphatase-to-COOH-terminal telopeptide of type I collagen ratio). At the end of the patrol (day 58), a partial compensation of CRA episode, combined with a recovered normal bone remodeling coupling, was observed in SP, not, however, in WP submariners. The mild CRA episode displayed over the initial 41-day submersion period was mainly induced by a hypercapnia resulting from the submarine-enriched CO(2) level. The correlated impaired bone remodeling may imply a physiological attempt to compensate this acidosis via bone buffering. On patrol day 58, the discrepancy observed in terms of CRA compensation between SP and WP may result from the seasonal influence on vitamin D status.


Asunto(s)
Acidosis Respiratoria/metabolismo , Huesos/metabolismo , Estaciones del Año , Medicina Submarina , Deficiencia de Vitamina D/metabolismo , Adulto , Fosfatasa Alcalina/sangre , Biomarcadores/sangre , Calcio/sangre , Colágeno Tipo I/sangre , Homeostasis , Humanos , Masculino , Personal Militar , Hormona Paratiroidea/sangre , Péptidos/sangre , Vitamina D/sangre
13.
Biomaterials ; 32(14): 3547-54, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21324523

RESUMEN

Optimization of implant osseointegration in patients with reduced bone healing potential is a challenge remaining in implant dentistry. Identification of the genes that are modulated during implant osseointegration in normal versus osteopenic bone is needed to successfully address these pertinent clinical needs. The present study aimed to assess the initial and early molecular events following titanium implant installation in normal and compromised bone in a rat tibia model. Peri-implant tissue from a well-defined tissue regeneration compartment was analyzed at 2 and 7 days post-surgery for the expression of select markers of inflammation, angiogenesis, bone resorption and bone formation. Impaired bone was induced by hindlimb unloading and validated using µCT. The essential step of angiogenesis preceding bone regeneration was evidenced for the peri-implant setting in healthy bone. Compromised bone significantly affected the angiogenesis-osteogenesis coupling in the initial phase (2 days post-surgery), with altered expressions of Vegfa and Epas1 coinciding with downregulated expressions of Col1a1, Bmp2, Bmp4, Alpl and Bglap. At 7 days post-implantation, differences between normal and compromised peri-implant bone were no longer observed. This in vivo molecular evidence of delayed implant osseointegration in compromised bone reassert modern strategies in implant development, such as surface modifications and bioengineered approaches, to improve implant osseointegration in compromised conditions.


Asunto(s)
Oseointegración/fisiología , Titanio/química , Animales , Femenino , Miembro Posterior/diagnóstico por imagen , Prótesis e Implantes , Ratas , Ratas Wistar , Tibia/diagnóstico por imagen , Tomografía Computarizada por Rayos X
14.
Tissue Eng Part C Methods ; 17(3): 311-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20874261

RESUMEN

Shortening of the healing time before loading risks impeding successful titanium implant anchorage into compromised bone. A thorough understanding at the genetic scale of the early phases of bone regeneration at the implant interface is required before the development of strategies to enhance implant osseointegration. In this study a new in vivo implant model to explore the mechanism by which titanium implant osseointegration is affected by the host bone properties is presented. An implant was conceptualized enabling standardized harvesting of peri-implant tissue for quantitative molecular analysis while preserving the mimicking of the clinical setting. The implant is partly indented to provide a well-defined healing compartment from where tissue differentiation and de novo bone formation can be investigated and partly screw-threaded to provide a good implant anchorage into the bone. The feasibility of the implant design was assessed in osteopenic bone conditions, evoked by simulated weightlessness. Wistar rats were either hindlimb unloaded by tail suspension (HU) for 9 days or acted as controls (CTL). The status of compromised bone tissue through 9-days HU was confirmed by micro-X-ray computed tomography. The implant was installed in the proximal tibial bone 7 days after the onset of HU or CTL. Two days postimplantation, the peri-implant regenerating tissue responses were recorded by measuring expression of inflammatory, angiogenic, and bone resorption parameters (hypoxia-inducible factor 1, alpha subunit; vascular endothelial growth factor A; angiopoietin 1; endothelial PAS domain protein 1; fibroblast growth factor 2; tumor necrosis factor; interleukin 11; acid phosphatase 5, tartrate resistant; tumor necrosis factor (ligand) superfamily, member 11/RANKL). We successfully demonstrated that HU-associated bone conditions evoked a significant alteration of expression of the angiogenic markers in the peri-implant regenerative tissue during initial implant osseointegration, whereas the expression levels of the inflammatory and bone resorption parameters remained unchanged. We concluded that this in vivo implant model provides a well-designed and controlled method to examine molecular responses in implant osseointegration to impaired bone conditions. This model may serve to explore the application of anabolic strategies in peri-implant osteogenesis.


Asunto(s)
Implantes Experimentales , Modelos Animales , Oseointegración/efectos de los fármacos , Tibia/efectos de los fármacos , Titanio/farmacología , Adaptación Fisiológica/efectos de los fármacos , Animales , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Suspensión Trasera , Ratas , Ratas Wistar , Tibia/diagnóstico por imagen , Cicatrización de Heridas/efectos de los fármacos , Microtomografía por Rayos X
15.
Med Sci Sports Exerc ; 43(1): 54-63, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20473218

RESUMEN

PURPOSE: The aim of the present study was to investigate if wheel running exercise could offset the detrimental influences of independent or combined high-phosphorus and low-calcium diets on bone tissue in rats. METHODS: Forty male dark Agouti rats were randomly assigned to eight groups of five animals. Four sedentary groups (SED) and four voluntary trained groups (TR) were fed over 6 wk of either a standard food or a modified diet, namely, high phosphorus (HP), low calcium (LCa), or high phosphorus combined with low calcium (HP/LCa). After sacrifice, blood samples were collected to determine parathyroid hormone, Ca(2+), and Pi levels. Both tibiae were removed for bone mass determination and extended histomorphometric analyses. RESULTS: In SED rats, all unbalanced diets induced a sizeable bone volume decrease, up to 56%. Interestingly, steady training partially compensates for this bone volume loss, regardless of the considered modified diets. At the cellular level, only independent LCa diet induced a 38% decrease in osteoblastic surface in both SED and TR rat groups, generating thereby a reduction in bone neosynthesis. In terms of osteoclastic surface, an increase in this parameter was evidenced only in HP diets (both HP and HP-LCa), implying heightened bone resorption. The major effects of unbalanced diets are mainly observed on bone tissue because serum parameters (parathyroid hormone, Ca(2+), and Pi levels) remained only slightly modified. CONCLUSIONS: Training induced a positive effect on unbalanced diet-altered bone tissue formation but remained inadequate to reach standard bone mass measured in SED rats fed with balanced food. Further, we suggest that the nature of the diet influences the balance between bone formation and resorption: LCa diet decreases bone formation, whereas HP and HP-LCa increase bone resorption.


Asunto(s)
Remodelación Ósea/efectos de los fármacos , Huesos/fisiología , Calcio de la Dieta/farmacología , Calcio/deficiencia , Dieta/efectos adversos , Actividad Motora/fisiología , Fósforo/farmacología , Condicionamiento Físico Animal/fisiología , Animales , Densidad Ósea/efectos de los fármacos , Calcio/sangre , Calcio de la Dieta/administración & dosificación , Masculino , Hormona Paratiroidea/sangre , Fósforo/administración & dosificación , Fósforo/sangre , Distribución Aleatoria , Ratas , Tibia/fisiología
16.
Am J Physiol Endocrinol Metab ; 294(4): E794-801, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18378961

