Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
ACS Appl Mater Interfaces ; 16(31): 40411-40427, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39044386

RESUMEN

The treatment of critical-sized bone defects caused by tumor removal, skeletal injuries, or infections continues to pose a major clinical challenge. A popular potential alternative solution to autologous bone grafts is a tissue-engineered approach that utilizes the combination of mesenchymal stromal/stem cells (MSCs) with synthetic biomaterial scaffolds. This approach aims to support new bone formation by mimicking many of the biochemical and biophysical cues present within native bone. Regrettably, osteocyte cells, crucial for bone maturation and homeostasis, are rarely produced within MSC-seeded scaffolds, thereby restricting the development of fully mature cortical bone from these synthetic implants. In this work, we have constructed a multimodal scaffold by combining electrospun poly(lactic-co-glycolic acid) (PLGA) fibrous scaffolds with poly(ethylene glycol) (PEG)-based hydrogels that mimic the functional unit of cortical bone, osteon (osteon-mimetic) scaffolds. These scaffolds were decorated with a novel bone morphogenic protein-6 (BMP6) peptide (BMP6p) after our findings revealed that the BMP6p drives higher levels of Smad signaling than the full-length protein counterpart, soluble or when bound to the PEG hydrogel backbone. We show that our osteon-mimetic scaffolds, in presenting concentric layers of BMP6p-PEG hydrogel overlaid on MSC-seeded PLGA nanofibers, promoted the rapid formation of osteocyte-like cells with a phenotypic dendritic morphology, producing early osteocyte markers, including E11/gp38 (E11). Maturation of these osteocyte-like cells was further confirmed by the observation of significant dentin matrix protein 1 (DMP1) throughout our bilayered scaffolds after 3 weeks, even when cultured in a medium without dexamethasone (DEX) or any other osteogenic supplements. These results demonstrate that these osteon-mimetic scaffolds, in presenting biochemical and topographical cues reminiscent of the forming osteon, can drive the formation of osteocyte-like cells in vitro from hBMSCs without the need for any osteogenic factor media supplementation.


Asunto(s)
Materiales Biomiméticos , Células Madre Mesenquimatosas , Nanofibras , Osteocitos , Osteogénesis , Andamios del Tejido , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Andamios del Tejido/química , Nanofibras/química , Humanos , Osteogénesis/efectos de los fármacos , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Osteocitos/citología , Osteocitos/metabolismo , Osteocitos/efectos de los fármacos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Proteína Morfogenética Ósea 6/química , Proteína Morfogenética Ósea 6/farmacología , Proteína Morfogenética Ósea 6/metabolismo , Polietilenglicoles/química , Diferenciación Celular/efectos de los fármacos , Ingeniería de Tejidos/métodos , Hidrogeles/química , Hidrogeles/farmacología
2.
Cell Tissue Res ; 396(1): 103-117, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38403744

RESUMEN

The formation of the epiretinal fibrotic membrane by retinal pigment epithelial (RPE) cells is a primary pathological change for proliferative vitreoretinopathy (PVR). Bone morphogenetic protein 6 (BMP6) is an antifibrogenic factor in various cells. To date, it is still unknown whether BMP6 can interfere with the fibrogenesis of RPE cells during the progression of PVR. This work aimed to address the relationship between BMP6 and transforming growth factor-ß2 (TGF-ß2)-elicited fibrogenesis of RPE cells, an experimental model for studying PVR in vitro. The BMP6 level was down-regulated, while the TGF-ß2 level was up-regulated in the vitreous humor of PVR patients. The BMP6 level was down-regulated in human RPE cells challenged with TGF-ß2. The treatment of RPE cells with TGF-ß2 resulted in significant increases in proliferation, migration, epithelial-to-mesenchymal transition (EMT), and extracellular matrix (ECM) remodelling. These effects were found to be inhibited by the overexpression of BMP6 or exacerbated by the knockdown of BMP6. BMP6 overexpression reduced the phosphorylation of p38 and JNK in TGF-ß2-stimulated RPE cells, while BMP6 knockdown showed the opposite effects. The inhibition of p38 or JNK partially reversed the BMP6-silencing-induced promoting effects on TGF-ß2-elicited fibrogenesis in RPE cells. Taken together, BMP6 demonstrates the ability to counteract the proliferation, migration, EMT, and ECM remodelling of RPE cells induced by TGF-ß2. This is achieved through the regulation of the p38 and JNK MAPK pathways. These findings imply a potential connection between BMP6 and PVR, and highlight the potential application of BMP6 in therapeutic interventions for PVR.


Asunto(s)
Vitreorretinopatía Proliferativa , Humanos , Vitreorretinopatía Proliferativa/tratamiento farmacológico , Vitreorretinopatía Proliferativa/metabolismo , Vitreorretinopatía Proliferativa/patología , Epitelio Pigmentado de la Retina , Factor de Crecimiento Transformador beta2/farmacología , Factor de Crecimiento Transformador beta2/metabolismo , Factor de Crecimiento Transformador beta2/uso terapéutico , Proteína Morfogenética Ósea 6/farmacología , Proteína Morfogenética Ósea 6/metabolismo , Proteína Morfogenética Ósea 6/uso terapéutico , Transición Epitelial-Mesenquimal , Células Epiteliales/metabolismo , Pigmentos Retinianos/metabolismo , Pigmentos Retinianos/farmacología , Pigmentos Retinianos/uso terapéutico , Movimiento Celular
3.
J Assist Reprod Genet ; 41(1): 31-48, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37930517

RESUMEN

PURPOSE: To evaluate whether PTX3 is differentially expressed in the granulosa lutein cells derived from women with PCOS and whether BMP6 can regulate the expression of PTX3 in hGL cells. METHODS: The expression levels of BMP6 and PTX3 in granulosa lutein cells were evaluated by RT-qPCR. The correlation between the expression levels of BMP6 /PTX3 and oocyte quality indexes were analyzed using clinical samples. The cells were incubated with BMP6 at different concentrations and times to check the expression of PTX3 in KGN cells. TGF-ß type I inhibitors and small interfering RNA targeting ALK2/3/6,SMAD1/5/8 and SMAD4 were used to study the involvement of SMAD dependent pathways in KGN cells. RESULTS: The levels of BMP6 in hGL cells were negatively correlated with the corresponding oocyte maturation rate and high-quality embryo rate, whereas the levels of PTX3 were positively correlated with the corresponding oocyte maturation rate in PCOS. Additionally, the in vitro cell cultured results showed BMP6 significantly inhibited the expression of PTX3 in KGN cells. Furthermore, using a dual inhibition approach (kinase inhibitors and small interfering RNAs), we identified the ALK2/ALK3 type I receptors and BMPR2/ACVR2A type II receptors and the downstream SMAD1/SMAD5-SMAD4 signaling pathway were responsible for the BMP6-induced cellular activities in KGN cells. CONCLUSIONS: The suppressive effect of BMP6 on PTX3 was mediated by ALK2/ALK3 type I receptors and BMPR2/ACVR2A type II receptors in granulosa cells through the SMAD1/5-SMAD4 dependent signaling pathway in PCOS.Our findings provides new insights into the understanding of the pathogenesis of PCOS-related ovulatory disorders.


Asunto(s)
Proteína C-Reactiva , Células Lúteas , Síndrome del Ovario Poliquístico , Componente Amiloide P Sérico , Femenino , Humanos , Proteína Morfogenética Ósea 6/genética , Proteína Morfogenética Ósea 6/metabolismo , Proteína Morfogenética Ósea 6/farmacología , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Regulación hacia Abajo/genética , Células de la Granulosa/metabolismo , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/metabolismo
4.
J Dent Res ; 102(1): 93-102, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36281063

RESUMEN

T-cell dysfunction has been shown to play an important role in the pathogenesis of Sjögren's syndrome (SS). In recent studies, the increased expression of BMP6 has been reported to be related to SS. However, the roles that BMP6 plays in immune homeostasis in the development of SS as well as the downstream signals activated by BMP6 remain unclear. In this study, we investigated the effects and molecular mechanisms of BMP6 on naive CD4+ T cells, showing that BMP6 could upregulate interferon (IFN)-γ secretion from CD4+ T cells through a ceramide/nuclear factor-κB pathway, with no effect on T-cell activation or proliferation. Moreover, an in vivo study showed that anticeramide treatment (myriocin) for an SS animal model (NOD/LtJ mice) could significantly decrease the IFN-γ expression and Th1 frequency in the salivary glands and suppress the inflammation infiltration in salivary glands and maintain the salivary flow rates, both of which reflect SS-like symptoms. This study identifies a promising target that could effectively attenuate the abnormal state of CD4+ T cells and reverse the progression of SS.


Asunto(s)
Síndrome de Sjögren , Células TH1 , Animales , Ratones , Proteína Morfogenética Ósea 6/metabolismo , Proteína Morfogenética Ósea 6/farmacología , Interferón gamma/farmacología , Ratones Endogámicos NOD , Glándulas Salivales/metabolismo , Síndrome de Sjögren/tratamiento farmacológico , Linfocitos T
5.
PLoS One ; 16(6): e0253475, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34161397

RESUMEN

Hepcidin is a peptide hormone that targets the iron exporter ferroportin, thereby limiting iron entry into the bloodstream. It is generated in hepatocytes mainly in response to increased body iron stores or inflammatory cues. Iron stimulates expression of bone morphogenetic protein 6 (BMP6) from liver sinusoidal endothelial cells, which in turn binds to BMP receptors on hepatocytes and induces the SMAD signaling cascade for transcriptional activation of the hepcidin-encoding HAMP mRNA. SMAD signaling is also essential for inflammatory HAMP mRNA induction by the IL-6/STAT3 pathway. Herein, we utilized human Huh7 hepatoma cells and primary murine hepatocytes to assess the effects of iron perturbations on signaling to hepcidin. Iron chelation appeared to slightly impair signaling to hepcidin. Subsequent iron supplementation not only failed to reverse these effects, but drastically reduced basal HAMP mRNA and inhibited HAMP mRNA induction by BMP6 and/or IL-6. Thus, treatment of cells with excess iron inhibited basal and BMP6-mediated SMAD5 phosphorylation and induction of HAMP, ID1 and SMAD7 mRNAs in a dose-dependent manner. Iron also inhibited IL-6-mediated STAT3 phosphorylation and induction of HAMP and SOCS3 mRNAs. These responses were accompanied by induction of GCLC and HMOX1 mRNAs, known markers of oxidative stress. We conclude that hepatocellular iron overload suppresses hepcidin by inhibiting the SMAD and STAT3 signaling pathways downstream of their respective ligands.


Asunto(s)
Deferoxamina/farmacología , Hepatocitos/metabolismo , Hepcidinas/metabolismo , Sobrecarga de Hierro/metabolismo , Sideróforos/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Proteína Morfogenética Ósea 6/farmacología , Línea Celular Tumoral , Hepatocitos/efectos de los fármacos , Humanos , Interleucina-6/farmacología , Ratones , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Proteínas Smad/metabolismo
6.
Gene ; 788: 145662, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33887373

RESUMEN

INTRODUCTION: Culture conditions and differentiation cocktails may facilitate cell maturation and extracellular matrix (ECM) secretion and support the production of engineered fibroblastic tissues with applications in ligament regeneration. The objective of this study is to investigate the potential of two connective tissue-related ligands (i.e., BMP6 and GDF5) to mediate collagenous ECM synthesis and tissue maturation in vitro under normoxic and hypoxic conditions based on the hypothesis that BMP6 and GDF5 are components of normal paracrine signalling events that support connective tissue homeostasis. METHODS: Human adipose-derived MSCs were seeded on 3D-printed medical-grade polycaprolactone (PCL) scaffolds using a bioreactor and incubated in media containing GDF5 and/or BMP6 for 21 days in either normoxic (5% oxygen) or hypoxic (2% oxygen) conditions. Constructs were harvested on Day 3 and 21 for cell viability analysis by live/dead staining, structural analysis by scanning electron microscopy, mRNA levels by RTqPCR analysis, and in situ deposition of proteins by immunofluorescence microscopy. RESULTS: Pro-fibroblastic gene expression is enhanced by hypoxic culture conditions compared to normoxic conditions. Hypoxia renders cells more responsive to treatment with BMP6 as reflected by increased expression of ECM mRNA levels on Day 3 with sustained expression until Day 21. GDF5 was not particularly effective either in the absence or presence of BMP6. CONCLUSIONS: Fibroblastic differentiation of MSCs is selectively enhanced by BMP6 and not GDF5. Environmental factors (i.e., hypoxia) also influenced the responsiveness of cells to this morphogen.


Asunto(s)
Proteína Morfogenética Ósea 6/farmacología , Técnicas de Cultivo de Célula/métodos , Fibroblastos/citología , Factor 5 de Diferenciación de Crecimiento/farmacología , Células Madre Mesenquimatosas/citología , Reactores Biológicos , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibroblastos/química , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/efectos de los fármacos , Andamios del Tejido
7.
Vet J ; 269: 105605, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33593496

RESUMEN

Mesenchymal stromal cells (MSC) are used for cell-based treatment for canine osteoarthritis (OA). Compared with human MSCs, detailed information on the functional characterisation of canine MSCs is limited. In particular, the chondrogenic differentiation of canine adipose tissue-derived MSCs (cAT-MSCs) is challenging. In this study, we aimed to compare cAT-MSCs with bone marrow-derived MSCs (cBM-MSCs), focusing specifically on their in vitro chondrogenic potential, with or without bone morphogenetic proteins (BMP). cBM-MSCs and cAT-MSCs were characterised using flow cytometry and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The chondrogenic differentiation potential of all cMSC preparations in the presence of TGF-ß1 alone or when supplemented with 10, 100, or 250 ng/mL BMP-2 or BMP-6 was investigated using RT-qPCR, and biochemical, histochemical and immunohistological analyses. Both cBM-MSCs and cAT-MSCs expressed the surface markers CD90, CD73, and CD29, and were negative for CD45 and CD34, although the expression of CD73 and CD271 varied with donor and tissue origin. Interestingly, expression of ACAN and SOX9 was higher in cBM-MSCs than cAT-MSCs. In contrast with cBM-MSCs, cAT-MSCs could not differentiate toward the chondrogenic lineage without BMP-2/-6, and their in vitro chondrogenesis was inferior to cBM-MSCs with BMP-2/-6. Thus, cAT-MSCs have lower in vitro chondrogenic capacity than cBM-MSC under the studied culture conditions with 10, 100, or 250 ng/mL BMP-2 or BMP-6. Therefore, further characterisation is necessary to explore the potential of cAT-MSCs for cell-based OA treatments.


Asunto(s)
Células de la Médula Ósea/fisiología , Proteína Morfogenética Ósea 2/farmacología , Proteína Morfogenética Ósea 6/farmacología , Condrogénesis/fisiología , Células Madre Mesenquimatosas/fisiología , Animales , Antígenos de Superficie/análisis , Técnicas de Cultivo de Célula/veterinaria , Diferenciación Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias/veterinaria , Enfermedades de los Perros/terapia , Perros , Trasplante de Células Madre Mesenquimatosas , Osteoartritis/terapia , Osteoartritis/veterinaria , Factor de Crecimiento Transformador beta1/farmacología
8.
J Gene Med ; 23(3): e3311, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33527563

RESUMEN

BACKGROUND: Fragments of subcutaneous adipose tissue that have been genetically modified to express bone morphogenetic protein-2 (BMP-2) regenerate large segmental osseous lesions in rodents. Gene-activated adipose tissue can be implanted into osseous defects without prior cell extraction and cell culture. The present study aimed to explore whether the heterodimers BMP-2/6 or BMP-2/7 exceed the osteoinductive effect of BMP-2 on adipose tissue. METHODS: In an in vitro tissue culture system, freshly harvested rat subcutaneous adipose tissue was cultivated in the presence of either BMP-2 or BMP-2/6 or BMP-2/7 at a high (200 ng/ml) and low (50 ng/ml) concentration. Gene expression analysis as well as histological and immunohistochemical methods were applied to test for osteoinduction. RESULTS: A concentration of 200 ng/ml of homodimeric BMP-2 induced osteogenic differentiation most potently, showing more calcification and a higher expression level of bone markers than both concentrations of BMP-2/6 or -2/7. A concentration of 50 ng/ml of BMP-2 was a significantly stronger osteogenic inducer than both concentrations of BMP-2/6 and the low concentration of BMP-2/7. The most potent heterodimeric driver of osteoinduction was BMP-2/7 at a high concentration, demonstrating effects similar to those of BMP-2 at a low concentration. CONCLUSIONS: Homodimeric BMP-2 evoked osteoinduction within adipose tissue more potently and at a lower concentration than heterodimeric BMP-2/6 or BMP-2/7. This result agrees well with the fact that it might be easier to translate adipose grafts activated by homodimeric BMP-2 clinically. Preclinical in vivo gene transfer studies are necessary to confirm the results of the present study.


Asunto(s)
Proteína Morfogenética Ósea 2/farmacología , Proteína Morfogenética Ósea 6/farmacología , Proteína Morfogenética Ósea 7/farmacología , Regeneración Ósea/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Grasa Subcutánea/efectos de los fármacos , Grasa Subcutánea/metabolismo , Animales , Biomarcadores/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratas , Ratas Endogámicas F344 , Proteínas Recombinantes/farmacología , Técnicas de Cultivo de Tejidos
9.
Stem Cell Rev Rep ; 16(3): 596-611, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32112264

RESUMEN

Osteoarthritis (OA) is a general joint disease. Cartilage damage is associated with a decrease in the density of chondrocytes. Mesenchymal stem cells (MSCs) differentiate into adipocytes, osteocytes and chondrocytes, and are an excellent source of cell therapy. Cartilage-derived extracellular matrix (ECM) promotes chondrogenesis of MSCs. However, the role of MSCs stimulated by ECM is not well known in OA. The purpose of this study is to determine the role of specific factors generated by the application of ECM and umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) in managing OA symptoms. Cartilage acellular matrix (CAM), which is a cartilage-derived ECM, was used to promote the chondrogenesis of UCB-MSCs. Induced MSCs were analyzed using chondrogenic markers (aggrecan, collagen type 2, and SOX9) and bone morphogenic protein 6 (BMP6). BMP6 is known to be involved in early chondrogenesis of MSCs. As a result, treatment with CAM significantly increased the expression of chondrogenic markers and BMP6 in UCB-MSCs. Treatment with recombinant human BMP6 also dramatically increased the levels of chondrogenic markers in UCB-MSCs. In addition, UCB-MSCs and CAM were used to evaluate OA symptom improvement in a rabbit articular cruciate ligament transection (ACLT) model. Application of UCB-MSCs and CAM enhanced not only the structure and synthesis of proteoglycan and collagen type 2 but also anti-inflammatory effects in both rabbit joint and synovial fluid. Moreover, the detection of human cells and involvement of BMP6 were confirmed in rabbit cartilage tissues. This study indicates that therapeutic potential of UCB-MSCs with CAM is mediated via BMP6 in OA.


Asunto(s)
Lesiones del Ligamento Cruzado Anterior/terapia , Proteína Morfogenética Ósea 6/farmacología , Cartílago Articular/patología , Matriz Extracelular/metabolismo , Sangre Fetal/citología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Animales , Lesiones del Ligamento Cruzado Anterior/diagnóstico por imagen , Lesiones del Ligamento Cruzado Anterior/patología , Conducta Animal , Rastreo Celular , Condrogénesis , Modelos Animales de Enfermedad , Humanos , Osteoartritis/patología , Comunicación Paracrina , Conejos , Líquido Sinovial/metabolismo
11.
J Mater Sci Mater Med ; 31(1): 4, 2019 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-31832785

RESUMEN

In the present study, a chitosan-based, multifunctional and double-faced barrier membrane was developed for the periodontitis therapy. The porous surface of the membrane was coated with bone-like hydroxyapatite (HA) produced by microwave-assisted biomimetic method and enriched with bone morphogenetic factor 6 (BMP-6) to enhance the bioactivity of chitosan. This surface of the membrane was designed to be in contact with the hard tissue that was damaged due to periodontitis. Otherwise the nonporous surface of membrane, which is in contact with the inflammatory soft tissue, was coated with electrospun polycaprolactone (PCL) fibers to prevent the migration of epithelial cells to the defect area. PrestoBlue, Scanning Electron Microscope (SEM) and real-time PCR results demonstrated that while porous surface of the membrane was enhancing the proliferation and differentiation of MC3T3-E1 preosteoblasts, nonporous surface of membrane did not allow migration of epithelial Madine Darby Bovine Kidney (MDBK) cells. The barrier membrane developed here is biodegradable and can be easily manipulated, has osteogenic activity and inactivity for epithelial cells. Thus, by implanting this membrane to the damaged periodontal tissue, bone regeneration will take place and integrity of periodontal tissues will be preserved.


Asunto(s)
Proteína Morfogenética Ósea 6/farmacología , Quitosano/química , Membranas Artificiales , Nanoestructuras/química , Animales , Materiales Biocompatibles , Prótesis Vascular , Bovinos , Línea Celular , Supervivencia Celular , Humanos , Ratones , Nanoestructuras/ultraestructura , Osteoblastos , Propiedades de Superficie , Andamios del Tejido
12.
Am J Hematol ; 94(11): 1227-1235, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31400017

RESUMEN

The erythroferrone (ERFE) is the erythroid regulator of hepatic iron metabolism by suppressing the expression of hepcidin. Congenital dyserythropoietic anemia type II (CDAII) is an inherited hyporegenerative anemia due to biallelic mutations in the SEC23B gene. Patients with CDAII exhibit marked clinical variability, even among individuals sharing the same pathogenic variants. The ERFE expression in CDAII is increased and related to abnormal erythropoiesis. We identified a recurrent low-frequency variant, A260S, in the ERFE gene in 12.5% of CDAII patients with a severe phenotype. We demonstrated that the ERFE-A260S variant leads to increased levels of ERFE, with subsequently marked impairment of iron regulation pathways at the hepatic level. Functional characterization of ERFE-A260S in the hepatic cell system demonstrated its modifier role in iron overload by impairing the BMP/SMAD pathway. We herein described for the first time an ERFE polymorphism as a genetic modifier variant. This was with a mild effect on disease expression, under a multifactorial-like model, in a condition of iron-loading anemia due to ineffective erythropoiesis.


Asunto(s)
Anemia Diseritropoyética Congénita/genética , Proteínas Morfogenéticas Óseas/fisiología , Sobrecarga de Hierro/etiología , Hígado/metabolismo , Hormonas Peptídicas/genética , Transducción de Señal/genética , Proteínas Smad/fisiología , Adolescente , Adulto , Anemia Diseritropoyética Congénita/complicaciones , Anemia Diseritropoyética Congénita/metabolismo , Transfusión Sanguínea , Proteína Morfogenética Ósea 6/farmacología , Línea Celular , Niño , Eritropoyesis/genética , Femenino , Estudios de Asociación Genética , Hepcidinas/biosíntesis , Hepcidinas/sangre , Hepcidinas/genética , Humanos , Masculino , Hormonas Peptídicas/sangre , Hormonas Peptídicas/farmacología , Hormonas Peptídicas/fisiología , Proteínas Recombinantes/farmacología , Índice de Severidad de la Enfermedad , Proteínas Smad/biosíntesis , Proteínas Smad/genética , Adulto Joven
13.
Acta Biomater ; 96: 258-270, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31302300

RESUMEN

Multiple myeloma is a malignant disease characterized by accumulation of clonal plasma cells in the bone marrow. Uncoupling of bone formation and resorption by myeloma cells leads to osteolytic lesions. These are prone to fracture and represent a possible survival space for myeloma cells under treatment causing disease relapse. Here we report on a novel approach suitable for local treatment of multiple myeloma based on hyaluronic acid (HA) hydrogels mimicking the physical properties of the bone marrow. The HA hydrogels are complexed with heparin to achieve sustained presentation and controlled release of bone morphogenetic protein 6 (BMP-6). Others and we have shown that BMP-6 induces myeloma cell apoptosis and bone formation. Using quartz crystal microbalance and enzyme-linked immunosorbent assay, we measured an initial surface density of 400 ng BMP6/cm2, corresponding to two BMP-6 per heparin molecule, with 50% release within two weeks. HA-hydrogels presenting BMP-6 enhanced the phosphorylation of Smad 1/5 while reducing the activity of BMP-6 antagonist sclerostin. These materials induced osteogenic differentiation of mesenchymal stromal cells and decreased the viability of myeloma cell lines and primary myeloma cells. BMP-6 functionalized HA-hydrogels represent a promising material for local treatment of myeloma-induced bone disease and residual myeloma cells within lesions to minimize disease relapse or fractures. STATEMENT OF SIGNIFICANCE: Multiple myeloma is a hematological cancer characterized by the accumulation of clonal plasma cells in the bone marrow and local suppression of bone formation, resulting in osteolytic lesions and fractures. Despite recent advances in systemic treatment of multiple myeloma, it is rare to achieve a targeted suppression of myeloma cells and healing of bone lesions. Here we present hydrogels which mimic the physico-chemical properties of the bone marrow, consisting of hyaluronic acid with crosslinked heparin for the controlled presentation of bioactive BMP-6. The hydrogels decrease the viability of myeloma cell lines and primary myeloma cells and induces osteogenic differentiation of mesenchymal stromal cells. The presentation of BMP-6 in the hyaluronan hydrogels enhances the phosphorylation of Smad1/5 while reducing the activity of the BMP-6 antagonist sclerostin. As such, BMP-6 functionalized hyaluronan hydrogels represent a promising material for the localized eradication of myeloma cells.


Asunto(s)
Proteína Morfogenética Ósea 6 , Diferenciación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Ácido Hialurónico , Hidrogeles , Células Madre Mesenquimatosas , Mieloma Múltiple/metabolismo , Osteogénesis/efectos de los fármacos , Proteína Morfogenética Ósea 6/química , Proteína Morfogenética Ósea 6/farmacología , Humanos , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Hidrogeles/química , Hidrogeles/farmacología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Mieloma Múltiple/patología , Células Tumorales Cultivadas
14.
Biol Reprod ; 101(2): 445-456, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31210269

RESUMEN

Connective tissue growth factor (also known as CTGF or CCN2) is a secreted matricellular protein that belongs to the CCN family. With wide-ranging biological activities and tissue expression patterns, CTGF plays a critical role in regulating various cellular functions. In the female reproductive system, CTGF is highly expressed in granulosa cells in growing ovarian follicles and is involved in the regulation of follicular development, ovulation, and luteal function. In the mammalian ovary, bone morphogenetic protein 6 (BMP6) is an important intraovarian modulator of follicular development. In this study, we demonstrated that BMP6 treatment significantly increased the expression of CTGF in both primary and immortalized human granulosa cells. Using both pharmacological inhibitors and Small interfering RNA-mediated knockdown approaches, we showed that ALK2 and ALK3 type I receptors are required for BMP6-induced cellular activities. Furthermore, this effect is most likely mediated by a Sma- and Mad-related protein (SMAD)-dependent pathway. Our studies provide novel insight into the molecular mechanisms by which an intraovarian growth factor affects the production of another factor via a paracrine effect in human granulosa cells.


Asunto(s)
Proteína Morfogenética Ósea 6/farmacología , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Células de la Granulosa/metabolismo , Proteínas Smad/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Proteína 61 Rica en Cisteína/genética , Proteína 61 Rica en Cisteína/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Células de la Granulosa/efectos de los fármacos , Humanos , Luteinización , Hormona Luteinizante , Transducción de Señal , Proteínas Smad/genética
15.
Sci Transl Med ; 11(489)2019 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-31019025

RESUMEN

Bone morphogenetic protein (BMP)/carriers approved for orthopedic procedures achieve efficacy superior or equivalent to autograft bone. However, required supraphysiological BMP concentrations have been associated with potential local and systemic adverse events. Suboptimal BMP/receptor binding and rapid BMP release from approved carriers may contribute to these outcomes. To address these issues and improve efficacy, we engineered chimeras with increased receptor binding by substituting BMP-6 and activin A receptor binding domains into BMP-2 and optimized a carrier for chimera retention and tissue ingrowth. BV-265, a BMP-2/BMP-6/activin A chimera, demonstrated increased binding affinity to BMP receptors, including activin-like kinase-2 (ALK2) critical for bone formation in people. BV-265 increased BMP intracellular signaling, osteogenic activity, and expression of bone-related genes in murine and human cells to a greater extent than BMP-2 and was not inhibited by BMP antagonist noggin or gremlin. BV-265 induced larger ectopic bone nodules in rats compared to BMP-2 and was superior to BMP-2, BMP-2/6, and other chimeras in nonhuman primate bone repair models. A composite matrix (CM) containing calcium-deficient hydroxyapatite granules suspended in a macroporous, fenestrated, polymer mesh-reinforced recombinant human type I collagen matrix demonstrated improved BV-265 retention, minimal inflammation, and enhanced handling. BV-265/CM was efficacious in nonhuman primate bone repair models at concentrations ranging from 1/10 to 1/30 of the BMP-2/absorbable collagen sponge (ACS) concentration approved for clinical use. Initial toxicology studies were negative. These results support evaluations of BV-265/CM as an alternative to BMP-2/ACS in clinical trials for orthopedic conditions requiring augmented healing.


Asunto(s)
Activinas/química , Proteína Morfogenética Ósea 6/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Activinas/farmacología , Animales , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 6/farmacología , Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular/efectos de los fármacos , Humanos , Osteogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
16.
J Tissue Eng Regen Med ; 13(5): 846-856, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30815997

RESUMEN

Nude mice have been extensively used to investigate the potency of tissue engineering strategies for bone repair. However, the contribution of pro-inflammatory and proregenerative stimuli of the host for the process of new bone formation and integration remains poorly understood. In this study, ectopic bone formation was investigated in nude (Nu) versus wild-type (WT) mice. Calcium phosphate (CaP) scaffolds (CopiOs [Zimmer] and Bio-Oss [Geistlich]) were loaded with different concentrations of rhBMP6 (40, 120, and 240 ng/mm3 rhBMP6) and implanted subcutaneously in Nu (BALB/c and NMR1) and WT (BALB/c and c57BL/6) mice. CaP scaffolds loaded with rhBMP6 did not form bone in WT mice. However, in Nu mice, 40 ng/mm3 rhBMP6 was sufficient to generate relevant volumes of new bone at 6 weeks after implantation. Looking into potential underlying mechanisms, TNF-α blocking antibodies were injected intraperitoneally but could not restore bone formation. Also, mouse periosteal cells (mPDCs) seeded in CopiOs loaded with rhBMP6 did not significantly improve the outcome. Abrogation of bone formation was associated with dense cellular infiltration, in particular with the presence of CD3+ T-lymphocytes. To probe a correlation between calcium ions and impaired bone formation in WT mice, type 1 collagen gels were loaded with rhBMP6 and calcium chloride and injected subcutaneously. These gels generated new bone in WT mice despite the increased percentage of CD3+ cells at Day 3 after implantation as compared with control gels. Overall, this study illustrated the negative effect of the inflammatory host response on the bone-forming capacity of rhBMP6 coated on bioceramic scaffolds.


Asunto(s)
Proteína Morfogenética Ósea 6 , Fosfatos de Calcio/química , Osteogénesis/efectos de los fármacos , Andamios del Tejido/química , Animales , Proteína Morfogenética Ósea 6/química , Proteína Morfogenética Ósea 6/farmacocinética , Proteína Morfogenética Ósea 6/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología
17.
Nat Microbiol ; 4(2): 339-351, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30510168

RESUMEN

Understanding the control of viral infections is of broad importance. Chronic hepatitis C virus (HCV) infection causes decreased expression of the iron hormone hepcidin, which is regulated by hepatic bone morphogenetic protein (BMP)/SMAD signalling. We found that HCV infection and the BMP/SMAD pathway are mutually antagonistic. HCV blunted induction of hepcidin expression by BMP6, probably via tumour necrosis factor (TNF)-mediated downregulation of the BMP co-receptor haemojuvelin. In HCV-infected patients, disruption of the BMP6/hepcidin axis and genetic variation associated with the BMP/SMAD pathway predicted the outcome of infection, suggesting that BMP/SMAD activity influences antiviral immunity. Correspondingly, BMP6 regulated a gene repertoire reminiscent of type I interferon (IFN) signalling, including upregulating interferon regulatory factors (IRFs) and downregulating an inhibitor of IFN signalling, USP18. Moreover, in BMP-stimulated cells, SMAD1 occupied loci across the genome, similar to those bound by IRF1 in IFN-stimulated cells. Functionally, BMP6 enhanced the transcriptional and antiviral response to IFN, but BMP6 and related activin proteins also potently blocked HCV replication independently of IFN. Furthermore, BMP6 and activin A suppressed growth of HBV in cell culture, and activin A inhibited Zika virus replication alone and in combination with IFN. The data establish an unappreciated important role for BMPs and activins in cellular antiviral immunity, which acts independently of, and modulates, IFN.


Asunto(s)
Activinas/farmacología , Antivirales/farmacología , Proteína Morfogenética Ósea 6/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Antivirales/metabolismo , Células Cultivadas , Endopeptidasas/genética , Hepacivirus/efectos de los fármacos , Hepatitis C/tratamiento farmacológico , Hepatitis C/metabolismo , Hepcidinas/genética , Humanos , Factores Reguladores del Interferón/genética , Interferón-alfa/farmacología , Interferón-alfa/uso terapéutico , ARN Viral/metabolismo , Transducción de Señal/genética , Proteína Smad1/genética , Ubiquitina Tiolesterasa , Replicación Viral/efectos de los fármacos , Virus Zika/efectos de los fármacos
18.
JAMA Facial Plast Surg ; 21(2): 110-117, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30520953

RESUMEN

IMPORTANCE: Osseous craniofacial defects are currently reconstructed with bone grafting, rigid fixation, free tissue transfer, and/or recombinant human bone morphogenetic protein 2. Although these treatment options often have good outcomes, they are associated with substantial morbidity, and many patients are not candidates for free tissue transfer. OBJECTIVE: To assess whether polysaccharide-based scaffold (PS) constructs that are cross-linked with smoothened agonist (SAG), vascular endothelial growth factor (VEGF), and bone morphogenetic protein 6 (BMP-6) would substantially increase bone regeneration. DESIGN, SETTING, AND PARTICIPANTS: This animal model study was conducted at the University of Virginia School of Medicine Cui Laboratory from March 1, 2017, to June 30, 2017. Thirty-three 10-week-old female Lewis rats were acquired for the study. Bilateral nonsegmental critical-sized defects were created in the angle of rat mandibles. The defects were either left untreated or filled with 1 of the 9 PSs. The rats were killed after 8 weeks, and bone regeneration was evaluated using microcomputed tomographic imaging and mechanical testing. Analysis of variance testing was used to compare the treatment groups. MAIN OUTCOMES AND MEASURES: Blinded analysis and computer analysis of the microcomputed tomographic images were used to assess bone regeneration. RESULTS: In the 33 female Lewis rats, minimal healing was observed in the untreated mandibles. Addition of SAG was associated with increases in bone regeneration and bone density in all treatment groups, and maximum bone healing was seen in the group with BMP-6, VEGF, and SAG cross-linked to PS. For each of the 5 no scaffold group vs BMP-6, VEGF, and SAG cross-linked to PS group comparisons, mean defect bone regeneration was 4.14% (95% CI, 0.94%-7.33%) vs 66.19% (95% CI, 54.47%-77.90%); mean bone volume, 14.52 mm3 (95% CI, 13.07-15.97 mm3) vs 20.87 mm3 (95% CI, 14.73- 27.01 mm3); mean bone surface, 68.97 mm2 (95% CI, 60.08-77.85 mm2) vs 96.77 mm2 (95% CI, 76.11-117.43 mm2); mean ratio of bone volume to total volume, 0.11 (95% CI, 0.10-0.11) vs 0.15 (95% CI, 0.10-0.19); and mean connectivity density 0.03 (95% CI, 0.02-0.05) vs 0.32 (95% CI, 0.25-0.38). On mechanical testing, mandibles with untreated defects broke with less force than control mandibles in which no defect was made, although this force did not reach statistical significance. No significant difference in force to fracture was observed among the treatment groups. CONCLUSIONS AND RELEVANCE: In this rat model study, activation of the hedgehog signaling pathway using smoothened agonist was associated with increased craniofacial bone regeneration compared with growth factors alone, including US Food and Drug Administration-approved recombinant human bone morphogenetic protein 2. Pharmaceuticals that target this pathway may offer a new reconstructive option for bony craniofacial defects as well as nonunion and delayed healing fractures. LEVEL OF EVIDENCE: NA.


Asunto(s)
Regeneración Ósea/fisiología , Proteínas Hedgehog/metabolismo , Mandíbula/cirugía , Animales , Densidad Ósea , Proteína Morfogenética Ósea 2/farmacología , Proteína Morfogenética Ósea 6/farmacología , Sustitutos de Huesos/farmacología , Trasplante Óseo , Femenino , Modelos Animales , Ratas , Ratas Endogámicas Lew , Transducción de Señal , Andamios del Tejido , Factor A de Crecimiento Endotelial Vascular/farmacología , Cicatrización de Heridas , Microtomografía por Rayos X
19.
Acta Diabetol ; 56(3): 365-371, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30539233

RESUMEN

AIMS: Bone morphogenetic proteins (BMPs) are involved in the development and homeostasis of multiple organs and tissues. There has been a significant focus on understanding the role of BMPs in pancreatic ß-cell dysfunction associated with type 2 diabetes (T2D). Our objective was to investigate the relationship between BMP6 and glucose homeostasis. METHODS: Ob/ob mice were treated with BMP6 for 6 days and analyzed for insulin release, body weight, lipid parameters and glucose tolerance. Quantitative real-time PCR, chromatin immunoprecipitation and glucose output assays were used to assess BMP6 effect on gluconeogenesis in rat hepatoma H4IIE cells. Specificity of BMP6 receptors was characterized by the utilization of various receptor Fc fusion proteins in luciferase reporter gene and glucose output assays in INS1 and H4IIE cells. RESULTS: Treatment of ob/ob mice with BMP6 for 6 days resulted in a reduction of circulating glucose and lipid levels, followed by a significantly elevated plasma insulin level in a dose-dependent manner. In addition, BMP6 improved the glucose excursion during an oral glucose tolerance test, lowering the total glycemic response by 21%. In rat H4IIE hepatoma cells, BMP6 inhibited gluconeogenesis and glucose output via downregulation the PepCK expression. Moreover, BMP6 inhibited glucose production regardless of the presence of cAMP, antagonizing its glycogenolytic effect. BMP6 acted on pancreatic and liver cells utilizing Alk3, Alk6 and ActRIIA serine/threonine kinase receptors. CONCLUSIONS: Collectively, we demonstrate that BMP6 improves glycaemia in T2D mice and regulates glucose metabolism in hepatocytes representing an exciting prospect for future treatments of diabetes.


Asunto(s)
Proteína Morfogenética Ósea 6/farmacología , Diabetes Mellitus Experimental/metabolismo , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Animales , Glucemia/genética , Glucemia/metabolismo , Proteína Morfogenética Ósea 6/fisiología , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Homeostasis/genética , Insulina/metabolismo , Masculino , Ratones , Ratones Obesos , Ratas , Proteínas Recombinantes/farmacología
20.
Acta Biomater ; 80: 97-107, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30267882

RESUMEN

Cell based combination products with growth factors on optimal carriers represent a promising tissue engineering strategy to treat large bone defects. In this concept, bone morphogenetic protein (BMP) and calcium phosphate (CaP)-based scaffolds can act as potent components of the constructs to steer stem cell specification, differentiation and initiate subsequent in vivo bone formation. However, limited insight into BMP dosage and the cross-talk between BMP and CaP materials, hampers the optimization of in vivo bone formation and subsequent clinical translation. Herein, we combined human periosteum derived progenitor cells with different doses of BMP6 and with three types of clinical grade CaP-scaffolds (ChronOs®, ReproBone™, & CopiOs®). Comprehensive cellular and molecular analysis was performed based on in vitro cell metabolic activity and signaling pathway activation, as well as in vivo ectopic bone forming capacity after 2 weeks and 5 weeks in nude mice. Our data showed that cells seeded on CaP scaffolds with an intermediate Ca2+ release rate combined with low or medium dosage of BMP6 demonstrated a robust new bone formation after 5 weeks, which was contributed by both donor and host cells. This phenomenon might be due to the delicate balance between Ca2+ and BMP pathways, allowing an appropriate activation of the canonical BMP signaling pathway that is required for in vivo bone formation. For high BMP6 dosage, we found that the BMP6 dosage overrides the effect of the Ca2+ release rate and this appeared to be a dominant factor for ectopic bone formation. Taken together, this study illustrates the importance of matching BMP dosage and CaP properties to allow an appropriate activation of canonical BMP signaling that is crucial for in vivo bone formation. It also provides insightful knowledge with regard to clinical translation of cell-based constructs for bone regeneration. STATEMENT OF SIGNIFICANCE: The combination of bone morphogenetic proteins (BMP) and calcium phosphate (CaP)-based biomaterials with mesenchymal stromal cells represents a promising therapeutic strategy to treat large bone defects, an unmet medical need. However, there is limited insight into the optimization of these combination products, which hampers subsequent successful clinical translation. Our data reveal a delicate balance between Ca2+ and BMP pathways, allowing an appropriate activation of canonical BMP signaling required for in vivo bone formation. Our findings illustrate the importance of matching BMP dosage and CaP properties in the development of cell-based constructs for bone regeneration.


Asunto(s)
Proteína Morfogenética Ósea 6/farmacología , Fosfatos de Calcio/farmacología , Osteogénesis/efectos de los fármacos , Periostio/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Adulto , Recuento de Células , Glucosa/metabolismo , Humanos , Implantes Experimentales , Ácido Láctico/metabolismo , Análisis Multivariante , Osteocalcina/metabolismo , Análisis de Componente Principal , Microtomografía por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...