Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
J Periodontal Res ; 59(3): 611-621, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38500269

RESUMO

BACKGROUND AND OBJECTIVE: Forkhead box-O 1 (FOXO1) is a transcription factor actively involved in oral wound healing at the epithelial barrier. However, less is known regarding the role of FOXO1 during the tissue repair response in the connective tissue compartment. This study explored the involvement of FOXO1 in the modulation of fibroblast activity related to wound healing. METHODS: Primary cultures of human gingival fibroblasts were obtained from four healthy young donors. Myofibroblastic differentiation, collagen gel contraction, cell migration, cell spreading, and integrin activation were evaluated in the presence or absence of a FOXO1 inhibitor (AS1842856). Variations in mRNA and proteins of interest were evaluated through qRT-PCR and western blot, respectively. Distribution of actin, α-smooth muscle actin, and ß1 integrin was evaluated using immunofluorescence. FOXO1 and TGF-ß1 expression in gingival wound healing was assessed by immunohistochemistry in gingival wounds performed in C57BL/6 mice. Images were analyzed using ImageJ/Fiji. ANOVA or Kruskal-Wallis test followed by Tukey's or Dunn's post-hoc test was performed. All data are expressed as mean ± SD. p < .05 was considered statistically significant. RESULTS: FOXO1 inhibition caused a decrease in the expression of the myofibroblastic marker α-SMA along with a reduction in fibronectin, type I collagen, TGF-ß1, and ß1 integrin mRNA level. The FOXO1 inhibitor also caused decreases in cell migration, cell spreading, collagen gel contraction, and ß1 integrin activation. FOXO1 and TGF-ß1 were prominently expressed in gingival wounds in fibroblastic cells located at the wound bed. CONCLUSION: The present study indicates that FOXO1 plays an important role in the modulation of several wound-healing functions in gingival fibroblast. Moreover, our findings reveal an important regulatory role for FOXO1 on the differentiation of gingival myofibroblasts, the regulation of cell migration, and collagen contraction, all these functions being critical during tissue repair and fibrosis.


Assuntos
Actinas , Movimento Celular , Fibroblastos , Proteína Forkhead Box O1 , Gengiva , Cicatrização , Humanos , Gengiva/citologia , Gengiva/metabolismo , Cicatrização/fisiologia , Fibroblastos/metabolismo , Proteína Forkhead Box O1/metabolismo , Animais , Células Cultivadas , Diferenciação Celular , Camundongos Endogâmicos C57BL , Fator de Crescimento Transformador beta1/metabolismo , Camundongos , Integrina beta1 , Miofibroblastos , Quinolonas
2.
Front Endocrinol (Lausanne) ; 14: 1136117, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37576976

RESUMO

Introduction: Diabetes mellitus is associated with higher risks of long bone and jaw fractures. It is also associated with a higher incidence of delayed union or non-union. Our previous investigations concluded that a dominant mechanism was the premature loss of cartilage during endochondral bone formation associated with increased osteoclastic activities. We tested the hypothesis that FOXO1 plays a key role in diabetes-impaired angiogenesis and chondrocyte apoptosis. Methods: Closed fractures of the femur were induced in mice with lineage-specific FOXO1 deletion in chondrocytes. The control group consisted of mice with the FOXO1 gene present. Mice in the diabetic group were rendered diabetic by multiple streptozotocin injections, while mice in the normoglycemic group received vehicle. Specimens were collected 16 days post fracture. The samples were fixed, decalcified, and embedded in paraffin blocks for immunostaining utilizing anti cleaved caspase-3 or CD31 specific antibodies compared with matched control IgG antibody, and apoptosis by the TUNEL assay. Additionally, ATDC5 chondrocytes were examined in vitro by RT-PCR, luciferase reporter and chromatin immunoprecipitation assays. Results: Diabetic mice had ~ 50% fewer blood vessels compared to normoglycemic mice FOXO1 deletion in diabetic mice partially rescued the low number of blood vessels (p < 0.05). Additionally, diabetes increased caspase-3 positive and apoptotic chondrocytes by 50%. FOXO1 deletion in diabetic animals blocked the increase in both to levels comparable to normoglycemic animals (p < 0.05). High glucose (HG) and high advanced glycation end products (AGE) levels stimulated FOXO1 association with the caspase-3 promoter in vitro, and overexpression of FOXO1 increased caspase-3 promoter activity in luciferase reporter assays. Furthermore, we review previous mechanistic studies demonstrating that tumor necrosis factor (TNF) inhibition reverses impaired angiogenesis and reverses high levels of chondrocyte apoptosis that occur in fracture healing. Discussion: New results presented here, in combination with recent studies, provide a comprehensive overview of how diabetes, through high glucose levels, AGEs, and increased inflammation, impair the healing process by interfering with angiogenesis and stimulating chondrocyte apoptosis. FOXO1 in diabetic fractures plays a negative role by reducing new blood vessel formation and increasing chondrocyte cell death which is distinct from its role in normal fracture healing.


Assuntos
Condrócitos , Diabetes Mellitus Experimental , Proteína Forkhead Box O1 , Animais , Camundongos , Apoptose/genética , Caspase 3 , Condrócitos/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Consolidação da Fratura/fisiologia , Glucose/metabolismo , Proteína Forkhead Box O1/genética
4.
Curr Osteoporos Rep ; 21(2): 128-146, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36862360

RESUMO

PURPOSE OF REVIEW: To review the role of the immune cells and their interaction with cells found in gingiva, periodontal ligament, and bone that leads to net bone loss in periodontitis or bone remodeling in orthodontic tooth movement. RECENT FINDINGS: Periodontal disease is one of the most common oral diseases causing inflammation in the soft and hard tissues of the periodontium and is initiated by bacteria that induce a host response. Although the innate and adaptive immune response function cooperatively to prevent bacterial dissemination, they also play a major role in gingival inflammation and destruction of the connective tissue, periodontal ligament, and alveolar bone characteristic of periodontitis. The inflammatory response is triggered by bacteria or their products that bind to pattern recognition receptors that induce transcription factor activity to stimulate cytokine and chemokine expression. Epithelial, fibroblast/stromal, and resident leukocytes play a key role in initiating the host response and contribute to periodontal disease. Single-cell RNA-seq (scRNA-seq) experiments have added new insight into the roles of various cell types in the response to bacterial challenge. This response is modified by systemic conditions such as diabetes and smoking. In contrast to periodontitis, orthodontic tooth movement (OTM) is a sterile inflammatory response induced by mechanical force. Orthodontic force application stimulates acute inflammatory responses in the periodontal ligament and alveolar bone stimulated by cytokines and chemokines that produce bone resorption on the compression side. On the tension side, orthodontic forces induce the production of osteogenic factors, stimulating new bone formation. A number of different cell types, cytokines, and signaling/pathways are involved in this complex process. Inflammatory and mechanical force-induced bone remodeling involves bone resorption and bone formation. The interaction of leukocytes with host stromal cells and osteoblastic cells plays a key role in both initiating the inflammatory events as well as inducing a cellular cascade that results in remodeling in orthodontic tooth movement or in tissue destruction in periodontitis.


Assuntos
Reabsorção Óssea , Periodontite , Humanos , Osteoclastos/metabolismo , Técnicas de Movimentação Dentária , Reabsorção Óssea/metabolismo , Periodontite/metabolismo , Citocinas/metabolismo , Inflamação/metabolismo
5.
Front Immunol ; 13: 998244, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36304447

RESUMO

Periodontitis involves the loss of connective tissue attachment and alveolar bone. Single cell RNA-seq experiments have provided new insight into how resident cells and infiltrating immune cells function in response to bacterial challenge in periodontal tissues. Periodontal disease is induced by a combined innate and adaptive immune response to bacterial dysbiosis that is initiated by resident cells including epithelial cells and fibroblasts, which recruit immune cells. Chemokines and cytokines stimulate recruitment of osteoclast precursors and osteoclastogenesis in response to TNF, IL-1ß, IL-6, IL-17, RANKL and other factors. Inflammation also suppresses coupled bone formation to limit repair of osteolytic lesions. Bone lining cells, osteocytes and periodontal ligament cells play a key role in both processes. The periodontal ligament contains cells that exhibit similarities to tendon cells, osteoblast-lineage cells and mesenchymal stem cells. Bone lining cells consisting of mesenchymal stem cells, osteoprogenitors and osteoblasts are influenced by osteocytes and stimulate formation of osteoclast precursors through MCSF and RANKL, which directly induce osteoclastogenesis. Following bone resorption, factors are released from resorbed bone matrix and by osteoclasts and osteal macrophages that recruit osteoblast precursors to the resorbed bone surface. Osteoblast differentiation and coupled bone formation are regulated by multiple signaling pathways including Wnt, Notch, FGF, IGF-1, BMP, and Hedgehog pathways. Diabetes, cigarette smoking and aging enhance the pathologic processes to increase bone resorption and inhibit coupled bone formation to accelerate bone loss. Other bone pathologies such as rheumatoid arthritis, post-menopausal osteoporosis and bone unloading/disuse also affect osteoblast lineage cells and participate in formation of osteolytic lesions by promoting bone resorption and inhibiting coupled bone formation. Thus, periodontitis involves the activation of an inflammatory response that involves a large number of cells to stimulate bone resorption and limit osseous repair processes.


Assuntos
Reabsorção Óssea , Doenças Periodontais , Periodontite , Humanos , Proteínas Hedgehog/metabolismo , Osteoblastos/metabolismo , Reabsorção Óssea/patologia , Periodontite/patologia , Doenças Periodontais/metabolismo
6.
Angle Orthod ; 91(5): 664-671, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-33852725

RESUMO

OBJECTIVES: To investigate the role of NF-κB in osteoblast lineage cells and periodontal ligament (PDL) fibroblasts during orthodontic tooth movement (OTM). MATERIALS AND METHODS: Transgenic mice that expressed a dominant negative mutant of the inhibitor of kB kinase (IKK-DN) with lineage specific expression in osteoblastic cells and PDL fibroblasts driven by a response element in the collagen1α1 promoter and matched wild-type (WT) mice were examined. A 10-12 g force was applied by a NiTi coil and maintained for 5 or 12 days. OTM distance, PDL width, and bone volume fraction were measured using micro computed tomography. Osteoclast numbers were counted in tartrate-resistant acid phosphatase-stained sections. Activation of nuclear factor kappa B (NF-kB) was assessed by nuclear localization of p65, and the receptor activator of nuclear factor-κB ligand (RANKL) was measured by immunofluorescence and compared to control specimens with no orthodontic force. RESULTS: OTM-induced NF-kB activation (p65 nuclear localization) in WT mice was largely blocked in transgenic (TG) mice. OTM was significantly reduced in the TG mice compared to WT mice along with reduced osteoclastogenesis, narrower PDL width, higher bone volume fraction, and reduced RANKL expression. CONCLUSIONS: Osteoblast lineage cells and PDL fibroblasts are key contributors to alveolar bone remodeling in OTM through IKKß dependent NF-κB activation.


Assuntos
Ligamento Periodontal , Técnicas de Movimentação Dentária , Animais , Fibroblastos , Camundongos , NF-kappa B , Osteoblastos , Osteoclastos , Ligante RANK , Microtomografia por Raio-X
7.
Bone ; 148: 115905, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33662610

RESUMO

Fracture healing is a multistage process characterized by inflammation, cartilage formation, bone deposition, and remodeling. Chondrocytes are important in producing cartilage that forms the initial anlagen for the hard callus needed to stabilize the fracture site. We examined the role of FOXO1 by selective ablation of FOXO1 in chondrocytes mediated by Col2α1 driven Cre recombinase. Experimental mice with lineage-specific FOXO1 deletion (Col2α1Cre+FOXO1L/L) and negative control littermates (Col2α1Cre-FOXO1L/L) were used for in vivo, closed fracture studies. Unexpectedly, we found that in the early phases of fracture healing, FOXO1 deletion significantly increased the amount of cartilage formed, whereas, in later periods, FOXO1 deletion led to a greater loss of cartilage. FOXO1 was functionally important as its deletion in chondrocytes led to diminished bone formation on day 22. Mechanistically, the early effects of FOXO1 deletion were linked to increased proliferation of chondrocytes through enhanced expression of cell cycle genes that promote proliferation and reduced expression of those that inhibit it and increased expression of cartilage matrix genes. At later time points experimental mice with FOXO1 deletion had greater loss of cartilage, enhanced formation of osteoclasts, increased IL-6 and reduced numbers of M2 macrophages. These results identify FOXO1 as a transcription factor that regulates chondrocyte behavior by limiting the early expansion of cartilage and preventing rapid cartilage loss at later phases.


Assuntos
Condrócitos , Consolidação da Fratura , Animais , Calo Ósseo , Cartilagem , Proteína Forkhead Box O1/genética , Camundongos , Osteoclastos
8.
J Diabetes Res ; 2020: 3714704, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33195703

RESUMO

Diabetes has a significant and negative impact on wound healing, which involves complex interactions between multiple cell types. Keratinocytes play a crucial role in the healing process by rapidly covering dermal and mucosal wound surfaces to reestablish an epithelial barrier with the outside environment. Keratinocytes produce multiple factors to promote reepithelialization and produce factors that enhance connective tissue repair through the elaboration of mediators that stimulate angiogenesis and production of connective tissue matrix. Among the factors that keratinocytes produce to aid healing are transforming growth factor-ß (TGF-ß), vascular endothelial growth factor-A (VEGF-A), connective tissue growth factor (CTGF), and antioxidants. In a diabetic environment, this program is disrupted, and keratinocytes fail to produce growth factors and instead switch to a program that is detrimental to healing. Changes in keratinocyte behavior have been linked to high glucose and advanced glycation end products that alter the activities of the transcription factor, FOXO1. This review examines reepithelialization and factors produced by keratinocytes that upregulate connective tissue healing and angiogenesis and how they are altered by diabetes.


