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











Base de datos
Intervalo de año de publicación
1.
J Ethnopharmacol ; 335: 118702, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39168395

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Hepatic ischemia/reperfusion injury (HIRI) is a common occurrence during or after liver surgery, representing a major cause for postoperative complications or increased morbidity and mortality in liver diseases. Rehmanniae Radix Praeparata (RRP) is a traditional Chinese medicine frequently used and has garnered extensive attention for its therapeutic potential treating cardiovascular and hepatic ailments. Recent studies have indicated the possibility of RRP in regulating lipid accumulation and apoptosis in hepatocytes. AIM OF THE STUDY: This study aimed to investigate the specific mechanisms by which RRP may impede the progression of HIRI through the regulation of lipid metabolism. MATERIALS AND METHODS: High-performance liquid chromatography (HPLC) was used to identify the major components of RRP water extract. C57BL/6J mice were orally given RRP at doses of 2.5 g/kg, 5 g/kg, and 10 g/kg for a duration of 7 days before undergoing HIRI surgery. Furthermore, we established a lipid-loaded in vitro model by exposing hepatocytes to oleic acid and palmitic acid (OAPA). The anti-HIRI effect of RRP was determined through transcriptomics and various molecular biology experiments. RESULTS: After identifying active ingredients in RRP, we observed that RRP exerted lipid-lowering and hepatoprotective effects on HIRI mice and OAPA-treated hepatocytes. RRP activated AMP-activated protein kinase (AMPK) and inhibited mammalian target of rapamycin (mTOR), which further on the one hand, inhibited the cleavage and activation of sterol regulatory element binding protein 2 (SREBP2) by limiting the movement of SREBPs cleavage-activating protein (SCAP)-SREBP2 complex with the help of endoplasmic reticulum lipid raft-associated protein 1 (ERLIN1) and insulin-induced gene 1 (INSIG1), and on the other hand, promoted liver X receptor α (LXRα) nuclear transportation and subsequent cholesterol efflux. Meanwhile, the anti-lipotoxic effect of RRP can be partly reversed by an LXRα inhibitor but largely blocked by the application of compound C, an AMPK inhibitor. CONCLUSION: Our study elucidated that RRP served as a potential AMPK activator to alleviate HIRI by blocking SREBP2 activation and cholesterol synthesis, while also activating LXRα to facilitate cholesterol efflux. These findings shed new light on the potential therapeutic use of RRP for improving HIRI.

2.
Phytomedicine ; 133: 155923, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39094438

RESUMEN

BACKGROUND: Hepatic ischemia-reperfusion injury (HIRI) is commonly observed in cases of extensive hepatic resection and involves complex mechanisms. Cell senescence has been recognized as a factor in liver injury including HIRI, where it presents as a pro-inflammatory phenotype called senescence-associated secretory phenotype (SASP). Radix Rehmanniae Praeparata (RRP) is a commonly utilized traditional Chinese medicine known for its hepatoprotective, anti-aging and antioxidant qualities. Despite its recognized benefits, the specific mechanisms by which RRP may impede the progression of HIRI through the regulation of cell senescence and the identification of the most potent anti-aging extracts from RRP remain unclear. MATERIALS AND METHODS: Here, we first applied different chemical analysis methods to identify the RRP aqueous extract (RRPAE) and active fractions of RRP. Next, we constructed a surgically established mouse model and a hypoxia-reoxygenation (HR)-stimulated liver sinusoidal endothelial cells (LSECs) model to explore the underlying mechanism of RRP against HIRI through transcriptomics and multiple molecular biology experiments. RESULTS: After identifying active ingredients in RRP, we observed that RRP and its factions effectively restored LSECs fenestration and improved inflammation, cellular swelling and vascular continuity in the hepatic sinusoidal region during HIRI. Transcriptomic results revealed that RRP might reverse HIRI-induced senescence through the NOTCH signaling pathway and cell categorization further showed that the senescent cell population in HIRI liver was primarily LSECs rather than other cell types. Different RRPAE, especially RRP glucoside (RRPGLY), improved LSECs senescence and suppressed the expression of pro-inflammatory SASP genes either induced by HR insult or NOTCH1 activator, which was accompanied with the inhibition of LRP1-NOTCH1-C/EBPß pathways. Additionally, the specific inhibition of NOTCH1 by siRNA synergistically enhanced the hepatoprotective effect of RRPGLY. The ChIP-qPCR results further showed that C/EBPß was enriched at the promoter of a representative SASP, Il-1ß, in hypoxic LSECs but was significantly inhibited by RRPGLY. CONCLUSION: Our study not only clarified the potential mechanism of RRP active extractions in alleviating HIRI, but also highlighted RRPGLY was the main component of RRP that exerted anti-aging and anti-HIRI effects, providing a fresh perspective on the use of RRP to improve HIRI.


Asunto(s)
Senescencia Celular , Hígado , Receptor Notch1 , Rehmannia , Daño por Reperfusión , Animales , Daño por Reperfusión/tratamiento farmacológico , Ratones , Senescencia Celular/efectos de los fármacos , Masculino , Rehmannia/química , Receptor Notch1/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Células Endoteliales/efectos de los fármacos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones Endogámicos C57BL , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/química , Extractos Vegetales/farmacología , Modelos Animales de Enfermedad , Transducción de Señal/efectos de los fármacos
3.
Chin J Nat Med ; 22(7): 582-598, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39059828

RESUMEN

Liver fibrosis is characterized by chronic inflammatory responses and progressive fibrous scar formation. Macrophages play a central role in the pathogenesis of hepatic fibrosis by reconstructing the immune microenvironment. Picroside II (PIC II), extracted from Picrorhizae Rhizoma, has demonstrated therapeutic potential for various liver damage. However, the mechanisms by which macrophage polarization initiates immune cascades and contributes to the development of liver fibrosis, and whether this process can be influenced by PIC II, remain unclear. In the current study, RNA sequencing and multiple molecular approaches were utilized to explore the underlying mechanisms of PIC II against liver fibrosis in multidrug-resistance protein 2 knockout (Mdr2-/-) mice. Our findings indicate that PIC II activates M1-polarized macrophages to recruit natural killer cells (NK cells), potentially via the CXCL16-CXCR6 axis. Additionally, PIC II promotes the apoptosis of activated hepatic stellate cells (aHSCs) and enhances the cytotoxic effects of NK cells, while also reducing the formation of neutrophil extracellular traps (NETs). Notably, the anti-hepatic fibrosis effects associated with PIC II were largely reversed by macrophage depletion in Mdr2-/- mice. Collectively, our research suggests that PIC II is a potential candidate for halting the progression of liver fibrosis.


Asunto(s)
Apoptosis , Cinamatos , Células Estrelladas Hepáticas , Glucósidos Iridoides , Cirrosis Hepática , Macrófagos , Animales , Masculino , Ratones , Apoptosis/efectos de los fármacos , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Miembro 4 de la Subfamilia B de Casete de Unión a ATP/genética , Cinamatos/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Glucósidos Iridoides/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Cirrosis Hepática/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados
4.
J Ethnopharmacol ; 333: 118456, 2024 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-38878839

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Liver fibrosis is a generic fibrous scarring event resulting from accumulation of extracellular matrix (ECM) proteins, easily progressing to end-stage liver diseases. Tao-Hong-Si-Wu-Tang (THSWT) is a traditional Chinese medicine formula applied in clinics to treat gynecological and chronic liver diseases. However, the role of THSWT on thioacetamide (TAA)-induced hepatic fibrosis and the specific mechanisms remains unclear. AIM OF THE STUDY: To investigate the improving effects of THSWT on TAA-insulted hepatic fibrosis and the underlying mechanisms. MATERIALS AND METHODS: UHPLC-MS/MS was performed to explore the chemical characterization of THSWT. Mice were orally administered with THSWT once daily for 6 weeks along with TAA challenge. Liver function was reflected through serum biomarkers and histopathological staining. RNA sequencing, non-targeted metabolomics and molecular biology experiments were applied to investigate the underlying mechanisms. RESULTS: THSWT profoundly repaired lipid metabolism dysfunction and blocked collagen accumulation both in TAA-stimulated mice and in hepatocytes. Results of RNA sequencing and non-targeted metabolomics revealed that the anti-fibrotic effects of THSWT mostly relied on lipid metabolism repairment by increasing levels of acetyl-CoA, phosphatidylcholine, phosphatidylethanolamine, lysophosphatidylcholine and lysophosphatidylethanolamine, and decreasing relative abundances of acyl-CoA, total cholesterol, diacylglycerol, triacylglycerol and phosphatidylinositol. Mechanically, long-chain acyl-CoA synthetases 4 (ACSL4) was a key profibrotic target both in human and mice by disrupting lipid oxidation and metabolism in hepatic mitochondria. THSWT effectively blocked ACSL4 and promoted mitophagy to reverse above outcomes, which was verified by mitophagy depletion. CONCLUSION: THSWT may be a promising therapeutic option for treating hepatic fibrosis and its complications by modulating lipid metabolism and promoting mitophagy in livers.


Asunto(s)
Medicamentos Herbarios Chinos , Metabolismo de los Lípidos , Cirrosis Hepática , Mitofagia , Tioacetamida , Animales , Mitofagia/efectos de los fármacos , Tioacetamida/toxicidad , Ratones , Metabolismo de los Lípidos/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/metabolismo , Masculino , Ratones Endogámicos C57BL , Hígado/efectos de los fármacos , Hígado/patología , Hígado/metabolismo , Coenzima A Ligasas
6.
Clin Mol Hepatol ; 30(3): 303-325, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38414375

RESUMEN

Liver sinusoidal endothelial cells (LSECs) are liver-specific endothelial cells with the highest permeability than other mammalian endothelial cells, characterized by the presence of fenestrae on their surface, the absence of diaphragms and the lack of basement membrane. Located at the interface between blood and other liver cell types, LSECs mediate the exchange of substances between the blood and the Disse space, playing a crucial role in maintaining substance circulation and homeostasis of multicellular communication. As the initial responders to chronic liver injury, the abnormal LSEC activation not only changes their own physicochemical properties but also interrupts their communication with hepatic stellate cells and hepatocytes, which collectively aggravates the process of liver fibrosis. In this review, we have comprehensively updated the various pathways by which LSECs were involved in the initiation and aggravation of liver fibrosis, including but not limited to cellular phenotypic change, the induction of capillarization, decreased permeability and regulation of intercellular communications. Additionally, the intervention effects and latest regulatory mechanisms of anti-fibrotic drugs involved in each aspect have been summarized and discussed systematically. As we studied deeper into unraveling the intricate role of LSECs in the pathophysiology of liver fibrosis, we unveil a promising horizon that pave the way for enhanced patient outcomes.


Asunto(s)
Células Endoteliales , Cirrosis Hepática , Hígado , Humanos , Cirrosis Hepática/patología , Cirrosis Hepática/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Hígado/patología , Hígado/metabolismo , Animales , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Comunicación Celular , Hepatocitos/metabolismo , Hepatocitos/patología , Hepatocitos/citología
7.
Chin Herb Med ; 16(1): 82-93, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38375042

RESUMEN

Objective: Hepatic fibrosis has been widely considered as a conjoint consequence of almost all chronic liver diseases. Chuanxiong Rhizoma (Chuanxiong in Chinese, CX) is a traditional Chinese herbal product to prevent cerebrovascular, gynecologic and hepatic diseases. Our previous study found that CX extracts significantly reduced collagen contraction force of hepatic stellate cells (HSCs). Here, this study aimed to compare the protection of different CX extracts on bile duct ligation (BDL)-induced liver fibrosis and investigate plausible underlying mechanisms. Methods: The active compounds of CX extracts were identified by high performance liquid chromatography (HPLC). Network pharmacology was used to determine potential targets of CX against hepatic fibrosis. Bile duct hyperplasia and liver fibrosis were evaluated by serologic testing and histopathological evaluation. The expression of targets of interest was determined by quantitative real-time PCR (qPCR) and Western blot. Results: Different CX extracts were identified by tetramethylpyrazine, ferulic acid and senkyunolide A. Based on the network pharmacological analysis, 42 overlap targets were obtained via merging the candidates targets of CX and liver fibrosis. Different aqueous, alkaloid and phthalide extracts of CX (CXAE, CXAL and CXPHL) significantly inhibited diffuse severe bile duct hyperplasia and thus suppressed hepatic fibrosis by decreasing CCCTC binding factor (CTCF)-c-MYC-long non-coding RNA H19 (H19) pathway in the BDL-induced mouse model. Meanwhile, CX extracts, especially CXAL and CXPHL also suppressed CTCF-c-MYC-H19 pathway and inhibited ductular reaction in cholangiocytes stimulated with taurocholate acid (TCA), lithocholic acid (LCA) and transforming growth factor beta (TGF-ß), as illustrated by decreased bile duct proliferation markers. Conclusion: Our data supported that different CX extracts, especially CXAL and CXPHL significantly alleviated hepatic fibrosis and bile duct hyperplasia via inhibiting CTCF-c-MYC-H19 pathway, providing novel insights into the anti-fibrotic mechanism of CX.

8.
Acta Pharmacol Sin ; 45(6): 1115-1129, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38233527

RESUMEN

Numerous liver diseases, such as nonalcoholic fatty liver disease, hepatitis, hepatocellular carcinoma, and hepatic ischemia-reperfusion injury, have been increasingly prevalent, posing significant threats to global health. In recent decades, there has been increasing evidence linking the dysregulation of cyclic-GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING)-related immune signaling to liver disorders. Both hyperactivation and deletion of STING can disrupt the immune microenvironment dysfunction, exacerbating liver disorders. Consequently, there has been a surge in research investigating medical agents or mediators targeting cGAS-STING signaling. Interestingly, therapeutic manipulation of the cGAS-STING pathway has yielded inconsistent and even contradictory effects on different liver diseases due to the distinct physiological characteristics of intrahepatic cells that express and respond to STING. In this review, we comprehensively summarize recent advancements in understanding the dual roles of the STING pathway, highlighting that the benefits of targeting STING signaling depend on the specific types of target cells and stages of liver injury. Additionally, we offer a novel perspective on the suitability of STING agonists and antagonists for clinical assessment. In conclusion, STING signaling remains a highly promising therapeutic target, and the development of STING pathway modulators holds great potential for the treatment of liver diseases.


Asunto(s)
Hepatopatías , Proteínas de la Membrana , Nucleotidiltransferasas , Transducción de Señal , Humanos , Nucleotidiltransferasas/metabolismo , Proteínas de la Membrana/metabolismo , Hepatopatías/metabolismo , Hepatopatías/inmunología , Animales
9.
Chin J Nat Med ; 22(1): 31-46, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38278557

RESUMEN

Liver fibrosis is a dynamic wound-healing response characterized by the agglutination of the extracellular matrix (ECM). Si-Wu-Tang (SWT), a traditional Chinese medicine (TCM) formula, is known for treating gynecological diseases and liver fibrosis. Our previous studies demonstrated that long non-coding RNA H19 (H19) was markedly upregulated in fibrotic livers while its deficiency markedly reversed fibrogenesis. However, the mechanisms by which SWT influences H19 remain unclear. Thus, we established a bile duct ligation (BDL)-induced liver fibrosis model to evaluate the hepatoprotective effects of SWT on various cells in the liver. Our results showed that SWT markedly improved ECM deposition and bile duct reactions in the liver. Notably, SWT relieved liver fibrosis by regulating the transcription of genes involved in the cytoskeleton remodeling, primarily in hepatic stellate cells (HSCs), and influencing cytoskeleton-related angiogenesis and hepatocellular injury. This modulation collectively led to reduced ECM deposition. Through extensive bioinformatics analyses, we determined that H19 acted as a miRNA sponge and mainly inhibited miR-200, miR-211, and let7b, thereby regulating the above cellular regulatory pathways. Meanwhile, SWT reversed H19-related miRNAs and signaling pathways, diminishing ECM deposition and liver fibrosis. However, these protective effects of SWT were diminished with the overexpression of H19 in vivo. In conclusion, our study elucidates the underlying mechanisms of SWT from the perspective of H19-related signal networks and proposes a potential SWT-based therapeutic strategy for the treatment of liver fibrosis.


Asunto(s)
Medicamentos Herbarios Chinos , MicroARNs , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/genética , Hígado/metabolismo , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , MicroARNs/genética , MicroARNs/metabolismo , Matriz Extracelular/metabolismo
10.
Signal Transduct Target Ther ; 8(1): 424, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37935665

RESUMEN

Glioma is the most prevalent brain tumor, presenting with limited treatment options, while patients with malignant glioma and glioblastoma (GBM) have poor prognoses. The physical obstacle to drug delivery imposed by the blood‒brain barrier (BBB) and glioma stem cells (GSCs), which are widely recognized as crucial elements contributing to the unsatisfactory clinical outcomes. In this study, we found a small molecule, gambogic amide (GA-amide), exhibited the ability to effectively penetrate the blood-brain barrier (BBB) and displayed a notable enrichment within the tumor region. Moreover, GA-amide exhibited significant efficacy in inhibiting tumor growth across various in vivo glioma models, encompassing transgenic and primary patient-derived xenograft (PDX) models. We further performed a genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) knockout screen to determine the druggable target of GA-amide. By the combination of the cellular thermal shift assay (CETSA), the drug affinity responsive target stability (DARTS) approach, molecular docking simulation and surface plasmon resonance (SPR) analysis, WD repeat domain 1 (WDR1) was identified as the direct binding target of GA-amide. Through direct interaction with WDR1, GA-amide promoted the formation of a complex involving WDR1, MYH9 and Cofilin, which accelerate the depolymerization of F-actin to inhibit the invasion of patient-derived glioma cells (PDCs) and induce PDC apoptosis via the mitochondrial apoptotic pathway. In conclusion, our study not only identified GA-amide as an effective and safe agent for treating glioma but also shed light on the underlying mechanisms of GA-amide from the perspective of cytoskeletal homeostasis.


Asunto(s)
Glioma , Humanos , Simulación del Acoplamiento Molecular , Línea Celular Tumoral , Glioma/tratamiento farmacológico , Glioma/genética , Glioma/metabolismo , Citoesqueleto , Amidas , Proteínas de Microfilamentos/uso terapéutico
11.
Eur J Pharm Sci ; 190: 106581, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37696460

RESUMEN

Cancer immunotherapy has been recognized as a revolutionary breakthrough and has yielded impressive results. However, a major challenge facing immunotherapy is its limited efficacy, which may be largely due to the inadequate infiltration of immune cells into the tumor microenvironment (TME). Autophagy inhibition has been identified to enhance the recruitment of immune cells into the tumor by upregulating the expression and secretion of chemokines. Here, we verified a novel autophagy inhibitor tetramethylpyrazine (TMP) from natural products using a mCherry-GFP-LC3 probe-based autophagy flux reporter system. We then devised a liposomal system capable of co-delivering DOX and TMP using the thin-film dispersion method and modified the liposome with PD-L1 binding peptide JY4 (DOX-TMP-JY4LIPO). We found that DOX-TMP-JY4LIPO exhibited potent antitumor efficacy in vitro. In addition, DOX-TMP-JY4LIPO could effectively inhibit the autophagic flux to enhance the recruitment of immune cells into the tumor by upregulating CCL5 and CXCL10. The liposome exhibited favorable biocompatibility and safety while facilitating the accumulation of therapeutic drugs in tumors. DOX-TMP-JY4LIPO significantly inhibited tumor growth in LLC xenograft mice, accompanied by increased granzymes- and perforin-mediated cytotoxic immune responses. Our findings demonstrate that the TMP-loaded and PD-L1-targeting liposomal nanoparticles can significantly boost antitumor immunity by inhibiting autophagy, suggesting a novel natural product-based nanomedicine for immunotherapy.


Asunto(s)
Liposomas , Nanopartículas , Humanos , Animales , Ratones , Liposomas/farmacología , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Autofagia , Microambiente Tumoral
12.
Acta Pharmacol Sin ; 44(12): 2479-2491, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37580495

RESUMEN

Liver fibrosis is a wound-healing process characterized by excess formation of extracellular matrix (ECM) from activated hepatic stellate cells (HSCs). Previous studies show that both EZH2, an epigenetic regulator that catalyzes lysine 27 trimethylation on histone 3 (H3K27me3), and long non-coding RNA H19 are highly correlated with fibrogenesis. In the current study, we investigated the underlying mechanisms. Various models of liver fibrosis including Mdr2-/-, bile duct ligation (BDL) and CCl4 mice were adapted. We found that EZH2 was markedly upregulated and correlated with H19 and fibrotic markers expression in these models. Administration of EZH2 inhibitor 3-DZNeP caused significant protective effects in these models. Furthermore, treatment with 3-DZNeP or GSK126 significantly inhibited primary HSC activation and proliferation in TGF-ß-treated HSCs and H19-overexpreesing LX2 cells in vivo. Using RNA-pull down assay combined with RNA immunoprecipitation, we demonstrated that H19 could directly bind to EZH2. Integrated analysis of RNA-sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) further revealed that H19 regulated the reprogramming of EZH2-mediated H3K27me3 profiles, which epigenetically promoted several pathways favoring HSCs activation and proliferation, including epithelial-mesenchymal transition and Wnt/ß-catenin signaling. In conclusion, highly expressed H19 in chronic liver diseases promotes fibrogenesis by reprogramming EZH2-mediated epigenetic regulation of HSCs activation. Targeting the H19-EZH2 interaction may serve as a novel therapeutic approach for liver fibrosis.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2 , Histonas , Cirrosis Hepática , ARN Largo no Codificante , Animales , Ratones , Epigénesis Genética , Células Estrelladas Hepáticas/metabolismo , Histonas/metabolismo , Hígado/metabolismo , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Lisina/química , Lisina/metabolismo , Metilación , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo
13.
Adv Healthc Mater ; 12(11): e2202757, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36652763

RESUMEN

Immunotherapy has been regarded as a breakthrough in cancer treatment and achieved great success. However, the poor response rate is still a formidable challenge of current immunotherapies, especially in solid tumors without sufficient infiltration of immune cells, also known as "cold tumor." SAR405 is a highly specific VPS34 inhibitor and has been suggested as a potential approach converting "cold tumor" into "hot tumor" by inhibiting autophagy. In this study, a tri-functional doxorubicin (DOX) plus SAR405 liposome system is established and further modified with a novel anti-PD-L1 peptide JY4 for targeted delivery (DOX-SAR-JY4LIPO ). The data here demonstrate that in a lung cancer xenograft mouse model, by facilitating the tumoral enrichment of both SAR405 and DOX, DOX-SAR-JY4LIPO effectively increases the infiltration of cytotoxic lymphocytes in the tumor by synergizing DOX-induced immunogenic cell death (ICD) and SAR405-mediated upregulation of chemokines including CCL5 and CXCL10. As results, DOX-SAR-JY4LIPO significantly inhibits tumor growth, metastasis, and resurrection by re-educating immunosuppressive tumor microenvironment. In conclusion, this study not only proves the concept of inhibiting autophagy for better immune infiltration in the tumor but also presents a novel tri-functional liposomal system that overcomes the deficiencies of current therapies and holds great promise in cancer immunotherapy.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Animales , Ratones , Liposomas , Neoplasias/tratamiento farmacológico , Antineoplásicos/farmacología , Doxorrubicina/uso terapéutico , Inmunoterapia/métodos , Autofagia , Línea Celular Tumoral , Microambiente Tumoral , Antígeno B7-H1/uso terapéutico
14.
Imeta ; 2(1): e76, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38868343

RESUMEN

Metabolic-associated fatty liver disease (MAFLD) is a new emerging concept and is associated with metabolic dysfunction, generally replacing the name of nonalcoholic fatty liver disease (NAFLD) due to heterogeneous liver condition and inaccuracies in definition. The prevalence of MAFLD is rising by year due to dietary changes, metabolic disorders, and no approved therapy, affecting a quarter of the global population and representing a major economic problem that burdens healthcare systems. Currently, in addition to the common causative factors like insulin resistance, oxidative stress, and lipotoxicity, the role of immune cells, especially T cells, played in MAFLD is increasingly being emphasized by global scholars. Based on the diverse classification and pathophysiological effects of immune T cells, we comprehensively analyzed their bidirectional regulatory effects on the hepatic inflammatory microenvironment and MAFLD progression. This interaction between MAFLD and T cells was also associated with hepatic-intestinal immune crosstalk and gut microbiota homeostasis. Moreover, we pointed out several T-cell-based therapeutic approaches including but not limited to adoptive transfer of T cells, fecal microbiota transplantation, and drug therapy, especially for natural products and Chinese herbal prescriptions. Overall, this study contributes to a better understanding of the important role of T cells played in MAFLD progression and corresponding therapeutic options and provides a potential reference for further drug development.

15.
Pharm Biol ; 59(1): 1566-1575, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34767490

RESUMEN

CONTEXT: Gambogic amide (GA-amide) is a non-peptide molecule that has high affinity for tropomyosin receptor kinase A (TrkA) and possesses robust neurotrophic activity, but its effect on angiogenesis is unclear. OBJECTIVE: The study investigates the antiangiogenic effect of GA-amide on endothelial cells (ECs). MATERIALS AND METHODS: The viability of endothelial cells (ECs) treated with 0.1, 0.15, 0.2, 0.3, 0.4, and 0.5 µM GA-amide for 48 h was detected by MTS assay. Wound healing and angiogenesis assays were performed on cells treated with 0.2 µM GA-amide. Chicken eggs at day 7 post-fertilization were divided into the dimethyl sulfoxide (DMSO), bevacizumab (40 µg), and GA-amide (18.8 and 62.8 ng) groups to assess the antiangiogenic effect for 3 days. mRNA and protein expression in cells treated with 0.1, 0.2, 0.4, 0.8, and 1.2 µM GA-amide for 6 h was detected by qRT-PCR and Western blots, respectively. RESULTS: GA-amide inhibited HUVEC (IC50 = 0.1269 µM) and NhEC (IC50 = 0.1740 µM) proliferation, induced cell apoptosis, and inhibited the migration and angiogenesis at a relatively safe dose (0.2 µM) in vitro. GA-amide reduced the number of capillaries from 56 ± 14.67 (DMSO) to 20.3 ± 5.12 (62.8 ng) in chick chorioallantoic membrane (CAM) assay. However, inactivation of TrkA couldn't reverse the antiangiogenic effect of GA-amide. Moreover, GA-amide suppressed the expression of VEGF and VEGFR2, and decreased activation of the AKT/mTOR and PLCγ/Erk1/2 pathways. CONCLUSIONS: Considering the antiangiogenic effect of GA-amide, it might be developed as a useful agent for use in clinical combination therapies.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Células Endoteliales/efectos de los fármacos , Xantonas/farmacología , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Pollos , Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Receptor trkA/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Xantonas/administración & dosificación
16.
Toxicon ; 166: 24-33, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31047933

RESUMEN

Scars can have a dramatic impact on patients' physical appearance and function. Injection of botulinum toxin has been used to prevent scar formation at the site of injury and to remodel scars that have already formed. The purpose of this review is to assess the safety and efficacy of botulinum toxin for scar management compared to placebo or no treatment. MEDLINE, EMBASE, Web of Science, the Cochrane Central Register of Controlled Trials and PubMed Central were searched. Nine randomized controlled trials comparing injections of botulinum toxin versus placebo or no treatment for individuals of any age with scars were included and were assessed by two independent review authors. Cochrane risk of bias was used to assess trial quality, sensitivity analyses was conducted and trial sequential analysis was employed to assess the risk of random error. Mean difference (MD), standardized mean difference (SMD) and 95% CI were calculated for continuous outcomes. Risk ratios (RR) and corresponding 95% confidence intervals (CI) were calculated for all dichotomous outcomes. The MD for the Vancouver scar scale (VSS) was -0.87 (95%CI, -1.73 to -0.02), and for the visual analogue scale (VAS) was 1.30 (95%CI, 1.05 to 1.54). The SMD for scar width was -1.05 (95%CI, -1.29 to -0.81). The safety of botulinum toxin therapy was evaluated by calculating adverse events and complications (RR 0.36; 95%CI, 0.09 to 1.45). Evidence from this study has documented the clinical benefits of botulinum toxin versus placebo or no treatment in the prevention and remodeling of scars. Injection of botulinum toxin is both safe and effective. Future studies should focus on the different doses needed and differences in effects between pre-surgical and post-surgical injection periods.


Asunto(s)
Toxinas Botulínicas Tipo A/efectos adversos , Toxinas Botulínicas Tipo A/uso terapéutico , Cicatriz/prevención & control , Toxinas Botulínicas Tipo A/farmacología , Cicatriz/tratamiento farmacológico , Humanos , Inyecciones Intralesiones , Seguridad del Paciente , Ensayos Clínicos Controlados Aleatorios como Asunto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA