Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 191
Filtrar
1.
Mol Pain ; 20: 17448069241252384, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38631843

RESUMO

PD-1/PD-L1 inhibitors have been demonstrated to induce itch in both humans and experimental animals. However, whether the PD-1/PD-L1 pathway is involved in the regulation of chronic psoriatic itch remains unclear. This study aimed to investigate the role of the PD-1/PD-L1 pathway in imiquimod-induced chronic psoriatic itch. The intradermal injection of PD-L1 in the nape of neck significantly alleviated chronic psoriatic itch in imiquimod-treated skin. Additionally, we observed that spontaneous scratching behavior induced by imiquimod disappeared on day 21. Still, intradermal injection of PD-1/PD-L1 inhibitors could induce more spontaneous scratching for over a month, indicating that imiquimod-treated skin remained in an itch sensitization state after the spontaneous scratching behavior disappeared. During this period, there was a significant increase in PD-1 receptor expression in both the imiquimod-treated skin and the spinal dorsal horn in mice, accompanied by significant activation of microglia in the spinal dorsal horn. These findings suggest the potential involvement of the peripheral and central PD-1/PD-L1 pathways in regulating chronic itch and itch sensitization induced by imiquimod.


Assuntos
Antígeno B7-H1 , Imiquimode , Receptor de Morte Celular Programada 1 , Prurido , Psoríase , Animais , Imiquimode/farmacologia , Imiquimode/efeitos adversos , Prurido/induzido quimicamente , Prurido/metabolismo , Psoríase/induzido quimicamente , Psoríase/complicações , Psoríase/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Antígeno B7-H1/metabolismo , Masculino , Camundongos , Transdução de Sinais/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Camundongos Endogâmicos C57BL , Doença Crônica
2.
J Transl Med ; 22(1): 341, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38594751

RESUMO

BACKGROUND: Chemoimmunotherapy has shown promising advantages of eliciting immunogenic cell death and activating anti-tumor immune responses. However, the systemic toxicity of chemotherapy and tumor immunosuppressive microenvironment limit the clinical application. METHODS: Here, an injectable sodium alginate hydrogel (ALG) loaded with nanoparticle albumin-bound-paclitaxel (Nab-PTX) and an immunostimulating agent R837 was developed for local administration. Two murine hepatocellular carcinoma and breast cancer models were established. The tumor-bearing mice received the peritumoral injection of R837/Nab-PTX/ALG once a week for two weeks. The antitumor efficacy, the immune response, and the tumor microenvironment were investigated. RESULTS: This chemoimmunotherapy hydrogel with sustained-release character was proven to have significant effects on killing tumor cells and inhibiting tumor growth. Peritumoral injection of our hydrogel caused little harm to normal organs and triggered a potent antitumor immune response against both hepatocellular carcinoma and breast cancer. In the tumor microenvironment, enhanced immunogenic cell death induced by the combination of Nab-PTX and R837 resulted in 3.30-fold infiltration of effector memory T cells and upregulation of 20 biological processes related to immune responses. CONCLUSIONS: Our strategy provides a novel insight into the combination of chemotherapy and immunotherapy and has the potential for clinical translation.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas , Camundongos , Animais , Hidrogéis/farmacologia , Hidrogéis/uso terapêutico , Imiquimode/farmacologia , Imiquimode/uso terapêutico , Morte Celular Imunogênica , Linhagem Celular Tumoral , Neoplasias Hepáticas/tratamento farmacológico , Imunoterapia/métodos , Imunidade , Microambiente Tumoral
3.
Int Immunopharmacol ; 130: 111805, 2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38457930

RESUMO

OBJECTIVE: To elucidate the mechanism of Pentraxin 3 (PTX3) in the pathogenesis of psoriasiform dermatitis using Ptx3-knockout (Ptx3-KO) background mice. METHODS: An Imiquimod (IMQ)-induced murine psoriatic model was created using Ptx3-KO (Ptx3-/-) and wild-type (Ptx3+/+) mice. Skin lesion severity and expression of inflammatory mediators (IL-6 and TNFα) were assessed using PASI score and ELISA, respectively. Cutaneous tissues from the two mice groups were subjected to histological analyses, including HE staining, Masson staining, and Immunohistochemistry (IHC). The PTX3, iNOS, COX2, and Arg1 expressions were quantified and compared between the two groups. We used RNA-seq to clarify the underlying mechanisms of the disease. Flow cytometry was used to analyze systemic Th17 cell differentiation and macrophage polarization. RESULT: The psoriatic region exhibited a higher PTX3 expression than the normal cutaneous area. Moreover, PTX3 was upregulated in HaCaT cells post-TNFα stimulation. Upon IMQ stimulation, Ptx3-/- mice displayed a lower degree of the psoriasiform dermatitis phenotype compared to Ptx3+/+ mice. Consistent with the RNA-seq results, further experiments confirmed that compared to the wild-type group, the PTX3-KO group exhibited a generally lower IL-6, TNFα, iNOS, and COX2 expression and a contrasting trend in macrophage polarization. However, no significant difference in Th17 cell activation was observed between the two groups. CONCLUSIONS: This study revealed that PTX3 was upregulated in psoriatic skin tissues and TNFα-stimulated HaCaT cells. We also discovered that PTX3 deficiency in mice ameliorated the psoriasiform dermatitis phenotype upon IMQ stimulation. Mechanistically, PTX3 exacerbates psoriasiform dermatitis by regulating macrophage polarization rather than Th17 cell differentiation.


Assuntos
Proteína C-Reativa , Dermatite , Psoríase , Componente Amiloide P Sérico , Animais , Camundongos , Proteína C-Reativa/genética , Proteína C-Reativa/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dermatite/metabolismo , Dermatite/patologia , Modelos Animais de Doenças , Imiquimode/farmacologia , Interleucina-6/metabolismo , Macrófagos/patologia , Psoríase/metabolismo , Psoríase/patologia , Componente Amiloide P Sérico/genética , Componente Amiloide P Sérico/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Humanos , Progressão da Doença , Camundongos Knockout , Camundongos Endogâmicos C57BL
4.
Int Immunopharmacol ; 131: 111824, 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38461633

RESUMO

BACKGROUND: Psoriasis is an inflammatory skin disease that occurs repeatedly over time. The natural product of sesquiterpene lactones, Parthenolide (Par), is isolated from Tanacetum parthenium L. (feverfew) which has significant effects on anti-inflammatory. The therapeutic effect of the medication itself is crucial, but different routes of administration of the same drug can also produce different effects. PURPOSE: The aim of our research sought to investigate the ameliorating effects of Par in psoriasis-like skin inflammation and its related mechanism of action. RESULTS: In the IMQ-induced model, intragastric administration of Par reduced the Psoriasis Area and Severity Index (PASI) score, improved skin erythema, scaling, and other symptoms. And Par decreased the expression of Ki67, keratin14, keratin16 and keratin17, and increased the expression of keratin1. Par could reduce IL-36 protein expressions, meanwhile the expression of Il1b, Cxcl1 and Cxcl2 mRNA were also decreased. Par regulated the expression levels of F4/80, MPO and NE. However, skin transdermal administration of Par was more effective. Similarly, Par attenuated IL-36γ, IL-1ß and caspase-1 activated by Poly(I:C) in in vitro and ex vivo. In addition, Par also reduced NE, PR3, and Cathepsin G levels in explant skin tissues. CONCLUSION: Par ameliorated psoriasis-like skin inflammation in both in vivo and in vitro, especially after treatment with transdermal drug delivery, possibly by inhibiting neutrophil extracellular traps and thus by interfering IL-36 signaling pathway. It indicated that Par provides a new research strategy for the treatment of psoriasis-like skin inflammation and is expected to be a promising drug.


Assuntos
Dermatite , Armadilhas Extracelulares , Psoríase , Sesquiterpenos , Animais , Camundongos , Imiquimode/farmacologia , Administração Cutânea , Armadilhas Extracelulares/metabolismo , Pele , Psoríase/induzido quimicamente , Psoríase/tratamento farmacológico , Psoríase/metabolismo , Sesquiterpenos/uso terapêutico , Sesquiterpenos/farmacologia , Dermatite/tratamento farmacológico , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Modelos Animais de Doenças , Camundongos Endogâmicos BALB C
5.
Int Immunopharmacol ; 129: 111652, 2024 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-38335657

RESUMO

Psoriasis is a chronic, autoimmune skin disease characterized by the deregulated secretion of inflammatory factors in multiple organs. The aberrant activation of signal transducer and activator of transcription 3 (STAT3) signaling pathway mediated by cyclin-dependent kinase 9 (CDK9) is vital for the pathology of psoriasis, leading to the accumulation of inflammatory factors and the progression of skin damage. In this study, we explored the effect of CDK9 inhibition on attenuating the secretion of inflammatory factors and alleviating skin damage in psoriasis models both in vitro and in vivo. Results showed that Atuveciclib, a highly selective CDK9 inhibitor, significantly relieved skin lesions in Imiquimod (IMQ)-induced mice models by lowering the expression of CDK9 and p-RNA Pol II Ser2. Meanwhile, Atuveciclib significantly inhibited STAT3 phosphorylation in mice skin and reduced the levels of key inflammatory cytokines in mice skin, plasma and spleen. In addition to suppressing the secretion of inflammatory cytokines, Atuveciclib ablated the activation of STAT3 induced by tumor necrosis factor-α (TNF-α)/interferon-γ (IFN-γ). Overall, our findings indicated that the overexpression and hyperfunction of CDK9 promote the progression of psoriasis. Moreover, Atuveciclib interfered with the abnormal STAT3 signaling pathway through the inhibition of CDK9, which ultimately ameliorated psoriatic-like skin inflammation. These suggested that CDK9 inhibition is a potential strategy for batting psoriasis.


Assuntos
Dermatite , Psoríase , Dermatopatias , Sulfonamidas , Triazinas , Animais , Camundongos , Imiquimode/farmacologia , Fator de Transcrição STAT3/metabolismo , Quinase 9 Dependente de Ciclina , Psoríase/induzido quimicamente , Psoríase/tratamento farmacológico , Inflamação/induzido quimicamente , Transdução de Sinais , Citocinas/metabolismo
6.
Physiol Rep ; 12(3): e15949, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38346802

RESUMO

Toll-like receptor-7 (TLR7) activation promotes autoimmunity, and metabolic syndrome (MetS) is a common comorbidity in patients with autoimmune disease. We previously demonstrated hyperinsulinemia in TLR7 agonist imiquimod (IMQ)-treated, high-fat diet (HFD)-fed female C57BL/6 mice. Since mouse strains differ in susceptibility to MetS and target organ damage, this study investigated whether 12 weeks of exposure to HFD and IMQ promoted MetS, autoimmunity, and target organ damage in female FVB/N mice. Supporting early-stage autoimmunity, spleen-to-tibia ratio, and anti-nuclear antibodies (ANA) were significantly increased by IMQ. No significant effect of IMQ on urinary albumin excretion or left ventricular hypertrophy was observed. HFD increased liver-to-tibia ratio, which was further exacerbated by IMQ. HFD increased fasting blood glucose levels at the end of 12 weeks, but there was no significant effect of IMQ treatment on fasting blood glucose levels at 6 or 12 weeks of treatment. However, oral glucose tolerance testing at 12 weeks revealed impaired glucose tolerance in HFD-fed mice compared to control diet mice together with IMQ treatment exacerbating the impairment. Accordingly, these data suggest TLR7 activation also exacerbates HFD-induced dysregulation of glucose handling FVB/N mice, supporting the possibility that endogenous TLR7 activation may contribute to dysglycemia in patients with autoimmune disease.


Assuntos
Doenças Autoimunes , Síndrome Metabólica , Humanos , Feminino , Camundongos , Animais , Imiquimode/farmacologia , Dieta Hiperlipídica/efeitos adversos , Glicemia/metabolismo , Receptor 7 Toll-Like/metabolismo , Controle Glicêmico , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos
7.
ACS Appl Mater Interfaces ; 16(7): 8403-8416, 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38334116

RESUMO

Cancer immunotherapy is expected to achieve tumor treatment mainly by stimulating the patient's own immune system to kill tumor cells. However, the low immunogenicity of the tumor and the poor efficiency of tumor antigen presentation result in a variety of solid tumors that do not respond to immunotherapy. Herein, we designed a proton-gradient-driven porphyrin-based liposome (PBL) with highly efficient Toll-like receptor 7 (TLR7) agonist (imiquimod, R837) encapsulation (R837@PBL). R837@PBL rapidly released R837 in the acid microenvironment to activate the TLR in the endosome inner membrane to promote bone-marrow-derived dendritic cell maturation and enhance antigen presentation. R837@PBL upon laser irradiation triggered immunogenic cell death of tumor cells and tumor-associated antigen release after subcutaneous injection, activated TLR7, formed in situ tumor nanoadjuvants, and enhanced the antigen presentation efficiency. Photoimmunotherapy promoted the infiltration of cytotoxic T lymphocytes into tumor tissues, inhibited the growth of the treated and abscopal tumors, and exerted highly effective photoimmunotherapeutic effects. Hence, our designed in situ tumor nanoadjuvants are expected to be an effective treatment for treated and abscopal tumors, providing a novel approach for synergistic photoimmunotherapy of tumors.


Assuntos
Neoplasias , Porfirinas , Humanos , Imiquimode/farmacologia , Lipossomos/farmacologia , Receptor 7 Toll-Like/agonistas , Prótons , Porfirinas/farmacologia , Neoplasias/terapia , Imunoterapia , Adjuvantes Imunológicos/farmacologia , Antígenos de Neoplasias , Microambiente Tumoral , Linhagem Celular Tumoral
8.
Adv Healthc Mater ; 13(9): e2303305, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38277491

RESUMO

Nanomedicine in combination with immunotherapy has shown great potential in the cancer treatment, but phototherapeutic nanomaterials that specifically activate the immunopharmacological effects in deep tumors have rarely been developed due to limited laser penetration depth and tumor immune microenvironment. Herein, this work reports a newly synthesized semiconducting polymer (SP) grafted with imiquimod R837 and indoxmid encapsulated micelle (SPRIN-micelle) with strong absorption in the second near infrared window (NIR-II) that can relieve tumor immunosuppression and enhance the photothermal immunotherapy and catabolic modulation on tumors. Immune agonists (Imiquimod R837) and immunometabolic modulators (indoxmid) are covalently attached to NIR-II SP sensors via a glutathione (GSH) responsive self-immolation linker and then loaded into Pluronic F127 (F127) micelles by a temperature-sensitive critical micelle concentration (CMC)-switching method. Using this method, photothermal effect of SPRIN-micelles in deep-seated tumors can be activated, leading to effective tumor ablation and immunogenic cell death (ICD). Meanwhile, imiquimod and indoxmid are tracelessly released in response to the tumor microenvironment, resulting in dendritic cell (DC) maturation by imiquimod R837 and inhibition of both indoleamine 2,3-dioxygenase (IDO) activity and Treg cell expression by indoxmid. Ultimately, cytotoxic T-lymphocyte infiltration and tumor metastasis inhibition in deep solid tumors (9 mm) are achieved. In summary, this work demonstrates a new strategy for the combination of photothermal immunotherapy and metabolic modulation by developing a dual functional polymer system including activable SP and temperature-sensitive F127 for the treatment of deep solid tumors.


Assuntos
Nanopartículas , Neoplasias , Polietilenos , Polipropilenos , Humanos , Imiquimode/farmacologia , Polímeros/farmacologia , Micelas , Fototerapia/métodos , Neoplasias/tratamento farmacológico , Imunoterapia/métodos , Linhagem Celular Tumoral , Microambiente Tumoral
9.
Adv Healthc Mater ; 13(12): e2303256, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38207170

RESUMO

Janus kinase (JAK) inhibitors are approved for many dermatologic disorders, but their use is limited by systemic toxicities including serious cardiovascular events and malignancy. To overcome these limitations, injectable hydrogels are engineered for the local and sustained delivery of baricitinib, a representative JAK inhibitor. Hydrogels are formed via disulfide crosslinking of thiolated hyaluronic acid macromers. Dynamic thioimidate bonds are introduced between the thiolated hyaluronic acid and nitrile-containing baricitinib for drug tethering, which is confirmed with 1H and 13C nuclear magnetic resonance (NMR). Release of baricitinib is tunable over six weeks in vitro and active in inhibiting JAK signaling in a cell line containing a luciferase reporter reflecting interferon signaling. For in vivo activity, baricitinib hydrogels or controls are injected intradermally into an imiquimod-induced mouse model of psoriasis. Imiquimod increases epidermal thickness in mice, which is unaffected when treated with baricitinib or hydrogel alone. Treatment with baricitinib hydrogels suppresses the increased epidermal thickness in mice treated with imiquimod, suggesting that the sustained and local release of baricitinib is important for a therapeutic outcome. This study is the first to utilize a thioimidate chemistry to deliver JAK inhibitors to the skin through injectable hydrogels, which has translational potential for treating inflammatory disorders.


Assuntos
Azetidinas , Hidrogéis , Purinas , Pirazóis , Pele , Sulfonamidas , Animais , Hidrogéis/química , Purinas/química , Purinas/farmacologia , Sulfonamidas/química , Sulfonamidas/farmacologia , Sulfonamidas/administração & dosagem , Camundongos , Pirazóis/química , Pirazóis/farmacologia , Azetidinas/química , Azetidinas/farmacologia , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Humanos , Psoríase/tratamento farmacológico , Psoríase/patologia , Psoríase/induzido quimicamente , Imiquimode/química , Imiquimode/farmacologia , Inibidores de Janus Quinases/química , Inibidores de Janus Quinases/farmacologia , Feminino
10.
Int Immunopharmacol ; 127: 111344, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38086269

RESUMO

OBJECTIVES: Psoriasis is a prevalent chronic inflammatory skin disease in humans that is characterized by frequent relapses and challenging to cure. WB518 is a novel small molecule compound with an undisclosed structure. Therefore, our study aimed to investigate the therapeutic potential of WB518 in vitro and in vivo for the treatment of psoriasis, specifically targeting the abnormal proliferation, aberrant differentiation of epidermal keratinocytes, and pathogenic inflammatory response. MATERIALS AND METHODS: We employed dual luciferase reporter assay to screen compounds capable of inhibiting STAT3 gene transcription. Flow cytometry was utilized to analyze CD3-positive cells. Protein and mRNA levels were assessed through Western blotting, immunofluorescence, immunohistochemistry, and real-time PCR. Cell viability was measured using the MTS assay, while in vivo models of psoriasis induced by IMQ and TPA were employed to study the anti-psoriasis effect of WB518. RESULTS: WB518 was found to significantly reduce the mRNA and protein levels of Keratin 17 (K17) in HaCaT cells by inhibiting the phosphorylation of STAT3 Tyr705 (Y705). In the IMQ and TPA-induced psoriasis mouse model, WB518 effectively improved scaling, epidermal hyperplasia, and inflammation. WB518 also suppressed the expression of inflammatory cytokines, such as interleukin (IL)-1ß, IL-6, IL-17, and IL-23. Furthermore, WB518 decreased the proportion of CD3-positive cells in the psoriatic skin of mice. CONCLUSIONS: WB518 exhibits promising potential as a treatment candidate for psoriasis.


Assuntos
Queratina-17 , Psoríase , Humanos , Animais , Camundongos , Queratina-17/metabolismo , Fosforilação , Imiquimode/farmacologia , Psoríase/induzido quimicamente , Psoríase/tratamento farmacológico , Psoríase/patologia , Pele/patologia , Queratinócitos , RNA Mensageiro/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos BALB C , Proliferação de Células , Fator de Transcrição STAT3/metabolismo
11.
Int Immunopharmacol ; 126: 111293, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-38056199

RESUMO

Psoriasis is a devastating autoimmune illness resulting from excessive keratinocyte growth and leukocyte infiltration into the dermis/epidermis. In the pathogenesis of psoriasis, different immune cells such as myeloid cells and CD4 + T cells play a key role. Th17/Th1 immune responses and oxidant-antioxidant responses are critical in regulation of psoriatic inflammation. Di-2-ethylhexyl phthalate (DEHP) is one of the well-known plasticizers and has widespread use worldwide. DEHP exposure through ingestion may produce harmful effects on the skin through systemic inflammation and oxidative stress, which may modify psoriatic inflammation. However, the effect of oral DEHP exposure on inflammatory cytokines and Nrf2/iNOS signaling in myeloid cells and CD4 + T cells in the context of psoriatic inflammation has not been investigated earlier. Therefore, this study explored the effect of DEHP on systemic inflammation in myeloid cells (IL-6, IL-17A, IL-23), Th17 (p-STAT3, IL-17A, IL-23R, TNF-α), Th1 (IFN-γ), Treg (Foxp3, IL-10), and Nrf2/iNOS signaling in imiquimod (IMQ)-induced mouse model of psoriasis-like inflammation. Our study showed increased Th17 signaling in imiquimod model which was further aggravated by DEHP exposure. Further, Nrf2 and iNOS signaling were also elevated in IMQ model where DEHP exposure further increased iNOS expression but did not modify the Nrf2 expression. Most importantly, IL-17A levels were also elevated in myeloid cells along with IL-6 which were further elevated by DEHP exposure. Overall, this study shows that IL-17A signaling is upregulated, whereas there is deficiency of Nrf2/HO-1 signaling by DEHP exposure in mice with psoriasiform inflammation. These observations suggest that DEHP aggravates IL-17A-mediated signaling both in CD4 + T cells as well as myeloid cells which is linked to exacerbation of IMQ-induced psoriatic inflammation in mice. Strategies that counteract the effect of DEHP exposure in the context of psoriatic inflammation through downregulation of IL-17A may be fruitful.


Assuntos
Dietilexilftalato , Poluentes Ambientais , Psoríase , Animais , Camundongos , Imiquimode/farmacologia , Interleucina-17/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Interleucina-6/metabolismo , Poluentes Ambientais/efeitos adversos , Dietilexilftalato/toxicidade , Pele/patologia , Inflamação/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Modelos Animais de Doenças
12.
Int J Pharm ; 651: 123729, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38142016

RESUMO

Cancer immunotherapy has shown promise in treating various malignancies. However, the presence of an immunosuppressive tumor microenvironment (TME) triggered by M2 tumor-associated macrophages (TAMs) and the limited tumor cell antigenicity have hindered its broader application. To address these challenges, we developed DOX/R837@ManL, a liposome loaded with imiquimod (R837) and doxorubicin (DOX), modified with mannose-polyethylene glycol (Man-PEG). DOX/R837@ManL employed a mannose receptor (MRC1)-mediated targeting strategy, allowing it to accumulate selectively at M2 Tumor associated macrophages (TAMs) and tumor sites. R837, an immune adjuvant, promoted the conversion of immunosuppressive M2 TAMs into immunostimulatory M1 TAMs, and reshaped the immunosuppressive TME. Simultaneously, DOX release induced immunogenic cell death (ICD) in tumor cells and enhanced tumor cell antigenicity by promoting dendritic cells (DCs) maturation. Through targeted delivery, the synergistic action of R837 and DOX activated innate immunity and coordinated adaptive immunity, enhancing immunotherapy efficacy. In vivo experiments have demonstrated that DOX/R837@ManL effectively eliminated primary tumors and lung metastases, while also preventing tumor recurrence post-surgery. These findings highlighted the potential of DOX/R837@ManL as a promising strategy for cancer immunotherapy.


Assuntos
Lipossomos , Neoplasias , Humanos , Lipossomos/farmacologia , Macrófagos Associados a Tumor , Imiquimode/farmacologia , Morte Celular Imunogênica , Macrófagos , Linhagem Celular Tumoral , Doxorrubicina , Neoplasias/patologia , Imunoterapia , Microambiente Tumoral
13.
Int Immunopharmacol ; 125(Pt A): 111033, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38149569

RESUMO

BACKGROUND: Jueyin granules (JYG) is effective against psoriasis, but its utility components are not clear. Rutin is the main monomer of JYG, its therapeutic effect and mechanism on psoriasis need to be further clarified. PURPOSE: To explore the potential mechanisms of rutin on psoriasis through network pharmacology and experiments. METHODS: In vitro, cell viability was determined using the CCK8 assay, and inflammatory factors were identified using RT-qPCR. The hub genes and kernel pathways of action were identified by modular pharmacology analysis. In vivo, a BALB/c mice model of psoriasis was induced by Imiquimod (IMQ). The therapeutic effect and action pathway were detected through Western Blotting, RT-qPCR, histopathologic and immunohistochemical analysis. RESULTS: Rutin inhibited cell proliferation and expression of TNF-α and IL-6 in HaCaT cells. The hub genes include APP, INS, and TNF, while the kernel pathways contain the AGE-RAGE signaling pathway. In IMQ-induced psoriasis-like mice, rutin ameliorated skin lesions and inhibited cell proliferation. Rutin could attenuate inflammation by downregulating the AGE-RAGE signaling pathway. CONCLUSION: This study suggests that rutin can reduce IMQ-induced psoriasis like skin inflammation in mice, and regulation of AGE-RAGE signaling pathway may be one of its potential anti-inflammatory mechanisms. Rutin has a promising therapeutic use for the treatment of psoriasis.


Assuntos
Psoríase , Rutina , Animais , Camundongos , Rutina/farmacologia , Rutina/uso terapêutico , Farmacologia em Rede , Psoríase/induzido quimicamente , Psoríase/tratamento farmacológico , Psoríase/patologia , Inflamação/induzido quimicamente , Transdução de Sinais , Imiquimode/farmacologia , Camundongos Endogâmicos BALB C , Modelos Animais de Doenças , Pele/patologia , Queratinócitos
14.
Lupus Sci Med ; 10(2)2023 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-38154828

RESUMO

OBJECTIVES: Methylprednisolone (mPSL) pulse therapy is an essential option for patients with active systemic lupus erythematosus, but there is a risk of adverse events related to microcirculation disorders, including idiopathic osteonecrosis of the femoral head (ONFH). Recent studies have revealed that excessive neutrophil extracellular traps (NETs) are involved in microcirculation disorders. This study aimed to demonstrate that mPSL pulse could induce NETs in lupus mice and identify the factors contributing to this induction. METHODS: Six mice with imiquimod (IMQ)-induced lupus-like disease and six normal mice were intraperitoneally injected with mPSL on days 39 to 41, and five mice with IMQ-induced lupus-like disease and six normal mice were injected with phosphate-buffered saline. Pathological examinations were conducted to evaluate the ischaemic state of the femoral head and tissue infiltration of NET-forming neutrophils. Proteome analysis was performed to extract plasma proteins specifically elevated in mPSL-administered mice with IMQ-induced lupus-like disease, and their effects on NET formation were assessed in vitro. RESULTS: Mice with IMQ-induced lupus-like disease that received mPSL pulse demonstrated ischaemia of the femoral head cartilage with tissue infiltration of NET-forming neutrophils. Proteome analysis suggested that prenylcysteine oxidase 1 (PCYOX1) played a role in this phenomenon. The reaction of PCYOX1-containing very low-density lipoproteins (VLDL) with its substrate farnesylcysteine (FC) induced NETs in vitro. The combined addition of IMQ and mPSL synergistically enhanced VLDL-plus-FC-induced NET formation. CONCLUSION: PCYOX1 and related factors are worthy of attention to understand the underlying mechanisms and create novel therapeutic strategies for mPSL-mediated microcirculation disorders, including ONFH.


Assuntos
Armadilhas Extracelulares , Lúpus Eritematoso Sistêmico , Camundongos , Humanos , Animais , Metilprednisolona/uso terapêutico , Metilprednisolona/metabolismo , Metilprednisolona/farmacologia , Cabeça do Fêmur/patologia , Imiquimode/metabolismo , Imiquimode/farmacologia , Imiquimode/uso terapêutico , Lúpus Eritematoso Sistêmico/induzido quimicamente , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Proteoma/metabolismo , Proteoma/farmacologia , Cartilagem , Isquemia/metabolismo , Isquemia/patologia
15.
Biomed Pharmacother ; 168: 115823, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37924792

RESUMO

Psoriasis is a chronic inflammatory skin disease characterized by thickening the epidermis with erythema, scaling, and proliferation. Noscapine (NOS) has several anti-inflammatory, anti-angiogenic, and anti-fibrotic effects, but its low solubility and large size results in its lower efficacy in the clinic. In this regard, solid lipid nanoparticles (SLN) encapsulated NOS (SLN-NOS) were fabricated using the well-known response surface method based on the central composite design and modified high-shear homogenization and ultrasound method. As a result, Precirol® was selected as the best lipid base for the SLN formulation based on Hildebrand-Hansen solubility parameters, in which SLN-NOS 1 % had the best zeta potential (-35.74 ± 2.59 mV), average particle size (245.66 ± 17 nm), polydispersity index (PDI, 0.226 ± 0.09), high entrapment efficiency (89.77 %), and ICH-based stability results. After 72 h, the SLN-NOS 1 % released 83.23 % and 58.49 % of the NOS at pH 5.8 and 7.4, respectively. Moreover, Franz diffusion cell's results indicated that the skin levels of NOS for SLN and cream formulations were 46.88 % and 13.5 % of the total amount, respectively. Our pharmacological assessments revealed that treatment with SLN-NOS 1 % significantly attenuated clinical parameters, namely ear thickness, length, and psoriasis area and severity index, compared to the IMQ group. Interestingly, SLN-NOS 1 % reduced the levels of interleukin (IL)-17, tumor necrosis factor-α, and transforming growth factor-ß, while elevating IL-10, compared to the IMQ group. Histology studies also showed that topical application of SLN-NOS 1 % significantly decreased parakeratosis, hyperkeratosis, acanthosis, and inflammation compared to the IMQ group. Taken together, SLN-NOS 1 % showed a high potential to attenuate skin inflammation.


Assuntos
Nanopartículas , Noscapina , Psoríase , Humanos , Imiquimode/farmacologia , Noscapina/farmacologia , Lipídeos/química , Pele , Psoríase/induzido quimicamente , Psoríase/tratamento farmacológico , Inflamação/tratamento farmacológico
16.
Allergol Immunopathol (Madr) ; 51(6): 16-22, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37937491

RESUMO

OBJECTIVE: To investigate the possible role of La ribonucleoprotein 7 (LARP7) in psoriasis through a mouse model and uncover its underlying mechanism. METHODS: The back skin of C57BL/6 mice was smeared with IMquimod (IMQ) cream for 7 days to induce psoriasis. Immunoblot kit was used to detect the deacetylase activity of SIRT1 (member of sirtuin family). Hematoxylin and eosin staining was used to assess the degree of psoriasis in mouse. Flow cytometry assays were performed to confirm effects on Th1/Th17 cell differentiation. Enzyme-linked-immunosorbent serologic assays were used to detect the level of secreted cytokines. RESULTS: LARP7 upregulated SIRT1 deacetylase activity. LARP7 alleviated psoriasis symptoms in mice by upregulating SIRT1 deacetylase activity. In addition, LARP7 regulated Th1/Th17 cell differentiation in psoriatic mice. We further found that LARP7 inhibited Th1/Th17 cytokine. CONCLUSION: LARP7 upregulated SIRT1 activity and inhibited Th1/Th17 cytokine response in psoriatic mice.


Assuntos
Citocinas , Psoríase , Animais , Camundongos , Imiquimode/farmacologia , Camundongos Endogâmicos C57BL , Psoríase/tratamento farmacológico , Sirtuína 1/genética , Células Th17
17.
Photodiagnosis Photodyn Ther ; 44: 103834, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37802276

RESUMO

Immunotherapy has some shortcomings such as off-target toxicity, treatment time and poor immunogenicity, which limit its therapeutic effect. Nanomaterials are particularly attractive in immunotherapy due to their drug delivery capabilities. Nano drug delivery system loaded with Toll-like receptor (TLR) agonist imiquimod (IMQ) and CD3 immune antibody OKT3 is constructed by using polydopamine (PDA) and CaCO3. While PDA-IMQ@CaCO3-OKT3 (PICO NPs) drug delivery system has the advantages of high biocompatibility, low toxicity, degradability. Antitumor studies in vitro and in vivo have shown that the system can effectively inhibit the proliferation of mouse breast cancer cells and the activity of Regulatory T Cells (Tregs), activate immunogenic cell death (ICD), and enhance the activity of antigen-presenting cells (APCs). Effectively eliminate tumor immunosuppression and fully activate immune function.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Animais , Camundongos , Receptor 7 Toll-Like , Muromonab-CD3 , Adjuvantes Imunológicos/farmacologia , Linhagem Celular Tumoral , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/uso terapêutico , Sistemas de Liberação de Medicamentos , Imunoterapia , Imiquimode/farmacologia , Imiquimode/uso terapêutico , Neoplasias/tratamento farmacológico
18.
J Mater Chem B ; 11(37): 9005-9018, 2023 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-37712149

RESUMO

Photothermal therapy (PTT) is a noninvasive and effective thermal therapeutic approach. Near-infrared (NIR) light responsive organic nanoparticles (NPs) have been shown to enhance the efficacy of cancer PTT. However, photothermal ablation induced NPs are currently more effective in treating primary and metastatic cancer. Herein, we designed a NIR light responsive theranostic nanosystem that combines PTT with immunotherapy. The caffeic acid doped polyaniline NPs (CA-PANi) were explored for their potential as PTT agents and their ability to mediate immunogenic cell death (ICD). The nano-theranostic agent of CA-PANi functionalized with the RGD (Arg-Gly-Asp) peptide plays a functional role in targeting integrin receptor overexpressed cancer cells. Furthermore, to enhance the immune response in the immune suppressive tumor microenvironment (iTME), imiquimod (R837) a Toll-like receptor 7 agonist that can promote dendritic cell (DC) maturation greatly inhibits tumor growth and tumor recurrence by initiating a strong antitumor immune response. Therefore, combination of PTT and immunotherapy involving CA-PANi-R837-RGD (denoted as CPRR) to improve the therapeutic effect will provide a nanovaccine strategy for targeted antitumor therapy.


Assuntos
Neoplasias , Receptor 7 Toll-Like , Imiquimode/farmacologia , Imunoterapia , Adjuvantes Imunológicos , Diferenciação Celular , Neoplasias/tratamento farmacológico
19.
Front Immunol ; 14: 1197650, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37545524

RESUMO

Imiquimod (IMQ) is a topical agent that induces local inflammation via the Toll-like receptor 7 pathway. Recently, an IMQ-driven skin inflammation model was developed in healthy volunteers for proof-of-pharmacology trials. The aim of this study was to profile the cellular, biochemical, and clinical effects of the marketed anti-inflammatory compound prednisolone in an IMQ model. This randomized, double-blind, placebo-controlled study was conducted in 24 healthy volunteers. Oral prednisolone (0.25 mg/kg/dose) or placebo (1:1) was administered twice daily for 6 consecutive days. Two days after treatment initiation with prednisolone or placebo, 5 mg imiquimod (IMQ) once daily for two following days was applied under occlusion on the tape-stripped skin of the back for 48 h in healthy volunteers. Non-invasive (imaging and biophysical) and invasive (skin punch biopsies and blister induction) assessments were performed, as well as IMQ ex vivo stimulation of whole blood. Prednisolone reduced blood perfusion and skin erythema following 48 h of IMQ application (95% CI [-26.4%, -4.3%], p = 0.0111 and 95% CI [-7.96, -2.13], p = 0.0016). Oral prednisolone suppressed the IMQ-elevated total cell count (95% CI [-79.7%, -16.3%], p = 0.0165), NK and dendritic cells (95% CI [-68.7%, -5.2%], p = 0.0333, 95% CI [-76.9%, -13.9%], p = 0.0184), and classical monocytes (95% CI [-76.7%, -26.6%], p = 0.0043) in blister fluid. Notably, TNF, IL-6, IL-8, and Mx-A responses in blister exudate were also reduced by prednisolone compared to placebo. Oral prednisolone suppresses IMQ-induced skin inflammation, which underlines the value of this cutaneous challenge model in clinical pharmacology studies of novel anti-inflammatory compounds. In these studies, prednisolone can be used as a benchmark.


Assuntos
Vesícula , Dermatite , Humanos , Imiquimode/farmacologia , Voluntários Saudáveis , Prednisolona/farmacologia , Prednisolona/uso terapêutico , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico
20.
Biomed Pharmacother ; 166: 115307, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37573659

RESUMO

The chronic disease psoriasis is associated with severe inflammation and abnormal keratinocyte propagation in the skin. Tranexamic acid (TXA), a plasmin inhibitor, is used to cure serious bleeding. We investigated whether TXA ointment mitigated Imiquimod (IMQ)-induced psoriasis-like inflammation. Furthermore, this study investigated the effect of noncytotoxic concentrations of TXA on IL-17-induced human keratinocyte (HaCaT) cells to determine the status of proliferative psoriatic keratinocytes. We found that TXA reduced IMQ-induced psoriasis-like erythema, thickness, scaling, and cumulative scores (erythema plus thickness plus scaling) on the back skin of BALB/c mice. Additionally, TXA decreased ear thickness and suppressed hyperkeratosis, hyperplasia, and inflammation of the ear epidermis in IMQ-induced BALB/c mice. Furthermore, TXA inhibited IMQ-induced splenomegaly in BALB/c mouse models. In IL-17-induced HaCaT cells, TXA inhibited ROS production and IL-8 secretion. Interestingly, TXA suppressed the IL-17-induced NFκB signaling pathway via IKK-mediated IκB degradation. TXA inhibited IL-17-induced activation of the NLRP3 inflammasome through caspase-1 and IL1ß expression. TXA inhibited IL-17-induced NLRP3 inflammasome activation by enhancing autophagy, as indicated by LC3-II accumulation, p62/SQSTM1 expression, ATG4B inhibition, and Beclin-1/Bcl-2 dysregulation. Notably, TXA suppressed IL-17-induced Nrf2-mediated keratin 17 expression. N-acetylcysteine pretreatment reversed the effects of TXA on NFκB, NLRP3 inflammasomes, and the Nrf2-mediated keratin 17 pathway in IL-17-induced HaCaT cells. Results further confirmed that in the ear skin of IMQ-induced mice, psoriasis biomarkers such as NLRP3, IL1ß, Nrf2, and keratin 17 expression were downregulated by TXA treatment. TXA improves IMQ-induced psoriasis-like inflammation in vivo and psoriatic keratinocytes in vitro. Tranexamic acid is a promising future treatment for psoriasis.


Assuntos
Dermatite , Psoríase , Ácido Tranexâmico , Humanos , Animais , Camundongos , Interleucina-17/metabolismo , Ácido Tranexâmico/farmacologia , Ácido Tranexâmico/uso terapêutico , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Queratina-17 , Fator 2 Relacionado a NF-E2 , Psoríase/induzido quimicamente , Psoríase/tratamento farmacológico , Psoríase/metabolismo , Pele , Queratinócitos , Inflamação/tratamento farmacológico , Inflamação/induzido quimicamente , Imiquimode/farmacologia , NF-kappa B/metabolismo , Camundongos Endogâmicos BALB C , Modelos Animais de Doenças
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA