30 results on '"Peihua Luo"'
Search Results
2. Cutaneous toxicity of FDA-approved small-molecule kinase inhibitors
- Author
-
Bo Yang, Peihua Luo, Hao Yan, Zhifei Xu, Qiaojun He, Jiangxia Du, and Xiaohong Wang
- Subjects
Pharmacology ,Frequency of occurrence ,United States Food and Drug Administration ,Kinase ,Mechanism (biology) ,business.industry ,Cutaneous toxicity ,General Medicine ,Toxicology ,Bioinformatics ,Skin Diseases ,United States ,Expert opinion ,Humans ,Medicine ,business ,Drug Approval ,Protein Kinase Inhibitors - Abstract
INTRODUCTION By 1 January 2021, the FDA has approved a total of 62 small-molecule kinase inhibitors (SMKIs). The increasing clinical use of small-molecule kinase inhibitors has led to some side effects, the most common of which is cutaneous toxicity, as reflected by approximately 90% (57 of 62) of the FDA-approved SMKIs have reported treatment-related cutaneous toxicities. Since these cutaneous toxicities may have a crucial influence on the emotional, physical and psychosocial health of the patients, it is of great importance for doctors, patients, oncologists and interrelated researchers to be aware of the cutaneous side effects of these drugs in order to make the diagnosis accurate and the treatment appropriate. AREAS COVERED This review aims to summarize the potential cutaneous toxicities and the frequency of occurrence of FDA-approved 62 SMKIs, and provide a succinct overview of the potential mechanisms of certain cutaneous toxicities. The literature review was performed based on PubMed database and FDA official website. EXPERT OPINION It is significant to determine the risk factors for SMKI-induced cutaneous toxicity. The mechanisms underlying SMKI-induced cutaneous toxicities remain unclear at present. Future research should focus on the mechanisms of SMKI-induced cutaneous toxicities to find out mechanistically driven therapies.
- Published
- 2021
3. JAK-STAT signaling as an ARDS therapeutic target: Status and future trends
- Author
-
Yuanteng Zhang, Zizheng Gao, Feng Jiang, Hao Yan, Bo Yang, Qiaojun He, Peihua Luo, Zhifei Xu, and Xiaochun Yang
- Subjects
Pharmacology ,Biochemistry - Abstract
Acute respiratory distress syndrome (ARDS) is characterized by noncardiogenic pulmonary edema. It has a high mortality rate and lacks effective pharmacotherapy. With the outbreak of COVID-19 worldwide, the mortality of ARDS has increased correspondingly, which makes it urgent to find effective targets and strategies for the treatment of ARDS. Recent clinical trials of Janus kinase (JAK) inhibitors in treating COVID-19-induced ARDS have shown a positive outcome, which makes the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway a potential therapeutic target for treating ARDS. Here, we review the complex cause of ARDS, the molecular JAK/STAT pathway involved in ARDS pathology, and the progress that has been made in strategies targeting JAK/STAT to treat ARDS. Specifically, JAK/STAT signaling directly participates in the progression of ARDS or colludes with other pathways to aggravate ARDS. We summarize JAK and STAT inhibitors with ARDS treatment benefits, including inhibitors in clinical trials and preclinical studies and natural products, and discuss the side effects of the current JAK inhibitors to reveal future trends in the design of JAK inhibitors, which will help to develop effective treatment strategies for ARDS in the future.
- Published
- 2022
4. Discovery of N-((3S,4S)-4-(3,4-Difluorophenyl)piperidin-3-yl)-2-fluoro-4-(1-methyl-1H-pyrazol-5-yl)benzamide (Hu7691), a Potent and Selective Akt Inhibitor That Enables Decrease of Cutaneous Toxicity
- Author
-
Xiaoyang Dai, Yang Lu, Zizheng Gao, Wenhu Zhan, Yongzhou Hu, Dan Li, Xiaowu Dong, Sheng Haichao, Binhui Chen, Bo Yang, Qinjie Weng, Zegao Jin, Jinxin Che, Peihua Luo, Qiaojun He, and Jian Gao
- Subjects
chemistry.chemical_compound ,HaCaT ,chemistry ,In vivo ,Kinase ,Cell growth ,Drug Discovery ,Molecular Medicine ,AKT1 ,AKT2 ,Pharmacology ,Benzamide ,Protein kinase B - Abstract
Rash is one of the primary dose-limiting toxicities of Akt (protein kinase B) inhibitors in clinical trials. Here, we demonstrate the inhibition of Akt2 isozyme may be a driver for keratinocyte apoptosis, which promotes us to search for new selective Akt inhibitors with an improved cutaneous safety property. According to our previous research, compound 2 is selected for further optimization for overcoming the disadvantages of compound 1, including high Akt2 inhibition and high toxicity against HaCaT keratinocytes. The dihedral angle-based design and molecular dynamics simulation lead to the identification of Hu7691 (B5) that achieves a 24-fold selectivity between Akt1 and Akt2. Hu7691 exhibits low activity in inducing HaCaT apoptosis, promising kinase selectivity, and excellent anticancer cell proliferation potencies. Based on the superior results of safety property, pharmacokinetic profile, and in vivo efficacy, the National Medical Products Administration (NMPA) approved the investigational new drug (IND) application of Hu7691.
- Published
- 2021
5. Decreased HMGB1 expression contributed to cutaneous toxicity caused by lapatinib
- Author
-
Liyu Jiang, Yan Zeng, Leilei Ai, Hao Yan, Xiaochun Yang, Peihua Luo, Bo Yang, Zhifei Xu, and Qiaojun He
- Subjects
Pharmacology ,Receptor, ErbB-2 ,Antineoplastic Agents ,Apoptosis ,Lapatinib ,Biochemistry ,Mice ,Cell Line, Tumor ,Neoplasms ,Quinazolines ,Animals ,Humans ,HMGB1 Protein ,Protein Kinase Inhibitors - Abstract
The application of lapatinib, a widely used dual inhibitor of human epidermal growth factor receptor 1 (EGFR/ERBB1) and 2 (HER2/ERBB2), has been seriously limited due to cutaneous toxicity. However, the specific mechanism of lapatinib-induced cutaneous toxicity has not been clarified, leading to the lack of an effective strategy to improve clinical safety. Here, we found that lapatinib could induce mitochondrial dysfunction, lead to DNA damage and ultimately cause apoptosis of keratinocytes. In addition, we found that lapatinib could induce an aberrant immune response and promote the release of inflammatory factors in vitro and in vivo. Mechanistically, downregulated expression of the DNA repair protein HMGB1 played a critical role in these toxic reaction processes. Overexpression of HMGB1 inhibited keratinocyte apoptosis and inflammatory reactions. Therefore, restoring HMGB1 expression might be an effective remedy against lapatinib-induced cutaneous toxicity. Finally, we found that saikosaponin A could significantly rescue the reduced HMGB1 transcription, which could alleviate lapatinib-induced DNA damage, inhibit keratinocyte apoptosis and further prevent the toxicity of lapatinib in mice. Collectively, our study might bring new hope to clinicians and tumor patients and shed new light on the prevention of cutaneous adverse drug reactions induced by EGFR inhibitors.
- Published
- 2022
6. Research Status and Outlook of PD-1/PD-L1 Inhibitors for Cancer Therapy
- Author
-
Xiaochun Yang, Peihua Luo, Hao Yan, Qiaojun He, Leilei Ai, Jian Chen, and Zhifei Xu
- Subjects
0301 basic medicine ,Pharmacology ,medicine.medical_specialty ,biology ,business.industry ,Immune checkpoint inhibitors ,Cancer therapy ,Pharmaceutical Science ,Cancer ,medicine.disease ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,PD-L1 ,Drug Discovery ,medicine ,biology.protein ,Intensive care medicine ,business ,Adverse effect - Abstract
PD-1/PD-L1 inhibitors are a group of immune checkpoint inhibitors as front-line treatment of multiple types of cancer. However, the serious immune-related adverse reactions limited the clinical application of PD-1/PD-L1 monoclonal antibodies, despite the promising curative effects. Therefore, it is urgent to develop novel inhibitors, such as small molecules, peptides or macrocycles, targeting the PD-1/PD-L1 axis to meet the increasing clinical demands. Our review discussed the mechanism of action of PD-1/PD-L1 inhibitors and presented clinical trials of currently approved PD-1/PD-L1 targeted drugs and the incidence of related adverse reactions, helping clinicians pay more attention to them, better formulate their intervention and resolution strategies. At last, some new inhibitors whose patent have been published are listed, which provide development ideas and judgment basis for the efficacy and safety of novel PD-1/PD-L1 inhibitors.
- Published
- 2020
7. Bisdemethoxycurcumin alleviates vandetanib-induced cutaneous toxicity in vivo and in vitro through autophagy activation
- Author
-
Xiaofei Shen, Ying Jin, Zhifei Xu, Xueqin Chen, Zizheng Gao, Peihua Luo, Bo Yang, Qiaojun He, Hao Yan, Huangxi Fu, and Zezheng Pan
- Subjects
Keratinocytes ,DNA damage ,Antineoplastic Agents ,Apoptosis ,RM1-950 ,Vandetanib ,Bisdemethoxycurcumin ,Skin Diseases ,chemistry.chemical_compound ,Piperidines ,In vivo ,Diarylheptanoids ,medicine ,Autophagy ,Animals ,HaCaT Cells ,Humans ,Protein Kinase Inhibitors ,Skin ,Pharmacology ,chemistry.chemical_classification ,Membrane Potential, Mitochondrial ,Reactive oxygen species ,General Medicine ,Mitochondria ,Mice, Inbred C57BL ,HaCaT ,Disease Models, Animal ,chemistry ,Cancer research ,Quinazolines ,Therapeutics. Pharmacology ,Apoptosis Regulatory Proteins ,Reactive Oxygen Species ,Cutaneous toxicity ,medicine.drug ,DNA Damage - Abstract
High incidence of cutaneous toxicity ranging from 29.2% to 71.2% has been reported during clinical use of vandetanib, which is a multi-target kinase inhibitor indicated for the treatment of unresectable medullary thyroid carcinoma. The cutaneous toxicity of vandetanib has limited its clinical benefits, but the underlying mechanisms and protective strategies are not well studied. Hence, we firstly established an in vivo model by continuously administrating vandetanib at 55 mg/kg/day to C57BL/6 for 21 days and verified that vandetanib could induce skin rash in vivo, which was consistent with the clinical study. We further cultured HaCaT and NHEK cells, the immortalized or primary human keratinocyte line, and investigated vandetanib (0–10 μM, 0–24 h)-caused alteration in cellular survival and death processes. The western blot showed that the expression level of apoptotic-related protein, c-PARP, c-Caspase 3 and Bax were increased, while the anti-apoptotic protein Bcl2 and MCL1 level were decreased. Meanwhile, vandetanib downregulated mitochondrial membrane potential which in turn caused the release of Cytochrome C, excessive production of reactive oxygen species and DNA damage. Furthermore, we found that 5 μM bisdemethoxycurcumin partially rescued vandetanib-induced mitochondria pathway-dependent keratinocyte apoptosis via activation of autophagy in vivo and in vitro, thereby ameliorated cutaneous toxicity. Conclusively, our study revealed the mechanisms of vandetanib-induced apoptosis in keratinocytes during the occurrence of cutaneous toxicity, and suggested bisdemethoxycurcumin as a potential protective drug. This work provided a potentially promising therapeutic strategy for the treatment of vandetanib-induced cutaneous toxicity.
- Published
- 2021
8. Molecular basis for class side effects associated with PI3K/AKT/mTOR pathway inhibitors
- Author
-
Peihua Luo, Bo Yang, Ying Zhang, Zhifei Xu, Qiaojun He, and Hao Yan
- Subjects
Antineoplastic Agents ,Toxicology ,Bioinformatics ,030226 pharmacology & pharmacy ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,Animals ,Humans ,Medicine ,Clinical significance ,Adverse effect ,Protein Kinase Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Pharmacology ,business.industry ,Drug discovery ,TOR Serine-Threonine Kinases ,General Medicine ,Clinical trial ,Drug development ,030220 oncology & carcinogenesis ,Signal transduction ,business ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Introduction: The phosphatidylinositide 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway has emerged as an important target in cancer therapy. Numerous PI3K/AKT/mTOR pathway inhibitors are extensively studied; some are used clinically, but most of these drugs are undergoing clinical trials. Potential adverse effects, such as severe hepatotoxicity and pneumonitis, have largely restricted the application and clinical significance of these inhibitors. A summary of mechanisms underlying the adverse effects is not only significant for the development of novel PI3K/AKT/mTOR inhibitors but also beneficial for the optimal use of existing drugs. Areas covered: We report a profile of the adverse effects, which we consider the class effects of PI3K/AKT/mTOR inhibitors. This review also discusses potential molecular toxicological mechanisms of these agents, which might drive future drug discovery. Expert opinion: Severe toxicities associated with PI3K/AKT/mTOR inhibitors hinder their approval and limit long-term clinical application of these drugs. A better understanding regarding PI3K/AKT/mTOR inhibitor-induced toxicities is needed. However, the mechanisms underlying these toxicities remain unclear. Future research should focus on developing strategies to reduce toxicities of approved inhibitors as well as accelerating new drug development. This review will be useful to clinical, pharmaceutical, and toxicological researchers.
- Published
- 2019
9. Bisdemethoxycurcumin protects against renal fibrosis via activation of fibroblast apoptosis
- Author
-
Ying Jin, Jiangxia Du, Ziying Zhao, Bo Yang, Liyu Jiang, Fuquan Jin, Peihua Luo, Qiaojun He, and Ying Zhang
- Subjects
Male ,0301 basic medicine ,Curcumin ,Apoptosis ,Kidney ,Protective Agents ,urologic and male genital diseases ,Cell Line ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacotherapy ,Diarylheptanoids ,In vivo ,Bisdemethoxycurcumin ,medicine ,Renal fibrosis ,Animals ,Humans ,Renal Insufficiency, Chronic ,Urinary Tract ,Pharmacology ,Biological Products ,business.industry ,Therapeutic effect ,Fibroblasts ,Fibrosis ,In vitro ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Mechanism of action ,Cancer research ,Female ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Ureteral Obstruction - Abstract
Renal fibrosis is the common final outcome of nearly all progressive chronic kidney diseases (CKD) that eventually develop into end-stage renal failure, which threatens the lives of patients. Currently, there are no effective drugs for the treatment of renal fibrosis. However, studies have shown that certain plant natural products have a fibrosis-alleviating effect. Thus, we have screened a large number of natural products for their ability to protect against renal fibrosis and found that bisdemethoxycurcumin has a good therapeutic effect in renal fibrosis according to the data obtained in a mouse model of unilateral ureteral obstruction (UUO). The results indicate that bisdemethoxycurcumin can efficiently attenuate renal fibrosis induced by UUO. Additional studies of the bisdemethoxycurcumin mechanism of action in the treatment of renal fibrosis demonstrated that the therapeutic effect of bisdemethoxycurcumin is mediated by the specific induction of fibroblast apoptosis at a concentration of 20 μM. bisdemethoxycurcumin can efficiently protect against renal fibrosis both in vitro and in vivo. This discovery will provide new ideas for renal fibrosis treatment in clinics and a new direction for the development of effective drug therapy of renal fibrosis.
- Published
- 2019
10. Defining therapeutic targets for renal fibrosis: Exploiting the biology of pathogenesis
- Author
-
Zhifei Xu, Qiaojun He, Jiangxin Xu, Bo Yang, Peihua Luo, and Hao Yan
- Subjects
Pathology ,medicine.medical_specialty ,Renal function ,Inflammation ,RM1-950 ,Biology ,urologic and male genital diseases ,Kidney ,Renal Agents ,Extracellular matrix ,Pathogenesis ,Renal fibrosis ,Chronic kidney disease ,medicine ,Renal tubular epithelial cells ,Animals ,Humans ,Molecular Targeted Therapy ,Pathological ,Pharmacology ,General Medicine ,medicine.disease ,Fibrosis ,Extracellular Matrix ,medicine.anatomical_structure ,Therapeutic strategy ,Kidney Diseases ,Therapeutics. Pharmacology ,medicine.symptom ,Kidney disease ,Signal Transduction - Abstract
Renal fibrosis is a failed wound-healing process of the kidney tissue after chronic, sustained injury, which is a common pathway and pathological marker of virtually every type of chronic kidney disease (CKD), regardless of cause. However, there is a lack of effective treatment specifically targeting against renal fibrosis per se to date. The main pathological feature of renal fibrosis is the massive activation and proliferation of renal fibroblasts and the excessive synthesis and secretion of extracellular matrix (ECM) deposited in the renal interstitium, leading to structural damage, impairment of renal function, and eventually end-stage renal disease. In this review, we summarize recent advancements regarding the participation and interaction of many types of kidney residents and infiltrated cells during renal fibrosis, attempt to comprehensively discuss the mechanism of renal fibrosis from the cellular level and conclude by highlighting novel therapeutic targets and approaches for development of new treatments for patients with renal fibrosis.
- Published
- 2021
11. Cardiovascular toxicity induced by anti-VEGF/VEGFR agents: a special focus on definitions, diagnoses, mechanisms and management
- Author
-
Xiaochun Yang, Li Ping, Peihua Luo, Zhifei Xu, Hao Yan, Qiaojun He, and Liyu Jiang
- Subjects
Cardiovascular toxicity ,Vascular Endothelial Growth Factor A ,VEGF receptors ,Cancer therapy ,Angiogenesis Inhibitors ,Antineoplastic Agents ,Toxicology ,030226 pharmacology & pharmacy ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Drug Development ,Neoplasms ,Medicine ,Animals ,Humans ,Pharmacology ,Anti vegf ,Cardiovascular safety ,integumentary system ,biology ,business.industry ,General Medicine ,Vascular endothelial growth factor ,Receptors, Vascular Endothelial Growth Factor ,chemistry ,Cardiovascular Diseases ,030220 oncology & carcinogenesis ,embryonic structures ,Cancer research ,Molecular mechanism ,biology.protein ,business - Abstract
Vascular endothelial growth factor (VEGF) is a key target in cancer therapy. However, cardiovascular safety has been one of the most challenging aspects of anti-VEGF/VEGF receptor (VEGFR) agent development and therapy. While accurate diagnostic modalities for assessment of cardiac function have been developed over the past few decades, a lack of an optimal definition and precise mechanism still places a significant limit on the effective management of cardiovascular toxicity.Here, we report the cardiovascular toxicity profile associated with anti-VEGF/VEGFR agents and summarize the clinical diagnoses as well as management that are already performed in clinical practice or are currently being investigated. Furthermore, the review discusses the potential molecular toxicological mechanisms, which may provide strategies to prevent toxicity and drive drug discovery.Cardiovascular toxicity associated with anti-VEGF/VEGFR agents has been a substantial risk for cancer treatment. To improve its management, the development of guidelines for prevention, monitoring and treatment of cardiovascular toxicity has become a hot topic. The summary of cardiovascular toxicity profile, mechanisms and management given in this review is not only significant for the optimal use of existing anti-VEGF/VEGFR agents to protect patients predisposed to cardiovascular toxicity but is also beneficial for drug development.
- Published
- 2020
12. A Comprehensive Review of Clinical Cardiotoxicity Incidence of FDA-Approved Small-Molecule Kinase Inhibitors
- Author
-
Hao Yan, Xiaochun Yang, Ying Jin, Zhifei Xu, Peihua Luo, and Qiaojun He
- Subjects
0301 basic medicine ,Oncology ,Drug ,medicine.medical_specialty ,media_common.quotation_subject ,cardiotoxicity ,Review ,QT interval ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Pharmacology (medical) ,Myocardial infarction ,Adverse effect ,media_common ,Pharmacology ,Cardiotoxicity ,protein kinases ,business.industry ,lcsh:RM1-950 ,medicine.disease ,targeted therapies ,small-molecule kinase inhibitor ,Discontinuation ,Clinical trial ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,Systematic review ,030220 oncology & carcinogenesis ,incidence ,business - Abstract
Numerous protein kinases encoded in the genome have become attractive targets for the treatment of different types of cancer. As of January 2020, a total of 52 small-molecule kinase inhibitors (SMKIs) have been approved by the FDA. With the numerous clinical trials and a heavy focus on drug safety, SMKI-induced cardiotoxicity, which is a life-threatening risk, has greatly attracted the attention of researchers. In this review, the SMKIs with cardiotoxicity incidence were described exhaustively. The data were collected from 42 clinical trials, 25 FDA-published documents, seven meta-analysis/systematic reviews, three case reports and more than 50 other types of articles. To date, 73% (38 of 52) of SMKIs have reported treatment-related cardiotoxicity. Among the 38 SMKIs with known cardiotoxicity, the rates of incidence of cardiac adverse events were QT prolongation: 47% (18 of 38), hypertension: 40% (15 of 38), left ventricular dysfunction: 34% (13 of 38), arrhythmia: 34% (13 of 38), heart failure: 26% (10 of 38) and ischemia or myocardial infarction: 29% (11 of 38). In the development process of novel SMKIs, more attention should be paid to balancing the treatment efficacy and the risk of cardiotoxicity. In preclinical drug studies, producing an accurate and reliable cardiotoxicity evaluation model is of key importance. To avoid the clinical potential cardiotoxicity risk and discontinuation of a highly effective drug, patients treated with SMKIs should be proactively monitored on the basis of a global standard. Moreover, the underlying mechanisms of SMKI-induced cardiotoxicity need to be further studied to develop new therapies for SMKI-induced cardiotoxicity.
- Published
- 2020
13. The contribution of keratinocytes in capecitabine-stimulated hand-foot-syndrome
- Author
-
Qiaojun He, Guanqun Li, Jian Chen, Jinjin Shao, Yuanrun Zhu, Bo Yang, Min Chen, Hong Zhu, Peihua Luo, Zhifei Xu, and Xueming Peng
- Subjects
Keratinocytes ,Male ,0301 basic medicine ,Antimetabolites, Antineoplastic ,medicine.medical_specialty ,Health, Toxicology and Mutagenesis ,Apoptosis ,Stimulation ,Toxicology ,Cell Line ,Capecitabine ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Internal medicine ,medicine ,Animals ,Humans ,Cell Proliferation ,Membrane Potential, Mitochondrial ,Pharmacology ,Membrane potential ,Mice, Inbred ICR ,integumentary system ,Chemistry ,General Medicine ,humanities ,Pathophysiology ,In vitro ,body regions ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Hand-Foot Syndrome ,Keratinocyte ,Intracellular ,medicine.drug - Abstract
Capecitabine, as the first-line treatment for multiple tumor types, has a serious drawback of hand-foot-syndrome (HFS) that limits its clinical use. However, the pathophysiology and mechanism of capecitabine-induced HFS is rarely known. Here we built the experimental mouse model of HFS induced by capecitabine at first and it was shown that 3 of 6 mice appeared HFS in the 5th day and 5 mice occurred HFS in the 30th day. The corneous layer was reduced in capecitabine-induced HFS in vivo. Moreover, we found that capecitabine could significantly induce keratinocytes cells death in vitro through activated apoptosis pathway and decreased mitochondrial membrane potential. In conclusion, these results suggested that HFS of capecitabine may be developed from reduction of corneous layer through stimulation of intracellular mitochondrial dysfunction following activation of caspase-dependent apoptosis pathway.
- Published
- 2017
14. Enhanced proliferation inhibition and apoptosis in glioma cells elicited by combination of irinotecan and imatinib
- Author
-
Yuhuai Hu, Xiaochun Yang, Jiabin Lu, Renyun Qian, Peihua Luo, and Yuqian Zhang
- Subjects
0301 basic medicine ,Cyclin-Dependent Kinase Inhibitor p21 ,Combination therapy ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Irinotecan ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Glioma ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,neoplasms ,Protein Kinase Inhibitors ,Cell Proliferation ,Pharmacology ,Gene knockdown ,Mice, Inbred BALB C ,medicine.diagnostic_test ,Chemistry ,Imatinib ,medicine.disease ,Rats ,Tumor Burden ,030104 developmental biology ,Cancer research ,Imatinib Mesylate ,Female ,Tumor Suppressor Protein p53 ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Glioma is a kind of lethal malignant tumor, and lacks efficient therapies. Combination therapy has been claimed to be a promising approach to combat cancer, due to its increased anti-cancer effects and reduced side effects. This study aimed to investigate the anti-cancer effect and mechanism of combining imatinib with irinotecan or its active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38). First, we found that this drug combination exerted synergistic antitumor effects against glioma in vitro and in vivo. In addition, flow cytometry results proved that the SN-38-induced apoptosis was further enhanced by imatinib, and similar results were observed by determining the protein expression levels of apoptosis biomarkers. Interestingly, p53 expression was elevated by the SN-38 mono-treatment, and was not further increased after the co-treatment; besides, knockdown of p53 could only reduce the expression of cleaved-PARP partially, and weaken the enhanced proliferation inhibition induced by SN-38 plus imatinib, indicating that there might be other factors involved in the synergistic effects besides p53. Meanwhile, the markedly elevated p21 expression was observed only in the combination group, instead of the mono-treated groups. According to the results of p21 knockdown, we found that p21 was also required for the synergistic inhibitory effects. Moreover, we explored and ruled out the possibility of imatinib enhancing the sensitivity of irinotecan by inhibiting drug efflux pumps. Thus, our findings collectively suggest that combining irinotecan with imatinib could be a promising new strategy to fight against glioma.
- Published
- 2019
15. ROS-dependent DNA damage contributes to crizotinib-induced hepatotoxicity via the apoptotic pathway
- Author
-
Hao Yan, Xiaochun Yang, Qiaojun He, Jiangxia Du, Xueqin Chen, Bo Yang, and Peihua Luo
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.drug_class ,DNA damage ,Cell Survival ,Apoptosis ,Toxicology ,Proto-Oncogene Mas ,Tyrosine-kinase inhibitor ,Receptor tyrosine kinase ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Crizotinib ,medicine ,ROS1 ,Anaplastic lymphoma kinase ,Humans ,Protein Kinase Inhibitors ,Pharmacology ,biology ,Dose-Response Relationship, Drug ,Chemistry ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Hepatocyte ,biology.protein ,Cancer research ,Hepatocytes ,Reactive Oxygen Species ,medicine.drug ,DNA Damage - Abstract
Crizotinib is an oral small-molecule tyrosine kinase inhibitor targeting anaplastic lymphoma kinase (ALK), ROS proto-oncogene 1, receptor tyrosine kinase (ROS1) and MET proto-oncogene, receptor tyrosine kinase (MET). Unfortunately, hepatotoxicity is a serious limitation in its clinical application, and the reason remains largely unknown. In this study, we tested the effect of crizotinib in human hepatocyte cell line HL-7702 and human primary hepatocytes, and the results showed that crizotinib treatment caused hepatocyte damage, suggesting that crizotinib induced liver injury by causing hepatocyte death, consistent with the clinical cases. Mechanistically, crizotinib induced hepatocyte death via the apoptotic pathway, and cleaved PARP (c-PARP) was observed as a signaling protein. Moreover, mitochondrial membrane potential (MMP) decrease contributed to crizotinib-induced hepatocyte apoptosis accompanied by hepatocyte DNA damage and reactive oxygen species (ROS) generation. Importantly, crizotinib induced hepatocyte apoptosis independent of its targets, ALK, ROS1 and MET. In conclusion, our data showed that crizotinib induced liver injury through hepatocyte death via the apoptotic pathway which was independent of ALK, ROS1 and MET. And we also found that MMP decrease, DNA damage and ROS generation were involved in the process.
- Published
- 2019
16. Regulation of p53 stability as a therapeutic strategy for cancer
- Author
-
Yuhuai Hu, Wentong Wu, Hao Yan, Qiaojun He, Peihua Luo, and Zhifei Xu
- Subjects
0301 basic medicine ,Mutant ,Antineoplastic Agents ,medicine.disease_cause ,Biochemistry ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Neoplasms ,Autophagy ,medicine ,Animals ,Humans ,Pharmacology ,biology ,Protein Stability ,Neurodegeneration ,Ubiquitination ,Wild type ,Cancer ,medicine.disease ,030104 developmental biology ,Proteasome ,030220 oncology & carcinogenesis ,Mutation ,Proteolysis ,Cancer research ,biology.protein ,Tumor Suppressor Protein p53 ,Carcinogenesis - Abstract
The tumor suppressor protein p53 participates in the control of key biological functions such as cell death, metabolic homeostasis and immune function, which are closely related to various diseases such as tumors, metabolic disorders, infection and neurodegeneration. The p53 gene is also mutated in approximately 50% of human cancer cells. Mutant p53 proteins escape from the ubiquitination-dependent degradation, gain oncogenic function and promote the carcinogenesis, malignant progression, metastasis and chemoresistance. Therefore, the stability of both wild type and mutant p53 needs to be precisely regulated to maintain normal functions and targeting the p53 stability is one of the therapeutic strategies against cancer. Here, we focus on compound-induced degradation of p53 by both the ubiquitination-dependent proteasome and autophagy-lysosome degradation pathways. We also review other posttranslational modifications which control the stability of p53 and the biological functions involved in these processes. This review provides the current theoretical basis for the regulation of p53 abundance and its possible applications in different diseases.
- Published
- 2021
17. Bisdemethoxycurcumin attenuates cisplatin-induced renal injury through anti-apoptosis, anti-oxidant and anti-inflammatory
- Author
-
Zhifei Xu, Qiaojun He, Hao Yan, Fuquan Jin, Peihua Luo, Bo Yang, and Xueqin Chen
- Subjects
Male ,0301 basic medicine ,NF-E2-Related Factor 2 ,medicine.drug_class ,Anti-Inflammatory Agents ,Antineoplastic Agents ,Apoptosis ,Pharmacology ,Kidney ,medicine.disease_cause ,Antioxidants ,Anti-inflammatory ,Cell Line ,Nephrotoxicity ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Diarylheptanoids ,Bisdemethoxycurcumin ,Animals ,Humans ,Medicine ,Chemokine CCL2 ,Cisplatin ,Mice, Inbred ICR ,business.industry ,Monocyte ,Transcription Factor RelA ,Intercellular Adhesion Molecule-1 ,Oxidative Stress ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Kidney Diseases ,Tumor Suppressor Protein p53 ,business ,030217 neurology & neurosurgery ,Oxidative stress ,medicine.drug - Abstract
Cisplatin is a widely used chemotherapy drug that is first-line therapy for a variety of tumors. Unfortunately, its adverse effects on various normal tissues and organs, especially nephrotoxicity, threaten the life of patients. Although the mechanism of cisplatin nephrotoxicity has been confirmed to be related to oxidative stress, apoptosis of renal tubular epithelial cells and inflammatory response, there is no effective prevention strategy in the clinic. Here, we found that bisdemethoxycurcumin (BDMC), a natural compound, can significantly attenuates cisplatin-induced apoptosis of renal tubular epithelial cells in vitro at the concentration of 5–20 μM and has a significant protective effect on cisplatin-induced kidney injury in mice at the dose of 50 mg/kg. Mechanistically, BDMC attenuates cisplatin-induced apoptosis of renal tubular epithelial cells by inhibiting cisplatin-induced up-regulation of p53. Meanwhile, BDMC counteracts oxidative stress by inhibiting cisplatin-induced down-regulation of nuclear factor erythroid-2-related factor 2 (Nrf2). BDMC also significantly reduced the expression of intercellular adhesion molecule-1 (ICAM-1) and monocyte chemoattractant protein-1 (MCP-1) proteins, as well as the expression and translocation of the p65 subunit of nuclear factor-κB (NF-κB p65) into the nucleus, all of which were increased in the kidney by cisplatin treatment. Collectively, BDMC might be an effective prevention strategy which could against cisplatin-induced nephrotoxicity, and our research may shed a new light on treatment of drug toxicity.
- Published
- 2020
18. All-trans retinoic acid synergizes with topotecan to suppress AML cells via promoting RARα-mediated DNA damage
- Author
-
Qiaojun He, Bo Yang, Hao Yan, Guanqun Li, Peihua Luo, Jinjin Shao, Min Chen, Zhifei Xu, Lin Li, and Xueming Peng
- Subjects
0301 basic medicine ,Cancer Research ,Receptors, Retinoic Acid ,HL60 ,DNA damage ,Retinoic acid ,Apoptosis ,Tretinoin ,Pharmacology ,Biology ,Small hairpin RNA ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,AML ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,Genetics ,medicine ,Animals ,Humans ,RNA, Small Interfering ,neoplasms ,All-trans retinoic acid ,Gene Expression Regulation, Leukemic ,Retinoic Acid Receptor alpha ,Cell Differentiation ,Drug Synergism ,Xenograft Model Antitumor Assays ,RARα ,Comet assay ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Oncology ,chemistry ,Retinoic acid receptor alpha ,030220 oncology & carcinogenesis ,Cancer research ,Topotecan ,Research Article ,medicine.drug - Abstract
Background Chemotherapy is the only therapy option for the majority of AML patients, however, there are several limitations for this treatment. Our aim was to find a new chemotherapy strategy that is more effective and less toxic. Methods MTT assays and a xenograft mouse model were employed to evaluate the synergistic activity of all-trans retinoic acid (ATRA) combined with topotecan (TPT). Drug-induced DNA damage and apoptosis were determined by flow cytometry analysis with PI and DAPI staining, the comet assay and Western blots. Short hairpin RNA (shRNA) and a RARα plasmid were used to determine whether RARα expression influenced DNA damage and apoptosis. Results We found that ATRA exhibited synergistic activity in combination with Topotecan in AML cells, and the enhanced apoptosis induced by Topotecan plus ATRA resulted from caspase pathway activation. Mechanistically, ATRA dramatically down regulated RARα protein levels and led to more DNA damage and ultimately resulted in the synergism of these two agents. In addition, the increased antitumor efficacy of Topotecan combined with ATRA was further validated in the HL60 xenograft mouse model. Conclusions Our data demonstrated, for the first time, that the combination of TPT and ATRA showed potential benefits in AML, providing a novel insight into clinical treatment strategies. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-2010-6) contains supplementary material, which is available to authorized users.
- Published
- 2016
19. Oxidative stress is involved in Dasatinib-induced apoptosis in rat primary hepatocytes
- Author
-
Qiaojun He, Hong Zhu, Youping Wu, Renhua Gai, Yuqin Zhao, Honghai Wu, Xiaochun Yang, Peihua Luo, Tao Xue, and Bo Yang
- Subjects
Male ,Indoles ,Blotting, Western ,Dasatinib ,Apoptosis ,Mitochondria, Liver ,Cell Separation ,Pharmacology ,Toxicology ,medicine.disease_cause ,Antioxidants ,Rats, Sprague-Dawley ,Superoxide dismutase ,Lipid peroxidation ,chemistry.chemical_compound ,hemic and lymphatic diseases ,In Situ Nick-End Labeling ,medicine ,Animals ,Protein Kinase Inhibitors ,Fluorescent Dyes ,Membrane Potential, Mitochondrial ,chemistry.chemical_classification ,Reactive oxygen species ,TUNEL assay ,L-Lactate Dehydrogenase ,biology ,Alanine Transaminase ,DNA ,Glutathione ,Malondialdehyde ,Molecular biology ,Acetylcysteine ,Electrophoresis, Gel, Pulsed-Field ,Rats ,Oxidative Stress ,Thiazoles ,Pyrimidines ,chemistry ,Hepatocytes ,biology.protein ,Mitochondrial Swelling ,Reactive Oxygen Species ,Oxidative stress ,medicine.drug - Abstract
Dasatinib, a multitargeted inhibitor of BCR-ABL and SRC kinases, exhibits antitumor activity and extends the survival of patients with chronic myeloid leukemia (CML) and Philadelphia chromosome-positive acute lymphoblastic leukemia (ALL). However, some patients suffer from hepatotoxicity, which occurs through an unknown mechanism. In the present study, we found that Dasatinib could induce hepatotoxicity both in vitro and in vivo. Dasatinib reduced the cell viability of rat primary hepatocytes, induced the release of alanine aminotransferase (ALT) and lactate dehydrogenase (LDH) in vitro, and triggered the ballooning degeneration of hepatocytes in Sprague-Dawley rats in vivo. Apoptotic markers (chromatin condensation, cleaved caspase-3 and cleaved PARP) were detected to indicate that the injury induced by Dasatinib in hepatocytes in vitro was mediated by apoptosis. This result was further validated in vivo using terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assays. Here we found that Dasatinib dramatically increased the level of reactive oxygen species (ROS) in hepatocytes, reduced the intracellular glutathione (GSH) content, attenuated the activity of superoxide dismutase (SOD), generated malondialdehyde (MDA), a product of lipid peroxidation, decreased the mitochondrial membrane potential, and activated nuclear factor erythroid 2-related factor 2 (Nrf2) and mitogen-activated protein kinases (MAPK) related to oxidative stress and survival. These results confirm that oxidative stress plays a pivotal role in Dasatinib-mediated hepatotoxicity. N-acetylcysteine (NAC), a typical antioxidant, can scavenge free radicals, attenuate oxidative stress, and protect hepatocytes against Dasatinib-induced injury. Thus, relieving oxidative stress is a viable strategy for reducing Dasatinib-induced hepatotoxicity.
- Published
- 2012
20. The ubiquitin-proteasome pathway plays essential roles in ATRA-induced leukemia cells G0/G1phase arrest and transition into granulocytic differentiation
- Author
-
Peihua Luo, Qiaojun He, Bo Yang, Meidan Ying, Xinglu Zhou, Like Zhong, Yanfen Fang, Hui Jing, and Meihua Lin
- Subjects
Acute promyelocytic leukemia ,Proteasome Endopeptidase Complex ,Cancer Research ,Cellular differentiation ,Antineoplastic Agents ,HL-60 Cells ,Tretinoin ,Cell Growth Processes ,Resting Phase, Cell Cycle ,Leukemia, Promyelocytic, Acute ,Differentiation therapy ,medicine ,Humans ,neoplasms ,Pharmacology ,biology ,Ubiquitin ,organic chemicals ,Cyclin-dependent kinase 2 ,G1 Phase ,Myeloid leukemia ,Cell Differentiation ,medicine.disease ,biological factors ,Up-Regulation ,Cell biology ,Leukemia ,Oncology ,Proteasome ,biology.protein ,Molecular Medicine ,Granulocytes ,medicine.drug - Abstract
All-trans retinoic acid (ATRA) has been successfully used in differentiation therapy for acute promyelocytic leukemia (APL) in the clinic. ATRA-induced differentiation of leukemia cells is accompanied by a G0/G1 arrest, yet how ATRA couples cell cycle arrest to differentiation remains largely unknown. Here we observed that the ubiquitin-proteasome pathway (UPP) was activated upon ATRA treatment in the human acute myeloid leukemia cell lines, NB4 and HL-60, as represented by the accumulation of ubiquitinated proteins, the up-regulation of ubiquitin mRNA and increased 20S proteasome activity. Interestingly, we found that complete inhibition of proteasome activity suppressed ATRA-induced proliferation/differentiation (P/D) transition in both cell lines. Furthermore, we demonstrate that the exact protein contributing to this phenomenon is different in these two cell lines. Cyclin-dependent kinase 2 (CDK2) and Cyclin E were degraded by the UPP; they accumulated significantly after complete inhibition of the proteasome in ATRA-treated NB4 and HL-60 cells, respectively. These findings suggested that the UPP might be indispensable in the ATRA-induced G0/G1 arrest and differentiation of leukemia cells. The exact protein degraded by the UPP to promote the myeloid maturation program set in motion by the retinoid may be cell type dependent.
- Published
- 2010
21. Antimetastatic activity of MONCPT in preclinical melanoma mice model
- Author
-
Hong Zhu, Wei Lu, Peihua Luo, Xinglu Zhou, Difeng Zhu, Honghai Wu, Qiaojun He, Chongxing Tu, Bo Yang, and Xiaochun Yang
- Subjects
Vascular Endothelial Growth Factor A ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,Green Fluorescent Proteins ,Cell Separation ,Biology ,Metastasis ,Mice ,Cell Movement ,Transduction, Genetic ,In vivo ,Cell Line, Tumor ,Cell Adhesion ,medicine ,Animals ,Neoplasm Invasiveness ,Pharmacology (medical) ,Neoplasm Metastasis ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Melanoma ,Cell Proliferation ,Pharmacology ,Cell growth ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,Cell Hypoxia ,In vitro ,Mice, Inbred C57BL ,Disease Models, Animal ,Vascular endothelial growth factor A ,Oncology ,Cell culture ,Cancer research ,Camptothecin ,Female ,Drug Screening Assays, Antitumor ,Proto-Oncogene Proteins c-akt ,Signal Transduction ,medicine.drug - Abstract
Previous study demonstrated that MONCPT, a topoisomerase I inhibitor, exhibited potent anti-proliferation and anti-angiogenesis activity in vitro and in vivo. In this study, we report the efficacy of MONCPT against the development of melanoma metastasis by an intravenous injection of green fluorescent protein-transfected mice melanoma carcinoma (B16F10-GFP) cells in C57BL/6 mice. MONCPT (2.0, 5.0 and 12.5 mg/kg/2 days) markedly decreased B16F10-GFP pulmonary metastases by 12.8%, 53.1% and 76.3%, respectively; whereas higher doses of MONCPT (31.0 mg/kg/2 days) significantly inhibited the tumor growth of B16F10 xenograft model. In the in vitro experiment, MONCPT suppressed the B16F10-GFP cell invasion and migration without affecting cell survival. Further studies demonstrated that MONCPT decreased the secretion of matrix metalloproteinase (MMP)-9 and VEGF, and reduced the protein expression of HIF-1α as well as the phosphorylation level of ERK in B16F10-GFP cells. These in vivo and in vitro results indicate that MONCPT possesses both the potent antimetastatic ability and the tumor growth-inhibition activity, and the dual function promises MONCPT as a potential therapeutic agent for tumor metastasis and tumor growth of melanoma carcinoma.
- Published
- 2009
22. Autophagy protects against dasatinib-induced hepatotoxicity via p38 signaling
- Author
-
Chao Chen, Shenglin Ma, Xiaochun Yang, Jinjin Shao, Jian Ma, Bo Yang, Jincheng Wang, Peihua Luo, Jiabin Dai, and Qiaojun He
- Subjects
Agonist ,medicine.drug_class ,MAP Kinase Signaling System ,p38 mitogen-activated protein kinases ,Dasatinib ,Mice, Nude ,Antineoplastic Agents ,Pharmacology ,Biology ,medicine.disease_cause ,p38 Mitogen-Activated Protein Kinases ,Mice ,Random Allocation ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Autophagy ,Gene silencing ,Animals ,Humans ,Drug Interactions ,Pharmaceutical sciences ,Liver injury ,Mice, Inbred BALB C ,Mice, Inbred ICR ,p38 signaling ,Hepatotoxicity ,Isoproterenol ,medicine.disease ,Oncology ,Hepatocytes ,Female ,Chemical and Drug Induced Liver Injury ,Oxidative stress ,medicine.drug ,Research Paper - Abstract
// Xiaochun Yang 1 , Jincheng Wang 1 , Jiabin Dai 1 , Jinjin Shao 1 , Jian Ma 2 , Chao Chen 2 , Shenglin Ma 3 , Qiaojun He 1 , Peihua Luo 1 and Bo Yang 1 1 Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China 2 Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, China 3 Nanjing Medical University, Affiliated Hangzhou Hospital, Hangzhou First People’s Hospital, Hangzhou, China Correspondence: Peihua Luo, email: // Bo Yang, email: // Keywords : Autophagy, p38 signaling, Hepatotoxicity, Dasatinib Received : October 14, 2014 Accepted : January 14, 2015 Published : January 31, 2015 Abstract Liver dysfunction is a common side effect associated with the treatment of dasatinib and its mechanism is poorly understood. Autophagy has been thought to be a potent survival or death factor for liver dysfunction, which may shed the light on a novel strategy for the intervention of hepatotoxicity caused by dasatinib. In this study, we show for the first time that autophagy is induced, which is consistent with the formation of liver damage. Autophagy inhibition exacerbated dasatinib-induced liver failure, suggesting that autophagy acted as a self-defense mechanism to promote survival. Oxidative stress has been shown to be an important stimulus for autophagy and hepatotoxicity. Interestingly, dasatinib increased the activity of p38, which is a critical modulator of the oxidative stress related to liver injury and autophagy. p38 silencing significantly blocked LC3-II induction and p62 reduction by dasatinib, which was accompanied by increased caspase-3 and PARP cleavage, indicating that autophagy alleviated dasatinib-induced hepatotoxicity via p38 signaling. Finally, the p38 agonist isoproterenol hydrochloride (ISO) alleviated dasatinib-induced liver failure by enhancing autophagy without affecting the anticancer activity of dasatinib. Thus, this study revealed that p38-activated autophagy promoted survival during liver injury, which may provide novel approaches for managing the clinical applications of dasatinib.
- Published
- 2014
23. Dihydromyricetin prevents cardiotoxicity and enhances anticancer activity induced by adriamycin
- Author
-
Hong Zhu, Bo Yang, Qinjie Weng, Jiabin Dai, Yingying Fu, Jincheng Wang, Peihua Luo, Qiaojun He, Lin-Lin Chang, Jinjin Shao, and Xiaochun Yang
- Subjects
Time Factors ,Flavonols ,Apoptosis ,Pharmacology ,medicine.disease_cause ,Rats, Sprague-Dawley ,Myocytes, Cardiac ,Cardioprotection ,Mice, Inbred BALB C ,Mice, Inbred ICR ,Antibiotics, Antineoplastic ,Dihydromyricetin ,Proto-Oncogene Proteins c-mdm2 ,U937 Cells ,Cytoprotection ,ARC ,Tumor Burden ,Oncology ,Lymphoma, Large B-Cell, Diffuse ,medicine.drug ,Signal Transduction ,Research Paper ,Anthracycline ,Heart Diseases ,cardiotoxicity ,HL-60 Cells ,Nerve Tissue Proteins ,Protective Agents ,anticancer ,Adriamycin ,medicine ,Animals ,Humans ,Doxorubicin ,Cardioprotective Agent ,Cell Proliferation ,Cardiotoxicity ,Dose-Response Relationship, Drug ,business.industry ,Xenograft Model Antitumor Assays ,Rats ,Cytoskeletal Proteins ,Oxidative Stress ,Animals, Newborn ,Tumor Suppressor Protein p53 ,business ,K562 Cells ,Oxidative stress - Abstract
Adriamycin, a widely used anthracycline antibiotic in multiple chemotherapy regimens, has been challenged by the cardiotoxicity leading to fatal congestive heart failure in the worst condition. The present study demonstrated that Dihydromyricetin, a natural product extracted from ampelopsis grossedentat, exerted cardioprotective effect against the injury in Adriamycin-administrated ICR mice. Dihydromyricetin decreased ALT, LDH and CKMB levels in mice serum, causing a significant reduction in the toxic death triggered by Adriamycin. The protective effects were also indicated by the alleviation of abnormal electrocardiographic changes, the abrogation of proliferation arrest and apoptotic cell death in primary myocardial cells. Further study revealed that Dihydromyricetin-rescued loss of anti-apoptosis protein ARC provoked by Adriamycin was involved in the cardioprotection. Intriguingly, the anticancer activity of Adriamycin was not compromised upon the combination with Dihydromyricetin, as demonstrated by the enhanced anticancer effect achieved by Adriamycin plus Dihydromyricetin in human leukemia U937 cells and xenograft models, in a p53-dependent manner. These results collectively promised the potential value of Dihydromyricetin as a rational cardioprotective agent of Adriamycin, by protecting myocardial cells from apoptosis, while potentiating anticancer activities of Adriamycin, thus further increasing the therapeutic window of the latter one.
- Published
- 2014
24. Autophagy blockade sensitizes the anticancer activity of CA-4 via JNK-Bcl-2 pathway
- Author
-
Qiaojun He, Xiaochun Yang, Yangling Li, Jiabin Dai, Honghai Wu, Jincheng Wang, Peihua Luo, and Bo Yang
- Subjects
Programmed cell death ,MAP Kinase Signaling System ,ATG5 ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Biology ,Toxicology ,Piperazines ,Autophagy-Related Protein 5 ,Nitrophenols ,chemistry.chemical_compound ,Mice ,Cell Line, Tumor ,Stilbenes ,Autophagy ,Animals ,Humans ,Phosphorylation ,RNA, Small Interfering ,Pharmacology ,Combretastatin ,Sulfonamides ,Adenine ,Biphenyl Compounds ,Membrane Proteins ,Xenograft Model Antitumor Assays ,Cell biology ,chemistry ,Proto-Oncogene Proteins c-bcl-2 ,Cancer cell ,Beclin-1 ,Macrolides ,Apoptosis Regulatory Proteins ,Microtubule-Associated Proteins ,Intracellular - Abstract
Combretastatin A-4 (CA-4) has already entered clinical trials of solid tumors over ten years. However, the limited anticancer activity and dose-dependent toxicity restrict its clinical application. Here, we offered convincing evidence that CA-4 induced autophagy in various cancer cells, which was demonstrated by acridine orange staining of intracellular acidic vesicles, the degradation of p62, the conversion of LC3-I to LC3-II and GFP-LC3 punctate fluorescence. Interestingly, CA-4-mediated apoptotic cell death was further potentiated by pretreatment with autophagy inhibitors (3-methyladenine and bafilomycin A1) or small interfering RNAs against the autophagic genes (Atg5 and Beclin 1). The enhanced anticancer activity of CA-4 and 3-MA was further confirmed in the SGC-7901 xenograft tumor model. These findings suggested that CA-4-elicited autophagic response played a protective role that impeded the eventual cell death while autophagy inhibition was expected to improve chemotherapeutic efficacy of CA-4. Meanwhile, CA-4 treatment led to phosphorylation/activation of JNK and JNK-dependent phosphorylation of Bcl-2. Importantly, JNK inhibitor or JNK siRNA inhibited autophagy but promoted CA-4-induced apoptosis, indicating a key requirement of JNK-Bcl-2 pathway in the activation of autophagy by CA-4. We also identified that pretreatment of Bcl-2 inhibitor (ABT-737) could significantly enhance anticancer activity of CA-4 due to inhibition of autophagy. Taken together, our data suggested that the JNK-Bcl-2 pathway was considered as the critical regulator of CA-4-induced protective autophagy and a potential drug target for chemotherapeutic combination.
- Published
- 2013
25. Bortezomib sensitizes human acute myeloid leukemia cells to all-trans-retinoic acid-induced differentiation by modifying the RARα/STAT1 axis
- Author
-
Like Zhong, Nengming Lin, Meidan Ying, Qiaojun He, Hui Jing, Hua Song, Bo Yang, Peihua Luo, Xiaochun Yang, and Xinglu Zhou
- Subjects
Acute promyelocytic leukemia ,Cancer Research ,Myeloid ,Receptors, Retinoic Acid ,Cellular differentiation ,Mice, Nude ,Apoptosis ,HL-60 Cells ,Tretinoin ,Biology ,Pharmacology ,Bortezomib ,Mice ,Random Allocation ,Differentiation therapy ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,neoplasms ,Cell Proliferation ,Retinoic Acid Receptor alpha ,Myeloid leukemia ,Cell Differentiation ,Drug Synergism ,medicine.disease ,Boronic Acids ,Xenograft Model Antitumor Assays ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,STAT1 Transcription Factor ,Oncology ,Pyrazines ,Proteasome inhibitor ,medicine.drug - Abstract
All-trans-retinoic acid (ATRA) has held great promise for differentiation-based therapy but reportedly downregulates retinoic acid receptor-α (RARα) in a proteasome-dependent manner, which leads to decreased acute myeloid leukemia (AML) cell differentiation efficiency. Therefore, research strategies that seek to further sensitize cells to retinoids and extend the range of retinoid-affected myeloid malignancies beyond acute promyelocytic leukemia (APL) are key investigative avenues. Here, we show that bortezomib, the first proteasome inhibitor approved for newly diagnosed and relapsed multiple myeloma, exhibited strong synergism with ATRA to promote HL60 and NB4 AML cell differentiation. We observed that bortezomib sensitized AML cells to ATRA-induced morphologic, biochemical, and functional changes, indicative of myeloid differentiation without cell death. In addition, treatment of human leukemia HL60 xenografts with bortezomib and ATRA together did not increase bortezomib-induced progressive weight loss but resulted in significant tumor growth inhibition in addition to increased differentiation (P < 0.05). These enhanced differentiation effects were accompanied by RARα stabilization and STAT1 activation. Taken together, our study was the first to evaluate bortezomib and ATRA synergy in AML cell differentiation and to assess new opportunities for bortezomib and ATRA combination as a promising approach for future differentiation therapy. Mol Cancer Ther; 12(2); 195–206. ©2012 AACR.
- Published
- 2012
26. Gefitinib Synergizes with Irinotecan to Suppress Hepatocellular Carcinoma via Antagonizing Rad51-Mediated DNA-Repair
- Author
-
Yuanrun Zhu, Qiaojun He, Min Chen, Hong Zhu, Li Xu, Zhifei Xu, Xueming Peng, Jinjin Shao, Peihua Luo, and Bo Yang
- Subjects
0301 basic medicine ,DNA Repair ,medicine.medical_treatment ,lcsh:Medicine ,Apoptosis ,Pharmacology ,Mice ,0302 clinical medicine ,heterocyclic compounds ,RNA, Small Interfering ,lcsh:Science ,Multidisciplinary ,Liver Neoplasms ,Drug Synergism ,Gefitinib ,Hep G2 Cells ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Research Article ,medicine.drug ,Carcinoma, Hepatocellular ,DNA damage ,DNA repair ,Blotting, Western ,Mice, Nude ,Antineoplastic Agents ,Irinotecan ,03 medical and health sciences ,Cell Line, Tumor ,Carcinoma ,medicine ,Animals ,Humans ,neoplasms ,Cell Proliferation ,Chemotherapy ,business.industry ,lcsh:R ,medicine.disease ,digestive system diseases ,030104 developmental biology ,Drug Resistance, Neoplasm ,Quinazolines ,Camptothecin ,lcsh:Q ,Rad51 Recombinase ,Topoisomerase I Inhibitors ,business ,Neoplasm Transplantation - Abstract
Chemotherapy is the only choice for most of the advanced hepatocellular carcinoma (HCC) patients, while few agents were available, making it an urgent need to develop new chemotherapy strategies. A phase II clinical trial suggested that the efficacy of irinotecan in HCC was limited due to dose-dependent toxicities. Here, we found that gefitinib exhibited synergistic activity in combination with SN-38, an active metabolite of irinotecan, in HCC cell lines. And the enhanced apoptosis induced by gefitinib plus SN-38 was a result from caspase pathway activation. Mechanistically, gefitinib dramatically promoted the ubiquitin–proteasome-dependent degradation of Rad51 protein, suppressed the DNA repair, gave rise to more DNA damages, and ultimately resulted in the synergism of these two agents. In addition, the increased antitumor efficacy of gefitinib combined with irinotecan was further validated in a HepG2 xenograft mice model. Taken together, our data demonstrated for the first time that the combination of irinotecan and gefitinib showed potential benefit in HCC, which suggests that Rad51 is a promising target and provides a rationale for clinical trials investigating the efficacy of the combination of topoisomerase I inhibitors and gefitinib in HCC.
- Published
- 2016
27. The proteasome inhibitor bortezomib enhances ATRA-induced differentiation of neuroblastoma cells via the JNK mitogen-activated protein kinase pathway
- Author
-
Bo Yang, Peihua Luo, Lin Li, Qiaojun He, and Meili Lin
- Subjects
MAP Kinase Signaling System ,Cellular differentiation ,Science ,Cell ,Cancer Treatment ,Tretinoin ,Biology ,Pharmacology ,Bortezomib ,Mice ,Neuroblastoma ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,Basic Cancer Research ,medicine ,Animals ,Humans ,MTT assay ,Protease Inhibitors ,Neurological Tumors ,neoplasms ,Multidisciplinary ,Cell growth ,JNK Mitogen-Activated Protein Kinases ,Cancers and Neoplasms ,Cell Differentiation ,Drug Synergism ,Chemotherapy and Drug Treatment ,medicine.disease ,Boronic Acids ,Xenograft Model Antitumor Assays ,Rats ,medicine.anatomical_structure ,Oncology ,Mitogen-activated protein kinase ,Pyrazines ,biology.protein ,Proteasome inhibitor ,Medicine ,medicine.drug ,Research Article - Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in childhood. Differentiated human NBs are associated with better outcome and lower stage; induction of differentiation is considered to be therapeutically advantageous. All-trans retinoic acid (ATRA) has been shown to induce the differentiation of neuroblastoma (NB) cell lines. The proteasome inhibitor bortezomib inhibits cell growth and angiogenesis in NBs. Here, we investigated the synergistic effect between bortezomib and ATRA in inducing NB cell differentiation in different NB cell lines. Bortezomib combined with ATRA had a significantly enhanced antiproliferative effect. This inhibition was characterized by a synergistic increase in neuronal differentiation. At the same time, the combination therapy showed little neuronal toxicity which was assessed in primary cultures of rat cerebellar granule cells by the MTT assay, PI staining. The combination of bortezomib and ATRA triggered increased differentiation through the activation of proteins, including RARα, RARβ, RARγ, p-JNK and p21, compared with ATRA treatment alone. Using JNK inhibitor SP600125 to block JNK-dependent activity, the combination therapy-induced neuronal differentiation was partially attenuated. In addition, p21 shRNA had no effect on the combination therapy-induced neuronal differentiation. The in vivo antitumor activities were examined in human NB cell xenografts and GFP-labeled human NB cell xenografts. Treatment of human NB cell CHP126-bearing nude mice with ATRA plus bortezomib resulted in more significant tumor growth inhibition than mice treated with either drug alone. These findings provide the rationale for the development of a new therapeutic strategy for NB based on the pharmacological combination of ATRA and bortezomib.
- Published
- 2011
28. Potent antitumor activity of 10-methoxy-9-nitrocamptothecin
- Author
-
Yongzhou Hu, Peihua Luo, Bo Yang, Xungui He, Wei Lu, Qiaojun He, and Yiyu Cheng
- Subjects
Male ,Cancer Research ,Lung Neoplasms ,Cell Survival ,Transplantation, Heterologous ,Pharmacology ,Biology ,Mice ,In vivo ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Cytotoxicity ,Cyclin-dependent kinase 1 ,Cell Cycle ,Prostatic Neoplasms ,Cell cycle ,Antineoplastic Agents, Phytogenic ,In vitro ,Transplantation ,Oncology ,Cell culture ,Camptothecin ,medicine.drug - Abstract
The present data showed that 10-methoxy-9-nitrocamptothecin (MONCPT), a family of camptothecin analogues, possessed high antitumor activity in vitro and in vivo. Cytotoxicity assays showed that MONCPT was a potential and highly efficient antitumor compound with IC50 values of 0.1 to 500 nmol/L in nine tumor cell lines. The high cytotoxic potency of MONCPT was paralleled with its ability to increase the cellular accumulation of DNA damage. DNA relaxation assay also showed that MONCPT exerted high potency as a topoisomerase I inhibitor. Moreover, administration of MONCPT (5–20 mg/kg) for 15 to 17 days significantly inhibited tumor growth in human androgen–independent prostate tumor (PC3) and human non–small cell lung tumor (A549) xenografts; the inhibition rates ranged from 29.6% to 98%. The cytotoxic effect of 1,000 nmol/L of MONCPT in PC3 cells was associated with causing an arrest in G0-G1 phase, whereas that of 10 and 100 nmol/L MONCPT was relative to a persistent block in G2-M phase. Furthermore, down-regulation of CDK2, CDK4, and cyclin D1 was observed in PC3 cells treated with 1,000 nmol/L of MONCPT, whereas overexpression of CDK7, CDK1, and cyclin B1 was seen in PC3 cells treated with 10 and 100 nmol/L of MONCPT. These results suggested that cell cycle regulation might contribute to the anticancer properties of MONCPT and strongly support the further anticancer development of MONCPT. [Mol Cancer Ther 2006;5(4):962–8]
- Published
- 2006
29. Antiangiogenesis response of endothelial cells to the antitumour drug 10-methoxy-9-nitrocamptothecin
- Author
-
Xiaochun Yang, Qiaojun He, Bo Yang, and Peihua Luo
- Subjects
Angiogenesis ,Angiogenesis Inhibitors ,Apoptosis ,Chick Embryo ,Pharmacology ,Topoisomerase-I Inhibitor ,Biology ,Flow cytometry ,Cell Line ,Cell Movement ,medicine ,Animals ,Humans ,MTT assay ,Enzyme Inhibitors ,Cell Proliferation ,Tube formation ,Matrigel ,medicine.diagnostic_test ,Endothelial Cells ,Molecular biology ,Chorioallantoic membrane ,Camptothecin ,Topoisomerase I Inhibitors - Abstract
10-Methoxy-9-nitrocamptothecin (MONCPT), a topoisomerase I inhibitor, exhibited high anticancer activity in solid tumour xenograft animal models in our previous study. We hypothesized that this phenomenon was associated with antiangiogenesis response. In the present study, we found that MONCPT exhibited high antiproliferation action in human EA.hy926 endothelial cells and the IC 50 value was 0.13 ± 0.04 μM (MTT assay). With AO/EB stain, MONCPT (50–5000 nM)-mediated apoptosis was observed in EA.hy926 cells, and the similar results were shown in flow cytometry assay, the percentage of apoptotic cells induced by MONCPT (50–5000 nM) was 9.2–58.5%. In Chick embryo chorioallantoic membrane (CAM) assay, MONCPT (1–5 μg) resulted in a dose-dependent angiogenic inhibition. In addition, MONCPT significantly inhibited chemotactic-migration invasion on gelatin and tube formation on Matrigel of HUVECs. These results suggest that MONCPT has potential property for inhibiting angiogenesis which is involved in its antitumour activity.
- Published
- 2006
30. Erratum to: Antimetastatic activity of MONCPT in preclinical melanoma mice model
- Author
-
Honghai Wu, Peihua Luo, Xinglu Zhou, Bo Yang, Xiaochun Yang, Hong Zhu, Wei Lu, Qiaojun He, Chongxing Tu, and Difeng Zhu
- Subjects
Pharmacology ,Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,Melanoma ,Pharmacology toxicology ,medicine ,Pharmacology (medical) ,Pharmaceutical sciences ,business ,medicine.disease - Abstract
The online version of the original article can be found at http://dx.doi. org/10.1007/s10637-009-9323-8. X.-C. Yang : C.-X. Tu : P.-H. Luo :H. Zhu :D.-F. Zhu : H.-H. Wu :X.-L. Zhou :Q.-J. He (*) : B. Yang (*) Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China e-mail: qiaojunhe@zju.edu.cn e-mail: yang924@zju.edu.cn
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.