Assuntos
Complicações do Diabetes/metabolismo , Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica , Queratinócitos/citologia , Animais , Antioxidantes/metabolismo , Glicemia/análise , Glicemia/metabolismo , Linhagem da Célula , Movimento Celular , Proliferação de Células , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Células Epiteliais/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Neovascularização Patológica , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização
9.
Theranostics ; 10(14): 6500-6516, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32483466

RESUMO

Microbiome, considered as the "second genome" of the host, is altered in type 1 diabetes mellitus (T1DM) patients to a state of dysbiosis. Mesenchymal stem cell (MSC) transplantation is a promising treatment for T1DM but is limited by several factors in the diabetic host. In this study, we tested the hypothesis that dysbiotic gut microbiota may limit MSC therapy, and modulating gut microbiota may help to improve the effects of MSC transplantation. Methods: NOD/Ltj mice, treated with adipose-derived stem cells (ADSCs), were fed with an antibiotics cocktails (Abx) for 1 week. The blood glucose levels, insulitis, intestinal permeability and gut bacteria translocation to the pancreas were evaluated. 16s rRNA and colon tissue transcription sequencing were performed to analyze beneficial bacteria and reactive host biomolecules in the ADSCs+Abx group. Based on the sequencing results, specific bacteria were gavaged orally to diabetic mice to confirm their effect on ADSCs transplantation in T1DM was determined. Results: We found that the recolonized the diabetic gut microbiota abolished the therapeutic effect of ADSCs. On the contrary, depletion of the diabetic gut microbiota by antibiotics treatment in diabetic mice significantly enhanced the therapeutic effects of ADSCs as measured by reversal of hyperglycemia, insulitis, and increased insulin output. Mechanistically, treatment with antibiotics increased the abundance of Bifidobacterium in the gut and reduced bacterial translocation to the pancreas by promoting Mucin2 expression and thickening the mucus layer through TRPM7. The mechanism was confirmed the re-colonization of the gut by B.breve through oral gavage that produced similar results. Conclusions: These results provide the rationale for a new approach to improve MSC therapy for T1DM by altering the gut microbiota.


Assuntos
Diabetes Mellitus Tipo 1 , Microbioma Gastrointestinal , Transplante de Células-Tronco Mesenquimais , Animais , Antibacterianos/farmacologia , Bifidobacterium/crescimento & desenvolvimento , Células Cultivadas/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/microbiologia , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/microbiologia , Diabetes Mellitus Tipo 1/terapia , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Humanos , Células-Tronco Mesenquimais , Camundongos , Camundongos Endogâmicos NOD , RNA Ribossômico 16S/genética
10.
Periodontol 2000 ; 82(1): 214-224, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31850631

RESUMO

The susceptibility and severity of periodontal diseases is made more severe by diabetes, with the impact on the disease process inversely proportional to the level of glycemic control. Although type 1 diabetes mellitus and type 2 diabetes mellitus have different etiologies, and their impact on bone is not identical, they share many of the same complications. Studies in animals and humans agree that both forms of diabetes increase inflammatory events in periodontal tissue, impair new bone formation, and increase expression of RANKL in response to bacterial challenge. High levels of glucose, reactive oxygen species, and advanced glycation end-products are found in the periodontium of diabetic individuals and lead to increased activation of nuclear factor-kappa B and expression of inflammatory cytokines such as tumor necrosis factor and interleukin-1. Studies in animals, moreover, suggest that there are multiple cell types in periodontal tissues that are affected by diabetes, including leukocytes, vascular cells, mesenchymal stem cells, periodontal ligament fibroblasts, osteoblasts, and osteocytes. The etiology of periodontal disease involves the host response to bacterial challenge that is affected by diabetes, which increases the expression of RANKL and reduces coupled bone formation. In addition, the inflammatory response also modifies the oral microbiota to render it more pathogenic, as demonstrated by increased inflammation and bone loss in animals where bacteria are transferred from diabetic donors to germ-free hosts compared with transfer from normoglycemic donors. This approach has the advantage of not relying upon limited knowledge of the specific bacterial taxa to determine pathogenicity, and examines the overall impact of the microbiota rather than the presumed pathogenicity of a few bacterial groups. Thus, animal studies have provided new insights into pathogenic mechanisms that identify cause-and-effect relationships that are difficult to perform in human studies.


Assuntos
Diabetes Mellitus Tipo 2 , Doenças Periodontais , Periodontite , Animais , Citocinas , Humanos , Periodonto
11.
Front Immunol ; 10: 2530, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31849924

RESUMO

FOXO1 transcription factors affect a number of cell types that are important in the host response. Cell types whose functions are modulated by FOXO1 include keratinocytes in the skin and mucosal dermis, neutrophils and macrophages, dendritic cells, Tregs and B-cells. FOXO1 is activated by bacterial or cytokine stimulation. Its translocation to the nucleus and binding to promoter regions of genes that have FOXO response elements is stimulated by the MAP kinase pathway and inhibited by the PI3 kinase/AKT pathway. Downstream gene targets of FOXO1 include pro-inflammatory signaling molecules (TLR2, TLR4, IL-1ß, and TNF-α), wound healing factors (TGF-ß, VEGF, and CTGF) adhesion molecules (integrins-ß1, -ß3, -ß6, αvß3, CD11b, CD18, and ICAM-1), chemokine receptors (CCR7 and CXCR2), B cell regulators (APRIL and BLYS), T-regulatory modulators (Foxp3 and CTLA-4), antioxidants (GPX-2 and cytoglobin), and DNA repair enzymes (GADD45α). Each of the above cell types are found in oral mucosa and modulated by bacteria or an inflammatory microenvironment. FOXO1 contributes to the regulation of these cells, which collectively maintain and repair the epithelial barrier, formation and activation of Tregs that are needed to resolve inflammation, mobilization, infiltration, and activation of anti-bacterial defenses in neutrophils, and the homing of dendritic cells to lymph nodes to induce T-cell and B-cell responses. The goal of the manuscript is to review how the transcription factor, FOXO1, contributes to the activation and regulation of key leukocytes needed to maintain homeostasis and respond to bacterial challenge in oral mucosal tissues. Examples are given with an emphasis on lineage specific deletion of Foxo1 to explore the impact of FOXO1 on cell behavior, inflammation and susceptibility to infection.


Assuntos
Proteína Forkhead Box O1/metabolismo , Imunidade nas Mucosas , Mucosa/imunologia , Mucosa/metabolismo , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Suscetibilidade a Doenças , Proteína Forkhead Box O1/genética , Regulação da Expressão Gênica , Humanos , Linfócitos/imunologia , Linfócitos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Doenças Periodontais/etiologia , Doenças Periodontais/metabolismo , Doenças Periodontais/patologia , Transdução de Sinais
12.
Diabetes ; 68(11): 2095-2106, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31439641

RESUMO

Type 1 diabetes (T1D) imposes a significant health burden by negatively affecting tissue regeneration during wound healing. The adverse effect of diabetes is attributed to high levels of inflammation, but the cellular mechanisms responsible remain elusive. In this study, we show that intrinsic skeletal stem cells (SSCs), a subset of mesenchymal stem cells, are essential for resolution of inflammation to occur during osseous healing by using genetic approaches to selectively ablate SSCs. T1D caused aberrant nuclear factor-κB (NF-κB) activation in SSCs and substantially enhanced inflammation in vivo. Constitutive or tamoxifen-induced inhibition of NF-κB in SSCs rescued the impact of diabetes on inflammation, SSC expansion, and tissue formation. In contrast, NF-κB inhibition in chondrocytes failed to reverse the effect of T1D. Mechanistically, diabetes caused defective proresolving macrophage (M2) polarization by reducing TGF-ß1 expression by SSCs, which was recovered by NF-κB inhibition or exogenous TGF-ß1 treatment. These data identify an underlying mechanism for altered healing in T1D and demonstrate that diabetes induces NF-κB hyperactivation in SSCs to disrupt their ability to modulate M2 polarization and resolve inflammation.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Inflamação/metabolismo , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , NF-kappa B/metabolismo , Animais , Fraturas do Fêmur/metabolismo , Consolidação da Fratura/fisiologia , Camundongos , Camundongos Transgênicos , Transdução de Sinais
13.
Bone ; 123: 159-167, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30904630

RESUMO

Diabetes increases the risk of fracture, impairs fracture healing and causes rapid loss of the fracture callus cartilage, which was linked to increased FOXO1 expression in chondrocytes. We recently demonstrated that deletion of FOXO1 in chondrocytes blocked the premature removal of cartilage associated with endochondral bone formation during fracture healing. However, the ultimate impact of this deletion on mechanical strength was not investigated and remains unknown. Closed fractures were induced in Col2α1Cre+.FOXO1L/L mice with lineage specific deletion of FOXO1 in chondrocytes compared to littermate controls. Type 1 diabetes was induced by multiple low dose streptozotocin treatment. Thirty-five days after fracture micro CT analysis showed that diabetes significantly reduced callus volume and bone volume (P < 0.05), both which were reversed by FOXO1 deletion in chondrocytes. Diabetes significantly reduced mechanical strength measured by maximum torque, stiffness, modulus of rigidity and toughness and FOXO1 deletion in diabetic mice rescued each parameter (P < 0.05). Diabetes also reduced both bone volume and mechanical strength in non-fractured femurs. However, FOXO1 deletion did not affect bone volume or strength in non-fractured bone. These results point to the important effect that diabetes has on chondrocytes and show for the first time that the premature removal of cartilage induced by FOXO1 in chondrocytes has a significant impact on the mechanical strength of the healing bone.