RESUMEN

Loss of mechanical loading induces rapid bone loss resulting from reduced osteoblastogenesis and decreased bone formation. The signaling mechanisms involved in this deleterious effect on skeletal metabolism remain poorly understood. We have previously shown that hindlimb suspension in rats increases osteoblast apoptosis associated with decreased phosphatidylinositol 3-kinase (PI3K) signaling. In this study, we investigated whether transforming growth factor (TGF)-beta2 may prevent the altered signaling and osteoblast apoptosis induced by skeletal unloading in vivo. Hindlimb suspension-induced decreased bone volume was associated with reduced alpha(5)beta(1)-integrin protein levels and PI3K/Akt signaling in unloaded bone. Continuous administration of TGF-beta2 using osmotic minipumps prevented the decreased alpha(5)beta(1)-integrin expression and the reduced PI3K/Akt signaling in unloaded bone, resulting in the prevention of osteoblast apoptosis. We also show that TGF-beta2 prevented the decreased Bcl-2 levels induced by unloading, which suggests that TGF-beta2 targets Bcl-2 via PI3K/Akt to prevent osteoblast apoptosis in unloaded bone. Furthermore, we show that TGF-beta2 prevented the decrease in phosphorylated Bad, the inactive form of the proapoptotic protein Bad, induced by unloading. These results identify a protective role for TGF-beta2 in osteoblast apoptosis induced by mechanical unloading via the alpha(5)beta(1)/PI3K/Akt signaling cascade and downstream Bcl-2 and phospho-Bad survival proteins. We thus propose a novel role for TGF-beta2 in protection from unloading-induced apoptosis in vivo.


Asunto(s)
Apoptosis/fisiología , Suspensión Trasera , Osteoblastos/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta2/metabolismo , Animales , Apoptosis/efectos de los fármacos , Huesos/efectos de los fármacos , Huesos/metabolismo , Masculino , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Osteoblastos/efectos de los fármacos , Osteoblastos/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta2/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Proteína Letal Asociada a bcl/metabolismo
17.
Leuk Lymphoma ; 48(10): 2032-41, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17917971

RESUMEN

Osteolytic bone lesions are common in patients with multiple myeloma (MM), a clonal plasma cell disorder, and result from increased osteoclastic bone resorption and decreased osteoblastic bone formation. Because mesenchymal stem cells (MSCs) are committed towards cells of the osteoblast lineage, we compared the in vitro characteristics of MSCs from the bone marrow of 18 MM patients (MM-MSCs) and eight normal donors (ND-MSCs). MM-MSCs displayed deficient growth that could be explained in part by the reduced expression of several growth factor receptors on the surface of MM-MSCs compared with ND-MSCs. Receptor downregulation was observed on RT-PCR analysis. A major finding was an approximately fivefold higher expression of osteoblast inhibitor DKK1 at transcript and protein levels in MM-MSCs than ND-MSCs. These data suggest that defective osteoblast function in patients with advanced MM may be related not only to factors released by tumor myeloma cells but also to MSC abnormalities.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Células Madre Mesenquimatosas/citología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Anciano , Huesos/metabolismo , Diferenciación Celular , Femenino , Humanos , Inmunofenotipificación , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-6/metabolismo , Masculino , Células Madre Mesenquimatosas/patología , Persona de Mediana Edad , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteólisis , Sindecano-1/biosíntesis
18.
J Immunol ; 176(12): 7761-7, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16751424

RESUMEN

Several reports have suggested that mesenchymal stem cells (MSCs) could exert a potent immunosuppressive effect in vitro, and thus may have a therapeutic potential for T cell-dependent pathologies. We aimed to establish whether MSCs could be used to control graft-vs-host disease (GVHD), a major cause of morbidity and mortality after allogeneic hemopoietic stem cell transplantation. From C57BL/6 and BALB/c mouse bone marrow cells, we purified and expanded MSCs characterized by the lack of expression of CD45 and CD11b molecules, their typical spindle-shaped morphology, together with their ability to differentiate into osteogenic, chondrogenic, and adipogenic cells. These MSCs suppressed alloantigen-induced T cell proliferation in vitro in a dose-dependent manner, independently of their MHC haplotype. However, when MSCs were added to a bone marrow transplant at a MSCs:T cells ratio that provided a strong inhibition of the allogeneic responses in vitro, they yielded no clinical benefit on the incidence or severity of GVHD. The absence of clinical effect was not due to MSC rejection because they still could be detected in grafted animals, but rather to an absence of suppressive effect on donor T cell division in vivo. Thus, in these murine models, experimental data do not support a significant immunosuppressive effect of MSCs in vivo for the treatment of GVHD.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Trasplante de Médula Ósea/patología , Proliferación Celular , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Terapia de Inmunosupresión , Linfocitos/inmunología , Trasplante de Células Madre Mesenquimatosas , Animales , Trasplante de Médula Ósea/efectos adversos , Separación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Enfermedad Injerto contra Huésped/prevención & control , Inmunofenotipificación , Prueba de Cultivo Mixto de Linfocitos , Linfocitos/patología , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Quimera por Radiación , Bazo/citología , Bazo/inmunología , Bazo/metabolismo
19.
J Cell Physiol ; 205(2): 228-36, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15887229

RESUMEN

Mesenchymal stem cells (MSCs) are considered as emergent "universal" cells and various tissue repair programs using MSCs are in development. In vitro expansion of MSCs is conventionally achieved in medium containing fetal calf serum (FCS) and is increased by addition of growth factors. However, for widespread clinical applications, contact of MSCs with FCS must be minimized since it is a putative source of prion or virus transmission. Therefore, because platelets are a natural source of growth factors, we sought to investigate in vitro MSC expansion in response to platelet lysates (PL) obtained from platelet-rich plasma. Human MSCs were expanded in FCS (+/-bFGF)- or PL-supplemented medium through a process of subculture. We demonstrated that PL-containing medium is enriched by growth factors (platelet-derived growth factors (PDGFs), basic fibroblast growth factor (bFGF), transforming growth factor (TGF-beta), insulin-like growth factor-1 (IGF-1) ...) and showed that PL is able to promote MSC expansion, to decrease the time required to reach confluence, and to increase CFU-F size, as compared to the FCS medium. Furthermore, we demonstrated that MSCs cultured in the presence of PL maintain their osteogenic, chondrogenic, and adipogenic differentiation properties and retain their immunosuppressive activity. Therefore, we propose that PL may be a powerful and safe substitute for FCS in development of tissue- and cellular-engineered products in clinical settings using MSCs.


Asunto(s)
Plaquetas/fisiología , Sustitutos Sanguíneos/efectos adversos , Extractos Celulares/farmacología , Proliferación Celular/efectos de los fármacos , Tratamiento Basado en Trasplante de Células y Tejidos , Células Madre Mesenquimatosas/fisiología , Animales , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Sustancias de Crecimiento/análisis , Humanos , Inmunofenotipificación , Cinética , Factores de Tiempo
20.
J Appl Physiol (1985) ; 93(3): 1047-56, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12183502

RESUMEN

Using analyses of iliac crest cell and tissue, back-scattered electron imaging, and biochemical techniques, we characterized the effects of a 14-day spaceflight (Bion 11) on bone structure and bone formation in two 3- to 4-yr-old male rhesus monkeys compared with eight age-matched Earth-control monkeys. We found that postflight bone volume was 35% lower than preflight values in flight monkeys. This was associated with reduced osteoid (-40%) and mineralizing (-32%) surfaces and decreased bone formation rate (-53%). Moreover, flight monkeys exhibited trends to lower values of mineralization profile in iliac bone (back-scattered electron imaging) and to decreased osteocalcin serum levels (P = 0.08). The initial number of trabecular bone cells yielded in cultures did not differ in flight and control animals before or after the flight. However, osteoblastic cell proliferation was markedly lower in postflight vs. preflight at 9 and 14 days of culture in one flight monkey. This study suggests that a 14-day spaceflight reduces iliac bone formation, osteoblastic activity, and/or recruitment in young rhesus monkeys, resulting in decreased trabecular bone volume.


Asunto(s)
Osteogénesis/fisiología , Vuelo Espacial , Animales , Densidad Ósea , Huesos/anatomía & histología , División Celular , Células Cultivadas , Ilion/citología , Ilion/metabolismo , Macaca mulatta , Masculino , Tamaño de los Órganos , Osteoblastos/citología , Osteocalcina/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...