Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
JCI Insight ; 8(17)2023 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-37681413

RESUMO

Osteoarthritis (OA) is the most common joint disorder, and disease-modifying OA drugs (DMOADs) represent a major need in OA management. Krüppel-like factor 4 (KLF4) is a central transcription factor upregulating regenerative and protective functions in joint tissues. This study was aimed to identify small molecules activating KLF4 expression and to determine functions and mechanisms of the hit compounds. High-throughput screening (HTS) with 11,948 clinical-stage compounds was performed using a reporter cell line detecting endogenous KLF4 activation. Eighteen compounds were identified through the HTS and confirmed in a secondary screen. After testing in SW1353 chondrosarcoma cells and human chondrocytes, mocetinostat - a class I selective histone deacetylase (HDAC) inhibitor - had the best profile of biological activities. Mocetinostat upregulated cartilage signature genes in human chondrocytes, meniscal cells, and BM-derived mesenchymal stem cells, and it downregulated hypertrophic, inflammatory, and catabolic genes in those cells and synoviocytes. I.p. administration of mocetinostat into mice reduced severity of OA-associated changes and improved pain behaviors. Global gene expression and proteomics analyses revealed that regenerative and protective effects of mocetinostat were dependent on peroxisome proliferator-activated receptor γ coactivator 1-α. These findings show therapeutic and protective activities of mocetinostat against OA, qualifying it as a candidate to be used as a DMOAD.


Assuntos
Neoplasias Ósseas , Osteoartrite , Humanos , Animais , Camundongos , Fator 4 Semelhante a Kruppel , Osteoartrite/tratamento farmacológico , Inflamação , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico
2.
Ann Rheum Dis ; 82(5): 710-718, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36627169

RESUMO

OBJECTIVES: CHRFAM7A is a uniquely human fusion gene that functions as a dominant negative regulator of alpha 7 acetylcholine nicotinic receptor (α7nAChR) in vitro. This study determined the impact of CHRFAM7A on α7nAChR agonist responses, osteoarthritis (OA) severity and pain behaviours and investigated mechanisms. METHODS: Transgenic CHRFAM7A (TgCHRFAM7A) mice were used to determine the impact of CHRFAM7A on knee OA histology, pain severity in OA and other pain models, response to nAchR agonist and IL-1ß. Mouse and human cells were used for mechanistic studies. RESULTS: Transgenic (Tg) TgCHRFAM7A mice developed more severe structural damage and increased mechanical allodynia than wild type (WT) mice in the destabilisation of medial meniscus model of OA. This was associated with a decreased suppression of inflammation by α7nAchR agonist. TgCHRFAM7A mice displayed a higher basal sensitivity to pain stimuli and increased pain behaviour in the monoiodoacetate and formalin models. Dorsal root ganglia of TgCHRFAM7A mice showed increased macrophage infiltration and expression of the chemokine fractalkine and also had a compromised antinociceptive response to the α7nAchR agonist nicotine. Both native CHRNA7 and CHRFAM7A subunits were expressed in human joint tissues and the CHRFAM7A/CHRNA7 ratio was increased in OA cartilage. Human chondrocytes with two copies of CHRFAM7A had reduced anti-inflammatory responses to nicotine. CONCLUSION: CHRFAM7A is an aggravating factor for OA-associated inflammation and tissue damage and a novel genetic risk factor and therapeutic target for pain.


Assuntos
Osteoartrite do Joelho , Receptor Nicotínico de Acetilcolina alfa7 , Animais , Humanos , Camundongos , Receptor Nicotínico de Acetilcolina alfa7/genética , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Inflamação/genética , Camundongos Transgênicos , Nicotina , Osteoartrite do Joelho/genética , Dor/genética
3.
Ann Rheum Dis ; 2022 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-35534137

RESUMO

OBJECTIVES: Analysing expression patterns of Krüppel-like factor (KLF) transcription factors in normal and osteoarthritis (OA) human cartilage, and determining functions and mechanisms of KLF4 and KLF2 in joint homoeostasis and OA pathogenesis. METHODS: Experimental approaches included human joint tissues cells, transgenic mice and mouse OA model with viral KLF4 gene delivery to demonstrate therapeutic benefit in structure and pain improvement. Mechanistic studies applied global gene expression analysis and chromatin immunoprecipitation sequencing (ChIP-seq). RESULTS: Several KLF genes were significantly decreased in OA cartilage. Among them, KLF4 and KLF2 were strong inducers of cartilage collagen genes and Proteoglycan-4. Cartilage-specific deletion of Klf2 in mature mice aggravated severity of experimental OA. Transduction of human chondrocytes with Adenovirus (Ad) expressing KLF4 or KLF2 enhanced expression of major cartilage extracellular matrix (ECM) genes and SRY-box transcription factor-9, and suppressed mediators of inflammation and ECM-degrading enzymes. Ad-KLF4 and Ad-KLF2 enhanced similar protective functions in meniscus cells and synoviocytes, and promoted chondrocytic differentiation of human mesenchymal stem cells. Viral KLF4 delivery into mouse knees reduced severity of OA-associated changes in cartilage, meniscus and synovium, and improved pain behaviours. ChIP-seq analysis suggested that KLF4 directly bound cartilage signature genes. Ras-related protein-1 signalling was the most enriched pathway in KLF4-transduced cells, and its signalling axis was involved in upregulating cartilage ECM genes by KLF4 and KLF2. CONCLUSIONS: KLF4 and KLF2 may be central transcription factors that increase protective and regenerative functions in joint tissue cells, suggesting that KLF gene transfer or molecules upregulating KLFs are therapeutic candidates for OA.

4.
Quintessence Int ; 53(7): 608-614, 2022 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-35274517

RESUMO

This case report presents a novel approach for minimally invasive fully guided apicoectomy of the palatal root of a maxillary first molar using a custom-made 3D-printed template. To date, the development of diagnostic radiographic tools such as high-resolution CBCT devices, as well as of CAD planning software and CAM technologies, like 3D printing, allow for increased application in endodontics. The patient (a 38-year-old woman) suffered from pain on the right side of the face since 4 weeks and was diagnosed with chronic apical periodontitis of the palatal root of the maxillary right first molar. The root treatment of this tooth was followed up recently and the buccal roots showed no pathologic findings. A guided apicoectomy with access from the palate was chosen as elective therapy. 3D radiographic and intraoral surface datasets were imported into an implant planning software and superimposed, and minimally invasive access to the palatal root apex was planned. Subsequently, a tooth-supported drilling template was designed and created by additive manufacturing. A flapless approach was adapted using a punch-drill and the access to the root apex as well as the apical resection were performed with a trephine drill. The connective tissue punch was finally replaced and sutured. No postoperative complication was reported and a complete remission of symptoms was reported after 2 weeks. The follow-up after 21 months showed clinically stable wound conditions and radiologically a slight reossification in the area of the palatal root tip. The presented technique may lead to novel minimally invasive approaches for the preservation of infected maxillary molars.


Assuntos
Apicectomia , Endodontia , Adulto , Tomografia Computadorizada de Feixe Cônico , Feminino , Humanos , Dente Molar/diagnóstico por imagem , Dente Molar/cirurgia , Raiz Dentária/diagnóstico por imagem , Raiz Dentária/cirurgia
5.
Swiss Dent J ; 132(2): 113-116, 2022 Feb 07.
Artigo em Alemão | MEDLINE | ID: mdl-35106988

RESUMO

This case report illustrates a new application of autologous dentin for defect augmentation. After cystectomy of a follicular cyst in the maxilla, autologous particulated dentin from the wisdom tooth is used for augmentation due to periodontal compromise of neighbouring teeth. The combination of simultaneous wisdom tooth removal and augmentation is a convenient option in this case. Dentin augmentation is a cost-effective and minimally invasive autologous graft technique. The decision which treatment method for a jaw cyst is the most effective for the patient is a well discussed topic. Factors as expansion, type of cyst, localisation, duration of treatment, and predictability have to be calculated when choosing the treatment strategy. Most cysts heal without grafting, however, periodontal situation, sinus association and predictability in aesthetics led to the decision of grafting necessity in this presented case.


Assuntos
Aumento do Rebordo Alveolar , Cistectomia , Processo Alveolar , Transplante Ósseo , Dentina , Humanos
6.
J Craniomaxillofac Surg ; 49(7): 556-561, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33726950

RESUMO

The aim of this study was to investigate the accuracy of a previously described technique for guided biopsy of osseous pathologies of the jawbone in a clinical setting. The data sets of patients who had undergone guided biopsy procedures were retrospectively examined for accuracy. Digital planning of the biopsies and manufacturing of the tooth-supported drilling template were performed with superimposed cone beam computed tomography and intraoral scans using implant planning software. After a trephine biopsy was taken using the template, the postoperative low-dose cone beam computed tomography was analyzed for accuracy using the planning software with the corresponding (digitally-planned) biopsy cylinder. The mean angular deviation was 4.35 ± 2.5°. The mean depth deviation was -1.40 ± 1.41 mm. Guided biopsy seems to be an alternative to a conventional approach for minimally invasive and highly accurate jawbone biopsy.


Assuntos
Implantes Dentários , Cirurgia Assistida por Computador , Biópsia , Desenho Assistido por Computador , Tomografia Computadorizada de Feixe Cônico , Implantação Dentária Endóssea , Humanos , Imageamento Tridimensional , Planejamento de Assistência ao Paciente , Estudos Retrospectivos
7.
Sci Transl Med ; 12(567)2020 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-33115953

RESUMO

Meniscus tears are common knee injuries and a major osteoarthritis (OA) risk factor. Knowledge gaps that limit the development of therapies for meniscus injury and degeneration concern transcription factors that control the meniscus cell phenotype. Analysis of RNA sequencing data from 37 human tissues in the Genotype-Tissue Expression database and RNA sequencing data from meniscus and articular cartilage showed that transcription factor Mohawk (MKX) is highly enriched in meniscus. In human meniscus cells, MKX regulates the expression of meniscus marker genes, OA-related genes, and other transcription factors, including Scleraxis (SCX), SRY Box 5 (SOX5), and Runt domain-related transcription factor 2 (RUNX2). In mesenchymal stem cells (MSCs), the combination of adenoviral MKX (Ad-MKX) and transforming growth factor-ß3 (TGF-ß3) induced a meniscus cell phenotype. When Ad-MKX-transduced MSCs were seeded on TGF-ß3-conjugated decellularized meniscus scaffold (DMS) and inserted into experimental tears in meniscus explants, they increased glycosaminoglycan content, extracellular matrix interconnectivity, cell infiltration into the DMS, and improved biomechanical properties. Ad-MKX injection into mouse knee joints with experimental OA induced by surgical destabilization of the meniscus suppressed meniscus and cartilage damage, reducing OA severity. Ad-MKX injection into human OA meniscus tissue explants corrected pathogenic gene expression. These results identify MKX as a previously unidentified key transcription factor that regulates the meniscus cell phenotype. The combination of Ad-MKX with TGF-ß3 is effective for differentiation of MSCs to a meniscus cell phenotype and useful for meniscus repair. MKX is a promising therapeutic target for meniscus tissue engineering, repair, and prevention of OA.


Assuntos
Cartilagem Articular , Proteínas de Homeodomínio/metabolismo , Menisco , Células-Tronco Mesenquimais , Osteoartrite , Animais , Proteínas de Homeodomínio/genética , Camundongos , Fenótipo , Fatores de Transcrição
8.
Arthritis Rheumatol ; 72(9): 1514-1523, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32281255

RESUMO

OBJECTIVE: Osteoarthritis (OA) is the most common age-related joint disease. With aging and in OA, the expression of FoxO transcription factors is reduced, diminishing their chondroprotective actions. In order to elucidate the molecular mechanisms by which FoxO1 protects chondrocytes, we sought to identify the genome-wide occupancy profile of FoxO1. METHODS: We performed FoxO1 chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-Seq) on human primary chondrocytes. ChIP-Seq data were integrated with RNA sequencing (RNA-Seq) data sets. Bioinformatics results were confirmed in primary chondrocytes that were treated with a FoxO1 inhibitor. RESULTS: Analysis of FoxO1 ChIP-Seq on human primary chondrocytes showed that pathways implicated in OA pathogenesis are mainly regulated by FoxO1 binding to tissue-specific enhancers with suboptimal binding sites (20% of the peaks), while more ubiquitous FoxO1 pathways are regulated at the promoter level through interaction with its canonical binding motif (7% of the peaks). Integrating FoxO1 occupancy data with RNA-Seq data comparing OA and healthy human cartilage revealed 428 putative FoxO1 target genes that are dysregulated in OA. Pathway analysis showed enrichment for genes belonging to the senescence pathway (logP = -6.73), extracellular matrix (ECM) pathway (logP = -12.97), and circadian clock pathway (logP = -6.30), which suggests that FoxO1 dysregulation plays an important role in their abnormal expression in OA. Using an inhibitor of FoxO1, we confirmed that FoxO1 regulates these pathways in cultured human chondrocytes. CONCLUSION: FoxO1 regulates ubiquitous and cartilage-specific genes in chondrocytes by using different mechanisms. The FoxO1 transcriptional network is a key player in regulating homeostasis, ECM, and circadian clock genes and plays an important role in the abnormal expression of these pathways observed in OA pathogenesis.


Assuntos
Condrócitos/metabolismo , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/genética , Matriz Extracelular/genética , Proteína Forkhead Box O1/genética , Osteoartrite/genética , Idoso , Cartilagem Articular/citologia , Cartilagem Articular/metabolismo , Senescência Celular/genética , Condrócitos/efeitos dos fármacos , Imunoprecipitação da Cromatina , Sequenciamento de Cromatina por Imunoprecipitação , Relógios Circadianos/genética , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Matriz Extracelular/metabolismo , Feminino , Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica , Humanos , Masculino , Osteoartrite/metabolismo , Quinolonas/farmacologia , RNA-Seq
9.
Proc Natl Acad Sci U S A ; 117(6): 3135-3143, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-31980519

RESUMO

The objective of this study was to examine FoxO expression and FoxO function in meniscus. In menisci from human knee joints with osteoarthritis (OA), FoxO1 and 3 expression were significantly reduced compared with normal menisci from young and old normal donors. The expression of FoxO1 and 3 was also significantly reduced in mouse menisci during aging and OA induced by surgical meniscus destabilization or mechanical overuse. Deletion of FoxO1 and combined FoxO1, 3, and 4 deletions induced abnormal postnatal meniscus development in mice and these mutant mice spontaneously displayed meniscus pathology at 6 mo. Mice with Col2Cre-mediated deletion of FoxO3 or FoxO4 had normal meniscus development but had more severe aging-related damage. In mature AcanCreERT2 mice, the deletion of FoxO1, 3, and 4 aggravated meniscus lesions in all experimental OA models. FoxO deletion suppressed autophagy and antioxidant defense genes and altered several meniscus-specific genes. Expression of these genes was modulated by adenoviral FoxO1 in cultured human meniscus cells. These results suggest that FoxO1 plays a key role in meniscus development and maturation, and both FoxO1 and 3 support homeostasis and protect against meniscus damage in response to mechanical overuse and during aging and OA.


Assuntos
Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Articulação do Joelho/metabolismo , Menisco/metabolismo , Osteoartrite/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Modelos Animais de Doenças , Feminino , Proteína Forkhead Box O1/análise , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O3/análise , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Humanos , Masculino , Menisco/crescimento & desenvolvimento , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Adulto Jovem
10.
Int J Comput Dent ; 22(4): 363-369, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31840144

RESUMO

AIM: Modern microsurgical techniques have increased the success rate of apicoectomy relative to that of traditional approaches. This case report introduces a novel workaround for guided apicoectomy using a patient-specific computer-aided design/computer-aided manufacturing (CAD/CAM) three-dimensional (3D)-printed template. MATERIALS AND METHODS: Apicoectomy was performed on the mesial root of tooth 36 using template-guided trephine drilling, followed by retrograde filling with mineral trioxide aggregate (MTA). Initially, a cone beam computed tomography (CBCT) scan and an intraoral surface scan were imported into the planning software. After superimposition, virtual planning was performed to determine the exact localization for root resection. Subsequently, a tooth-supported drilling template was designed and 3D printed. Endodontic microsurgical approaches, including root-end cavity preparation and root-end filling, completed the surgical treatment. RESULT: The apical resection was easily feasible. There were no postoperative complications. Radiological assessment after a 6-month period showed signs of reossification. CONCLUSION: Guided apicoectomy allowed precise root resection, suggesting that this technique may be advantageous in complex anatomical situations.


Assuntos
Apicectomia , Dente , Desenho Assistido por Computador , Tomografia Computadorizada de Feixe Cônico , Humanos , Impressão Tridimensional
11.
J Biol Chem ; 294(46): 17555-17569, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31601652

RESUMO

The forkhead box O (FOXO) proteins are transcription factors involved in the differentiation of many cell types. Type II collagen (Col2) Cre-Foxo1-knockout and Col2-Cre-Foxo1,3,4 triple-knockout mice exhibit growth plate malformation. Moreover, recent studies have reported that in some cells, the expressions and activities of FOXOs are promoted by transforming growth factor ß1 (TGFß1), a growth factor playing a key role in chondrogenic differentiation. Here, using a murine chondrogenic cell line (ATDC5), mouse embryos, and human mesenchymal stem cells, we report the mechanisms by which FOXOs affect chondrogenic differentiation. FOXO1 expression increased along with chondrogenic differentiation, and FOXO1 inhibition suppressed chondrogenic differentiation. TGFß1/SMAD signaling promoted expression and activity of FOXO1. In ATDC5, FOXO1 knockdown suppressed expression of sex-determining region Y box 9 (Sox9), a master regulator of chondrogenic differentiation, resulting in decreased collagen type II α1 (Col2a1) and aggrecan (Acan) expression after TGFß1 treatment. On the other hand, chemical FOXO1 inhibition suppressed Col2a1 and Acan expression without suppressing Sox9 To investigate the effects of FOXO1 on chondrogenic differentiation independently of SOX9, we examined FOXO1's effects on the cell cycle. FOXO1 inhibition suppressed expression of p21 and cell-cycle arrest in G0/G1 phase. Conversely, FOXO1 overexpression promoted expression of p21 and cell-cycle arrest. FOXO1 inhibition suppressed expression of nascent p21 RNA by TGFß1, and FOXO1 bound the p21 promoter. p21 inhibition suppressed expression of Col2a1 and Acan during chondrogenic differentiation. These results suggest that FOXO1 is necessary for not only SOX9 expression, but also cell-cycle arrest during chondrogenic differentiation via TGFß1 signaling.


Assuntos
Condrogênese/genética , Proteína Forkhead Box O1/genética , Fatores de Transcrição SOX9/genética , Fator de Crescimento Transformador beta1/genética , Agrecanas/genética , Animais , Pontos de Checagem do Ciclo Celular/genética , Diferenciação Celular/genética , Colágeno Tipo II/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Proteína Forkhead Box O1/antagonistas & inibidores , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Proteínas Smad/genética , Fator de Crescimento Transformador beta1/farmacologia
12.
JCI Insight ; 4(19)2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31465300

RESUMO

Excessive vascular remodeling is characteristic of hemophilic arthropathy (HA) and may contribute to joint bleeding and the progression of HA. Mechanisms for pathological vascular remodeling after hemophilic joint bleeding are unknown. In hemophilia, activation of thrombin-activatable fibrinolysis inhibitor (TAFI) is impaired, which contributes to joint bleeding and may also underlie the aberrant vascular remodeling. Here, hemophilia A (factor VIII-deficient; FVIII-deficient) mice or TAFI-deficient mice with transient (antibody-induced) hemophilia A were used to determine the role of FVIII and TAFI in vascular remodeling after joint bleeding. Excessive vascular remodeling and vessel enlargement persisted in FVIII-deficient and TAFI-deficient mice, but not in transient hemophilia WT mice, after similar joint bleeding. TAFI-overexpression in FVIII-deficient mice prevented abnormal vessel enlargement and vascular leakage. Age-related vascular changes were observed with FVIII or TAFI deficiency and correlated positively with bleeding severity after injury, supporting increased vascularity as a major contributor to joint bleeding. Antibody-mediated inhibition of uPA also prevented abnormal vascular remodeling, suggesting that TAFI's protective effects include inhibition of uPA-mediated plasminogen activation. In conclusion, the functional TAFI deficiency in hemophilia drives maladaptive vascular remodeling in the joints after bleeding. These mechanistic insights allow targeted development of potentially new strategies to normalize vascularity and control rebleeding in HA.


Assuntos
Carboxipeptidase B2/genética , Carboxipeptidase B2/metabolismo , Fator VIII/genética , Hemartrose/complicações , Hemofilia A/complicações , Hemofilia A/genética , Remodelação Vascular/fisiologia , Animais , Modelos Animais de Doenças , Fator VIII/metabolismo , Feminino , Predisposição Genética para Doença/genética , Hemartrose/patologia , Hemofilia A/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transcriptoma
13.
EBioMedicine ; 45: 588-605, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31285188

RESUMO

BACKGROUND: Ageing-related failure of homeostasis mechanisms contributes to articular cartilage degeneration and osteoarthritis (OA), for which disease-modifying treatments are not available. Our objective was to identify molecules to prevent OA by regulating chondrocyte senescence and autophagy. METHODS: Human chondrocytes with IL-6 induced senescence and autophagy suppression and SA-ß-gal as a reporter of senescence and LC3 as reporter of autophagic flux were used to screen the Prestwick Chemical Library of approved drugs. Preclinical cellular, tissue and blood from OA and blood from OA and ageing models were used to test the efficacy and relevance of activating PPARα related to cartilage degeneration. FINDINGS: Senotherapeutic molecules with pro-autophagic activity were identified. Fenofibrate (FN), a PPARα agonist used for dyslipidaemias in humans, reduced the number of senescent cells via apoptosis, increased autophagic flux, and protected against cartilage degradation. FN reduced both senescence and inflammation and increased autophagy in both ageing human and OA chondrocytes whereas PPARα knockdown conferred the opposite effect. Moreover, PPARα expression was reduced through both ageing and OA in mice and also in blood and cartilage from knees of OA patients. Remarkably, in a retrospective study, fibrate treatment improved OA clinical conditions in human patients from the Osteoarthritis Initiative (OAI) Cohort. INTERPRETATION: These results demonstrate that FDA-approved fibrate drugs targeting lipid metabolism protect against cartilage degeneration seen with ageing and OA. Thus, these drugs could have immediate clinically utility for age-related cartilage degeneration and OA treatment. FUND: This study was supported by Instituto de Salud Carlos III- Ministerio de Ciencia, Innovación y Universidades, Spain, Plan Estatal 2013-2016 and Fondo Europeo de Desarrollo Regional (FEDER), "Una manera de hacer Europa", PI14/01324 and PI17/02059, by Innopharma Pharmacogenomics platform applied to the validation of targets and discovery of drugs candidates to preclinical phases, Ministerio de Economía y Competitividad, by grants of the National Instiutes of Health to PDR (P01 AG043376 and U19 AG056278). We thank FOREUM Foundation for Research in Rheumatology for their support.


Assuntos
Envelhecimento/efeitos dos fármacos , Doenças das Cartilagens/tratamento farmacológico , Fenofibrato/farmacologia , Osteoartrite/tratamento farmacológico , PPAR alfa/genética , Envelhecimento/genética , Animais , Apoptose , Autofagia/efeitos dos fármacos , Doenças das Cartilagens/genética , Doenças das Cartilagens/patologia , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Humanos , Interleucina-6/genética , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Osteoartrite/genética , Osteoartrite/patologia , PPAR alfa/agonistas
14.
Nat Commun ; 10(1): 2429, 2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31160553

RESUMO

The WW domain-containing protein 2 (Wwp2) gene, the host gene of miR-140, codes for the Wwp2 protein, which is an HECT-type E3 ubiquitin ligases abundantly expressed in articular cartilage. However, its function remains unclear. Here, we show that mice lacking Wwp2 and mice in which the Wwp2 E3 enzyme is inactivated (Wwp2-C838A) exhibit aggravated spontaneous and surgically induced osteoarthritis (OA). Consistent with this phenotype, WWP2 expression level is downregulated in human OA cartilage. We also identify Runx2 as a Wwp2 substrate and Adamts5 as a target gene, as similar as miR-140. Analysis of Wwp2-C838A mice shows that loss of Wwp2 E3 ligase activity results in upregulation of Runx2-Adamts5 signaling in articular cartilage. Furthermore, in vitro transcribed Wwp2 mRNA injection into mouse joints reduces the severity of experimental OA. We propose that Wwp2 has a role in protecting cartilage from OA by suppressing Runx2-induced Adamts5 via Runx2 poly-ubiquitination and degradation.


Assuntos
Proteína ADAMTS5/metabolismo , Cartilagem Articular/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Osteoartrite/genética , Ubiquitina-Proteína Ligases/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Artrite Experimental/genética , Artrite Experimental/metabolismo , Cartilagem Articular/diagnóstico por imagem , Modelos Animais de Doenças , Humanos , Articulação do Joelho/diagnóstico por imagem , Meniscos Tibiais/cirurgia , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Osteoartrite/metabolismo , RNA Mensageiro/farmacologia , Transdução de Sinais , Crânio/diagnóstico por imagem , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Microtomografia por Raio-X , Adulto Jovem
15.
J Autoimmun ; 94: 90-98, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30077426

RESUMO

T-cell resilience is critical to the immune pathogenesis of human autoimmune arthritis. Autophagy is essential for memory T cell generation and associated with pathogenesis in rheumatoid arthritis (RA). Our aim here was to delineate the role and molecular mechanism of autophagy in resilience and persistence of pathogenic T cells from autoimmune arthritis. We demonstrated "Autophagic memory" as elevated autophagy levels in CD4+ memory T cells compared to CD4+ naive T cells and in Jurkat Human T cell line trained with starvation stress. We then showed increased levels of autophagy in pathogenic CD4+ T cells subsets from autoimmune arthritis patients. Using RNA-sequencing, transcription factor gene regulatory network and methylation analyses we identified MYC as a key regulator of autophagic memory. We validated MYC levels using qPCR and further demonstrated that inhibiting MYC increased autophagy. The present study proposes the novel concept of autophagic memory and suggests that autophagic memory confers metabolic advantage to pathogenic T cells from arthritis and supports its resilience and long term survival. Particularly, suppression of MYC imparted the heightened autophagy levels in pathogenic T cells. These studies have a direct translational valency as they identify autophagy and its metabolic controllers as a novel therapeutic target.


Assuntos
Artrite Juvenil/imunologia , Artrite Reumatoide/imunologia , Autofagia/imunologia , Redes Reguladoras de Genes/imunologia , Memória Imunológica , Proteínas Proto-Oncogênicas c-myc/genética , Adolescente , Adulto , Animais , Artrite Juvenil/genética , Artrite Juvenil/patologia , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Autofagia/genética , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Estudos de Casos e Controles , Metilação de DNA , Feminino , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Células Jurkat , Masculino , Camundongos , Camundongos Endogâmicos DBA , Oxidiazóis/farmacologia , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/imunologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia , Fatores de Transcrição/classificação , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia
16.
Aging Cell ; 17(5): e12800, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29963746

RESUMO

Intervertebral disk (IVD) degeneration is a prevalent age-associated musculoskeletal disorder and a major cause of chronic low back pain. Aging is the main risk factor for the disease, but the molecular mechanisms regulating IVD homeostasis during aging are unknown. The aim of this study was to investigate the function of FOXO, a family of transcription factors linked to aging and longevity, in IVD aging and age-related degeneration. Conditional deletion of all FOXO isoforms (FOXO1, 3, and 4) in IVD using the Col2a1Cre and AcanCreER mouse resulted in spontaneous development of IVD degeneration that was driven by severe cell loss in the nucleus pulposus (NP) and cartilaginous endplates (EP). Conditional deletion of individual FOXO in mature mice showed that FOXO1 and FOXO3 are the dominant isoforms and have redundant functions in promoting IVD homeostasis. Gene expression analyses indicated impaired autophagy and reduced antioxidant defenses in the NP of FOXO-deficient IVD. In primary human NP cells, FOXO directly regulated autophagy and adaptation to hypoxia and promoted resistance to oxidative and inflammatory stress. Our findings demonstrate that FOXO are critical regulators of IVD homeostasis during aging and suggest that maintaining or restoring FOXO expression can be a therapeutic strategy to promote healthy IVD aging and delay the onset of IVD degeneration.


Assuntos
Envelhecimento/metabolismo , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O3/metabolismo , Disco Intervertebral/metabolismo , Animais , Células Cultivadas , Proteína Forkhead Box O1/deficiência , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O3/deficiência , Proteína Forkhead Box O3/genética , Homeostase , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
17.
Sci Rep ; 8(1): 9601, 2018 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-29942000

RESUMO

Although various surgical procedures have been developed for chronic rotator cuff tear repair, the re-tear rate remains high with severe fat infiltration. However, little is known about the molecular regulation of this process. Mesenchymal stem cells (MSCs) in the intra-muscular space are origin of ectopic fat cells in skeletal muscle. We have previously shown that high-mobility group box 2 (HMGB2), which is a nuclear protein commonly associated with mesenchymal differentiation, is involved in the early articular cartilage degeneration. In this study, we addressed the role of HMGB2 in adipogenesis of MSCs and fat infiltration into skeletal muscles. HMGB2 was highly expressed in undifferentiated MSCs and co-localized with platelet-derived growth factor receptor α (PDGFRA) known as an MSC-specific marker, while their expressions were decreased during adipocytic differentiation. Under the deficiency of HMGB2, the expressions of adipogenesis-related molecules were reduced, and adipogenic differentiation is substantially impaired in MSCs. Moreover, HMGB2+ cells were generated in the muscle belly of rat supraspinatus muscles after rotator cuff transection, and some of these cells expressed PDGFRA in intra-muscular spaces. Thus, our findings suggest that the enhance expression of HMGB2 induces the adipogenesis of MSCs and the fat infiltration into skeletal muscles through the cascade of HMGB2-PDGFRA.


Assuntos
Adipogenia , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Proteína HMGB2/metabolismo , Músculo Esquelético/citologia , Animais , Diferenciação Celular , Regulação da Expressão Gênica , Proteína HMGB2/genética , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Ratos , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo
18.
Hum Cell ; 31(1): 1-9, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28916968

RESUMO

The high-mobility group box (HMGB) family includes four members: HMGB1, 2, 3 and 4. HMGB proteins have two functions. In the nucleus, HMGB proteins bind to DNA in a DNA structure-dependent but nucleotide sequence-independent manner to function in chromatin remodeling. Extracellularly, HMGB proteins function as alarmins, which are endogenous molecules released upon tissue damage to activate the immune system. HMGB1 acts as a late mediator of inflammation and contributes to prolonged and sustained systemic inflammation in subjects with rheumatoid arthritis. By contrast, Hmgb2 -/- mice represent a relevant model of aging-related osteoarthritis (OA), which is associated with the suppression of HMGB2 expression in cartilage. Hmgb2 mutant mice not only develop early-onset OA but also exhibit a specific phenotype in the superficial zone (SZ) of articular cartilage. Given the similar expression and activation patterns of HMGB2 and ß-catenin in articular cartilage, the loss of these pathways in the SZ of articular cartilage may lead to altered gene expression, cell death and OA-like pathogenesis. Moreover, HMGB2 regulates chondrocyte hypertrophy by mediating Runt-related transcription factor 2 expression and Wnt signaling. Therefore, one possible mechanism explaining the modulation of lymphoid enhancer binding factor 1 (LEF1)-dependent transactivation by HMGB2 is that a differential interaction between HMGB2 and nuclear factors affects the transcription of genes containing LEF1-responsive elements. The multiple functions of HMGB proteins reveal the complex roles of these proteins as innate and endogenous regulators of inflammation in joints and their cooperative roles in cartilage hypertrophy as well as in the maintenance of joint tissue homeostasis.


Assuntos
Artrite Reumatoide/genética , Proteína HMGB1/fisiologia , Proteína HMGB2/fisiologia , Osteoartrite/genética , Alarminas , Animais , Artrite Reumatoide/imunologia , Cartilagem Articular/metabolismo , Condrócitos/patologia , Montagem e Desmontagem da Cromatina/genética , Subunidade alfa 1 de Fator de Ligação ao Core , DNA/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Proteína HMGB1/metabolismo , Proteína HMGB2/metabolismo , Humanos , Hidroxietilrutosídeo , Hipertrofia/genética , Mediadores da Inflamação , Fator 1 de Ligação ao Facilitador Linfoide , Camundongos , Osteoartrite/imunologia , Ligação Proteica , Via de Sinalização Wnt
19.
J Orthop Res ; 35(12): 2682-2691, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28430387

RESUMO

Aging is a main risk factor for intervertebral disc (IVD) degeneration, the main cause of low back pain. FOXO transcription factors are important regulators of tissue homeostasis and longevity. Here, we determined the expression pattern of FOXO in healthy and degenerated human IVD and the associations with IVD degeneration during mouse aging. FOXO expression was assessed by immunohistochemistry in normal and degenerated human IVD samples and in cervical and lumbar IVD from 6-, 12-, 24-, and 36-month-old C57BL/6J mice. Mouse spines were graded for key histological features of disc degeneration in all the time points and expression of two key FOXO downstream targets, sestrin 3 (SESN3) and superoxide dismutase (SOD2), was assessed by immunohistochemistry. Histological analysis revealed that FOXO proteins are expressed in all compartments of human and mouse IVD. Expression of FOXO1 and FOXO3, but not FOXO4, was significantly deceased in human degenerated discs. In mice, degenerative changes in the lumbar spine were seen at 24 and 36 months of age whereas cervical IVD showed increased histopathological scores at 36 months. FOXO expression was significantly reduced in lumbar IVD at 12-, 24-, and 36-month-old mice and in cervical IVD at 36-month-old mice when compared with the 6-month-old group. The reduction of FOXO expression in lumbar IVD was concomitant with a decrease in the expression of SESN3 and SOD2. These findings suggest that reduced FOXO expression occurs in lumbar IVD during aging and precedes the major histopathological changes associated with lumbar IVD degeneration. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2682-2691, 2017.


Assuntos
Envelhecimento/metabolismo , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O3/metabolismo , Degeneração do Disco Intervertebral/metabolismo , Disco Intervertebral/metabolismo , Idoso , Animais , Humanos , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade
20.
Sci Rep ; 7: 42990, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28220902

RESUMO

The objective was to investigate the levels of TWIST1 in normal and OA cartilage and examine its role in regulating gene expression in chondrocytes. Human cartilage tissues and chondrocytes were obtained at autopsy from normal knee joints and from OA-affected joints at the time of total knee arthroplasty. TWIST1 expression was increased in human OA knee cartilage compared to normal knee cartilage. TWIST1 induced matrix metalloproteinase 3 (MMP3) expression without direct binding to MMP3 promoter and increased the 5-hydroxymethylcytosine (5hmC) level at the MMP3 promoter. The effect of TWIST1 on expression of TET family (TET1, 2 and 3) was measured in stable TWIST1 transfected TC28 cells, and TET1 expression was up-regulated. TWIST1 dependent upregulation of Mmp3 expression was suppressed in Tet triple KO fibroblast derived from mouse ES cells. Increased TWIST1 expression is a feature of OA-affected cartilage. We identified a novel mechanism of catabolic reaction where TWIST1 up-regulates MMP3 expression by enriching 5hmC levels at the MMP3 promoter via TET1 induction. These findings implicate TWIST1 as an important factor regulating OA related gene expression. Clarifying epigenetic mechanisms of 5hmC induced by TWIST1 is a critical molecule to understanding OA pathogenesis.


Assuntos
Metaloproteinase 3 da Matriz/metabolismo , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Adulto , Idoso , Animais , Cartilagem Articular/citologia , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Células Cultivadas , Condrócitos/citologia , Condrócitos/metabolismo , Metilação de DNA , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Metaloproteinase 3 da Matriz/genética , Camundongos , Pessoa de Meia-Idade , Proteínas Nucleares/genética , Osteoartrite do Joelho/metabolismo , Osteoartrite do Joelho/patologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Proteína 1 Relacionada a Twist/genética , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA