Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(7): e2310264121, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38319963

RESUMEN

Epigenetic regulation plays a crucial role in the pathogenesis of autoimmune diseases such as inflammatory arthritis. DNA hypomethylating agents, such as decitabine (DAC), have been shown to dampen inflammation and restore immune homeostasis. In the present study, we demonstrate that DAC elicits potent anti-inflammatory effects and attenuates disease symptoms in several animal models of arthritis. Transcriptomic and epigenomic profiling show that DAC-mediated hypomethylation regulates a wide range of cell types in arthritis, altering the differentiation trajectories of anti-inflammatory macrophage populations, regulatory T cells, and tissue-protective synovial fibroblasts (SFs). Mechanistically, DAC-mediated demethylation of intragenic 5'-Cytosine phosphate Guanine-3' (CpG) islands of the transcription factor Irf8 (interferon regulatory factor 8) induced its re-expression and promoted its repressor activity. As a result, DAC restored joint homeostasis by resetting the transcriptomic signature of negative regulators of inflammation in synovial macrophages (MerTK, Trem2, and Cx3cr1), TREGs (Foxp3), and SFs (Pdpn and Fapα). In conclusion, we found that Irf8 is necessary for the inhibitory effect of DAC in murine arthritis and that direct expression of Irf8 is sufficient to significantly mitigate arthritis.


Asunto(s)
Artritis , Azacitidina , Ratones , Animales , Decitabina/farmacología , Azacitidina/farmacología , Epigénesis Genética , Metilación de ADN , Factores Reguladores del Interferón/metabolismo , Inflamación/genética , Artritis/genética , Antiinflamatorios , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/genética
2.
Proc Natl Acad Sci U S A ; 120(15): e2210409120, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37023130

RESUMEN

Stimulator of interferon genes (STING) is a key mediator of type-I interferon (IFN-I) signaling in response to a variety of stimuli, but the contribution of STING to homeostatic processes is not fully characterized. Previous studies showed that ligand activation of STING limits osteoclast differentiation in vitro through the induction of IFNß and IFN-I interferon-stimulated genes (ISGs). In a disease model (SAVI) driven by the V154M gain-of-function mutation in STING, fewer osteoclasts form from SAVI precursors in response to receptor activator of NF-kappaB ligand (RANKL) in an IFN-I-dependent manner. Due to the described role of STING-mediated regulation of osteoclastogenesis in activation settings, we sought to determine whether basal STING signaling contributes to bone homeostasis, an unexplored area. Using whole-body and myeloid-specific deficiency, we show that STING signaling prevents trabecular bone loss in mice over time and that myeloid-restricted STING activity is sufficient for this effect. STING-deficient osteoclast precursors differentiate with greater efficiency than wild types. RNA sequencing of wild-type and STING-deficient osteoclast precursor cells and differentiating osteoclasts reveals unique clusters of ISGs including a previously undescribed ISG set expressed in RANKL naïve precursors (tonic expression) and down-regulated during differentiation. We identify a 50 gene tonic ISG signature that is STING dependent and shapes osteoclast differentiation. From this list, we identify interferon-stimulated gene 15 (ISG15) as a tonic STING-regulated ISG that limits osteoclast formation. Thus, STING is an important upstream regulator of tonic IFN-I signatures shaping the commitment to osteoclast fates, providing evidence for a nuanced and unique role for this pathway in bone homeostasis.


Asunto(s)
Osteoclastos , Transducción de Señal , Animales , Ratones , Diferenciación Celular/fisiología , Interferones/metabolismo , Ligandos , Ratones Endogámicos C57BL , Osteoclastos/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo
3.
Proc Natl Acad Sci U S A ; 112(1): 154-9, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25535389

RESUMEN

The MAP kinase TGFß-activated kinase (TAK1) plays a crucial role in physiologic and pathologic cellular functions including cell survival, differentiation, apoptosis, inflammation, and oncogenesis. However, the entire repertoire of its mechanism of action has not been elucidated. Here, we found that ablation of Tak1 in myeloid cells causes osteopetrosis in mice as a result of defective osteoclastogenesis. Mechanistically, Tak1 deficiency correlated with increased NUMB-like (NUMBL) levels. Accordingly, forced expression of Numbl abrogated osteoclastogenesis whereas its deletion partially restored osteoclastogenesis and reversed the phenotype of Tak1 deficiency. Tak1 deletion also down-regulated Notch intracellular domain (NICD), but increased the levels of the transcription factor recombinant recognition sequence binding protein at Jκ site (RBPJ), consistent with NUMBL regulating notch signaling through degradation of NICD, a modulator of RBPJ. Accordingly, deletion of Rbpj partially corrected osteopetrosis in Tak1-deficient mice. Furthermore, expression of active IKK2 in RBPJ/TAK1-deficient cells significantly restored osteoclastogenesis, indicating that activation of NF-κB is essential for complete rescue of the pathway. Thus, we propose that TAK1 regulates osteoclastogenesis by integrating activation of NF-κB and derepression of NOTCH/RBPJ in myeloid cells through inhibition of NUMBL.


Asunto(s)
FN-kappa B/metabolismo , Osteopetrosis/enzimología , Osteopetrosis/patología , Receptores Notch/metabolismo , Transducción de Señal , Animales , Diferenciación Celular , Eliminación de Gen , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Quinasas Quinasa Quinasa PAM/deficiencia , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones Endogámicos C57BL , Células Mieloides/enzimología , Células Mieloides/patología , Proteínas del Tejido Nervioso/metabolismo , Osteoclastos/patología , Osteogénesis , Fenotipo , Células Madre/metabolismo
4.
Plant Biotechnol J ; 12(1): 69-80, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24102754

RESUMEN

Isoflavones, a group of flavonoids, restricted almost exclusively to family Leguminosae are known to exhibit anticancerous and anti-osteoporotic activities in animal systems and have been a target for metabolic engineering in commonly consumed food crops. Earlier efforts based on the expression of legume isoflavone synthase (IFS) genes in nonlegume plant species led to the limited success in terms of isoflavone content in transgenic tissue due to the limitation of substrate for IFS enzyme. In this work to overcome this limitation, the activation of multiple genes of flavonoid pathway using Arabidopsis transcription factor AtMYB12 has been carried out. We developed transgenic tobacco lines constitutively co-expressing AtMYB12 and GmIFS1 (soybean IFS) genes or independently and carried out their phytochemical and molecular analyses. The leaves of co-expressing transgenic lines were found to have elevated flavonol content along with the accumulation of substantial amount of genistein glycoconjugates being at the highest levels that could be engineered in tobacco leaves till date. Oestrogen-deficient (ovariectomized, Ovx) mice fed with leaf extract from transgenic plant co-expressing AtMYB12 and GmIFS1 but not wild-type extract exhibited significant conservation of trabecular microarchitecture, reduced osteoclast number and expression of osteoclastogenic genes, higher total serum antioxidant levels and increased uterine oestrogenicity compared with Ovx mice treated with vehicle (control). The skeletal effect of the transgenic extract was comparable to oestrogen-treated Ovx mice. Together, our results establish an efficient strategy for successful pathway engineering of isoflavones and other flavonoids in crop plants and provide a direct evidence of improved osteoprotective effect of transgenic plant extract.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Flavonoles/metabolismo , Isoflavonas/metabolismo , Nicotiana/metabolismo , Oxigenasas/metabolismo , Plantas Modificadas Genéticamente/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Arabidopsis/genética , Oxigenasas/genética , Plantas Modificadas Genéticamente/genética , Nicotiana/genética , Factores de Transcripción/genética
5.
Connect Tissue Res ; 53(5): 398-406, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22420753

RESUMEN

Osteoblasts play a pivotal role in load-driven bone formation by activating Wnt signaling through a signal from osteocytes as a mechanosensor. Osteoblasts are also sensitive to mechanical stimulation, but the role of RhoA, a small GTPase involved in the regulation of cytoskeleton adhesion complexes, in mechanotransduction of osteoblasts is not completely understood. Using MC3T3-E1 osteoblast-like cells under 1 hr flow treatment at 10 dyn/cm(2), we examined a hypothesis that RhoA signaling mediates the cellular responses to flow-induced shear stress. To test the hypothesis, we conducted genome-wide pathway analysis and evaluated the role of RhoA in molecular signaling. Activity of RhoA was determined with a RhoA biosensor, which determined the activation state of RhoA based on a fluorescence resonance energy transfer between CFP and YFP fluorophores. A pathway analysis indicated that flow treatment activated phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) signaling as well as a circadian regulatory pathway. Western blot analysis revealed that in response to flow treatment phosphorylation of Akt in PI3K signaling and phosphorylation of p38 and ERK1/2 in MAPK signaling were induced. FRET measurement showed that RhoA was activated by flow treatment, and an inhibitor to a Rho kinase significantly reduced flow-induced phosphorylation of p38, ERK1/2, and Akt as well as flow-driven elevation of the mRNA levels of osteopontin and cyclooxygenase-2. Collectively, the result demonstrates that in response to 1 hr flow treatment to MC3T3-E1 cells at 10 dyn/cm(2), RhoA plays a critical role in activating PI3K and MAPK signaling as well as modulating the circadian regulatory pathway.


Asunto(s)
Mecanotransducción Celular , Osteoblastos/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Amidas/farmacología , Animales , Línea Celular , Ritmo Circadiano/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Mecanotransducción Celular/efectos de los fármacos , Ratones , Modelos Biológicos , Osteoblastos/efectos de los fármacos , Osteoblastos/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Reología/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
6.
Bone Res ; 10(1): 12, 2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-35145063

RESUMEN

Osteoarthritis is a joint disease characterized by a poorly-defined inflammatory response that does not encompass a massive immune cell infiltration yet contributes to cartilage degradation and loss of joint mobility, suggesting a chondrocyte intrinsic inflammatory response. Using primary chondrocytes from joints of osteoarthritic mice and patients, we first show that these cells express ample pro-inflammatory markers and RANKL in an NF-κB dependent manner. The inflammatory phenotype of chondrocytes was recapitulated by exposure of chondrocytes to IL-1ß and bone particles, which were used to model bone matrix breakdown products revealed to be present in synovial fluid of OA patients, albeit their role was not defined. We further show that bone particles and IL-1ß can promote senescent and apoptotic changes in primary chondrocytes due to oxidative stress from various cellular sources such as the mitochondria. Finally, we provide evidence that inflammation, oxidative stress and senescence converge upon IκB-ζ, the principal mediator downstream of NF-κB, which regulates expression of RANKL, inflammatory, catabolic, and SASP genes. Overall, this work highlights the capacity and mechanisms by which inflammatory cues, primarily joint degradation products, i.e., bone matrix particles in concert with IL-1ß in the joint microenvironment, program chondrocytes into an "inflammatory phenotype" which inflects local tissue damage.

7.
Elife ; 112022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35916374

RESUMEN

Osteoarthritis is the most common joint disease in the world with significant societal consequences but lacks effective disease-modifying interventions. The pathophysiology consists of a prominent inflammatory component that can be targeted to prevent cartilage degradation and structural defects. Intracellular metabolism has emerged as a culprit of the inflammatory response in chondrocytes, with both processes co-regulating each other. The role of glutamine metabolism in chondrocytes, especially in the context of inflammation, lacks a thorough understanding and is the focus of this work. We display that mouse chondrocytes utilize glutamine for energy production and anabolic processes. Furthermore, we show that glutamine deprivation itself causes metabolic reprogramming and decreases the inflammatory response of chondrocytes through inhibition of NF-κB activity. Finally, we display that glutamine deprivation promotes autophagy and that ammonia is an inhibitor of autophagy. Overall, we identify a relationship between glutamine metabolism and inflammatory signaling and display the need for increased study of chondrocyte metabolic systems.


Asunto(s)
Condrocitos , Osteoartritis , Animales , Cartílago , Condrocitos/metabolismo , Glutamina/metabolismo , Ratones , FN-kappa B/metabolismo , Osteoartritis/metabolismo
8.
Bioorg Med Chem Lett ; 21(15): 4617-21, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21719287

RESUMEN

Phytochemical investigation of ethanol extracts of the Pterospermum acerifolium flowers led to the isolation and identification of two new flavones, 4'-(2-methoxy-4-(1,2,3-trihydroxypropyl) phenoxy luteolin (1) and 5,7,3'-trihydroxy-6-O-ß-D-glucopyranosyl flavone (2), and one new lactone, 3,5-dihydroxyfuran-2(5H)-one (3) along with 14 known compounds (4-17). The structure of compounds 1-17 was established based on MS, 1D and 2D NMR, spectroscopic analysis. Eight of these compounds (1-6, 8 and 9) were assessed for osteogenic activity by using primary cultures of rat osteoblast. The compounds 1, 3 and 4 significantly stimulated osteoblast differentiation and mineralization as evident from a marked increase in expression of alkaline phosphatase and alizarin red-S staining of osteoblasts.


Asunto(s)
4-Butirolactona/análogos & derivados , Flavonas/química , Malvaceae/química , Osteoblastos/efectos de los fármacos , 4-Butirolactona/química , 4-Butirolactona/aislamiento & purificación , 4-Butirolactona/farmacología , Fosfatasa Alcalina/metabolismo , Animales , Antraquinonas/química , Flavonas/aislamiento & purificación , Flavonas/farmacología , Flores/química , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Conformación Molecular , Osteoblastos/metabolismo , Ratas
9.
Methods Mol Biol ; 2366: 267-282, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34236644

RESUMEN

The skeletal system is constantly undergoing turnover in order to create strong, organized structures, requiring the bone breakdown and building properties by osteoclasts and osteoblasts, respectively. However, in pathological disease states, excessive osteoclast activity can cause bone loss leading to increase in morbidity and mortality. Osteoclasts differentiate from macrophages in the presence of various factors. M-CSF is a cytokine that is required to maintain the survival of macrophages. However, RANKL is the critical factor required for differentiation of osteoclasts. RANKL is produced from a variety of different cell types such as osteoblasts and osteocytes. RANKL binds to RANK, its receptor, on the surface of osteoclast precursors, which activates various signaling pathways to drive the transcription and production of genes important for osteoclast formation. The major signaling pathway activated by RANKL-RANK interaction is the NF-κB pathway. The NF-κB pathway is the principle inflammatory response pathway activated by a variety of stimuli such as inflammatory cytokines, genotoxic stress, and other factors. This likely explains the finding that inflammatory diseases often present with some component of increased osteoclast formation and activity, driving bone loss. Determining the signaling mechanisms downstream of RANKL can provide valuable therapeutic targets for the treatment of bone loss in various disease states.


Asunto(s)
Transducción de Señal , Diferenciación Celular , FN-kappa B/metabolismo , Osteoclastos/metabolismo , Osteogénesis , Ligando RANK/metabolismo , Factor 6 Asociado a Receptor de TNF
10.
Bone Res ; 9(1): 29, 2021 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099632

RESUMEN

Atrophic fracture nonunion poses a significant clinical problem with limited therapeutic interventions. In this study, we developed a unique nonunion model with high clinical relevance using serum transfer-induced rheumatoid arthritis (RA). Arthritic mice displayed fracture nonunion with the absence of fracture callus, diminished angiogenesis and fibrotic scar tissue formation leading to the failure of biomechanical properties, representing the major manifestations of atrophic nonunion in the clinic. Mechanistically, we demonstrated that the angiogenesis defect observed in RA mice was due to the downregulation of SPP1 and CXCL12 in chondrocytes, as evidenced by the restoration of angiogenesis upon SPP1 and CXCL12 treatment in vitro. In this regard, we developed a biodegradable scaffold loaded with SPP1 and CXCL12, which displayed a beneficial effect on angiogenesis and fracture repair in mice despite the presence of inflammation. Hence, these findings strongly suggest that the sustained release of SPP1 and CXCL12 represents an effective therapeutic approach to treat impaired angiogenesis and fracture nonunion under inflammatory conditions.

11.
Arthritis Res Ther ; 23(1): 286, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34784954

RESUMEN

BACKGROUND: Gasdermin D (GSDMD) is cleaved by several proteases including by caspase-1, a component of intracellular protein complexes called inflammasomes. Caspase-1 also converts pro-interleukin-1ß (pro-IL-1ß) and pro-IL-18 into bioactive IL-1ß and IL-18, respectively. GSDMD amino-terminal fragments form plasma membrane pores, which mediate the secretion of IL-1ß and IL-18 and cause the inflammatory form of cell death pyroptosis. Here, we tested the hypothesis that GSDMD contributes to joint degeneration in the K/BxN serum transfer-induced arthritis (STIA) model in which autoantibodies against glucose-6-phosphate isomerase promote the formation of pathogenic immune complexes on the surface of myeloid cells, which highly express the inflammasomes. The unexpected outcomes with the STIA model prompted us to determine the role of GSDMD in the post-traumatic osteoarthritis (PTOA) model caused by meniscus ligamentous injury (MLI) based on the hypothesis that this pore-forming protein is activated by signals released from damaged joint tissues. METHODS: Gsdmd +/+ and Gsdmd-/- mice were injected with K/BxN mouse serum or subjected to MLI to cause STIA or PTOA, respectively. Paw and ankle swelling and DXA scanning were used to assess the outcomes in the STIA model whereas histopathology and micro-computed tomography (µCT) were utilized to monitor joints in the PTOA model. Murine and human joint tissues were also examined for GSDMD, IL-1ß, and IL-18 expression by qPCR, immunohistochemistry, or immunoblotting. RESULTS: GSDMD levels were higher in serum-inoculated paws compared to PBS-injected paws. Unexpectedly, ablation of GSDMD failed to reduce joint swelling and osteolysis, suggesting that GSDMD was dispensable for the pathogenesis of STIA. GSDMD levels were also higher in MLI compared to sham-operated joints. Importantly, ablation of GSDMD attenuated MLI-associated cartilage degradation (p = 0.0097), synovitis (p = 0.014), subchondral bone sclerosis (p = 0.0006), and subchondral bone plate thickness (p = 0.0174) based on histopathological and µCT analyses. CONCLUSION: GSDMD plays a key role in the pathogenesis of PTOA, but not STIA, suggesting that its actions in experimental arthropathy are tissue context-specific.


Asunto(s)
Complejo Antígeno-Anticuerpo , Artritis , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de Unión a Fosfato/genética , Heridas y Lesiones/complicaciones , Animales , Artritis/etiología , Autoanticuerpos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Noqueados , Microtomografía por Rayos X
12.
Elife ; 92020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-32202502

RESUMEN

Inflammatory osteolysis is governed by exacerbated osteoclastogenesis. Ample evidence points to central role of NF-κB in such pathologic responses, yet the precise mechanisms underpinning specificity of these responses remain unclear. We propose that motifs of the scaffold protein IKKγ/NEMO partly facilitate such functions. As proof-of-principle, we used site-specific mutagenesis to examine the role of NEMO in mediating RANKL-induced signaling in mouse bone marrow macrophages, known as osteoclast precursors. We identified lysine (K)270 as a target regulating RANKL signaling as K270A substitution results in exuberant osteoclastogenesis in vitro and murine inflammatory osteolysis in vivo. Mechanistically, we discovered that K270A mutation disrupts autophagy, stabilizes NEMO, and elevates inflammatory burden. Specifically, K270A directly or indirectly hinders binding of NEMO to ISG15, a ubiquitin-like protein, which we show targets the modified proteins to autophagy-mediated lysosomal degradation. Taken together, our findings suggest that NEMO serves as a toolkit to fine-tune specific signals in physiologic and pathologic conditions.


The human skeleton contains over 200 bones that together act as an internal framework for the body. Over our lifetime, the body constantly removes older bone tissue from the skeleton and replaces it with new bone tissue. This "bone remodeling" also controls how bones are repaired after being damaged by injuries, disease or normal wear and tear. Cells known as osteoclasts are responsible for breaking down old bone tissue and participate in repairing damaged bone. A cellular pathway known as NF-kB signaling stimulates other cells called "bone marrow macrophages" to become osteoclasts. A certain level of NF-kB signaling is required to maintain a healthy skeleton. However, under certain inflammatory conditions, the level of NF-kB signaling becomes too high causing hyperactive osteoclasts to accumulate and inflict severe bone breakdown. This abnormal osteoclast activity leads to eroded and fragile bones and joints, as is the case in diseases such as rheumatoid arthritis and osteoporosis. Previous studies have shown that a protein called NEMO is a core component of the NF-kB signal pathway, but the precise role of NEMO in the diseased response remained unclear. Adapala, Swarnkar, Arra et al. have now used site-directed mutagenesis approach to study the role of NEMO in bone marrow macrophages in mice. The experiments showed that one specific site within the NEMO protein, referred to as lysine 270, is crucial for its role in controlling osteoclasts and the breakdown of bone tissue. Mutating NEMO at lysine 270 led to uncontrolled NF-kB signaling in the bone marrow macrophages. Further experiments showed that lysine 270 served as a sensor to allow NEMO to bind another protein called ISG15, which in turn helped to decrease NF-kB signaling and slow down the erosion of the bone. These findings suggest that site-specific targeting of NEMO, rather than inhibiting the whole NF-kB pathway, may help to reduce the symptoms of bone disease while maintaining the beneficial roles of this essential pathway. However, additional research is required to identify NEMO sites responsible for controlling the inflammatory component.


Asunto(s)
Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Osteólisis/metabolismo , Animales , Células de la Médula Ósea , Regulación de la Expresión Génica , Inflamación/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Artropatías/metabolismo , Artropatías/patología , Ratones , Ratones Transgénicos , Mutación , FN-kappa B/genética , FN-kappa B/metabolismo , Osteoclastos/fisiología , Osteólisis/genética , Ligando RANK/genética , Ligando RANK/metabolismo
13.
Nat Commun ; 11(1): 3427, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32647171

RESUMEN

The contribution of inflammation to the chronic joint disease osteoarthritis (OA) is unclear, and this lack of clarity is detrimental to efforts to identify therapeutic targets. Here we show that chondrocytes under inflammatory conditions undergo a metabolic shift that is regulated by NF-κB activation, leading to reprogramming of cell metabolism towards glycolysis and lactate dehydrogenase A (LDHA). Inflammation and metabolism can reciprocally modulate each other to regulate cartilage degradation. LDHA binds to NADH and promotes reactive oxygen species (ROS) to induce catabolic changes through stabilization of IκB-ζ, a critical pro-inflammatory mediator in chondrocytes. IκB-ζ is regulated bi-modally at the stages of transcription and protein degradation. Overall, this work highlights the function of NF-κB activity in the OA joint as well as a ROS promoting function for LDHA and identifies LDHA as a potential therapeutic target for OA treatment.


Asunto(s)
Condrocitos/metabolismo , Lactato Deshidrogenasa 5/metabolismo , Terapia Molecular Dirigida , Osteoartritis/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aerobiosis , Animales , Cartílago Articular/patología , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/patología , Citoprotección/efectos de los fármacos , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Glucólisis/efectos de los fármacos , Humanos , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/farmacología , Articulación de la Rodilla/patología , Meniscos Tibiales/cirugía , Redes y Vías Metabólicas/efectos de los fármacos , Ratones Endogámicos C57BL , NAD/metabolismo , FN-kappa B/metabolismo , Osteoartritis/genética , Osteoartritis/patología
14.
Curr Med Chem ; 16(9): 1138-57, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19275617

RESUMEN

Substantial body of data generated from cultured bone cells and rat models of osteoporosis supports a significant bone-conserving effect of phytochemicals. Flavonoids including isoflavones, stilbenes and lignans with variable efficacy have shown promising therapeutic application in osteoporosis. Majority of the phytochemicals assessed for their effects on bone cells revealed multiple beneficial actions such as promoting osteoblast functions, and inhibiting osteoclast and adipocyte functions. A variety of molecular targets mediate multiple effects of phytochemicals in bone cells. In vivo, quite a few phytochemicals have been found to afford bone-sparing effect and in some cases even bone restoring effect. However, important pharmacokinetic and bioavailaibility studies associated with these phytochemicals are mostly lacking. As a result, translating these findings to the clinic has been challenging, and so far only a few clinical studies have attempted to evaluate the effect of phytochemicals in menopausal osteoporosis. Clinical studies so far performed are with dietary supplements rather than pure phytochemicals. Clinical trials with pure molecules necessitate preclinical regulatory and safety studies that are not available with the phytochemicals except ipriflavone with bone-conserving properties. Ipriflavone is the only marketed anti-osteoporosis agent that was obtained following a lead from natural substance. As phytochemicals have multiple beneficial influences on bone cells, making analogues of the most potent molecule for developing synthetic series with rational drug design approach could pay rich dividends in menopausal osteoporosis therapy.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Conservadores de la Densidad Ósea/farmacocinética , Huesos/efectos de los fármacos , Osteoporosis Posmenopáusica/prevención & control , Extractos Vegetales/farmacología , Extractos Vegetales/farmacocinética , Huesos/metabolismo , Femenino , Humanos , Estructura Molecular
15.
Bone ; 123: 86-91, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30904629

RESUMEN

Cytokines and growth factors mediate inflammatory osteolysis in response to particles released from bone implants. However, the mechanism by which this process develops is not entirely clear. Blood vessels and related factors may be required to deliver immune cells and soluble factors to the injury site. Therefore, in the current study we investigated if, vascular endothelial growth factor (VEGF), which is required for angiogenesis, mediates polymethylmethacrylate (PMMA) particles-induced osteolysis. Using bone marrow derived macrophages (BMMs) and ST2 stromal cell line, we show that PMMA particles increase VEGF expression. Further, using a murine calvarial osteolysis model, we found that PMMA injection over calvaria induce significant increase in VEGF expression as well as new vessel formation, represented by von Willebrand factor (vWF) staining. Co-treatment using a VEGF-neutralizing antibody abrogated expression of vWF, indicating decreased angiogenesis. Finally, VEGF neutralizing antibody reduced expression of Tumor necrosis factor (TNF) and decreased osteoclastogenesis induced by PMMA particles in calvariae. This work highlights the significance of angiogenesis, specifically VEGF, as key driver of PMMA particle-induced inflammatory osteolysis, inhibition of which attenuates this response.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Osteólisis/inducido químicamente , Osteólisis/prevención & control , Polimetil Metacrilato/toxicidad , Cráneo/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Animales , Cementos para Huesos/toxicidad , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Microesferas , Osteólisis/metabolismo , Distribución Aleatoria , Cráneo/metabolismo , Factor A de Crecimiento Endotelial Vascular/agonistas , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
16.
Sci Rep ; 9(1): 10429, 2019 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-31320680

RESUMEN

Chronic inflammatory insults compromise immune cell responses and ultimately contribute to pathologic outcomes. Clinically, it has been suggested that bone debris and implant particles, such as polymethylmethacrylate (PMMA), which are persistently released following implant surgery evoke heightened immune, inflammatory, and osteolytic responses that contribute to implant failure. However, the precise mechanism underlying this pathologic response remains vague. TREGS, the chief immune-suppressive cells, express the transcription factor Foxp3 and are potent inhibitors of osteoclasts. Using an intra-tibial injection model, we show that PMMA particles abrogate the osteoclast suppressive function of TREGS. Mechanistically, PMMA particles induce TREG instability evident by reduced expression of Foxp3. Importantly, intra-tibial injection of PMMA initiates an acute innate immune and inflammatory response, yet the negative impact on TREGS by PMMA remains persistent. We further show that PMMA enhance TH17 response at the expense of other T effector cells (TEFF), particularly TH1. At the molecular level, gene expression analysis showed that PMMA particles negatively regulate Nrp-1/Foxo3a axis to induce TREG instability, to dampen TREG activity and to promote phenotypic switch of TREGS to TH17 cells. Taken together, inflammatory cues and danger signals, such as bone and implant particles exacerbate inflammatory osteolysis in part through reprogramming TREGS.


Asunto(s)
Inflamación/inmunología , Neuropilina-1/inmunología , Linfocitos T Reguladores/inmunología , Animales , Femenino , Factores de Transcripción Forkhead/inmunología , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Masculino , Ratones , Osteoclastos/efectos de los fármacos , Osteoclastos/inmunología , Osteólisis/inmunología , Polimetil Metacrilato , Linfocitos T Reguladores/efectos de los fármacos , Células Th17/efectos de los fármacos , Células Th17/inmunología
17.
J Bone Miner Res ; 34(10): 1880-1893, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31107556

RESUMEN

Skeletal abnormalities are common comorbidities of inflammatory bowel disease (IBD). Patients suffering from IBD, including ulcerative colitis and Crohn's disease, present with skeletal complications. However, the mechanism underpinning IBD-associated bone loss remains vague. Intestinal inflammation generates an inflammatory milieu at the intestinal epithelium that leads to dysregulation of mucosal immunity through gut-residing innate lymphoid cells (ILCs) and other cell types. ILCs are recently identified mucosal cells considered as the gatekeeper of gut immunity and their function is regulated by intestinal epithelial cell (IEC)-secreted cytokines in response to the inflammatory microenvironment. We first demonstrate that serum as well as IECs collected from the intestine of dextran sulfate sodium (DSS)-induced colitis mice contain high levels of inflammatory and osteoclastogenic cytokines. Mechanistically, heightened inflammatory response of IECs was associated with significant intrinsic activation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) in IECs and increased frequency of ILC1, ILC3, and myeloid osteoclast progenitors. Validating the central role of IEC-specific NF-κB activation in this phenomenon, conditional expression of constitutively active inhibitor kappa B kinase 2 (IKK2) in IECs in mice recapitulates the majority of the cellular, inflammatory, and osteolytic phenotypes observed in the chemically induced colitis. Furthermore, conditional deletion of IKK2 from IECs significantly attenuated inflammation and bone loss in DSS-induced colitis. Finally, using the DSS-induced colitis model, pharmacologic inhibition of IKK2 was effective in reducing frequency of ILC1 and ILC3 cells, attenuated circulating levels of inflammatory cytokines, and halted colitis-associated bone loss. Our findings identify IKK2 in IECs as viable therapeutic target for colitis-associated osteopenia.


Asunto(s)
Resorción Ósea/metabolismo , Colitis/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , FN-kappa B/metabolismo , Animales , Resorción Ósea/etiología , Resorción Ósea/genética , Resorción Ósea/patología , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/genética , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Células Epiteliales/patología , Femenino , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Noqueados , FN-kappa B/genética
18.
Mol Cell Endocrinol ; 289(1-2): 85-93, 2008 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-18400372

RESUMEN

Kaempferol (K), a flavonol, is known to have anti-osteoclastogenic effect. We here show that K, from 0.2 to 5.0 microM, increased mineralized nodules in rat primary osteoblasts. K also significantly attenuated adipocyte formation from bone marrow cells (BMCs). A single oral dose of 1 mg/kg body weight of K in Sprague-Dawley (180-200 g) rats resulted in a peak serum level of 2.04+/-0.8 nM in 30 min (Tmax), suggesting its rapid absorption. The Cmax of K in bone marrow was 0.684 nM after 90 min. Rats were ovariectomized (OVx) along with sham-operated rats and left for 4 weeks. Daily oral administration of K (5 mg/kg body weight) was then started to one group of OVx rats, and continued for 10 weeks. K levels were found to be 0.311 and 0.838 nM at the end of 4 and 10 weeks, respectively. K exhibited no estrogenicity at the uterine level. The K-treated group exhibited significantly higher bone mineral density (BMD) in the trabecular regions (femur neck, proximal tibia and vertebrae) and lower serum ALP (bone turnover marker) compared with the OVx rats. The compressive energy of the vertebrae was significantly higher in the OVx+K-treated group compared with the OVx group. K treatment of OVx rats resulted in the increase in osteoprogenitor cells as well as inhibition of adipocyte differentiation from BMCs compared with the OVx group. Together we show that K is non-estrogenic in vivo and exerts bone anabolic activity with attendant inhibition of bone marrow adipogenesis.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Quempferoles/administración & dosificación , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/fisiología , Administración Oral , Animales , Densidad Ósea/fisiología , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/fisiología , Diferenciación Celular/efectos de los fármacos , Femenino , Cuello Femoral/fisiología , Quempferoles/farmacocinética , Vértebras Lumbares/fisiología , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Osteogénesis/efectos de los fármacos , Ovariectomía , Ratas , Ratas Sprague-Dawley , Tibia/fisiología
19.
Indian J Med Res ; 127(3): 274-86, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18497443

RESUMEN

Bone turnover helps accomplish long-term correction of the extracellular calcium (Ca2+ o) homeostasis by the actions of osteoblasts and osteoclasts. These processes are highly regulated by the actions of hormones, most prominently parathyroid hormone (PTH), the release of which is a function of the Ca2+ o, and is regulated by the action of the Ca2+ -sensing receptor (CaR) in the parathyroid gland. Various mutations of the CaR gene give rise to gain or loss of functions leading respectively to hypo- or hypercalcaemic conditions. CaR could conceivably be a target for local changes in the Ca2+ o in the bone microenvironment thereby acting as a 'growth factor' in various cells residing in the bone marrow. This review discusses about the roles of the CaR in bone. In osteoblasts, CaR promotes its proliferation, differentiation and mineralization. In osteoclasts, CaR mediates high Ca2+ o-stimulated osteoclast differentiation as well as osteoclast apoptosis. CaR regulates localization of haematopoietic stem cells from the foetal liver to endosteal niche, the socalled homing. Although the CaR plays a key role in the defense against hypercalcaemia, its function can be aberrant in humoral hypercalcaemia of malignancy in which CaR activation stimulates secretion of parathyroid hormone-related peptide (PTHrP) secretion. Increased levels of PTHrP cause a vicious hypercalcaemic state resulting from its increased bone-resorptive and positive renal calcium reabsorbing effects give rise to hypercalcaemia. CaR mediates a variety of functions of Ca2+ o in the bone microenvironment under both normal and pathological conditions.


Asunto(s)
Enfermedades Óseas/fisiopatología , Huesos/fisiología , Calcio/fisiología , Hipercalcemia/fisiopatología , Receptores Sensibles al Calcio/fisiología , Humanos
20.
Sci Rep ; 7(1): 12600, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974699

RESUMEN

NF-κB signaling is essential for osteoclast differentiation and skeletal homeostasis. We have reported recently that NUMB-like (NUMBL) protein modulates osteoclastogenesis by down regulating NF-κB activation. Herein, we decipher the mechanism underlying this phenomenon. We found that whereas NUMBL mRNA expression decreases upon stimulation of wild type (WT) bone marrow macrophages (BMMs) with RANKL, TAK1 deficiency in these cells leads to increased NUMBL and decreased TRAF6 and NEMO expression. These changes were restored upon WT-TAK1 expression, but not with catalytically inactive TAK1-K63W, suggesting that TAK1 enzymatic activity is required for these events. Forced expression of NUMBL inhibits osteoclast differentiation and function as evident by reduction in all hallmarks of osteoclastogenesis. Conversely, NUMBL-null BMMs, show increased osteoclast differentiation and mRNA expression of osteoclast marker genes. Post-translationally, K48-linked poly-ubiquitination of NUMBL is diminished in TAK1-null BMMs compared to elevated K48-poly-ubiquitination in WT cells, indicating increased stability of NUMBL in TAK1-null conditions. Further, our studies show that NUMBL directly interacts with TRAF6 and NEMO, and induces their K48-poly-ubiquitination mediated proteasomal degradation. Collectively, our data suggest that NUMBL and TAK1 are reciprocally regulated and that NUMBL acts as an endogenous regulator of NF-κB signaling and osteoclastogenesis by targeting the TAK1-TRAF6-NEMO axis.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas del Tejido Nervioso/genética , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/genética , Osteogénesis/genética , Factor 6 Asociado a Receptor de TNF/genética , Animales , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , FN-kappa B/genética , Osteoclastos/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , ARN Mensajero/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda