Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochem Pharmacol ; 220: 116006, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38142838

RESUMEN

Cancer immunotherapy, particularly with immune checkpoint inhibitors, has revolutionized the paradigm of cancer treatment. Nevertheless, the efficacy of cancer immunotherapy remains limited in most clinical settings due to the lack of a preexisting antitumor T-cell response in tumors. Therefore, the clinical outcomes of cancer immunotherapy must be improved crucially. With increased awareness of the importance of the innate immune response in the recruitment of T cells, as well as the onset and maintenance of the T cell response, great interest has been shown in activating the cGAS-STING signaling pathway to awaken the innate immune response, thereby orchestrating both innate and adaptive immune responses to induce tumor clearance. However, tumor cells have evolved to overexpress ectonucleotide pyrophosphate phosphodiesterase 1 (ENPP1), which degrades the immunotransmitter 2',3'-cGAMP and promotes the production of immune-suppressing adenosine, resulting in inhibition of the anticancer immune response in the tumor microenvironment. Clinically, ENPP1 overexpression is closely associated with poor prognosis in patients with cancer. Conversely, depleting or inhibiting ENPP1 has been verified to elevate extracellular 2',3'-cGAMP levels and inhibit the generation of adenosine, thereby reinvigorating the anticancer immune response for tumor elimination. A variety of ENPP1 inhibitors have recently been developed and have demonstrated significant promise for cancer immunotherapy. In this review, we provide an overview of ENPP1, dissect its immunosuppressive mechanisms, and discuss the development of ENPP1 inhibitors with the potential to further improve the efficacy of cancer immunotherapy.


Asunto(s)
Neoplasias , Hidrolasas Diéster Fosfóricas , Humanos , Adenosina , Difosfatos , Inmunoterapia , Neoplasias/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Inhibidores de Fosfodiesterasa/uso terapéutico , Hidrolasas Diéster Fosfóricas/metabolismo , Microambiente Tumoral
2.
Int Immunopharmacol ; 113(Pt A): 109304, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36252492

RESUMEN

Cancer immunotherapy with immune checkpoint inhibitors has achieved unprecedented success in cancer treatment; However, only a subset of patients achieved clinical benefit from this treatment, underscoring the urgent need to identify new strategies to enhance the clinical efficacy of immune checkpoint inhibitors. Given the essential role of innate immunity in cancer immune surveillance, tremendous effort has been focused on the innate immune pathways that can be pharmacologically modulated to improve the clinical outcome of checkpoint inhibitors. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway plays essential roles in host defense against cancers. Activation of the cGAS-STING signaling pathway induces the expression of type I interferons and proinflammatory cytokines, culminating in promotion of a robust adaptive antitumor immunity. As part of this innate immune signaling pathway, STING is ubiquitously expressed in immune and nonimmune cells. STING activation has been demonstrated to propagate the cancer immunity cycle, remodel the tumor microenvironment, and ultimately eliminate tumor cells. The immunomodulatory roles of STING enable it to be an appealing target for cancer immunotherapy. As such, STING agonists that are capable of triggering antitumor immune responses have been developed in recent years, and several of them have advanced into clinical trials. In this review, we first give an overview on the STING signaling pathway, then dissect the roles of STING activation in different steps of the cancer immunity cycle and finally discuss the development of STING agonists as well as challenges with STING activation, with the potential to make cancer immunotherapy with STING agonists more effective.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Neoplasias , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Proteínas de la Membrana/metabolismo , Inmunoterapia , Nucleotidiltransferasas/metabolismo , Neoplasias/patología , Inmunidad Innata , Microambiente Tumoral
3.
Mediators Inflamm ; 2020: 4346367, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33122966

RESUMEN

Atopic dermatitis (AD) is a relapsing, acute, and chronic skin disease featured by intractable itching, eczematous skin. Conventional therapies based on immunosuppression such as corticosteroids are associated with multiple adverse reactions. Periploca forrestii Schltr saponin (PFS) was shown to potently inhibit murine arthritis by protecting bone and cartilage injury and suppressing NF-κB activation. However, its therapeutic effect on oxazolone-induced atopic dermatitis (AD) and the underlying mechanisms on macrophage are still unclear. The AD-like dermatitis was induced by repeated oxazolone challenge to the skin of BALB/c mice in vivo. Blood and ears were biochemically or histologically processed. RT-PCR, western blotting, and ELISA were conducted to evaluate the expression of macrophage factors. Mouse bone marrow-derived macrophages (BMDMs) stimulated with lipopolysaccharide (LPS) were used as a model in vitro. PFS treatment inhibited AD-like dermatitis development. PFS downregulated epidermis thickness and cell infiltration, with histological analysis of the skin lesion. PFS alleviated plasma immunoglobulin (Ig) E, IgG2a, and IgG1 levels. PFS downregulated the expression of M1 macrophage factors, tumor necrosis factor- (TNF-) α, interleukin- (IL-) 6, monocyte chemotactic protein-1 (MCP-1), and nitric oxide synthase2 (NOS2), and M2 macrophage factors, IL-4, arginase1 (Arg1) and CD163 in AD-like skin, which were confirmed by western blot and ELISA analysis. In addition, PFS inhibited LPS-induced macrophage polarization via the inhibition of the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and nuclear translocation of NF-κB p65. These results suggest that PFS exerted an antidermatitis effect against oxazolone by modulating macrophage activation. PFS administration might be useful in the treatment of AD and inflammatory skin diseases.


Asunto(s)
Dermatitis Atópica/inducido químicamente , Dermatitis Atópica/tratamiento farmacológico , Oxazolona/toxicidad , Periploca/química , Saponinas/química , Saponinas/uso terapéutico , Animales , Supervivencia Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Activación de Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Células RAW 264.7 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
4.
Mediators Inflamm ; 2019: 5790291, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30906224

RESUMEN

BACKGROUND: The dry root of Flemingia philippinensis has been widely used in the treatment of rheumatism, arthropathy, and osteoporosis in traditional Chinese medicine; the therapeutic effects of Flemingia philippinensis are associated with antiarthritis in traditional Chinese medicine theory. This study was undertaken to investigate the mechanism of bone erosion protection and anti-inflammatory effect of Flemingia philippinensis flavonoids (FPF) in collagen-induced arthritis (CIA) in mice. METHODS: Flavonoids were extracted from the dry root of Flemingia philippinensis. Collagen-induced arthritis in C57BL/6 mice was used as a rheumatoid arthritis model, and the mice were orally fed with FPF prior to induction to mimic clinical prophylactic therapy for a total of 39 days. After treatment, histology and immunohistochemistry staining were performed, and the levels of anti-collagen type II (CII) antibody and inflammatory mediators, as well as the key proteins of nuclear factor kappa-B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways, were detected in the samples taken from ankle joints, plasma, and paws. RESULTS: FPF administration significantly suppressed the paw swelling and arthritic score in CIA mice. FPF reduced inflammatory infiltration and pannus formation, articular cartilage destruction and osteoclast infiltration, and the expression of MMP-9 and cathepsin K in the ankle joint. FPF inhibited plasma anti-CII antibody levels and the production of inflammatory cytokines and chemokines in CIA paws. FPF treatment suppressed the activation of NF-κB as indicated by downregulating the phosphorylation of NF-κB p65 and mitogen-activated protein kinases in CIA paws. Additionally, FPF significantly inhibited inflammation signaling by suppressing the activation of activator protein-1 subset and signal transducers and activators of transcription 3 (STAT3). CONCLUSIONS: Our data suggest that FPF might be an active therapeutic agent for rheumatoid arthritis and the preventive effect of FPF on arthritis is attributable to an anti-inflammatory effect on CIA by preventing bone destruction, regulating inflammatory mediators, and suppressing NF-κB and MAPK signaling pathways.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Fabaceae/química , Flavonoides/uso terapéutico , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Animales , Western Blotting , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos
5.
PLoS One ; 12(5): e0176672, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28463993

RESUMEN

OBJECTIVE: Periploca forrestii Schltr has been used as a Chinese folk medicine for the treatment of rheumatism, arthralgia and fractures. However, the anti-arthritic activity of Periploca forrestii saponin (PFS) and the active compound has still not been revealed. This study aimed to investigate the protective effects and mechanisms of PFS on collagen type II (CII) collagen-induced arthritis (CIA) mice. We sought to investigate whether PFS and Periplocin could regulate osteoclastogenesis, and if so, further investigation on its mechanism of action. METHODS: Arthritis was induced in female BALB/c mice by CIA method. PFS was administered at a dose of 50 mg/kg body weight once daily for five weeks. The effects of treatment in mice were assessed by histological and biochemical evaluation in sera and paws. Anti-osteoclastogenic action of PFS and Periplocin was identified using an osteoclast formation model induced by RANKL. RESULTS: PFS ameliorated paw erythema and swelling, inhibited bone erosion in ankle joint histopathological examination. PFS treatment resulted in decreased IgG2a, and increased IgG1 levels in the serum of CIA mice. Decreased TNF-α, and increased interleukin (IL)-4 and IL-22 levels were also found in PFS-treated mice. PFS inhibited the I-κBα phosphorylation, blocked nuclear factor (NF)-κB/p65 phosphorylation and abrogated AP-1/c-Fos activity. PFS downregulated toll-like receptor (TLR) 4, STAT3 and MMP-9 expression in CIA mice and RANKL-induced osteoclastogenesis. PFS and Periplocin inhibited RANKL-induced osteoclast formation in a dose dependent manner within nongrowth inhibitory concentration, and PFS decreased osteoclastogenesis-related marker expression, including cathepsin K and MMP-9. CONCLUSION: This study revealed that the protective mechanism of PFS on CIA was associated with regulatory effects on proinflammatory factors and further on the crosstalk between NF-κB and c-Fos/AP-1 in vivo and in vitro. Therefore, PFS is a promising therapeutic alternative for the treatment of RA, evidencing the need to conduct further studies that can identify their active components in treating and preventing RA.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Periploca , Fitoterapia , Saponinas/farmacología , Animales , Artritis Experimental/patología , Células Cultivadas , Citocinas/metabolismo , Femenino , Humanos , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Osteoclastos/efectos de los fármacos , Ligando RANK , Distribución Aleatoria , Proteínas Recombinantes , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...