1.Effects of forsythinol on apoptosis of hepatocellular carcinoma cells through the JAK2-STAT3 signaling pathway
Xin ZHANG ; Dong-Xiang HUANG ; Can-Hui TANG ; Zhi-Piao HUANG
The Chinese Journal of Clinical Pharmacology 2024;40(19):2837-2841
Objective To investigate the effects of forsythinol(Fo)on the expression of matrix metalloproteinase-2(MMP2)in hepatoma cells through Janus kinase 2/signal transduction and transcriptional activator 3(JAK2/STAT3)signaling pathway.Methods SMMC-7721 cells were divided into experimental-L,-M,-H groups,control group,inhibitor group and activator group.The control group was added with equal volume dimethyl sulfoxide(DMSO);the experimental-L,-M,-H groups were treated with 50,200,500 μg·mL-1 Fo;and the inhibitor group was added with 50 μmol·L-1 JAK2/STAT3 inhibitor AG490 based on the experimental-M group.In the activator group,10 μmol·L-1 JAK2/STAT3 activator Broussonin E was added to the experimental-M group.Apoptosis was detected by deoxynucleotide terminal transferase-mediated dUTP notch end labeling(TUNEL);protein expression was detected by Western blot;real-time quantitative polymerase chain reaction(qRT-PCR)was used to detect mRNA levels.Results The apoptosis rates of control group,experimental-M group,inhibitor group and activator group were(19.94±4.88)%,(27.04±5.27)%,(15.36±3.40)%and(46.66±7.89)%,respectively;the relative expression levels of phosphorylated JAK2 protein were 1.00±0.13,0.73±0.11,1.33±0.17 and 0.26±0.07,respectively;the relative expression levels of phosphorylated STAT3 protein were 1.00±0.12,0.27±0.04,0.88±0.13 and 0.12±0.04,respectively;the mRNA relative expression levels of MMP2 were 1.00±0.14,0.68±0.08,1.17±0.17 and 0.51±0.09,respectively.Compared with experimental-M group and control group,inhibitor group and activator group,there were statistically significant differences(P<0.05,P<0.001).Conclusion Fo promotes apoptosis of hepatocellular carcinoma cells,and its mechanism may be related to the effect of Fo on the expression of MMP2 by regulating JAK2-STAT3 signaling pathway.
2.Ferroptosis Is Crucial for Cisplatin Induced Sertoli Cell Injury via N6-Methyladenosine Dependent Manner
Zhongru FAN ; Peng XIN ; Lin ZHAO ; Chuize KONG ; Chiyuan PIAO ; Zhengqi WU ; Zhongkai QIU ; Wei ZHAO ; Zhe ZHANG
The World Journal of Men's Health 2024;42(4):865-880
Purpose:
This study aimed to investigate the effect of the N6-methyladenosine (m6A) dependent ferroptosis on cisplatininduced Sertoli cell injury.
Materials and Methods:
A cisplatin exposure mouse model was established by intraperitoneal injection of cisplatin in our study. TM4 cell lines was used for in vitro study. Ferroptosis was detected according to metabolomic analysis and a series of assays, including malondialdehyde, glutathione, and glutathione disulfide concentration detection, 2′,7′-dichlorodihydrofluorescein diacetate and BODIPY 581/591 C11 probe detection, and transmission electron microscope imaging. Key ferroptosis-related genes were identified via transcriptomic analysis, western blot and immunohistochemistry. The m6A modification was demonstrated via m6A RNA immunoprecipitation and luciferase reporter assays. Immune cell infiltration was detected by mass cytometry, and verified by flow cytometry and immunofluorescence.
Results:
Ferroptosis, but not other types of programmed cell death, is a significant phenomenon in cisplatin-induced testis damage and Sertoli cell loss. Ferroptosis induced by cisplatin in Sertoli cell/TM4 cell is GPX4 independent but is regulated by SLC7A11 and ALOX12. Both SLC7A11 and ALOX12 are regulated via m6A dependent manner by METTL3. Furthermore, overexpressed ALOX12-12HETE pathway may result in macrophage polarization and inflammatory response in cisplatin exposure testis.
Conclusions
Cisplatin-induced Sertoli cell injury via ferroptosis and promoted ferroptosis in an m6A dependent manner. m6A modification of both SLC7A11 and ALOX12 mRNA could result in ferroptosis in our in vitro model. Further, overexpressed ALOX12 can cause more production of 12-HETE, which may be responsible for testis inflammation caused by cisplatin.
3.Ferroptosis Is Crucial for Cisplatin Induced Sertoli Cell Injury via N6-Methyladenosine Dependent Manner
Zhongru FAN ; Peng XIN ; Lin ZHAO ; Chuize KONG ; Chiyuan PIAO ; Zhengqi WU ; Zhongkai QIU ; Wei ZHAO ; Zhe ZHANG
The World Journal of Men's Health 2024;42(4):865-880
Purpose:
This study aimed to investigate the effect of the N6-methyladenosine (m6A) dependent ferroptosis on cisplatininduced Sertoli cell injury.
Materials and Methods:
A cisplatin exposure mouse model was established by intraperitoneal injection of cisplatin in our study. TM4 cell lines was used for in vitro study. Ferroptosis was detected according to metabolomic analysis and a series of assays, including malondialdehyde, glutathione, and glutathione disulfide concentration detection, 2′,7′-dichlorodihydrofluorescein diacetate and BODIPY 581/591 C11 probe detection, and transmission electron microscope imaging. Key ferroptosis-related genes were identified via transcriptomic analysis, western blot and immunohistochemistry. The m6A modification was demonstrated via m6A RNA immunoprecipitation and luciferase reporter assays. Immune cell infiltration was detected by mass cytometry, and verified by flow cytometry and immunofluorescence.
Results:
Ferroptosis, but not other types of programmed cell death, is a significant phenomenon in cisplatin-induced testis damage and Sertoli cell loss. Ferroptosis induced by cisplatin in Sertoli cell/TM4 cell is GPX4 independent but is regulated by SLC7A11 and ALOX12. Both SLC7A11 and ALOX12 are regulated via m6A dependent manner by METTL3. Furthermore, overexpressed ALOX12-12HETE pathway may result in macrophage polarization and inflammatory response in cisplatin exposure testis.
Conclusions
Cisplatin-induced Sertoli cell injury via ferroptosis and promoted ferroptosis in an m6A dependent manner. m6A modification of both SLC7A11 and ALOX12 mRNA could result in ferroptosis in our in vitro model. Further, overexpressed ALOX12 can cause more production of 12-HETE, which may be responsible for testis inflammation caused by cisplatin.
4.Ferroptosis Is Crucial for Cisplatin Induced Sertoli Cell Injury via N6-Methyladenosine Dependent Manner
Zhongru FAN ; Peng XIN ; Lin ZHAO ; Chuize KONG ; Chiyuan PIAO ; Zhengqi WU ; Zhongkai QIU ; Wei ZHAO ; Zhe ZHANG
The World Journal of Men's Health 2024;42(4):865-880
Purpose:
This study aimed to investigate the effect of the N6-methyladenosine (m6A) dependent ferroptosis on cisplatininduced Sertoli cell injury.
Materials and Methods:
A cisplatin exposure mouse model was established by intraperitoneal injection of cisplatin in our study. TM4 cell lines was used for in vitro study. Ferroptosis was detected according to metabolomic analysis and a series of assays, including malondialdehyde, glutathione, and glutathione disulfide concentration detection, 2′,7′-dichlorodihydrofluorescein diacetate and BODIPY 581/591 C11 probe detection, and transmission electron microscope imaging. Key ferroptosis-related genes were identified via transcriptomic analysis, western blot and immunohistochemistry. The m6A modification was demonstrated via m6A RNA immunoprecipitation and luciferase reporter assays. Immune cell infiltration was detected by mass cytometry, and verified by flow cytometry and immunofluorescence.
Results:
Ferroptosis, but not other types of programmed cell death, is a significant phenomenon in cisplatin-induced testis damage and Sertoli cell loss. Ferroptosis induced by cisplatin in Sertoli cell/TM4 cell is GPX4 independent but is regulated by SLC7A11 and ALOX12. Both SLC7A11 and ALOX12 are regulated via m6A dependent manner by METTL3. Furthermore, overexpressed ALOX12-12HETE pathway may result in macrophage polarization and inflammatory response in cisplatin exposure testis.
Conclusions
Cisplatin-induced Sertoli cell injury via ferroptosis and promoted ferroptosis in an m6A dependent manner. m6A modification of both SLC7A11 and ALOX12 mRNA could result in ferroptosis in our in vitro model. Further, overexpressed ALOX12 can cause more production of 12-HETE, which may be responsible for testis inflammation caused by cisplatin.
5.Ferroptosis Is Crucial for Cisplatin Induced Sertoli Cell Injury via N6-Methyladenosine Dependent Manner
Zhongru FAN ; Peng XIN ; Lin ZHAO ; Chuize KONG ; Chiyuan PIAO ; Zhengqi WU ; Zhongkai QIU ; Wei ZHAO ; Zhe ZHANG
The World Journal of Men's Health 2024;42(4):865-880
Purpose:
This study aimed to investigate the effect of the N6-methyladenosine (m6A) dependent ferroptosis on cisplatininduced Sertoli cell injury.
Materials and Methods:
A cisplatin exposure mouse model was established by intraperitoneal injection of cisplatin in our study. TM4 cell lines was used for in vitro study. Ferroptosis was detected according to metabolomic analysis and a series of assays, including malondialdehyde, glutathione, and glutathione disulfide concentration detection, 2′,7′-dichlorodihydrofluorescein diacetate and BODIPY 581/591 C11 probe detection, and transmission electron microscope imaging. Key ferroptosis-related genes were identified via transcriptomic analysis, western blot and immunohistochemistry. The m6A modification was demonstrated via m6A RNA immunoprecipitation and luciferase reporter assays. Immune cell infiltration was detected by mass cytometry, and verified by flow cytometry and immunofluorescence.
Results:
Ferroptosis, but not other types of programmed cell death, is a significant phenomenon in cisplatin-induced testis damage and Sertoli cell loss. Ferroptosis induced by cisplatin in Sertoli cell/TM4 cell is GPX4 independent but is regulated by SLC7A11 and ALOX12. Both SLC7A11 and ALOX12 are regulated via m6A dependent manner by METTL3. Furthermore, overexpressed ALOX12-12HETE pathway may result in macrophage polarization and inflammatory response in cisplatin exposure testis.
Conclusions
Cisplatin-induced Sertoli cell injury via ferroptosis and promoted ferroptosis in an m6A dependent manner. m6A modification of both SLC7A11 and ALOX12 mRNA could result in ferroptosis in our in vitro model. Further, overexpressed ALOX12 can cause more production of 12-HETE, which may be responsible for testis inflammation caused by cisplatin.
6.Ferroptosis Is Crucial for Cisplatin Induced Sertoli Cell Injury via N6-Methyladenosine Dependent Manner
Zhongru FAN ; Peng XIN ; Lin ZHAO ; Chuize KONG ; Chiyuan PIAO ; Zhengqi WU ; Zhongkai QIU ; Wei ZHAO ; Zhe ZHANG
The World Journal of Men's Health 2024;42(4):865-880
Purpose:
This study aimed to investigate the effect of the N6-methyladenosine (m6A) dependent ferroptosis on cisplatininduced Sertoli cell injury.
Materials and Methods:
A cisplatin exposure mouse model was established by intraperitoneal injection of cisplatin in our study. TM4 cell lines was used for in vitro study. Ferroptosis was detected according to metabolomic analysis and a series of assays, including malondialdehyde, glutathione, and glutathione disulfide concentration detection, 2′,7′-dichlorodihydrofluorescein diacetate and BODIPY 581/591 C11 probe detection, and transmission electron microscope imaging. Key ferroptosis-related genes were identified via transcriptomic analysis, western blot and immunohistochemistry. The m6A modification was demonstrated via m6A RNA immunoprecipitation and luciferase reporter assays. Immune cell infiltration was detected by mass cytometry, and verified by flow cytometry and immunofluorescence.
Results:
Ferroptosis, but not other types of programmed cell death, is a significant phenomenon in cisplatin-induced testis damage and Sertoli cell loss. Ferroptosis induced by cisplatin in Sertoli cell/TM4 cell is GPX4 independent but is regulated by SLC7A11 and ALOX12. Both SLC7A11 and ALOX12 are regulated via m6A dependent manner by METTL3. Furthermore, overexpressed ALOX12-12HETE pathway may result in macrophage polarization and inflammatory response in cisplatin exposure testis.
Conclusions
Cisplatin-induced Sertoli cell injury via ferroptosis and promoted ferroptosis in an m6A dependent manner. m6A modification of both SLC7A11 and ALOX12 mRNA could result in ferroptosis in our in vitro model. Further, overexpressed ALOX12 can cause more production of 12-HETE, which may be responsible for testis inflammation caused by cisplatin.
7.Development of fluorine-substituted NH2-biphenyl-diarylpyrimidines as highly potent non-nucleoside reverse transcriptase inhibitors: Boosting the safety and metabolic stability.
Xin JIN ; Shuai WANG ; Limin ZHAO ; Wenjuan HUANG ; Yinxiang ZHANG ; Christophe PANNECOUQUE ; Erik DE CLERCQ ; Ge MENG ; Huri PIAO ; Fener CHEN
Acta Pharmaceutica Sinica B 2023;13(3):1192-1203
Our recent studies for nonnucleoside reverse transcriptase inhibitors identified a highly potent compound JK-4b against WT HIV-1 (EC50 = 1.0 nmol/L), but the poor metabolic stability in human liver microsomes (t 1/2 = 14.6 min) and insufficient selectivity (SI = 2059) with high cytotoxicity (CC50 = 2.08 μmol/L) remained major issues associated with JK-4b. The present efforts were devoted to the introduction of fluorine into the biphenyl ring of JK-4b, leading to the discovery of a novel series of fluorine-substituted NH2-biphenyl-diarylpyrimidines with noticeable inhibitory activity toward WT HIV-1 strain (EC50 = 1.8-349 nmol/L). The best compound 5t in this collection (EC50 = 1.8 nmol/L, CC50 = 117 μmol/L) was 32-fold in selectivity (SI = 66,443) compared to JK-4b and showed remarkable potency toward clinically multiple mutant strains, such as L100I, K103N, E138K, and Y181C. The metabolic stability of 5t was also significantly improved (t 1/2 = 74.52 min), approximately 5-fold higher than JK-4b in human liver microsomes (t 1/2 = 14.6 min). Also, 5t possessed good stability in both human and monkey plasma. No significant in vitro inhibition effect toward CYP enzyme and hERG was observed. The single-dose acute toxicity test did not induce mice death or obvious pathological damage. These findings pave the way for further development of 5t as a drug candidate.
8.Treatment and prognosis of severe hyperbilirubinemia in full-term infants meeting exchange transfusion criteria: a multicenter retrospective study
Ling LI ; Meihua PIAO ; Wei GUO ; Jingqun WANG ; Shuxia GENG ; Mei YANG ; Xin HE ; Shufen ZHAI ; Lili PING ; Baoli TIAN ; Lixia LIANG ; Fang LIU ; Shaoguang LYU ; Xueai FAN ; Liyuan HUI ; Liyan LIU ; Xiaohong GU ; Xiaojiao WANG ; Jing KANG
Chinese Journal of Perinatal Medicine 2021;24(6):454-460
Objective:To investigate the prognosis of severe hyperbilirubinemia in full-term infants who met the exchange transfusion criteria and were treated by blood exchange transfusion and phototherapy.Methods:A total of 168 full-term infants with severe hyperbilirubinemia who met the criteria for exchange transfusion and were hospitalized in the Neonatology Department of seven tertiary hospitals in Hebei Province from June 2017 to December 2018 were retrospectively included. According to the treatment protocol, they were divided into two groups: exchange transfusion group (38 cases) and phototherapy group (130 cases). Two independent sample t-test and Chi-square test were used to compare the clinical manifestations and follow-up results between the two groups. Multivariate logistic regression was used to analyze the risk factors for poor prognosis. Results:Neonatal severe hyperbilirubinemia in the exchange transfusion and phototherapy group were both mainly caused by hemolytic disease [42.1%(16/38) and 29.2%(38/130)], sepsis [28.9%(11/38) and 11.5%(15/130)] and early-onset breastfeeding jaundice [15.8%(6/38) and 11.5%(15/130)]. Total serum bilirubin level on admission in the exchange transfusion group was significantly higher than that in the phototherapy group [(531.7±141.3) vs (440.0±67.4) μmol/L, t=3.870, P<0.001]. Moreover, the percentage of patients with mild, moderate and severe acute bilirubin encephalopathy in the exchange transfusion group were higher than those in the phototherapy group [15.8%(6/38) vs 3.8%(5/130), 7.9%(3/38) vs 0.8%(1/130), 13.2%(5/38) vs 0.0%(0/130); χ2=29.119, P<0.001]. Among the 168 patients, 135 were followed up to 18-36 months of age and 12 showed poor prognosis (developmental retardation or hearing impairment) with four in the exchange transfusion group (12.9%, 4/31) and eight in the phototherapy group (7.7%, 8/104). Multivariate logistic regression analysis showed that for full-term infants with severe hyperbilirubinemia who met the exchange transfusion criteria, phototherapy alone without blood exchange transfusion as well as severe ABE were risk factors for poor prognosis ( OR=14.407, 95% CI: 1.101-88.528, P=0.042; OR=16.561, 95% CI: 4.042-67.850, P<0.001). Conclusions:Full-term infants who have severe hyperbilirubinemia and meet the exchange transfusion criteria should be actively treated with blood exchange transfusion, especially for those with severe ABE, so as to improve the prognosis.
9.Nuclear Factor-κB Signaling Mediates Antimony-induced Astrocyte Activation.
Tao ZHANG ; Yu Dan ZHENG ; Man JIAO ; Ye ZHI ; Shen Ya XU ; Piao Yu ZHU ; Xin Yuan ZHAO ; Qi Yun WU
Biomedical and Environmental Sciences 2021;34(1):29-39
Objective:
Antimony (Sb) has recently been identified as a novel nerve poison, although the cellular and molecular mechanisms underlying its neurotoxicity remain unclear. This study aimed to assess the effects of the nuclear factor kappa B (NF-κB) signaling pathway on antimony-induced astrocyte activation.
Methods:
Protein expression levels were detected by Western blotting. Immunofluorescence, cytoplasmic and nuclear fractions separation were used to assess the distribution of p65. The expression of protein in brain tissue sections was detected by immunohistochemistry. The levels of mRNAs were detected by Quantitative real-time polymerase chain reaction (qRT-PCR) and reverse transcription-polymerase chain reaction (RT-PCR).
Results:
Antimony exposure triggered astrocyte proliferation and increased the expression of two critical protein markers of reactive astrogliosis, inducible nitric oxide synthase (iNOS) and glial fibrillary acidic protein (GFAP), indicating that antimony induced astrocyte activation
Conclusion
Antimony activated astrocytes by activating the NF-κB signaling pathway.
Animals
;
Antimony/toxicity*
;
Astrocytes/metabolism*
;
Cell Line, Tumor
;
Cell Proliferation/drug effects*
;
Glial Fibrillary Acidic Protein/metabolism*
;
MAP Kinase Kinase Kinases
;
Male
;
Mice, Inbred ICR
;
NF-kappa B/metabolism*
;
Nitric Oxide Synthase Type II/metabolism*
;
Rats
;
Signal Transduction/drug effects*
10.Effect of Chinese Medicine Regulating Protein Phosphorylation on Tumorigenesis and Development
Shi-yuan JIANG ; Fu-chun SI ; Wen-bin WANG ; Zi-ti LIU ; Piao ZHANG ; Xin-jie ZHAO
Chinese Journal of Experimental Traditional Medical Formulae 2020;26(20):191-197
Protein phosphorylation is one of the main ways to activate protein bioactivity and make it participate in cell life activities. Researches have shown that approximately 30% of proteins in the human body are modified by phosphorylation at different levels and at different sites. If the protein phosphorylation modification level or site is abnormal, it will cause the occurrence and development of malignant tumors. Malignant tumors have always been a kind of diseases that endanger human life and health. According to statistics, as many as 18 million malignant tumors and more than 9.6 million deaths occur every year worldwide. With the continuous recognition on the abnormality of protein phosphorylation modification in tumorigenesis and development, the research and development of drugs for abnormality of protein phosphorylation modification has become a focus in the field of tumor therapy at present. Each traditional Chinese medicine(TCM) can be regarded as a natural molecular library, and it participates in the regulation of protein phosphorylation modification level with the advantages of multiple components and multiple targets, with slight side effect and low drug resistance, so TCM is one of the main sources of drug development for regulating protein phosphorylation modification levels. Through the search of multiple databases at home and abroad, it was found that certain monomers, parts extracted from TCM, single-TCM and TCM compounds can affect the tumor progression by regulating the level of protein phosphorylation and exert better anti-tumor effect. Based on the current research status of protein phosphorylation regulation by TCM at home and abroad, we found that TCM can inhibit tumor cell proliferation, invasion and metastasis, angiogenesis, and maintenance of stem cell stemness by regulating protein phosphorylation levels, and exert antitumor effects by promoting apoptosis. In order to clarify the molecular mechanism of TCM in regulating protein phosphorylation level and exerting antitumor effect, and provide evidences for the development and clinical application of antitumor TCM, the authors reviewed the mechanism of TCM in regulating protein phosphorylation level and exerting their antitumor effect from different ways in this paper.

Result Analysis
Print
Save
E-mail