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










Intervalo de año de publicación
1.
Aging (Albany NY) ; 16(8): 7217-7248, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38656880

RESUMEN

AIM: In 2019, to examine the functions of METTL3 in liver and underlying mechanisms, we generated mice with hepatocyte-specific METTL3 homozygous knockout (METTL3Δhep) by simultaneously crossing METTL3fl/fl mice with Alb-iCre mice (GPT) or Alb-Cre mice (JAX), respectively. In this study, we explored the potential reasons why hepatocyte-specific METTL3 homozygous disruption by Alb-iCre mice (GPT), but not by Alb-Cre mice (JAX), resulted in acute liver failure (ALF) and then postnatal lethality. MAIN METHODS: Mice with hepatocyte-specific METTL3 knockout were generated by simultaneously crossing METTL3fl/fl mice with Alb-iCre mice (GPT; Strain No. T003814) purchased from the GemPharmatech Co., Ltd., (Nanjing, China) or with Alb-Cre mice (JAX; Strain No. 003574) obtained from The Jackson Laboratory, followed by combined-phenotype analysis. The publicly available RNA-sequencing data deposited in the NCBI Gene Expression Omnibus (GEO) database under the accession No.: GSE198512 (postnatal lethality), GSE197800 (postnatal survival) and GSE176113 (postnatal survival) were mined to explore the potential reasons why hepatocyte-specific METTL3 homozygous deletion by Alb-iCre mice (GPT), but not by Alb-Cre mice (JAX), leads to ALF and then postnatal lethality. KEY FINDINGS: Firstly, we observed that hepatocyte-specific METTL3 homozygous deficiency by Alb-iCre mice (GPT) or by Alb-Cre mice (JAX) caused liver injury, abnormal lipid accumulation and apoptosis. Secondly, we are surprised to find that hepatocyte-specific METTL3 homozygous deletion by Alb-iCre mice (GPT), but not by Alb-Cre mice (JAX), led to ALF and then postnatal lethality. Our findings clearly demonstrated that METTL3Δhep mice (GPT), which are about to die, exhibited the severe destruction of liver histological structure, suggesting that METTL3Δhep mice (GPT) nearly lose normal liver function, which subsequently contributes to ALF, followed by postnatal lethality. Finally, we unexpectedly found that as the compensatory growth responses of hepatocytes to liver injury induced by METTL3Δhep (GPT), the proliferation of METTL3Δhep hepatocytes (GPT), unlike METTL3Δhep hepatocytes (JAX), was not evidenced by the significant increase of Ki67-positive hepatocytes, not accompanied by upregulation of cell-cycle-related genes. Moreover, GO analysis revealed that upregulated genes in METTL3Δhep livers (GPT), unlike METTL3Δhep livers (JAX), are not functionally enriched in terms associated with cell cycle, cell division, mitosis, microtubule cytoskeleton organization, spindle organization, chromatin segregation and organization, and nuclear division, consistent with the loss of compensatory proliferation of METTL3Δhep hepatocytes (GPT) observed in vivo. Thus, obviously, the loss of the compensatory growth capacity of METTL3Δhep hepatocytes (GPT) in response to liver injury might contribute to, at least partially, ALF and subsequently postnatal lethality of METTL3Δhep mice (GPT). SIGNIFICANCE: These findings from this study and other labs provide strong evidence that these phenotypes (i.e., ALF and postnatal lethality) of METTL3Δhep mice (GPT) might be not the real functions of METTL3, and closely related with Alb-iCre mice (GPT), suggesting that we should remind researchers to use Alb-iCre mice (GPT) with caution to knockout gene in hepatocytes in vivo.


Asunto(s)
Hepatocitos , Fallo Hepático Agudo , Metiltransferasas , Ratones Noqueados , Animales , Hepatocitos/metabolismo , Hepatocitos/patología , Fallo Hepático Agudo/genética , Fallo Hepático Agudo/patología , Fallo Hepático Agudo/metabolismo , Ratones , Metiltransferasas/genética , Metiltransferasas/metabolismo , Hígado/patología , Hígado/metabolismo
2.
Aging (Albany NY) ; 15(12): 5550-5568, 2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37335109

RESUMEN

AIMS: N6-methyladenosine (m6A), the most abundant and conserved epigenetic modification of mRNA, participates in various physiological and pathological processes. However, the roles of m6A modification in liver lipid metabolism have yet to be understood entirely. We aimed to investigate the roles of the m6A "writer" protein methyltransferase-like 3 (Mettl3) in liver lipid metabolism and the underlying mechanisms. MAIN METHODS: We assessed the expression of Mettl3 in liver tissues of diabetes (db/db) mice, obese (ob/ob) mice, high saturated fat-, cholesterol-, and fructose-induced non-alcoholic fatty liver disease (NAFLD) mice, and alcohol abuse and alcoholism (NIAAA) mice by quantitative reverse-transcriptase PCR (qRT-PCR). Hepatocyte-specific Mettl3 knockout mice were used to evaluate the effects of Mettl3 deficiency in mouse liver. The molecular mechanisms underlying the roles of Mettl3 deletion in liver lipid metabolism were explored by multi-omics joint analysis of public data from the Gene Expression Omnibus database and further validated by qRT-PCR and Western blot. KEY FINDINGS: Significantly decreased Mettl3 expression was associated with NAFLD progression. Hepatocyte-specific knockout of Mettl3 resulted in significant lipid accumulation in the liver, increased serum total cholesterol levels, and progressive liver damage in mice. Mechanistically, loss of Mettl3 significantly downregulated the expression levels of multiple m6A-modified mRNAs related to lipid metabolism, including Adh7, Cpt1a, and Cyp7a1, further promoting lipid metabolism disorders and liver injury in mice. SIGNIFICANCE: In summary, our findings demonstrate that the expression alteration of genes related to lipid metabolism by Mettl3-mediated m6A modification contributes to the development of NAFLD.


Asunto(s)
Trastornos del Metabolismo de los Lípidos , Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Metiltransferasas/genética , Metiltransferasas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Metabolismo de los Lípidos/genética , Expresión Génica
3.
Aging (Albany NY) ; 15(10): 4391-4410, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-37219449

RESUMEN

B-cell-specific Moloney murine leukemia virus integration site 1 (Bmi-1) is overexpressed in various cancer types. We found that Bmi-1 mRNA levels were elevated in nasopharyngeal carcinoma (NPC) cell lines. In immunohistochemical analyses, high Bmi-1 levels were observed in not only 5 of 38 non-cancerous nasopharyngeal squamous epithelial biopsies, but also in 66 of 98 NPC specimens (67.3%). High Bmi-1 levels were detected more frequently in T3-T4, N2-N3 and stage III-IV NPC biopsies than in T1-T2, N0-N1 and stage I-II NPC samples, indicating that Bmi-1 is upregulated in advanced NPC. In 5-8F and SUNE1 NPC cells, stable depletion of Bmi-1 using lentiviral RNA interference greatly suppressed cell proliferation, induced G1-phase cell cycle arrest, reduced cell stemness and suppressed cell migration and invasion. Likewise, knocking down Bmi-1 inhibited NPC cell growth in nude mice. Both chromatin immunoprecipitation and Western blotting assays demonstrated that Hairy gene homolog (HRY) upregulated Bmi-1 by binding to its promoter, thereby increasing the stemness of NPC cells. Immunohistochemistry and quantitative real-time PCR analyses revealed that HRY expression correlated positively with Bmi-1 expression in a cohort of NPC biopsies. These findings suggested that HRY promotes NPC cell stemness by upregulating Bmi-1, and that silencing Bmi-1 can suppress NPC progression.


Asunto(s)
Neoplasias Nasofaríngeas , Animales , Ratones , Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/patología , Ratones Desnudos , Línea Celular Tumoral , Nasofaringe/patología , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética
4.
Allergol Immunopathol (Madr) ; 51(1): 30-36, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36617819

RESUMEN

Psoriasis is a chronic multisystemic inflammatory disease with inflammatory cell infiltration, hyperproliferation of keratinocytes in skin lesions, and epidermal barrier dysfunction. Normal human epidermal keratinocytes (NHEKs) were stimulated with interleukin 17A (IL-17A). The expression levels of sirtuin-5 (SIRT5) were analyzed by RT-qPCR and western blot assay. The proliferation levels of NHEKs were assessed by EdU staining. The expression of ELOVL1 and ELOVL4 was analyzed by RT-Qpcr, and the expression levels of filaggrin, loricrin, and aquaporin-3 were analyzed by RT-qPCR and western blot. Extracellular signal-regulated kinase 1/2 (ERK1/2) activator t-butylhydroquinone was used to activate ERK1/2. Here, we show that SIRT5 overexpression reduces cell viability and cell proliferation, and improves barrier dysfunction in IL-17A-treated human epidermal keratinocytes, this effect of which is significantly blunted by the ERK1/2 activator. In epidermal keratinocytes, SIRT5 decreases cell proliferation and inflammation and improves barrier dysfunction via ERK/STAT3. This study reveals the role of SIRT5 in the pathogenesis of psoriasis, epidermal hyperplasia, keratinocyte-mediated inflammatory responses, and barrier dysfunction, the role of which is mediated by ERK/STAT3.


Asunto(s)
Interleucina-17 , Queratinocitos , Psoriasis , Sirtuinas , Humanos , Epidermis/metabolismo , Epidermis/patología , Inflamación/patología , Interleucina-17/farmacología , Queratinocitos/metabolismo , Queratinocitos/patología , Psoriasis/metabolismo , Psoriasis/patología , Sirtuinas/genética , Sirtuinas/metabolismo , Células Cultivadas
5.
Allergol. immunopatol ; 51(1): 30-36, ene. 2023. graf
Artículo en Inglés | IBECS | ID: ibc-214019

RESUMEN

Psoriasis is a chronic multisystemic inflammatory disease with inflammatory cell infiltration, hyperproliferation of keratinocytes in skin lesions, and epidermal barrier dysfunction. Normal human epidermal keratinocytes (NHEKs) were stimulated with interleukin 17A (IL-17A). The expression levels of sirtuin-5 (SIRT5) were analyzed by RT-qPCR and western blot assay. The proliferation levels of NHEKs were assessed by EdU staining. The expression of ELOVL1 and ELOVL4 was analyzed by RT-Qpcr, and the expression levels of filaggrin, loricrin, and aquaporin-3 were analyzed by RT-qPCR and western blot. Extracellular signal-regulated kinase 1/2 (ERK1/2) activator t-butylhydroquinone was used to activate ERK1/2. Here, we show that SIRT5 overexpression reduces cell viability and cell proliferation, and improves barrier dysfunction in IL-17A-treated human epidermal keratinocytes, this effect of which is significantly blunted by the ERK1/2 activator. In epidermal keratinocytes, SIRT5 decreases cell proliferation and inflammation and improves barrier dysfunction via ERK/STAT3. This study reveals the role of SIRT5 in the pathogenesis of psoriasis, epidermal hyperplasia, keratinocyte-mediated inflammatory responses, and barrier dysfunction, the role of which is mediated by ERK/STAT3 (AU)


Asunto(s)
Humanos , Sirtuinas/metabolismo , Interleucina-17 , Psoriasis/fisiopatología , Células Epiteliales/fisiología , Queratinocitos/fisiología , Reacción en Cadena de la Polimerasa , Western Blotting
6.
Pharmacol Res ; 176: 106059, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34998973

RESUMEN

Skp1-Cul1-F-box protein (SCF) ubiquitin E3 ligases play important roles in cancer development and serve as a promising therapeutic target in cancer therapy. Brusatol (Bru), a known Nrf2 inhibitor, holds promise for treating a wide range of tumors; however, the direct targets of Bru and its anticancer mode of action remain unclear. In our study, 793 Bru-binding candidate proteins were identified by using a biotin-brusatol conjugate (Bio-Bru) followed by streptavidin-affinity pull down-based mass spectrometry. We found that Bru can directly bind to Skp1 and disrupt the interactions of Skp1 with the F-box protein Skp2, leading to the inhibition of the Skp2-SCF E3 ligase. Bru inhibited both proliferation and migration via promoting the accumulation of the substrates p27 and E-cadherin; Skp1 overexpression attenuated while Skp1 knockdown enhanced these effects of Bru in non-small cell lung cancer (NSCLC) cells. Moreover, Bru binding to Skp1 also inhibited the ß-TRCP-SCF E3 ligase. In both subcutaneous and orthotopic NSCLC xenografts, Bru significantly inhibited the growth and metastasis of NSCLC through targeting SCF complex and upregulating p27 and E-cadherin protein levels. These data demonstrate that Bru is a Skp1-targeting agent that may have therapeutic potentials in lung cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Biotina/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Cuassinas/uso terapéutico , Proteínas Quinasas Asociadas a Fase-S/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Biotina/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Cuassinas/farmacología , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo
7.
Phytomedicine ; 91: 153689, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34446320

RESUMEN

BACKGROUND: Intrinsic and acquired chemoresistance remains a critical challenge in lung cancer chemotherapy. Fanconi anemia (FA) pathway plays an important role in antagonizing the cytotoxic effects of chemotherapeutics by repairing DNA damage. We recently demonstrated that the traditional Chinese medicinal herb, Centipeda minima (C. minima), possessed anti-inflammatory and antioxidant properties. However, the potential anticancer application of C. minima and the underlying mechanisms remain unclear. PURPOSE: We aimed to investigate the combined anticancer effects of the ethanol extract of C. minima (ECM) and DNA-crosslinking agents on non-small cell lung cancer (NSCLC) and elucidate the underlying mechanisms. METHODS: Cell viability and flow cytometry assay were performed to determine the synergistic cytotoxicity of ECM and DNA-crosslinking agents, cisplatin (CDDP) or mitomycin C (MMC), in NSCLC cells. Western blotting and immunofluorescence were conducted to examine the effects of ECM on protein expression in DNA damage repair pathway. Comet assay was applied to evaluate DNA damage levels. Subcutaneous xenografts of NSCLC were established to evaluate the combined anticancer effects of ECM and CDDP. RESULTS: Combined treatments with ECM and DNA-crosslinking agents exhibited synergistic cytotoxic effects against A549 and H1299 cells. FANCD2 was highly expressed in NSCLC that correlates with poor prognosis of NSCLC patients, based on the online database analysis. ECM significantly inhibited DNA damage-induced monoubiquitination and nuclear foci formation of FANCD2, thereby sensitizing NSCLC to CDDP- or MMC-induced DNA damage and apoptosis, as evidenced by increased expression of γ-H2AX, increased cleavage of caspases-3 and PARP, and enhanced Annexin V-FITC/PI staining. Further, ECM can also decrease the protein level of FANCD2 that contributes to the chemosensitizing effects. Moreover, ECM significantly attenuated CDDP-mediated S-phase arrest by antagonizing the activation of ATR/Chk1 pathway in NSCLC cells. Animal experiments further demonstrated that ECM and CDDP combination treatment synergistically inhibited tumor growth by decreasing FANCD2 protein level in tumor tissues. CONCLUSION: Our results demonstrated that ECM can inhibit DNA-crosslinking agents-induced activation of FA pathway by attenuating both the expression and monoubiquitination of FANCD2. ECM and CDDP combination therapy exhibited synergistic anticancer effects both in vitro and in vivo, indicating that ECM and its active components might serve as novel anticancer drugs in the combination chemotherapy.


Asunto(s)
Antineoplásicos Fitogénicos , Asteraceae/química , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Extractos Vegetales , Animales , Antineoplásicos Fitogénicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Extractos Vegetales/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Sci ; 112(8): 3218-3232, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34080260

RESUMEN

Skp2 is overexpressed in multiple cancers and plays a critical role in tumor development through ubiquitin/proteasome-dependent degradation of its substrate proteins. Drugs targeting Skp2 have exhibited promising anticancer activity. Here, we identified a plant-derived Skp2 inhibitor, betulinic acid (BA), via high-throughput structure-based virtual screening of a phytochemical library. BA significantly inhibited the proliferation and migration of non-small cell lung cancer (NSCLC) through targeting Skp2-SCF E3 ligase both in vitro and in vivo. Mechanistically, BA binding to Skp2, especially forming H-bonds with residue Lys145, decreases its stability by disrupting Skp1-Skp2 interactions, thereby inhibiting the Skp2-SCF E3 ligase and promoting the accumulation of its substrates; that is, E-cadherin and p27. In both subcutaneous and orthotopic xenografts, BA significantly inhibited the proliferation and metastasis of NSCLC through targeting Skp2-SCF E3 ligase and upregulating p27 and E-cadherin protein levels. Taken together, BA can be considered a valuable therapeutic candidate to inhibit metastasis of NSCLC.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Inhibidores Enzimáticos/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Triterpenos Pentacíclicos/administración & dosificación , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Células A549 , Animales , Antineoplásicos Fitogénicos/farmacología , Sitios de Unión , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Detección Precoz del Cáncer , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Triterpenos Pentacíclicos/farmacología , Unión Proteica/efectos de los fármacos , Proteínas Quinasas Asociadas a Fase-S/química , Ensayos Antitumor por Modelo de Xenoinjerto , Ácido Betulínico
9.
Biochem Pharmacol ; 190: 114593, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33964282

RESUMEN

Aberrant activation of the Hedgehog (Hh) pathway is implicated in the pathogenesis and development of multiple cancers, especially Hh-driven medulloblastoma (MB). Smoothened (SMO) is a promising therapeutic target of the Hh pathway in clinical cancer treatment. However, SMO mutations frequently occur, which leads to drug resistance and tumor relapse. Novel inhibitors that target both the wild-type and mutant SMO are in high demand. In this study, we identified a novel Hh pathway inhibitor, pseudolaric acid B (PAB), which significantly inhibited the expression of Gli1 and its transcriptional target genes, such as cyclin D1 and N-myc, thus inhibiting the proliferation of DAOY and Ptch1+/- primary MB cells. Mechanistically, PAB can potentially bind to the extracellular entrance of the heptahelical transmembrane domain (TMD) of SMO, based on molecular docking and the BODIPY-cyclopamine binding assay. Further, PAB also efficiently blocked ciliogenesis, demonstrating the inhibitory effects of PAB on the Hh pathway at multiple levels. Thus, PAB may overcome drug-resistance induced by SMO mutations, which frequently occurs in clinical setting. PAB markedly suppressed tumor growth in the subcutaneous allografts of Ptch1+/- MB cells. Together, our results identified PAB as a potent Hh pathway inhibitor to treat Hh-dependent MB, especially cases resistant to SMO antagonists.


Asunto(s)
Neoplasias Cerebelosas/tratamiento farmacológico , Diterpenos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Proteínas Hedgehog/antagonistas & inhibidores , Meduloblastoma/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Células A549 , Animales , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Diterpenos/química , Diterpenos/uso terapéutico , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Células HEK293 , Células HeLa , Proteínas Hedgehog/química , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Células 3T3 NIH , Estructura Secundaria de Proteína , Transducción de Señal/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
10.
J Zhejiang Univ Sci B ; 22(3): 190-203, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33719224

RESUMEN

The rapidly developing resistance of cancers to chemotherapy agents and the severe cytotoxicity of such agents to normal cells are major stumbling blocks in current cancer treatments. Most current chemotherapy agents have significant cytotoxicity, which leads to devastating adverse effects and results in a substandard quality of life, including increased daily morbidity and premature mortality. The death receptor of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can sidestep p53-dependent pathways to induce tumor cell apoptosis without damaging most normal cells. However, various cancer cells can develop resistance to TRAIL-induced apoptosis via different pathways. Therefore, it is critical to find an efficient TRAIL sensitizer to reverse the resistance of tumor cells to TRAIL, and to reinforce TRAIL's ability to induce tumor cell apoptosis. In recent years, traditional Chinese medicines and their active ingredients have shown great potential to trigger apoptotic cell death in TRAIL-resistant cancer cell lines. This review aims to collate information about Chinese medicines that can effectively reverse the resistance of tumor cells to TRAIL and enhance TRAIL's ability to induce apoptosis. We explore the therapeutic potential of TRAIL and provide new ideas for the development of TRAIL therapy and the generation of new anti-cancer drugs for human cancer treatment. This study involved an extensive review of studies obtained from literature searches of electronic databases such as Google Scholar and PubMed. "TRAIL sensitize" and "Chinese medicine" were the search keywords. We then isolated newly published studies on the mechanisms of TRAIL-induced apoptosis. The name of each plant was validated using certified databases such as The Plant List. This study indicates that TRAIL can be combined with different Chinese medicine components through intrinsic or extrinsic pathways to promote cancer cell apoptosis. It also demonstrates that the active ingredients of traditional Chinese medicines enhance the sensitivity of cancer cells to TRAIL-mediated apoptosis. This provides useful information regarding traditional Chinese medicine treatment, the development of TRAIL-based therapies, and the treatment of cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Medicina Tradicional China , Neoplasias/tratamiento farmacológico , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Bencilisoquinolinas/uso terapéutico , Clematis , Diterpenos/uso terapéutico , Humanos , Isoflavonas/uso terapéutico , Neoplasias/patología
11.
Phytomedicine ; 80: 153402, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33203590

RESUMEN

BACKGROUND: Although great achievements have been made in the field of cancer therapy, chemotherapy and radiotherapy remain the mainstay cancer therapeutic modalities. However, they are associated with various side effects, including cardiocytotoxicity, nephrotoxicity, myelosuppression, neurotoxicity, hepatotoxicity, gastrointestinal toxicity, mucositis, and alopecia, which severely affect the quality of life of cancer patients. Plants harbor a great chemical diversity and flexible biological properties that are well-compatible with their use as adjuvant therapy in reducing the side effects of cancer therapy. PURPOSE: This review aimed to comprehensively summarize the molecular mechanisms by which phytochemicals ameliorate the side effects of cancer therapies and their potential clinical applications. METHODS: We obtained information from PubMed, Science Direct, Web of Science, and Google scholar, and introduced the molecular mechanisms by which chemotherapeutic drugs and irradiation induce toxic side effects. Accordingly, we summarized the underlying mechanisms of representative phytochemicals in reducing these side effects. RESULTS: Representative phytochemicals exhibit a great potential in reducing the side effects of chemotherapy and radiotherapy due to their broad range of biological activities, including antioxidation, antimutagenesis, anti-inflammation, myeloprotection, and immunomodulation. However, since a majority of the phytochemicals have only been subjected to preclinical studies, clinical trials are imperative to comprehensively evaluate their therapeutic values. CONCLUSION: This review highlights that phytochemicals have interesting properties in relieving the side effects of chemotherapy and radiotherapy. Future studies are required to explore the clinical benefits of these phytochemicals for exploitation in chemotherapy and radiotherapy.


Asunto(s)
Antineoplásicos/efectos adversos , Fitoquímicos/farmacología , Sustancias Protectoras/farmacología , Radioterapia/efectos adversos , Animales , Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Estrés Oxidativo/efectos de los fármacos , Traumatismos por Radiación/prevención & control
12.
Phytomedicine ; 67: 153164, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31954258

RESUMEN

BACKGROUND: Centipeda minima (L.) A.Br. (C. minima) has been used in traditional Chinese herbal medicine to treat nasal allergy, diarrhea, asthma and malaria for centuries. Recent pharmacological studies have demonstrated that the ethanol extract of C. minima (ECM) and several active components possess anti-bacterial, anti-arthritis and anti-inflammatory properties. However, the effects of ECM on neuroinflammation and the underlying mechanisms have never been reported. PURPOSE: The study aimed to examine the potential inhibitory effects of ECM on neuroinflammation and illustrate the underlying mechanisms. METHODS: High performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) was performed to qualify the major components of ECM; BV2 and primary microglial cells were used to examine the anti-inflammatory activity of ECM in vitro. To evaluate the anti-inflammatory effects of ECM in vivo, the mice were orally administrated with ECM (100, 200 mg•kg-1•d-1) for 2 days before cotreatment with LPS (2 mg•kg-1•d-1, ip) for an additional 3 days. The mice were sacrificed the day after the last treatment and the hippocampus was dissected for further experiments. The expression of inflammatory proteins and the activation of microglia were respectively detected by real-time PCR, ELISA, Western blotting and immunofluorescence. RESULTS: HPLC-MS/MS analysis confirmed and quantified seven chemicals in ECM. In BV2 and primary microglial cells, ECM inhibited the LPS-induced production of tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), thus protecting HT22 neuronal cells from inflammatory damage. Furthermore, ECM inhibited the LPS-induced activation of NF-κB signaling pathway and subsequently attenuated the induction of inducible nitric oxide synthase (iNOS), cyclooxygenase 2 (COX2), NADPH oxidase 2 (NOX2) and NADPH oxidase 4 (NOX4), leading to the decreased production of nitrite oxide, prostaglandin E2 (PGE2) and reactive oxygen species (ROS). In an LPS-induced neuroinflammatory mouse model, ECM was found to exert anti-inflammatory activity by decreasing the production of proinflammatory mediators, inhibiting the phosphorylation of NF-κB, and reducing the expression of COX2, iNOS, NOX2 and NOX4 in the hippocampal tissue. Moreover, LPS-induced microglial activation was markedly attenuated in the hippocampus, while ECM at a high dose possesses a stronger anti-inflammatory activity than the positive drug dexamethansone (DEX). CONCLUSION: These findings demonstrate that ECM exerts antineuroinflammatory effects via attenuating the activation of NF-κB signaling pathway and inhibiting the production of proinflammatory mediators both in vitro and in vivo. C. minima might become a novel phytomedicine to treat neuroinflammatory diseases.


Asunto(s)
Antiinflamatorios/farmacología , Asteraceae/química , FN-kappa B/metabolismo , Extractos Vegetales/farmacología , Animales , Dinoprostona/metabolismo , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Enzimas/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/farmacología , Masculino , Ratones Endogámicos C57BL , Microglía/citología , Microglía/efectos de los fármacos , Microglía/metabolismo , Extractos Vegetales/química , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Espectrometría de Masas en Tándem , Factor de Necrosis Tumoral alfa/metabolismo
13.
Acta Pharmacol Sin ; 41(1): 10-21, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31213669

RESUMEN

Neuroinflammation is one of the critical events in neurodegenerative diseases, whereas microglia play an important role in the pathogenesis of neuroinflammation. In this study, we investigated the effects of a natural sesquiterpene lactone, 6-O-angeloylplenolin (6-OAP), isolated from the traditional Chinese medicine Centipeda minima (L.) A.Br., on neuroinflammation and the underlying mechanisms. We showed that treatment with lipopolysaccharide (LPS) caused activation of BV2 and primary microglial cells and development of neuroinflammation in vitro, evidenced by increased production of inflammatory cytokines TNF-α and IL-1ß, the phosphorylation and nuclear translocation of NF-κB, and the transcriptional upregulation of COX-2 and iNOS, leading to increased production of proinflammatory factors NO and PGE2. Moreover, LPS treatment induced oxidative stress through increasing the expression levels of NOX2 and NOX4. Pretreatment with 6-OAP (0.5-4 µM) dose-dependently attenuated LPS-induced NF-κB activation and oxidative stress, thus suppressed neuroinflammation in the cells. In a mouse model of LPS-induced neuroinflammation, 6-OAP (5-20 mg·kg-1·d-1, ip, for 7 days before LPS injection) dose-dependently inhibited the production of inflammatory cytokines, the activation of the NF-κB signaling pathway, and the expression of inflammatory enzymes in brain tissues. 6-OAP pretreatment significantly ameliorated the activation of microglia and astrocytes in the brains. 6-OAP at a high dose caused a much stronger antineuroinflammatory effect than dexamethansone (DEX). Furthermore, we demonstrated that 6-OAP pretreatment could inhibit LPS-induced neurite and synaptic loss in vitro and in vivo. In conclusion, our results demonstrate that 6-OAP exerts antineuroinflammatory effects and can be considered a novel drug candidate for the treatment of neuroinflammatory diseases.


Asunto(s)
Inflamación/tratamiento farmacológico , Lactonas/farmacología , Lipopolisacáridos/antagonistas & inhibidores , Enfermedades Neurodegenerativas/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Sesquiterpenos/farmacología , Animales , Asteraceae/química , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Inflamación/inducido químicamente , Inflamación/metabolismo , Lactonas/química , Lactonas/aislamiento & purificación , Lipopolisacáridos/farmacología , Masculino , Medicina Tradicional China , Ratones , Ratones Endogámicos C57BL , Conformación Molecular , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/metabolismo , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/aislamiento & purificación , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Oxidación-Reducción , Sesquiterpenos/química , Sesquiterpenos/aislamiento & purificación
14.
Int J Dermatol ; 56(2): 232-238, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28074522

RESUMEN

BACKGROUND: Oxidative damage may lead to the dysfunction of melanocytes (MCs) and is one of the causative mechanisms involved in the pathogenesis of vitiligo. OBJECTIVES: This study was designed to investigate the protective effects of the vitamin D3 analog tacalcitol on oxidative damage induced by hydrogen peroxide (H2 O2 ) in human epidermal MCs. METHODS: Human epidermal MCs were cultured and identified by l-DOPA staining and HMB-45 immunohistochemical staining. The model of oxidative damage induced by H2 O2 was established, and the cells were treated with tacalcitol. The viability of MCs was determined using an MTS assay. Morphological changes in cell dendrites were observed by microscopy, and the rate of change of dendrites was calculated. The reactive oxygen species (ROS) level in MCs was determined using immunofluorescence microscopy. Superoxide dismutase (SOD) activity and malondialdehyde (MDA) levels in MCs were determined using the WST-1 and TBA methods, respectively. RESULTS: In comparison with the control group, the viability of MCs and SOD activity were significantly decreased in the H2 O2 group (P < 0.05) and significantly increased in the tacalcitol group (P < 0.05). In comparison with the control group, the rate of change of cell dendrites and levels of ROS and MDA were significantly increased in the H2 O2 group (P < 0.05) and significantly decreased in the tacalcitol group (P < 0.05). CONCLUSIONS: Tacalcitol can reduce oxidative damage induced by H2 O2 in MCs by inhibiting intracellular ROS overproduction, increasing SOD activity, and decreasing the level of MDA, thereby reducing cell apoptosis.


Asunto(s)
Antiinflamatorios/farmacología , Dihidroxicolecalciferoles/farmacología , Melanocitos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Dendritas/patología , Células Epidérmicas , Humanos , Peróxido de Hidrógeno/farmacología , Malondialdehído/metabolismo , Superóxido Dismutasa/metabolismo
15.
Biomed Rep ; 4(5): 583-588, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27123251

RESUMEN

The present study aimed to investigate the effects of the Fructus Ligustri Lucidi (FLL) extract and its monomers quercetin and oleanolic acid on the adhesion and migration of human epidermal melanocytes (MCs) and intracellular actin. The human epidermal MCs were cultured and identified. The cells were treated with different concentrations of FLL extract, quercetin and oleanolic acid. The adhesion and migration abilities of the cells were determined by the fibronectin-coated culture experiment and Transwell assay, respectively. The structure and distribution of intracellular actin were observed by confocal laser microscopy, with semi-quantitative analysis. Results showed that compared with the control group, 0.0375-0.3 mg/ml of the FLL extract and 40 µM quercetin significantly improved the adhesion rate of MCs (P<0.05). The numbers of MCs permeating the microporous membrane in the 0.15 mg/ml FLL extract and 12 µM oleanolic acid groups were 43.7 and 30.3, respectively, significantly higher compared to the control group (P<0.01). In the control group, the intracellular actin was less, and the stress fiber structure was not clear. In the 0.15 mg/ml FLL extract, 12 µM oleanolic acid and 40 µM quercetin groups, there were numerous bunched stress fibers, indicating the aggregation of filamentous fibrous actin. The mean optical densities of actin expression in the 0.15 mg/ml FLL extract, 12 µM oleanolic acid and 40 µM quercetin groups were significantly higher compared to the control group (P<0.05). The FLL extract has a significant stimulatory effect on the adhesion and migration of human epidermal MCs. The mechanism may be associated with the promotion of intracellular actin cytoskeleton aggregation.

16.
Exp Ther Med ; 6(2): 317-320, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24137181

RESUMEN

Two brothers (case 1 and case 2) with erythema nodosum were diagnosed with thromboangiitis obliterans (TAO). The patients were treated with compounds including Danshen Dripping Pills, Fufang Danshen Diwan and Salvia tetramethylpyrazine. The patients were also treated with fibro-blast growth factor to promote epidermal growth and Bayaspirin enteric-coated tablets to reduce platelet aggregation. The polysaccharide nucleic acid fraction of Bacillus Calmette-Guérin and compound glycyrrhizin tablets were taken to improve immune function. Following treatment, case 2 had reduced pain levels in the left foot. The ulcer on the first toe of the left foot had decreased in size, with a reduction in pus secretions and inflammation. Case 1 demonstrated a reduction in pus secretion from the ulcer. However, the area of the ulcer had increased, spreading to the fifth toe with gangrene. A tendon had become exposed on the right foot, which was broken and induced severe pain.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...