Assuntos
Condrócitos/metabolismo , Diabetes Mellitus Experimental/metabolismo , Fraturas do Colo Femoral/metabolismo , Proteína Forkhead Box O1/deficiência , Consolidação da Fratura/fisiologia , Deleção de Genes , Animais , Fenômenos Biomecânicos/fisiologia , Condrócitos/patologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Fraturas do Colo Femoral/genética , Fraturas do Colo Femoral/patologia , Proteína Forkhead Box O1/genética , Camundongos , Camundongos Transgênicos
14.
J Bone Miner Res ; 34(3): 547-556, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30347467

RESUMO

Chondrocytes play an essential role in fracture healing by producing cartilage, which forms an anlage for endochondral ossification that stabilizes the healing fracture callus. More recently it has been appreciated that chondrocytes have the capacity to produce factors that may affect the healing process. We examined the role of chondrocytes in angiogenesis during fracture healing and the role of the transcription factor forkhead box-O 1 (FOXO1), which upregulates wound healing in soft tissue. Closed fractures were induced in experimental mice with lineage-specific FOXO1 deletion by Cre recombinase under the control of a collagen-2α1 promoter element (Col2α1Cre+ FOXO1L/L ) and Cre recombinase negative control littermates containing flanking loxP sites (Col2α1Cre- FOXO1L/L ). Experimental mice had significantly reduced CD31+ new vessel formation. Deletion of FOXO1 in chondrocytes in vivo suppressed the expression of vascular endothelial growth factor-A (VEGFA) at both the protein and mRNA levels. Overexpression of FOXO1 in chondrocytes in vitro increased VEGFA mRNA levels and VEGFA transcriptional activity whereas silencing FOXO1 reduced it. Moreover, FOXO1 interacted directly with the VEGFA promoter and a deacetylated FOXO1 mutant enhanced VEGFA expression whereas an acetylated FOXO1 mutant did not. Lastly, FOXO1 knockdown by siRNA significantly reduced the capacity of chondrocytes to stimulate microvascular endothelial cell tube formation in vitro. The results indicate that chondrocytes play a key role in angiogenesis which is FOXO1 dependent and that FOXO1 in chondrocytes regulates a potent angiogenic factor, VEGFA. These studies provide new insight into fracture healing given the important role of vessel formation in the fracture repair process. © 2018 American Society for Bone and Mineral Research.


Assuntos
Condrócitos/metabolismo , Proteína Forkhead Box O1/metabolismo , Consolidação da Fratura , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Linhagem Celular , Colágeno Tipo II/biossíntese , Colágeno Tipo II/genética , Regulação para Baixo , Células Endoteliais/patologia , Proteína Forkhead Box O1/genética , Deleção de Genes , Camundongos , Camundongos Transgênicos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Transcrição Gênica , Fator A de Crescimento do Endotélio Vascular/genética
15.
Diabetes ; 67(12): 2682-2694, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30279162

RESUMO

Type 1 diabetes impairs fracture healing. We tested the hypothesis that diabetes affects chondrocytes to impair fracture healing through a mechanism that involves the transcription factor FOXO1. Type 1 diabetes was induced by streptozotocin in mice with FOXO1 deletion in chondrocytes (Col2α1Cre+FOXO1L/L) or littermate controls (Col2α1Cre-FOXO1L/L) and closed femoral fractures induced. Diabetic mice had 77% less cartilage and 30% less bone than normoglycemics evaluated histologically and by micro-computed tomography. Both were reversed with lineage-specific FOXO1 ablation. Diabetic mice had a threefold increase in osteoclasts and a two- to threefold increase in RANKL mRNA or RANKL-expressing chondrocytes compared with normoglycemics. Both parameters were rescued by FOXO1 ablation in chondrocytes. Conditions present in diabetes, high glucose (HG), and increased advanced glycation end products (AGEs) stimulated FOXO1 association with the RANKL promoter in vitro, and overexpression of FOXO1 increased RANKL promoter activity in luciferase reporter assays. HG and AGE stimulated FOXO1 nuclear localization, which was reversed by insulin and inhibitors of TLR4, histone deacetylase, nitric oxide, and reactive oxygen species. The results indicate that chondrocytes play a prominent role in diabetes-impaired fracture healing and that high levels of glucose, AGEs, and tumor necrosis factor-α, which are elevated by diabetes, alter RANKL expression in chondrocytes via FOXO1.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Fraturas do Fêmur/metabolismo , Proteína Forkhead Box O1/metabolismo , Consolidação da Fratura/genética , Animais , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Diabetes Mellitus Experimental/genética , Fraturas do Fêmur/genética , Proteína Forkhead Box O1/genética , Consolidação da Fratura/efeitos dos fármacos , Regulação da Expressão Gênica , Glucose/farmacologia , Produtos Finais de Glicação Avançada/farmacologia , Camundongos , Camundongos Knockout , Ligante RANK/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Microtomografia por Raio-X
16.
J Oral Maxillofac Surg ; 76(8): 1677-1687, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29572133

RESUMO

PURPOSE: The increasing prevalence of obesity or metabolic syndrome (O/MS) and type 2 diabetes mellitus (DM) remains a global health concern. Clinically relevant and practical translational models mimicking human characteristics of these conditions are lacking. This study aimed to demonstrate proof of concept of the induction of stable O/MS and type 2 DM in a Göttingen minipig model and validate both of these disease-adjusted Göttingen minipig models as impaired healing models for the testing of dental implants. MATERIALS AND METHODS: Nine minipigs were split into 3 groups-control (normal diet), obese (cafeteria diet), and diabetic (cafeteria diet plus low-dosage streptozotocin)-followed by placement of dental implants. Inflammatory markers including tumor necrosis factor α, C-reactive protein, and cortisol were recorded for each study group. Removal torque was measured, and histomorphometric analysis (bone-to-implant contact and bone area fraction occupancy) was performed. RESULTS: O/MS pigs showed, on average, a 2-fold increase in plasma C-reactive protein (P < .05) and cortisol (P < .09) concentrations compared with controls; DM pigs showed, on average approximately, a 40-fold increase in plasma tumor necrosis factor α levels (P < .05) and a 2-fold increase in cortisol concentrations (P < .05) compared with controls. The impact of O/MS and DM on implants was determined. The torque to interface failure was highest in the control group (200 N-cm) and significantly lower in the O/MS (90 N-cm) and DM (60 N-cm) groups (P < .01). Bone formation around implants was significantly greater in the control group than in the O/MS and DM groups (P < .02). CONCLUSIONS: Both O/MS and DM minipigs express a human-like disease phenotype, and both presented bone-healing impairment around dental implants. Our finding of no significant difference between type 2 DM and O/MS in bone formation around implants provides evidence that further investigation of the impact of O/MS is warranted.


Assuntos
Implantes Dentários , Diabetes Mellitus Tipo 2/fisiopatologia , Síndrome Metabólica/fisiopatologia , Osseointegração/fisiologia , Animais , Biomarcadores/sangue , Modelos Animais de Doenças , Fenótipo , Projetos Piloto , Estudo de Prova de Conceito , Suínos , Porco Miniatura , Cicatrização/fisiologia
17.
J Pathol ; 245(3): 258-264, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29574902

RESUMO

Angiogenesis is a critical aspect of wound healing. We investigated the role of keratinocytes in promoting angiogenesis in mice with lineage-specific deletion of the transcription factor FOXO1. The results indicate that keratinocyte-specific deletion of Foxo1 reduces VEGFA expression in mucosal and skin wounds and leads to reduced endothelial cell proliferation, reduced angiogenesis, and impaired re-epithelialization and granulation tissue formation. In vitro FOXO1 was needed for VEGFA transcription and expression. In a porcine dermal wound-healing model that closely resembles healing in humans, local application of a FOXO1 inhibitor reduced angiogenesis. This is the first report that FOXO1 directly regulates VEGFA expression and that FOXO1 is needed for normal angiogenesis during wound healing. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Proteína Forkhead Box O1/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Gengiva/metabolismo , Mucosa Bucal/metabolismo , Neovascularização Fisiológica , Pele/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização , Ferimentos e Lesões/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Feminino , Proteína Forkhead Box O1/deficiência , Proteína Forkhead Box O1/genética , Fatores de Transcrição Forkhead/genética , Gengiva/lesões , Gengiva/patologia , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Masculino , Camundongos Knockout , Mucosa Bucal/lesões , Mucosa Bucal/patologia , Transdução de Sinais , Pele/lesões , Pele/patologia , Suínos , Porco Miniatura , Fator A de Crescimento do Endotélio Vascular/genética , Ferimentos e Lesões/genética , Ferimentos e Lesões/patologia
18.
Int J Oral Sci ; 10(1): 3, 2018 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-29483595

RESUMO

The bone remodeling process in response to orthodontic forces requires the activity of osteoclasts to allow teeth to move in the direction of the force applied. Receptor activator of nuclear factor-κB ligand (RANKL) is essential for this process although its cellular source in response to orthodontic forces has not been determined. Orthodontic tooth movement is considered to be an aseptic inflammatory process that is stimulated by leukocytes including T and B lymphocytes which are presumed to stimulate bone resorption. We determined whether periodontal ligament and bone lining cells were an essential source of RANKL by tamoxifen induced deletion of RANKL in which Cre recombinase was driven by a 3.2 kb reporter element of the Col1α1 gene in experimental mice (Col1α1.CreERTM+.RANKLf/f) and compared results with littermate controls (Col1α1.CreERTM-.RANKLf/f). By examination of Col1α1.CreERTM+.ROSA26 reporter mice we showed tissue specificity of tamoxifen induced Cre recombinase predominantly in the periodontal ligament and bone lining cells. Surprisingly we found that most of the orthodontic tooth movement and formation of osteoclasts was blocked in the experimental mice, which also had a reduced periodontal ligament space. Thus, we demonstrate for the first time that RANKL produced by periodontal ligament and bone lining cells provide the major driving force for tooth movement and osteoclastogenesis in response to orthodontic forces.


Assuntos
Remodelação Óssea/fisiologia , Osteoclastos/fisiologia , Ligamento Periodontal/metabolismo , Ligante RANK/metabolismo , Técnicas de Movimentação Dentária , Animais , Camundongos , Camundongos Transgênicos , Tamoxifeno/farmacologia
19.
J Clin Periodontol ; 45(3): 285-292, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29220094

RESUMO

AIM: Periodontitis results from bacteria-induced inflammation. A key cytokine, RANKL, is produced by a number of cell types. The cellular source of RANKL critical for periodontitis has not been established. METHODS: We induced periodontal bone loss by oral inoculation of Porphyromonas gingivalis and Fusobacterium nucleatum in both normoglycaemic and streptozotocin-induced type 1 diabetic mice. Experimental transgenic mice had osteocyte-specific deletion of floxed receptor activator of nuclear factor kappa-B ligand (RANKL) mediated by DMP-1-driven Cre recombinase. Outcomes were assessed by micro-CT, histomorphometric analysis, immunofluorescent analysis of RANKL and tartrate-resistant acid phosphatase staining for osteoclasts and osteoclast activity. RESULTS: Oral infection stimulated RANKL expression in osteocytes of wild-type mice, which was increased by diabetes and blocked in transgenic mice. Infected wild-type mice had significant bone loss and increased osteoclast numbers and activity, which were further enhanced by diabetes. No bone loss or increase in osteoclastogenesis or activity was detected in transgenic mice with RANKL deletion in osteocytes that were normoglycaemic or diabetic. CONCLUSIONS: This study demonstrates for the first time the essential role of osteocytes in bacteria-induced periodontal bone loss and in diabetes-enhanced periodontitis.


Assuntos
Perda do Osso Alveolar/microbiologia , Infecções por Bacteroidaceae/complicações , Diabetes Mellitus Experimental/complicações , Proteínas da Matriz Extracelular/genética , Osteócitos/metabolismo , Periodontite/metabolismo , Porphyromonas gingivalis , Ligante RANK/metabolismo , Animais , Proteínas da Matriz Extracelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Periodontite/complicações , Periodontite/microbiologia , Ligante RANK/deficiência
20.
Sci Rep ; 7(1): 10565, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28874756

RESUMO

Keratinocyte migration is a key aspect of re-epithelialization during wound healing. Matrix metalloproteinase 9 (MMP9) contributes to this process and deficiencies in the MMP9 lead to impaired healing. Inappropriate expression of MMP9 also contributes to impaired re-epithelialization. Previously we demonstrated that FOXO1 was activated in wound healing but to higher levels in diabetic wounds. To address mechanisms of impaired re-epithelialization we examined MMP9 expression in vivo in full thickness dermal scalp wounds created in experimental K14.Cre + .Foxo1 L/L mice with lineage-specific Cre recombinase deletion of floxed FOXO1 and compared the results to control littermates. MMP9 was induced during wound healing but at a significantly higher level in diabetic compared to normal wounds. FOXO1 deletion substantially blocked this increase. By chromatin immunoprecipitation FOXO1 was shown to bind to the MMP9 promoter, FOXO1 overexpression increased MMP9 transcriptional activity and increased MMP9 expression stimulated by high glucose was blocked by FOXO1 deletion or FOXO1 knockdown. We also show for the first time that high glucose impairs keratinocyte migration by inducing high levels of MMP9 expression and establish that it involves FOXO1. Thus, FOXO1 drives high levels of MMP9 expression in diabetic wound healing, which represents a novel mechanism for impaired re-epithelization in diabetic wounds.


Assuntos
Complicações do Diabetes/metabolismo , Proteína Forkhead Box O1/genética , Queratinócitos/metabolismo , Metaloproteinase 9 da Matriz/genética , Reepitelização , Dermatopatias/metabolismo , Animais , Células Cultivadas , Proteína Forkhead Box O1/metabolismo , Deleção de Genes , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA