Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int Immunopharmacol ; 136: 112343, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38810305

RESUMEN

Defective clearance of apoptotic cells due to impaired efferocytosis sustains error in self-tolerance that exacerbates rheumatoid arthritis (RA). However, the molecular determinant that directly or specifically impairs efferocytosis in RA is not yet studied. We identified a new perspective that IL-17A significantly impedes efferocytosis via preferential activation of the JAK/STAT-3/ADAM17 signaling axis. In contrast, disruption of the IL-17A/IL-17RA interaction using cyanidin or silencing of IL-17RA obstructed JAK/STAT-3 activation that further abolished ADAM17 expression. Subsequent depletion of ADAM17 inhibited the shedding of Mer tyrosine kinase receptor (MERTK), which significantly increased apoptotic cell intake and restored efferocytosis in adjuvant-induced arthritic (AA) model. Concomitantly, the amplification of the efferocytosis process due to IL-17A/IL-17RA interaction disruption was sensitive to mitochondrial fission mediated via Drp-1 phosphorylation downstream of STAT-3 inhibition. As expected, cyanidin treated AA synovial macrophages that exhibited increased efferocytosis demonstrated a phenotypic shift towards CD163 anti-inflammatory phenotype in a STAT-5 dependent manner. Similar results were obtained in IL-17A-sensitized AA synovial macrophages treated with S3I-201 (a STAT-3 inhibitor) indicating that IL-17A influences efferocytosis via the STAT-3 pathway. In view of our previous work where cyanidin restored Th17/Treg balance, our present investigation fulfils a critical gap by providing scientific validation that cyanidin escalated PD-L1 expression during the efferocytosis process that could have impacted the restoration of Th17/Treg balance in an AA model. Together, these data corroborate the hypothesis that IL-17A signaling can impair efferocytosis via regulating STAT-3/ADAM17/FL-MERTK axis and that its inhibition can amplify a pro-resolution signal against RA progression.


Asunto(s)
Artritis Reumatoide , Antígeno B7-H1 , Interleucina-17 , Macrófagos , Receptores de Interleucina-17 , Factor de Transcripción STAT3 , Transducción de Señal , Interleucina-17/metabolismo , Animales , Factor de Transcripción STAT3/metabolismo , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Antígeno B7-H1/metabolismo , Ratones , Receptores de Interleucina-17/metabolismo , Receptores de Interleucina-17/genética , Antocianinas/farmacología , Artritis Experimental/inmunología , Humanos , Modelos Animales de Enfermedad , Fagocitosis/efectos de los fármacos , Masculino , Membrana Sinovial/inmunología , Membrana Sinovial/patología , Ratones Endogámicos C57BL , Apoptosis/efectos de los fármacos , Eferocitosis
2.
3 Biotech ; 14(1): 25, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38164247

RESUMEN

Interleukin (IL)-21 is a major lineage-defining factor that promotes Tfh cell differentiation. The current study investigated the molecular basis of myricetin, a flavonoid that impedes IL-21-mediated differentiation of Tfh cells in RA. Through high-throughput virtual screening of natural compounds that inhibit IL-21, we found that myricetin binds to IL-21 and hampers its interaction with IL-21 receptor (IL-21R). Our in vivo studies demonstrated that myricetin treatment ameliorated the clinical manifestations in adjuvant-induced arthritis (AIA) mice by reducing paw thickness and cellular infiltration. In addition, myricetin inhibited splenic Tfh cell differentiation and IL-21 production in AIA mice. Myricetin negatively regulates JAK/STAT signaling and the downstream Bcl-6 transcription factor at the molecular level, which arrests Tfh cell differentiation. Our current research proposal to target IL-21 with myricetin inevitably represents a new molecular approach that expedites new alternative drugs for rheumatoid arthritis therapy. Supplementary Information: The online version contains supplementary material available at 10.1007/s13205-023-03880-w.

3.
Life Sci ; 332: 122115, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37739160

RESUMEN

The intriguing scientific relationship between autoimmunity and cancer immunology have been traditionally indulged to throw spotlight on novel pathological targets. Understandably, these "slowly killing" diseases are on the opposite ends of the immune spectrum. However, the immune regulatory mechanisms between autoimmunity and cancer are not always contradictory and sometimes mirror each other based on disease stage, location, and timepoint. Moreover, the blockade of immune checkpoint molecules or signalling pathways that unleashes the immune response against cancer is being leveraged to preserve self-tolerance and treat many autoimmune disorders. Therefore, understanding the common crucial factors involved in cancer is of paramount importance to paint the autoimmune disease spectrum and validate novel drug candidates. In the current review, we will broadly describe how ZEB1, or Zinc-finger E-box Binding Homeobox 1, reinforces immune exhaustion in cancer or contributes to loss of self-tolerance in auto-immune conditions. We made an effort to exchange information about the molecular pathways and pathological responses (immune regulation, cell proliferation, senescence, autophagy, hypoxia, and circadian rhythm) that can be regulated by ZEB1 in the context of autoimmunity. This will help untwine the intricate and closely postured pathogenesis of ZEB1, that is less explored from the perspective of autoimmunity than its counterpart, cancer. This review will further consider several approaches for targeting ZEB1 in autoimmunity.


Asunto(s)
Enfermedades Autoinmunes , Neoplasias , Humanos , Autoinmunidad , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Línea Celular Tumoral
4.
Int J Mol Sci ; 24(8)2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-37108809

RESUMEN

In recent years, the launch of clinical-grade exosomes is rising expeditiously, as they represent a new powerful approach for the delivery of advanced therapies and for diagnostic purposes for various diseases. Exosomes are membrane-bound extracellular vesicles that can act as biological messengers between cells, in the context of health and disease. In comparison to several lab-based drug carriers, exosome exhibits high stability, accommodates diverse cargo loads, elicits low immunogenicity and toxicity, and therefore manifests tremendous perspectives in the development of therapeutics. The efforts made to spur exosomes in drugging the untreatable targets are encouraging. Currently, T helper (Th) 17 cells are considered the most prominent factor in the establishment of autoimmunity and several genetic disorders. Current reports have indicated the importance of targeting the development of Th17 cells and the secretion of its paracrine molecule, interleukin (IL)-17. However, the present-day targeted approaches exhibit drawbacks, such as high cost of production, rapid transformation, poor bioavailability, and importantly, causing opportunistic infections that ultimately hamper their clinical applications. To overcome this hurdle, the potential use of exosomes as vectors seem to be a promising approach for Th17 cell-targeted therapies. With this standpoint, this review discusses this new concept by providing a snapshot of exosome biogenesis, summarizes the current clinical trials of exosomes in several diseases, analyzes the prospect of exosomes as an established drug carrier and delineates the present challenges, with an emphasis on their practical applications in targeting Th17 cells in diseases. We further decode the possible future scope of exosome bioengineering for targeted drug delivery against Th17 cells and its catastrophe.


Asunto(s)
Exosomas , Células Th17 , Sistemas de Liberación de Medicamentos/métodos , Portadores de Fármacos , Comunicación Celular
5.
Cytokine ; 163: 156136, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36716676

RESUMEN

Dendritic cells (DCs) are the critical players in the puzzle of rheumatoid arthritis (RA) disease pathogenesis. Blockade of DC activation has been shown to curtail Th17 cell differentiation and its aberrant function in RA. Recent studies have pointed to the role of the PI3K/AKT signaling axis in the maturation and activation of DCs. However, it is yet to be established how PI3K/AKT inhibition would lead to the abolishment of DC activation and Th17 cell plasticity in RA. Herein, our study decoded whether and how majoon chobchini, an unani compound, abated dendritic cell maturation and regulated the Th17/Treg paradigm in RA. Given our results, majoon chobchini conspicuously restrained MHC II, CD86 expression and, subsequently elevated PDL-1 levels in DCs in-vivo. Of note, inhibition of DC maturation by majoon chobchini, in turn, favoured suppression of the Th17 cell population while driving Treg cell development in adjuvant induced arthritic (AA) rats. Concurrently, majoon chobchini decreased the catabolic effects of IL-17 (Th17 associated cytokine) via a reciprocal increase in IL-10 (Treg associated cytokine) levels in AA rats. Mechanistically, majoon chobchini sustained FoxO1 nuclear localization signaled through dampened PI3K/AKT phosphorylation in-vitro. In concert, PDL-1 expression was heightened in majoon chobchini treated activated DCs that provides a framework for ablation of the DC-Th17 cell pathogenic axis in RA. Notwithstanding, the PI3K inhibitor LY294002 exhibited similar inhibitory effects. In essence, majoon chobchini enhanced PDL-1 expression that abolished DC maturation via regulation of the PI3K/AKT/FoxO1 axis, thereby hindering Th17 differentiation in an animal model of RA. This further warrants a clinical investigation that could validate majoon chobchini as a prospective therapeutic drug in the treatment of RA.


Asunto(s)
Artritis Reumatoide , Proteínas Proto-Oncogénicas c-akt , Ratas , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Artritis Reumatoide/metabolismo , Citocinas/metabolismo , Linfocitos T Reguladores/metabolismo , Modelos Animales de Enfermedad , Células Th17/metabolismo , Células Dendríticas/metabolismo
6.
Pharmacol Res ; 178: 106176, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35283302

RESUMEN

Rheumatoid arthritis (RA) is a chronic immune-mediated disorder, mainly characterized by synovial inflammation and joint damage. If insufficiently treated, RA can lead to irreversible joint destruction and decreased life expectancy. While better understanding of the pathologies and the development of new antirheumatic drugs have improved the outcome of individuals with RA, many patients still cannot achieve remission and experience progressive disability. Fibroblast-like synoviocytes (FLS) have gained attention due to its pivotal role in RA pathogenesis and thus targeting FLS has been suggested as an attractive therapeutic strategy. To identify candidate molecules with strong inhibitory activity against FLS inflammation, we tested the effect of 315 natural extracts against IL-17-mediated IL-6 production. Zingiber officinale was found as the top hit and further analysis on the active compound responsible led to the discovery of 8-shogaol as a potent molecule against synovitis. 8-Shogaol displayed significant inhibitory effects against TNF-α-, IL-1ß-, and IL-17-mediated inflammation and migration in RA patient-derived FLS (RA-FLS) and 3D synovial culture system. 8-Shogaol selectively and directly inhibited TAK1 activity and subsequently suppressed IKK, Akt, and MAPK signaling pathways. Moreover, treatment with 8-shogaol reduced paw thickness and improved walking performance in the adjuvant-induced arthritic (AIA) rat model. 8-Shogaol also reversed pathologies of joint structure in AIA rats and decreased inflammatory biomarkers in the joints. Collectively, we report a novel natural compound that inhibits RA through reversing pathologies of the inflamed synovium via targeting TAK1.


Asunto(s)
Artritis Reumatoide , Guayacol , Quinasas Quinasa Quinasa PAM , Sinoviocitos , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Guayacol/análogos & derivados , Guayacol/farmacología , Humanos , Interleucina-17/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Terapia Molecular Dirigida , Ratas , Sinoviocitos/efectos de los fármacos , Sinoviocitos/metabolismo , Sinoviocitos/patología
7.
Bioorg Chem ; 121: 105667, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35182886

RESUMEN

In search of potent and new anti-inflammatory agents, we explored a new class of isocoumarin derivatives possessing the 3-oxoalkyl moiety at C-4 position. These compounds were synthesized via the FeCl3 catalyzed construction of isocoumarin ring. The methodology involved coupling of 2-alkynyl benzamides with alkyl vinyl ketone and proceeded via a regioselective cyclization to give the desired compound as a result of formation of CO and CC bonds. A large number of isocoumarins were synthesized and assessed against PDE4B in vitro. While isocoumarins containing an aminosulfonyl moiety attached to the C-3 aryl ring showed encouraging inhibition of PDE4B, some of the derivatives devoid of aminosulfonyl moiety also showed considerable inhibition. According to the SAR analysis the C6H4NHSO2R2-m moiety at C-3 position of the isocoumarin ring was favorable when the R2 was chosen as an aryl or 2-thienyl group whereas the presence of F or OMe substituent at C-7 of the isocoumarin ring was found to be beneficial. The compound 5f with IC50 values 0.125 ± 0.032 and 0.43 ± 0.013 µM against PDE4B and 4D, respectively was identified as an initial hit. It showed in silico interaction with the PHE678 residue in the CR3 region of PDE4B and relatively less number of interactions with PDE4D. Besides showing the PDE4 selectivity over other PDEs and TNF-α inhibition in vitro the compound 5f at an intraperitoneal dose of 30 mg/kg demonstrated the protective effects against the development of arthritis and potent immunomodulatory activity in adjuvant induced arthritic (AIA) rats. Furthermore, no significant adverse effects were observed for this compound when evaluated in a systematic toxicity (e.g. teratogenicity, hepatotoxicity and cardiotoxicity) studies in zebrafish at various concentrations. Collectively, being a new, potent, moderately selective and safe inhibitor of PDE4B the isocoumarin 5f can be progressed into further pharmacological studies.


Asunto(s)
Compuestos Férricos , Isocumarinas , Animales , Catálisis , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/química , Isocumarinas/química , Ratas , Relación Estructura-Actividad , Pez Cebra/metabolismo
8.
Immunol Invest ; 51(6): 1582-1597, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34704880

RESUMEN

Fibroblast-like synoviocytes (FLS) are the critical effector cells primarily involved in rheumatoid arthritis (RA) disease pathogenesis. Interleukin (IL)-6, a proinflammatory cytokine most abundantly expressed in the rheumatoid synovium, promotes Janus kinase (JAK)/signal transducer and transcriptional activator (STAT) signaling cascade activation in RA-FLS, thus leading to its aggressive phenotype, invasiveness, and joint destruction. Momelotinib (CYT387) is a selective small-molecule inhibitor of JAK1/2 and is clinically approved to treat myelofibrosis. However, the therapeutic efficacy of CYT387 in FLS mediated RA pathogenesis is less known. In the present study, we investigated the modulatory effect of CYT387 on IL6/JAK/STAT signaling cascade in FLS induced RA pathogenesis. CYT387 treatment inhibited IL-6 induced high proliferative and migratory potential of FLS cells isolated from adjuvant-induced arthritic (AA) rats. CYT387 reduced the expression of PRMT5, survivin, and HIF-1α mediated by IL-6/sIL-6R in AA-FLS in a dose-dependent manner. The IL-6/sIL-6R induced expression of angiogenic factors such as VEGF and PIGF in AA-FLS cells was found downregulated by CYT387 treatment. Importantly, CYT387 significantly reduced IL-6/sIL-6R dependent activation of JAK1 and STAT3 and increased SOCS3 expression in AA-FLS cells. Next, the S3I-201 mediated blockade of STAT3 activation supported the inhibitory effect of CYT387 on IL-6/JAK1/STAT3 signaling cascade in AA-FLS. Overall, this study proves that CYT387 inhibits proliferation, migration, and pathogenic disease potential of FLS isolated from adjuvant-induced arthritic (AA) rats via targeting IL-6/JAK1/STAT3 signaling cascade.


Asunto(s)
Artritis Reumatoide , Sinoviocitos , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Benzamidas , Proliferación Celular , Células Cultivadas , Femenino , Fibroblastos , Interleucina-6/metabolismo , Janus Quinasa 1/metabolismo , Factor de Crecimiento Placentario/metabolismo , Factor de Crecimiento Placentario/farmacología , Factor de Crecimiento Placentario/uso terapéutico , Pirimidinas , Ratas , Factor de Transcripción STAT3/metabolismo , Membrana Sinovial/patología
9.
Int Immunopharmacol ; 101(Pt B): 108359, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34863656

RESUMEN

Disturbed Th17/Treg balance is a critical pathological event in the disease progression of rheumatoid arthritis (RA). Recently, emerging studies have demonstrated that CD4 + T helper follicular (Tfh) cells exacerbates the pathogenic manifestations of RA. Contrarily, our previous report has shown that cyanidin, a flavonoid compound, attenuates disease severity of RA. Howbeit, this study investigated the therapeutic efficacy of cyanidin in relation to Th17/Treg balance and pathogenic Tfh cells in RA. Onto results, cyanidin inhibited increased Th17 cell differentiation and reciprocally improved FoxP3 + Treg cells both in-vivo and in-vitro. Concomitantly, cyanidin abated the detrimental effects of IL-17 via restoration of IL-10 secretion in adjuvant induced arthritic (AIA) rats. Furthermore, cyanidin reduced Tfh cells proportion and IgG levels in AIA rats, thus rectifying Tfh and follicular regulatory T (Tfr) cell ratio. Mechanistically, the restoring effect of cyanidin was associated with blunted activation of ROCK2/STAT3 signaling axis and reciprocal increase in the level of STAT-5 activity. Notwithstanding, cyanidin therapeutic efficacy correlated with specific oral ROCK2 inhibitor KD025 in-vitro. Collectively, these results demonstrate a dual promising therapeutic role of cyanidin via regulating Th17/Treg ratio and Tfh cell differentiation in an experimental model of RA.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Células T Auxiliares Foliculares , Linfocitos T Reguladores , Células Th17 , Quinasas Asociadas a rho/metabolismo , Animales , Antocianinas , Artritis Experimental/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Quinasas Asociadas a rho/genética
10.
3 Biotech ; 11(10): 436, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34549015

RESUMEN

Majoon Chobchini, a polyherbal Unani compound, has been used holistically in India to treat rheumatoid arthritis. However, the potential mechanism underlying the antiarthritic efficacy of Majoon Chobchini has not been elucidated so far. This study was aimed to explore the underlying molecular mechanism and scientifically validate the therapeutic basis of Majoon Chobchini in rheumatoid arthritis (RA). The anti-arthritic efficacy of Majoon Chobchini was demonstrated in vivo using complete Freund's adjuvant-induced arthritic rat model and adjuvant-induced arthritic fibroblast-like synoviocytes (AA-FLS). The expression of pro-inflammatory mediators and enzymes was evaluated in the serum and synovial tissues of adjuvant-induced arthritis (AIA) rats. In-vitro, AA-FLS, and bone marrow macrophages (BMMs) were co-cultured to evaluate the formation and activity of osteoclasts using TRAP staining analysis and pit formation assay, respectively. RANKL and OPG levels were detected using western blotting and qRT-PCR analysis. Furthermore, the involvement of JAK-STAT-3 signaling in the therapeutic efficacy of Majoon Chobchini was evaluated both in vivo and in vitro. Majoon Chobchini significantly reversed the physical symptoms in AIA rats with reduced expression of pro-inflammatory cytokines and enzymes. Notably, Majoon Chobchini alleviated cartilage degradation and bone erosion in AIA rats via inhibiting the activation of the JAK-STAT-3 signaling pathway in the AIA rats. Consistent with its effect in vivo, Majoon Chobchini decreased osteoclast inducing potential of AA-FLS and thus attenuated osteoclast formation and bone resorption in vitro. Taken together, our findings suggest that the JAK/STAT-3 signaling inhibition may underlie the mechanism through which Majoon Chobchini provides relief against RA symptoms. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s13205-021-02985-4.

11.
Eur J Med Chem ; 221: 113514, 2021 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-33992926

RESUMEN

While anti-inflammatory properties of isocoumarins are known their PDE4 inhibitory potential was not explored previously. In our effort the non-PDE4 inhibitor isocoumarins were transformed into the promising inhibitors via introducing an aminosulfonyl/aminocarboxamide moiety to the C-3 benzene ring attached to the isocoumarin framework. This new class of isocoumarins were synthesized via a PdCl2-catalyzed construction of the 4-allyl substituted 3-aryl isocoumarin ring starting from the appropriate 2-alkynyl benzamide derivative. Several compounds showed good inhibition of PDE4B in vitro and the SAR indicated superiority of aminosulfonamide moiety over aminocarboxamide in terms of PDE4B inhibition. Two compounds 3q and 3u with PDE4B IC50 = 0.43 ± 0.11 and 0.54 ± 0.19 µM and ≥ 2-fold selectivity over PDE4D emerged as initial hits. The participation of aminosulfonamide moiety in PDE4B inhibition and the reason for selectivity though moderate shown by 3q and 3u was revealed by the in silico docking studies. In view of potential usefulness of moderately selective PDE4B inhibitors the compound 3u (that showed PDE4 selectivity over other PDEs) was further evaluated in adjuvant induced arthritic rats. At an intraperitoneal dose of 30 mg/kg the compound showed a significant reduction in paw swelling (in a dose dependent manner), inflammation and pannus formation (in the knee joints) as well as pro-inflammatory gene expression/mRNA levels and increase in body weight. Moreover, besides its TNF-α inhibition and no significant toxicity in an MTT assay the compound did not show any adverse effects in a thorough toxicity studies e.g. teratogenicity, hepatotoxicity, cardiotoxicity and apoptosis in zebrafish. Thus, the isocoumarin 3u emerged as a new, safe and moderately selective PDE4B inhibitor could be useful for inflammatory diseases possibly including COVID-19.


Asunto(s)
Antiinflamatorios/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Isocumarinas/uso terapéutico , Inhibidores de Fosfodiesterasa 4/uso terapéutico , Sulfonamidas/uso terapéutico , Animales , Antiinflamatorios/síntesis química , Antiinflamatorios/metabolismo , Antiinflamatorios/toxicidad , Artritis Experimental/patología , Catálisis , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Embrión no Mamífero/efectos de los fármacos , Femenino , Isocumarinas/síntesis química , Isocumarinas/metabolismo , Isocumarinas/toxicidad , Articulación de la Rodilla/efectos de los fármacos , Articulación de la Rodilla/patología , Masculino , Ratones , Simulación del Acoplamiento Molecular , Estructura Molecular , Paladio/química , Inhibidores de Fosfodiesterasa 4/síntesis química , Inhibidores de Fosfodiesterasa 4/metabolismo , Inhibidores de Fosfodiesterasa 4/toxicidad , Unión Proteica , Células RAW 264.7 , Ratas Wistar , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/metabolismo , Sulfonamidas/toxicidad , Pez Cebra
12.
Cytokine ; 142: 155502, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33810944

RESUMEN

Interleukin (IL)-17A signaling pathway plays a critical role in the initiation and progression of rheumatoid arthritis (RA) and represents a viable target for RA therapy. Cyanidin, a flavonoid compound, is a novel inhibitor of IL-17A/IL-17RA (receptor subunit A) interaction in several inflammatory diseases. However, the therapeutic efficacy of cyanidin on IL-17A cytokine signaling induced monocyte migration and fibroblast-like synoviocytes (FLS) released RANKL mediated osteoclastogenesis in RA has not yet been deciphered. In the present study, cyanidin impeded IL-17A induced migration of monocytes isolated from adjuvant-induced arthritic (AA) rats. At the molecular level, cyanidin blocked the activation of p38MAPK signaling in response to IL-17A. Importantly, cyanidin downregulated IL-17A induced expression of HSP27, CXCR4, and CCR7 in AA monocytes via modulating IL-17/p38 MAPK signaling axis. Alternatively, cyanidin significantly suppressed the formation of matured osteoclasts and bone resorption in a coculture system consisting of IL-17 treated AA-FLS and rat bone marrow-derived monocytes/macrophages. Further, cyanidin significantly inhibited the expression of RANKL and increased the expression of OPG in AA-FLS via blunted activation of IL-17A/STAT-3 signaling cascade. Interestingly, cyanidin impaired IL-17A induced overexpression of IL-17RA. Taken together, our study proposes a novel therapeutic function of cyanidin towards targeted inhibition of IL-17A/IL-17RA signaling mediated disease severity and bone erosion in RA.


Asunto(s)
Antocianinas/farmacología , Artritis Reumatoide/patología , Diferenciación Celular , Interleucina-17/metabolismo , Monocitos/patología , Osteoclastos/patología , Transducción de Señal , Animales , Antocianinas/química , Artritis Experimental/patología , Artritis Reumatoide/complicaciones , Resorción Ósea/complicaciones , Resorción Ósea/patología , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Proteínas de Choque Térmico HSP27/metabolismo , Masculino , Ratones , Modelos Biológicos , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Osteoprotegerina/metabolismo , Fosforilación/efectos de los fármacos , Ligando RANK/metabolismo , Ratas Wistar , Receptores CCR7/metabolismo , Receptores CXCR4/metabolismo , Receptores de Interleucina-17/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Toxicol Appl Pharmacol ; 391: 114917, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-32044269

RESUMEN

The hyperplastic phenotype of fibroblast-like synoviocytes (FLSs) plays an important role for synovitis, chronic inflammation and joint destruction in rheumatoid arthritis (RA). Interleukin 17A (IL-17A), a signature pro-inflammatory cytokine effectively influences the hyperplastic transformation of FLS cells and synovial pannus growth. IL-17A cytokine signalling participates in RA pathology by regulating an array of pro-inflammatory mediators and osteoclastogenesis. Cyanidin, a key flavonoid inhibits IL-17A/IL-17 receptor A (IL-17RA) interaction and alleviates progression and disease severity of psoriasis and asthma. However, the therapeutic efficacy of cyanidin on IL-17A cytokine signalling in RA remains unknown. In the present study, cyanidin inhibited IL-17A induced migratory and proliferative capacity of FLS cells derived from adjuvant-induced arthritis (AA) rats. Cyanidin treatment reduced IL-17A mediated reprogramming of AA-FLS cells to overexpress IL-17RA. In addition, significantly decreased expression of IL-17A dependent cyr61, IL-23, GM-CSF, and TLR3 were observed in AA-FLS cells in response to cyanidin. At the molecular level, cyanidin modulated IL-17/IL-17RA dependent JAK/STAT-3 signalling in AA-FLS cells. Importantly, cyanidin activated PIAS3 protein to suppress STAT-3 specific transcriptional activation in AA-FLS cells. Cyanidin treatment to AA rats attenuated clinical symptoms, synovial pannus growth, immune cell infiltration, and bone erosion. Cyanidin reduced serum level of IL-23 and GM-CSF and expression of Cyr 61 and TLR3 in the synovial tissue of AA rats. Notably, the level of p-STAT-3 protein was significantly decreased in the synovial tissue of AA rats treated with cyanidin. This study provides the first evidence that cyanidin can be used as IL-17/17RA signalling targeting therapeutic drug for the treatment of RA and this need to be investigated in RA patients.


Asunto(s)
Antocianinas/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Fibroblastos/efectos de los fármacos , Interleucina-17/metabolismo , Transducción de Señal/efectos de los fármacos , Líquido Sinovial/efectos de los fármacos , Sinoviocitos/efectos de los fármacos , Animales , Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/patología , Movimiento Celular/efectos de los fármacos , Progresión de la Enfermedad , Fibroblastos/patología , Regulación de la Expresión Génica/efectos de los fármacos , Quinasas Janus/biosíntesis , Quinasas Janus/genética , Masculino , Ratas , Ratas Wistar , Factor de Transcripción STAT3/biosíntesis , Factor de Transcripción STAT3/genética , Membrana Sinovial/patología , Sinoviocitos/patología
14.
Arthritis Res Ther ; 22(1): 16, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31973752

RESUMEN

BACKGROUND: Proper blocking of toll-like receptor (TLR) activation during disease progression has been reported to have inhibitory effect on the pathogenesis of rheumatoid arthritis (RA). We tested whether the TLR4 inhibitor TAK-242 had potential as a remedy for rheumatoid arthritis. METHODS: The therapeutic effect of TAK-242 was tested in vitro using the human rheumatoid fibroblast-like synoviocyte (FLS) line MH7A or primary human FLS and in an adjuvant-induced arthritis (AIA) rat model. RESULTS: TAK-242 dose dependently inhibited the increased expression of IL-6, IL-8, MMP-1, and VEGF in LPS-stimulated MH7A cells. It also inhibited the expression of IL-6 and IL-8 in poly(I:C), TLR3 activator-stimulated primary FLS, but not in IL-1ß-stimulated primary FLS. These findings suggest that TAK-242 blocks a specific signaling pathway to some degree. Further, TAK-242 slightly inhibited mobilization of NF-κB into nuclei. In the AIA rat model, TAK-242 significantly reversed the body weight and paw thickness of AIA rats to the normal state at a dose of 5 mg/kg, but not at 3 mg/kg, and reduced the increased serum level of IL-6 and VEGF in AIA rats. It also significantly ameliorated inflammatory symptoms of joint tissues at day 21 of treatment, according to histology and RT-PCR. CONCLUSIONS: Based on the drug repositioning concept, TAK-242, which is used for the treatment of TLR4-mediated inflammatory diseases, shows potential for cost-effective development as a remedy for rheumatoid arthritis or to control the progression of RA.


Asunto(s)
Antirreumáticos/farmacología , Artritis Reumatoide , Sulfonamidas/farmacología , Sinoviocitos/efectos de los fármacos , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Células Cultivadas , Femenino , Humanos , Ratas , Ratas Wistar
15.
Cell Immunol ; 332: 39-50, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30029761

RESUMEN

Interleukin 17 (IL-17) and hypoxia have been implicated to play a key role in rheumatoid arthritis (RA). In this study, the combined treatment of IL-17 and cobalt chloride (CoCl2), a hypoxia mimetic significantly increased the osteoclast formation and the expression of TRAP and MMP-9 in RAW 264.7 macrophage cells in the presence of RANKL and M-CSF. The unified effect of IL-17 and CoCl2 markedly increased osteoclast mediated bone erosion through the activation of RANKL/NF-κB/NFATc1 signaling pathway. The treatment of IL-17 in combination with CoCl2 further potentiated the protein and mRNA expression of HIF-1α and MMP-9 in rat synovial macrophages. Conversely, the blockage of HIF-1α expression with BAY87-2243 abrogated the IL-17 and CoCl2 mediated expression of HIF-1α and MMP-9. Further, the knockdown of IL-17RA using siRNA reversed the IL-17 and CoCl2 induced expression of HIF-1α in synovial macrophages. In conclusion, IL-17 synergizes with CoCl2 induced hypoxic condition to augment osteoclast mediated bone erosion and synovial macrophages mediated RA pathogenesis.


Asunto(s)
Huesos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Interleucina-17/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Osteoclastos/metabolismo , Animales , Artritis Reumatoide/metabolismo , Células Cultivadas , Cobalto/metabolismo , Macrófagos/metabolismo , Ratones , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Ligando RANK/metabolismo , Células RAW 264.7 , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Interleucina-17/metabolismo , Transducción de Señal/fisiología , Membrana Sinovial/metabolismo , Fosfatasa Ácida Tartratorresistente/metabolismo
16.
Life Sci ; 207: 284-295, 2018 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-29908722

RESUMEN

AIMS: Bone erosion induced by enhanced osteoclast formation is a debilitating pathological phenomenon in rheumatoid arthritis (RA). Recent finding has revealed that ferulic acid is associated with reduced osteoclast differentiation and bone erosion. However, the underlying mechanism through which ferulic acid inhibited osteoclast differentiation and bone erosion still remains to be elucidated. This study assessed the therapeutic effects of ferulic acid on osteoclast differentiation and bone erosion by targeting RANKL dependent NF-κB pathway. MAIN METHODS: RAW 264.7 monocyte/macrophage cells were left untreated/treated with 25, 50 and 100 µM ferulic acid prior to stimulation with/without RANKL and M-CSF. Osteoclast differentiation and formation was assessed by SEM and TRAP analysis whereas its functional activity of bone erosion was determined by pit formation assay. Crucial transcription factors (NF-κBp-65, NFATc1 and c-Fos) and osteoclast specific genes (TRAP, MMP-9 and Cathepsin K) were evaluated by quantitative RT-PCR. Further, the protein level expression of NF-κBp-65, NFAtc1, c-Fos and MMP-9 was assessed using western blot analysis. KEY FINDINGS: Our results demonstrated that ferulic acid significantly attenuated RANKL induced osteoclast differentiation as evidenced from SEM and TRAP staining analysis. A remarkable decrease in the bone resorption activity of osteoclasts was also noticed upon ferulic acid treatment. In addition, the down-regulation of RANKL induced NF-κB activation and its associated downstream factors like NFATc1, c-Fos, TRAP, Cathepsin K and MMP-9 was also observed upon ferulic acid treatment. SIGNIFICANCE: Thus, our findings evidence the anti-stimulatory and anti-resorptive role of ferulic acid via the inhibition of RANKL dependent NF-κB signalling pathway.


Asunto(s)
Ácidos Cumáricos/farmacología , Subunidad p50 de NF-kappa B/metabolismo , Osteoclastos/citología , Polifenoles/farmacología , Ligando RANK/metabolismo , Animales , Artritis Reumatoide/terapia , Células de la Médula Ósea/citología , Resorción Ósea , Huesos/metabolismo , Huesos/fisiopatología , Catepsina K/metabolismo , Diferenciación Celular , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Macrófagos/citología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Monocitos/citología , Células RAW 264.7 , Ratas , Ratas Wistar , Transducción de Señal , Fosfatasa Ácida Tartratorresistente/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...