Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38602626

RESUMEN

Psoriasis is a paradigmatic condition characterised by a heightened autoimmune response and chronic inflammation. However, the exact nature and the pathological causes behind it are still unknown. Growing evidence suggest dysregulated cytokine network as a result of over-activated T cells and plasmacytoid dendritic cells (pDCs) as the critical drivers in the development of psoriasis. In the present study, we aimed to investigate the therapeutic efficacy of 3,3'-diindolylmethane (DIM) on pDC activation and Th17 cell development in imiquimod (IMQ)-induced psoriasis mice. Our in vitro research investigated the IRF-7 signalling in pDCs that explained the reduced expression of the transcription factor IRF-7 responsible for pDC activation as a result of DIM treatment. Concurrently, DIM treatment decreased the release of Th17 cell polarising cytokines (IFN-α, IL-23, and IL-6) by pDCs which validated a reduction in differentiated pathogenic Th17 cell population and associated cytokine IL-17A in IMQ-induced psoriatic mice. Thus, our recent findings provide therapeutic evidence in targeting the early potential contributors for psoriasis treatment by preventing IRF-7-mediated pDC activation and Th17 cell development in IMQ-induced psoriasis mice.

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.
Immunol Invest ; 53(2): 115-159, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38054436

RESUMEN

Resistin, a cytokine hormone predominantly secreted by adipose tissue, is elevated in various metabolic disorders such as obesity, type 2 diabetes, and cardiovascular disease. In addition to its involvement in metabolic regulation, resistin has been implicated in the pathogenesis of psoriasis, a chronic inflammatory skin disorder. Numerous studies have reported increased resistin levels in psoriatic skin lesions, suggesting a possible association between resistin and psoriasis. Recent studies have suggested the potential involvement of resistin in the development and progression of certain cancers. Resistin is overexpressed in breast, colorectal, and gastric cancers. This suggests that it may play a role in the development of these cancers, possibly by inducing inflammation and cell growth. The link between resistin and cancer raises the possibility of shared underlying mechanisms driving the pathogenesis of psoriasis. Chronic inflammation, one such mechanism, is a hallmark of psoriasis and cancer. Further research is needed to fully understand the relationship between resistin and psoriasis. Identifying potential therapeutic targets is crucial for effective management of psoriasis. By doing so, we may be able to develop more effective treatment options for individuals living with psoriasis and ultimately improve their quality of life. Ultimately, a more comprehensive understanding of the mechanisms underlying the impact of resistin on psoriasis is essential for advancing our knowledge and finding new ways to treat and manage this challenging condition.


Asunto(s)
Psoriasis , Resistina , Humanos , Diabetes Mellitus Tipo 2/complicaciones , Inflamación , Neoplasias , Calidad de Vida , Resistina/fisiología
4.
In Vitro Cell Dev Biol Anim ; 59(10): 811-820, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38032403

RESUMEN

The synovial intimal lining is mainly governed by fibroblast-like synoviocytes (FLS), which portray a transformed tumor-like phenotype in rheumatoid arthritis (RA). Among the diverse cytokines that engender FLS, interleukin-21 (IL-21) was reported to stimulate hyperproliferation and perpetuate inflammation. Recently, choline kinase (ChoKα) has been reported to be an essential enzyme aiding RA-FLS hyperproliferation by altering phosphatidylcholine biosynthesis. The current study aimed to elucidate the therapeutic efficacy of myricetin, a flavonoid, in abating the IL-21-induced tumor-like phenotype of adjuvant-induced arthritis (AIA)-FLS via the ChoKα signaling cascade. Our results showed that myricetin suppressed IL-21 receptor expression and activation of the ChoKα signaling cascade (N-Ras, Ral-GDS, and PI3K) in IL-21-induced AIA-FLS. Consequently, myricetin treatment decreased ChoKα and PLD2 enzymatic activity and inhibited the proliferative, migratory, and invasive properties of AIA-FLSs. Our results demonstrated that myricetin could be a promising anti-arthritic compound by abating IL-21-induced hyperproliferation, migration, and invasive behavior of AIA-FLS by downregulating the ChoKα signaling cascade.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Neoplasias , Sinoviocitos , Animales , Sinoviocitos/metabolismo , Membrana Sinovial/metabolismo , Colina Quinasa/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Flavonoides/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Fibroblastos/metabolismo , Células Cultivadas
5.
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
6.
Bioorg Chem ; 138: 106606, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37210826

RESUMEN

The 12R-lipoxygenase (12R-LOX), a (non-heme) iron-containing metalloenzyme belonging to the lipoxygenase (LOX) family catalyzes the conversion of arachidonic acid (AA) to its key metabolites. Studies suggested that 12R-LOX plays a critical role in immune modulation for the maintenance of skin homeostasis and therefore can be considered as a potential drug target for psoriasis and other skin related inflammatory diseases. However, unlike 12-LOX (or 12S-LOX) the enzyme 12R-LOX did not receive much attention till date. In our effort, the 2-aryl quinoline derivatives were designed, synthesized and evaluated for the identification of potential inhibitors of 12R-hLOX. The merit of selection of 2-aryl quinolines was assessed by in silico docking studies of a representative compound (4a) using the homology model of 12R-LOX. Indeed, in addition to participating in H-bonding with THR628 and LEU635 the molecule formed a hydrophobic interaction with VAL631. The desired 2-aryl quinolines were synthesized either via the Claisen-Schmidt condensation followed by one-pot reduction-cyclization or via the AlCl3 induced heteroarylation or via the O-alkylation approach in good to high (82-95%) yield. When screened against human 12R-LOX (12R-hLOX) in vitro four compounds (e.g. 4a, 4d, 4e and 7b) showed encouraging (>45%) inhibition at 100 µM among which 7b and 4a emerged as the initial hits. Both the compounds showed selectivity towards 12R-hLOX over 12S-hLOX, 15-hLOX and 15-hLOXB and concentration dependent inhibition of 12R-hLOX with IC50 = 12.48 ± 2.06 and 28.25 ± 1.63 µM, respectively. The selectivity of 4a and 7b towards 12R-LOX over 12S-LOX was rationalized with the help of molecular dynamics simulations. The SAR (Structure-Activity Relationship) within the present series of compounds suggested the need of a o-hydroxyl group on the C-2 phenyl ring for the activity. The compound 4a and 7b (at 10 and 20 µM) reduced the hyper-proliferative state and colony forming potential of IMQ-induced psoriatic keratinocytes in a concentration dependent manner. Further, both compounds decreased the protein levels of Ki67 and the mRNA expression of IL-17A in the IMQ-induced psoriatic-like keratinocytes. Notably, 4a but not 7b inhibited the production of IL-6 and TNF-α in the keratinocyte cells. In the preliminary toxicity studies (i.e. teratogenicity, hepatotoxicity and heart rate assays) in zebrafish both the compounds showed low safety (<30 µM) margin. Overall, being the first identified inhibitors of 12R-LOX both 4a and 7b deserve further investigations.


Asunto(s)
Quinolinas , Pez Cebra , Animales , Humanos , Pez Cebra/metabolismo , Araquidonato 12-Lipooxigenasa/metabolismo , Piel/metabolismo , Quinolinas/farmacología , Relación Estructura-Actividad , Inhibidores de la Lipooxigenasa/farmacología , Simulación del Acoplamiento Molecular
7.
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
8.
Life Sci ; 309: 121031, 2022 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-36206833

RESUMEN

Choline kinase (ChoK) has been well documented as a major enzyme involved in the anomalous cellular lipid metabolic profile of chronic inflammatory disorders. However, new research has now been unveiled that helps us to better understand how changes in lipid metabolism influence the transformational phenotype, drug resistance, and antiapoptotic characteristics of invasive cells, leading to rheumatoid arthritis (RA) disease progression. It is still unknown how ChoK modulates the lipid metabolic aberrations that may promote altered cell phenotype and functionality in RA. Herein, we review the current understanding of ChoK's role in altered metabolism in diverse cell types involved in RA progression, and for the first time, we take a step forward to complete the puzzle and summarise striking facts that link choline metabolism to its transformed phenotype, in order to postulate ChoK as a robust therapeutic target in RA. This review forms a foundation on which ChoK can be tackled as a potential biomarker, opening doors for RA diagnosis and prognosis. It frameworks several ChoK inhibitors that rewire the lipid metabolic profile in the inflammatory disease landscape and envisages its being translated to clinics.


Asunto(s)
Artritis Reumatoide , Colina Quinasa , Humanos , Colina Quinasa/genética , Colina Quinasa/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Colina/metabolismo , Apoptosis , Lípidos , Membrana Sinovial/metabolismo
9.
Life Sci ; 298: 120516, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35367240

RESUMEN

Interleukin 6 (IL-6), a pleiotropic inflammatory cytokine, is produced transiently due to tissue damage and infections. Nonetheless, IL-6 contributes to the host regenerative defense mechanism via classical signaling at the basal physiological level. Although tightly regulated transcriptional and post-transcriptional mechanism modulates its expression, dysregulated continual production of IL-6 during inflammatory conditions negatively affects immune cells. Molecular evidence has substantiated the pernicious out-turn of IL-6 trans-signaling in developing one such autoimmune joint disorder, rheumatoid arthritis (RA). Significantly increased levels of IL-6 in RA, along with multiple growth factors mainly released by synovial-like fibroblasts (FLS) and macrophages, is crucial for clinical disease progression. Due to its pathogenicity, in mediating inflammation and context-driven signaling cassette, blockade of IL-6 could be a potent target in the therapeutic intervention of RA. The clinical trials of various humanized IL-6 and anti-IL-6 receptor antibodies have proved their efficacy. However, severe side effects like neutropenia, thrombocytopenia, and abnormal liver enzymes contributed to dysfunctional adaptive immunity. The JAK-STAT pathway has been majorly implicated in RA disease progression upon IL-6 stimulation, simultaneously paving the path for innovative therapeutic approaches. JAK inhibitors, namely Tofacitinib, Baricitinib, Decernotinib, Upadacitinib, Peficitinib, and Filgotinib, have demonstrated clinical efficacy in recent decades as an alternative therapeutic strategy to abrogate IL-6 mediated aberrant activity in RA. This approach substitutes for the side effects incurred due to the IL-6 targeted therapies. This review discusses the history of research into IL-6 biology and therapies that target the IL-6 driven JAK/STAT pathway, including the successes, challenges, and drawbacks, emphasizing RA.


Asunto(s)
Artritis Reumatoide , Interleucina-6 , Artritis Reumatoide/metabolismo , Biología , Progresión de la Enfermedad , Fibroblastos/metabolismo , Humanos , Interleucina-6/metabolismo , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Membrana Sinovial/metabolismo
10.
Int Immunopharmacol ; 109: 108795, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35487087

RESUMEN

Psoriasis is a highly inflammatory autoimmune-mediated skin disease. The strongest evidence has pointed to the influential role of interleukin (IL) -17A in the aberrant pathology of psoriasis. Henceforth, targeting the IL-17A cytokine is of prime importance in controlling the disease severity of psoriasis. Reportedly, 3, 3'- diindolylmethane (DIM) is a phytochemical that alleviated acute atopic dermatitis. Howbeit, the therapeutic intervention of DIM against IL-17A/IL-17RA interaction and its signaling mediated pathogenesis in psoriasis remains unexplored. In the current report, we decoded the molecular basis of DIM in psoriasis. Docking analysis has reported that DIM identified an IL-17A binding region in the functional fibronectin-III-like domain of IL-17RA and abrogated IL-17A/IL-17RA interaction. In-vitro experiments demonstrated that DIM impeded IL-17A mediated hyper-proliferative phenotype of psoriatic-like keratinocytes. Furthermore, DIM abated the catabolic effects of IL-17A stimulated expression of pathogenic mediators like HMGB-1, Cyr-61, CCL-20, and VEGF via blunted activation of JAK/STAT pathway in psoriatic like keratinocytes. Profoundly, DIM restricted the reprogramming of psoriatic-like keratinocytes to overexpress IL-17RA in concert with IL-17A stimulation. In line with in-vitro studies, DIM also ameliorated skin lesions and epidermal hyperplasia in an imiquimod-induced mice model of psoriasis. Additionally, DIM also reduced STAT-3 phosphorylation and associated expression of Cyr-61, CCL-20, and VEGF in psoriatic mice. However, if DIM has a direct effect on STAT-3 inhibition or it negatively regulates STAT-3 function via blockade of IL-17A/IL-17RA interaction needs to be investigated in the future. Conclusively, our studies demonstrated that the blockade of IL-17A/IL-17RA interaction is a novel therapeutic perspective of DIM against the progression of psoriasis disease.


Asunto(s)
Dermatitis , Psoriasis , Animales , Dermatitis/patología , Modelos Animales de Enfermedad , Imiquimod/farmacología , Indoles , Interleucina-17/metabolismo , Quinasas Janus/metabolismo , Queratinocitos , Ratones , Psoriasis/inducido químicamente , Psoriasis/tratamiento farmacológico , Psoriasis/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Piel/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
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
12.
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
13.
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
14.
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
15.
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.

16.
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
17.
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
18.
Inflammation ; 44(1): 229-242, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32876895

RESUMEN

Inflammation is a complex biological process which alters the normal physiological function of the immune system resulting in an abnormal microenvironment that leads to several clinical complications. The process of inflammation is mediated through various intracellular signaling factors inside the cells. Apoptosis signal-regulating kinase 1 (ASK1) is an inflammation-derived kinase that controls the activation of other family of kinases such as p38 mitogen-activated protein kinases (p38 MAPKs), which mediates various the inflammatory processes. In this study, we cultured THP-1 macrophage cells to undergo inflammatory proliferation with LPS (1 µg/ml) and TNFα (10 ng/ml) stimulation. Initial in silico analysis was utilized to predict novel microRNAs (miRNAs) that target ASK1 signaling and its expression levels in LPS and TNFα stimulated THP-1 cells were estimated. Among the miRNAs, miR-532-3p showcased the highest binding affinity towards ASK1 kinase. We witnessed that transient transfection of miR-532-3p diminished the levels of ASK1 and downstream phosphorylation/translocation of p38 MAPK. Furthermore, direct targeting of ASK1 resulted in regulation of uncontrolled release of cytokines (TNFα, IL-6, and IL-23) and chemokines (GM-CSF and MIP-2α). Overall, we suggest that miR-532-3p attenuates the pro-inflammatory nature of macrophages by targeting ASK1/p38 MAPK signaling pathway and can be used as a molecular intervention for treating inflammatory diseases.


Asunto(s)
Mediadores de Inflamación/metabolismo , MAP Quinasa Quinasa Quinasa 5/biosíntesis , Sistema de Señalización de MAP Quinasas/fisiología , Macrófagos/metabolismo , MicroARNs/biosíntesis , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Células THP-1
19.
J Cell Physiol ; 235(12): 9497-9509, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32372426

RESUMEN

Bone erosion is the major cause of deformities in autoimmune disease conditions such as osteoporosis and rheumatoid arthritis. Aberrant receptor activator of nuclear factor kappa B ligand (RANKL) secretion in bone disorders have been implicated to promote uncontrolled osteoclast differentiation through the regulation of nuclear factor of activated T cells 1 (NFATc1) transcription factor. This phenomenon is governed by several molecular factors including microRNAs, which are under-expressed during disease progression. This report focuses on elucidating the molecular mechanism of miR-506-3p towards the RANKL/NFATc1 pathway. miR-506-3p showed high binding affinity towards NFATc1 (ΔG = -22.4 kcal/mol). Bone marrow-derived macrophages (BMMs) isolated from rats stimulated with RANKL (100 ng/ml) showed active expression of NFATc1 which differentiated into mature osteoclasts. Moreover, NFATc1 activation resulted in downstream secretion of various bone resorptive enzymes (cathepsin K, carbonic anhydrase II, tartarate acid phosphatase, and matrix metalloproteinase 9) which lead to active bone resorption. However, transfection of miR-506-3p resulted in selective repression of NFATc1 inside the cells. This further resulted in the diminished release of bone resorptive enzymes that were essential for the degradation of the bone. Overall, we predict that miR-506-3p can be used as a molecular intervention for RANKL/NFATc1 mediated osteoclastogenesis.


Asunto(s)
MicroARNs/genética , Osteogénesis/genética , Ligando RANK/genética , Factores de Transcripción/genética , Animales , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Macrófagos/metabolismo , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Ligando RANK/farmacología , Ratas , Transducción de Señal/genética
20.
Int Immunopharmacol ; 82: 106328, 2020 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-32088641

RESUMEN

Fibroblast-like synoviocytes (FLS) lining the arthritic synovial joint region have been implicated to be a key player in bone remodeling. The uncontrolled proliferation of this cell subtype is strictly regulated by various molecular elements including microRNAs (miRNAs). The Wnt1/ß-catenin signaling pathway plays a crucial role in the survival of FLS cells. This study explores the underlying mechanism of miR-145-5p towards the Wnt1/ß-catenin pathway. MiR-145-5p depicted a strong binding affinity towards frizzled class receptor 4 (FZD4) 3' UTR, a key receptor complex essential for recognizing circulating Wnt1 molecules. Adjuvant induced arthritic fibroblast-like synoviocytes (AA-FLS) isolated from rats stimulated with Wnt1 (10 ng/ml) elicited active Wnt1/ß-catenin signaling. Transfection of miR-145-5p mimic (50 pmol) to AA-FLS stimulated with Wnt1 elicited reduced expression levels of various factors of Wnt1/ß-catenin signaling including low-density lipoprotein receptor-related protein 5 (LRP5), dishevelled segment polarity protein 1 (Dvl1) and ß-catenin transcription factor. Moreover, pro-inflammatory cytokines (TNFα, IL-1ß, IL-6 and IL-23) were regulated compared to the diseased groups. Furthermore, miR-145-5p counterbalanced the levels of receptor activator of nuclear factor kappa B ligand (RANKL) and osteoprotegerin (OPG) at the cellular level, essential for bone remodeling. Hence, we suggest that miR-145-5p regulates the survival/proliferation of FLS cells in RA disease condition through attenuation of Wnt1/ß-catenin signaling.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA