1
|
Wang L, Jia J, Yu X, Luo M, Li J, Khan GJ, He C, Duan H, Zhai K. Molecular mechanism of rapamycin-induced autophagy activation to attenuate smoking-induced COPD. Biochem Biophys Res Commun 2025; 764:151819. [PMID: 40253907 DOI: 10.1016/j.bbrc.2025.151819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/25/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the severe lung and respiratory airway disorders, with high prevalence rate in China. In this paper, we employed network pharmacology predictions to identify autophagy as a signaling pathway associated with COPD. To explore the protective effect of autophagy against COPD and its specific mechanism, we established a mouse model of COPD and administered 3-methyladenine (3-MA) and rapamycin (RAPA) to intervene in autophagy. The lung function of the mice was assessed using an animal pulmonary function analysis system, and lung tissue structure was evaluated through hematoxylin and eosin (HE) staining. The TUNEL staining method was employed to determine the level of apoptosis in lung tissue. Western blot analysis was conducted to measure the expression of autophagy and apoptosis-related proteins, while RT-qPCR was used to assess the expression of apoptosis-related mRNA. The results showed that RAPA effectively improved lung function, attenuated pathological lung injury and increased autophagy level in COPD mice. Apoptosis analysis showed that the apoptosis rate was elevated in COPD and 3- MA mice, whereas it was significantly reduced in RAPA mice. Our findings suggest that stimulation of autophagy may be a potential therapy for the future treatment of COPD.
Collapse
Affiliation(s)
- Li Wang
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Jianhu Jia
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Xinyan Yu
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui, 234000, China
| | - Mengmeng Luo
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui, 234000, China; College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui, 241000, China
| | - Jie Li
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui, 234000, China; School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241002, China
| | - Ghulam Jilany Khan
- Department of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, 54000, Pakistan
| | - Chenghui He
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China; Xinjiang Institute of Materia Medica, Key Laboratory of Xinjiang Uygur Medicine, Urumqi, 830004, China.
| | - Hong Duan
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China; School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui, 234000, China.
| | - Kefeng Zhai
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China; School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui, 234000, China.
| |
Collapse
|
2
|
Yuan R, Xu H, Wang M, Guo L, Yao Y, Zhang X, Wang X. Promoting the transition from pyroptosis to apoptosis in endothelial cells: a novel approach to alleviate methylglyoxal-induced vascular damage. J Transl Med 2025; 23:170. [PMID: 39930472 PMCID: PMC11809013 DOI: 10.1186/s12967-025-06195-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/01/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Methylglyoxal (MGO)-induced cell death in vascular endothelial cells (VECs) plays a critical role in the progression of diabetic vascular complications (DVCs). Previous studies have shown that MGO can induce inflammatory pyroptosis, leading to VEC damage. However, the underlying mechanism remains unclear, and effective interventions are yet to be developed. METHODS Human umbilical vein endothelial cells (HUVECs) were used for in vitro experiments. Cell death modes were assessed through morphological observations. Mechanistic investigations were performed using immunofluorescence, flow cytometry, Western blotting, and ELISA. Inhibitors and adenoviruses were employed to validate the mechanisms. Vascular organoids in conjunction with AngioTool plug-in assays were used to evaluate VEC damage and angiogenic capacity. Mouse blood pressure was measured using the tail-cuff method, and vascular morphology was examined through hematoxylin and eosin (H&E) staining as well as immunofluorescence staining. Data were analyzed using the GraphPad Prism software. RESULTS Our study revealed that MGO induces pyroptosis in VECs via the Caspase3/gasdermin E (GSDME) pathway. Furthermore, the saponin monomer 13 of dwarf lilyturf tuber (DT-13), inhibited MGO-induced pyroptosis and promoted the generation of apoptotic bodies, facilitating the transition from pyroptosis to apoptosis. Mechanistically, DT-13 suppressed the Caspase3-mediated cleavage of GSDME and non-muscle myosin heavy chain IIA (NMMHC IIA), while increasing the phosphorylation of myosin light chain 2 (MLC2), which facilitated apoptotic body formation. Additionally, DT-13 was shown to mitigate VEC damage, inhibit angiogenesis, reduce vascular remodeling, and alleviate MGO-induced hypertension. CONCLUSIONS This study uncovers a novel mechanism through which MGO induces VEC damage, highlighting the therapeutic significance of the transition from pyroptosis to apoptosis in this process. These findings suggest potential therapeutic strategies for managing diabetic angiopathy. Furthermore, DT-13 emerges as a promising compound for therapeutic intervention, offering new possibilities for clinical applications.
Collapse
Affiliation(s)
- Ruqiang Yuan
- College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Hu Xu
- Health Science Center, East China Normal University, Shanghai, 200241, China
| | - Mingqi Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Lina Guo
- College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Yang Yao
- College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Xiaoru Zhang
- College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Xiuli Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
3
|
Gou Z, Zhang D, Cao H, Li Y, Li Y, Zhao Z, Wang Y, Wang Y, Zhou H. Exploring the nexus between MYH9 and tumors: novel insights and new therapeutic opportunities. Front Cell Dev Biol 2024; 12:1421763. [PMID: 39149512 PMCID: PMC11325155 DOI: 10.3389/fcell.2024.1421763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024] Open
Abstract
The myosin heavy chain 9 (MYH9) gene, located on human chromosome 22, encodes non-muscle myosin heavy chain IIA (NM IIA). This protein is essential to various cellular events, such as generating intracellular chemomechanical force and facilitating the movement of the actin cytoskeleton. Mutations associated with thrombocytopenia in autosomal dominant diseases first highlighted the significance of the MYH9 gene. In recent years, numerous studies have demonstrated the pivotal roles of MYH9 in various cancers. However, its effects on cancer are intricate and not fully comprehended. Furthermore, the elevated expression of MYH9 in certain malignancies suggests its potential as a target for tumor therapy. Nonetheless, there is a paucity of literature summarizing MYH9's role in tumors and the therapeutic strategies centered on it, necessitating a systematic analysis. This paper comprehensively reviews and analyzes the pertinent literature in this domain, elucidating the fundamental structural characteristics, biological functions, and the nexus between MYH9 and tumors. The mechanisms through which MYH9 contributes to tumor development and its multifaceted roles in the tumorigenic process are also explored. Additionally, we discuss the relationship between MYH9-related diseases (MYH9-RD) and tumors and also summarize tumor therapeutic approaches targeting MYH9. The potential clinical applications of studying the MYH9 gene include improving early diagnosis, clinical staging, and prognosis of tumors. This paper is anticipated to provide novel insights for tumor therapy.
Collapse
Affiliation(s)
- Zixuan Gou
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Difei Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Hongliang Cao
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| | - Yao Li
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Yunkuo Li
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| | - Zijian Zhao
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Ye Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Honglan Zhou
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Raina S, Hübner E, Samuel E, Nagel G, Fuchs H. DT-13 attenuates inflammation by inhibiting NLRP3-inflammasome related genes in RAW264.7 macrophages. Biochem Biophys Res Commun 2024; 708:149763. [PMID: 38503169 DOI: 10.1016/j.bbrc.2024.149763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/21/2024]
Abstract
Plant derived saponins or other glycosides are widely used for their anti-inflammatory, antioxidant, and anti-viral properties in therapeutic medicine. In this study, we focus on understanding the function of the less known steroidal saponin from the roots of Liriope muscari L. H. Bailey - saponin C (also known as DT-13) in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages in comparison to the well-known saponin ginsenoside Rk1 and anti-inflammatory drug dexamethasone. We proved that DT-13 reduces LPS-induced inflammation by inhibiting nitric oxide (NO) production, interleukin-6 (IL-6) release, cycloxygenase-2 (COX-2), tumour necrosis factor-alpha (TNF-α) gene expression, and nuclear factor kappa-B (NFκB) translocation into the nucleus. It also inhibits the inflammasome component NOD-like receptor family pyrin domain containing protein 3 (NLRP3) regulating the inflammasome activation. This was supported by the significant inhibition of caspase-1 and interleukin-1 beta (IL-1β) expression and release. This study demonstrates the anti-inflammatory effect of saponins on LPS-stimulated macrophages. For the first time, an in vitro study shows the attenuating effect of DT-13 on NLRP3-inflammasome activation. In comparison to the existing anti-inflammatory drug, dexamethasone, and triterpenoid saponin Rk1, DT-13 more efficiently inhibits inflammation in the applied cell culture model. Therefore, DT-13 may serve as a lead compound for the development of new more effective anti-inflammatory drugs with minimised side effects.
Collapse
Affiliation(s)
- Shikha Raina
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Augustenburger Platz 1, D-13353, Berlin, Germany
| | - Emely Hübner
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Augustenburger Platz 1, D-13353, Berlin, Germany; Hochschule Bonn-Rhein Sieg, 53359, Rheinbach, Germany; HAN University of Applied Sciences, Groenewoudseweg, 6524, Nijmegen, Netherlands
| | - Esther Samuel
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Augustenburger Platz 1, D-13353, Berlin, Germany
| | - Gregor Nagel
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Augustenburger Platz 1, D-13353, Berlin, Germany
| | - Hendrik Fuchs
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Augustenburger Platz 1, D-13353, Berlin, Germany.
| |
Collapse
|
5
|
Zhan L, Su F, Li Q, Wen Y, Wei F, He Z, Chen X, Yin X, Wang J, Cai Y, Gong Y, Chen Y, Ma X, Zeng J. Phytochemicals targeting glycolysis in colorectal cancer therapy: effects and mechanisms of action. Front Pharmacol 2023; 14:1257450. [PMID: 37693915 PMCID: PMC10484417 DOI: 10.3389/fphar.2023.1257450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common malignant tumor in the world, and it is prone to recurrence and metastasis during treatment. Aerobic glycolysis is one of the main characteristics of tumor cell metabolism in CRC. Tumor cells rely on glycolysis to rapidly consume glucose and to obtain more lactate and intermediate macromolecular products so as to maintain growth and proliferation. The regulation of the CRC glycolysis pathway is closely associated with several signal transduction pathways and transcription factors including phosphatidylinositol 3-kinases/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR), adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), hypoxia-inducible factor-1 (HIF-1), myc, and p53. Targeting the glycolytic pathway has become one of the key research aspects in CRC therapy. Many phytochemicals were shown to exert anti-CRC activity by targeting the glycolytic pathway. Here, we review the effects and mechanisms of phytochemicals on CRC glycolytic pathways, providing a new method of drug development.
Collapse
Affiliation(s)
- Lu Zhan
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fangting Su
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiang Li
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Feng Wei
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhelin He
- Guang’an Hospital of Traditional Chinese Medicine, Guang’an, China
| | - Xiaoyan Chen
- Guang’an Hospital of Traditional Chinese Medicine, Guang’an, China
| | - Xiang Yin
- Guang’an Hospital of Traditional Chinese Medicine, Guang’an, China
| | - Jian Wang
- Guang’an Hospital of Traditional Chinese Medicine, Guang’an, China
| | - Yilin Cai
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuxia Gong
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Chen
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
6
|
He X, Liao Y, Liu J, Sun S. Research Progress of Natural Small-Molecule Compounds Related to Tumor Differentiation. Molecules 2022; 27:2128. [PMID: 35408534 PMCID: PMC9000768 DOI: 10.3390/molecules27072128] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 11/25/2022] Open
Abstract
Tumor differentiation is a therapeutic strategy aimed at reactivating the endogenous differentiation program of cancer cells and inducing cancer cells to mature and differentiate into other types of cells. It has been found that a variety of natural small-molecule drugs can induce tumor cell differentiation both in vitro and in vivo. Relevant molecules involved in the differentiation process may be potential therapeutic targets for tumor cells. Compared with synthetic drugs, natural small-molecule antitumor compounds have the characteristics of wide sources, structural diversity and low toxicity. In addition, natural drugs with structural modification and transformation have relatively concentrated targets and enhanced efficacy. Therefore, using natural small-molecule compounds to induce malignant cell differentiation represents a more targeted and potential low-toxicity means of tumor treatment. In this review, we focus on natural small-molecule compounds that induce differentiation of myeloid leukemia cells, osteoblasts and other malignant cells into functional cells by regulating signaling pathways and the expression of specific genes. We provide a reference for the subsequent development of natural small molecules for antitumor applications and promote the development of differentiation therapy.
Collapse
Affiliation(s)
- Xiaoli He
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; (X.H.); (Y.L.)
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha 410078, China
| | - Yongkang Liao
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; (X.H.); (Y.L.)
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha 410078, China
| | - Jing Liu
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; (X.H.); (Y.L.)
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha 410078, China
| | - Shuming Sun
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; (X.H.); (Y.L.)
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha 410078, China
| |
Collapse
|
7
|
Duan H, Khan GJ, Shang LJ, Peng H, Hu WC, Zhang JY, Hua J, Cassandra A, Rashed MM, Zhai KF. Computational pharmacology and bioinformatics to explore the potential mechanism of Schisandra against atherosclerosis. Food Chem Toxicol 2021; 150:112058. [DOI: 10.1016/j.fct.2021.112058] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/29/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022]
|
8
|
Baig MMFA, Zhang C, Akhtar MF, Saleem A, Mudassir J. The effective transfection of a low dose of negatively charged drug-loaded DNA-nanocarriers into cancer cells via scavenger receptors. J Pharm Anal 2021; 11:174-182. [PMID: 34012693 PMCID: PMC8116213 DOI: 10.1016/j.jpha.2020.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/13/2020] [Accepted: 10/19/2020] [Indexed: 12/26/2022] Open
Abstract
DNA-nanotechnology-based nano-architecture scaffolds based on circular strands were designed in the form of DNA-nanowires (DNA-NWs) as a polymer of DNA-triangles. Circularizing a scaffold strand (84-NT) was the critical step followed by annealing with various staple strands to make stiff DNA-triangles. Atomic force microcopy (AFM), native polyacrylamide gel electrophoresis (PAGE), UV-analysis, MTT-assay, flow cytometry, and confocal imaging were performed to assess the formulated DNA-NWs and cisplatin (CPT) loading. The AFM and confocal microscopy images revealed a uniform shape and size distribution of the DNA-NWs, with lengths ranging from 2 to 4 μm and diameters ranging from 150 to 300 nm. One sharp band at the top of the lane (500 bp level) with the loss of electrophoretic mobility during the PAGE (native) gel analysis revealed the successful fabrication of DNA-NWs. The loading efficiency of CPT ranged from 66.85% to 97.35%. MTT and flow cytometry results showed biocompatibility of the blank DNA-NWs even at 95% concentration compared with the CPT-loaded DNA-NWs. The CPT-loaded DNA-NWs exhibited enhanced apoptosis (22%) compared to the apoptosis (7%) induced by the blank DNA-NWs. The release of CPT from the DNA-NWs was sustained at < 75% for 6 h in the presence of serum, demonstrating suitability for systemic applications. The IC50 of CPT@DNA-NWs was reduced to 12.8 nM CPT, as compared with the free CPT solution exhibiting an IC50 of 51.2 nM. Confocal imaging revealed the targetability, surface binding, and slow internalization of the DNA-NWs in the scavenger-receptor-rich cancer cell line (HepG2) compared with the control cell line.
Collapse
Affiliation(s)
- Mirza Muhammad Faran Ashraf Baig
- Laboratory of Biomedical & Pharmaceutical Engineering of Stem Cells Research, Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, 999077, Hong Kong, PR China
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, PR China
- Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60000, Pakistan
| | - Chengfei Zhang
- Laboratory of Biomedical & Pharmaceutical Engineering of Stem Cells Research, Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, 999077, Hong Kong, PR China
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore, Pakistan
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Jahanzeb Mudassir
- Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60000, Pakistan
| |
Collapse
|
9
|
Baig MMFA, Zhang C, Akhtar MF, Saleem A, Nisar N. Treatment of Wilms’ nephroblastoma cancer cells via EGFR targeting of dactinomycin loaded DNA-nanowires. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2021; 51:233-242. [DOI: 10.1007/s40005-020-00509-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
|
10
|
Huang R, Guo L, Gao M, Li J, Xiang S. Research Trends and Regulation of CCL5 in Prostate Cancer. Onco Targets Ther 2021; 14:1417-1427. [PMID: 33664576 PMCID: PMC7921632 DOI: 10.2147/ott.s279189] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer (PCa) is considered as the most common cancer of urologic neoplasms, and its development and prognosis are associated with many factors. Chemokine receptor signaling combine with advances in advanced clinicopathological characteristics have provided new insights into the molecular landscape of prostate cancer. Chemokine (C-C motif) ligand 5 (CCL5) is an important member of the CC subfamily of chemokines. The expression of chemokine CCL5 is positively correlated with poor prognostic features in patients with PCa. Current study suggested that CCL5/CCR5 axis plays a significant role in the proliferation, metastasis, angiogenesis, drug resistance of prostate cancer cells and promotes self-renewal of prostate cancer stem cells (PCSCs). Due to the major domination in CCL5 by prostate cancer and the high cancer-specific mortality with prostate cancer, research on the CCL5/CCR5 axis effective antagonists is widespread application. However, challenges for precision oncology of CCL5/CCR5 axis and effective antagonists in CRPC remain. Herein, we summarized the crucial role of CCL5 in promoting the development of PCa and discussed the antitumor application of the antagonists of CCL5/CCR5 axis.
Collapse
Affiliation(s)
- Renlun Huang
- Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Lang Guo
- Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Menghan Gao
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Jing Li
- Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Songtao Xiang
- Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
11
|
Baig MMFA, Lai WF, Ashraf S, Saleem A, Akhtar MF, Mikrani R, Naveed M, Siddique F, Taleb A, Mudassir J, Khan GJ, Ansari MT. The integrin facilitated internalization of fibronectin-functionalized camptothecin-loaded DNA-nanofibers for high-efficiency anticancer effects. Drug Deliv Transl Res 2020; 10:1381-1392. [PMID: 32661832 DOI: 10.1007/s13346-020-00820-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Camptothecin (CMPT) in a free form is extremely cytotoxic as well as hydrophobic drug, and is considered to be highly contagious for systemic administration. The fibronectin (FN)-functionalized DNA-based nanocarrier has been designed to load CMPT and target integrin (αvβ3) receptors which are highly expressed on the A549 cancer cells. Here, we report DNA nanocarrier in the form of DNA-nanofibers (DNA-NFs) capable of loading CMPT via strand intercalation in the GC (base pairs)-rich regions of the DNA duplex. Hence, our keen purpose was to explore the potential of DNA-NFs to load CMPT and assess the improvements of the outcomes in terms of enhanced therapeutic effects to integrin-rich A549 cancer cells with reduced cytotoxic effects to integrin-lacking HEK293 cells. DNA-NFs were formulated as a polymer of DNA triangles. DNA triangles arranged in a programmed way through the complementary overhangs present at the vertices. DNA triangles were primarily obtained through the annealing of the freshly circularized scaffold strands with the three distinct staple strands of specific sequences. The polymerized triangular tiles instead of forming two-dimensional nanosheets underwent self-coiling to give rise to DNA-NF-shaped structures. Flow cytometry and MTT assays were performed to observe cytotoxic and apoptotic effects on integrin-rich A549 cancer cells compared with the integrin-deficient HEK293 cells. AFM, native-page, and confocal experiments confirmed the polymerization of DNA triangles and the morphology of the resulting nanostructures. AFM and confocal images revealed the length of DNA-NFs to be 3-6 μm and the width from 70 to 110 nm. CMPT loading (via strands intercalation) in GC-rich regions of DNA-NFs and the FN functionalization (TAMRA tagged; red fluorescence) via amide chemistry using amino-modified strands of DNA-NFs were confirmed through the UV-shift analysis (> 10 nm shift) and confocal imaging. Blank DNA-NFs were found to be highly biocompatible in 2-640 μM concentrations. MTT assay and flow cytometry experiments revealed that CMPT-loaded DNA-NFs showed a dose-dependent decrease in the cell viability to integrin-rich A549 cancer cells compared with the integrin-deficient HEK293 cells. Conclusively, FN-functionalized, CMPT-loaded DNA-NFs effectively destroyed integrin-rich A549 cancer cells in a targeted manner compared with integrin-deficient HEK293 cells. Grapical abstract.
Collapse
Affiliation(s)
- Mirza Muhammad Faran Ashraf Baig
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, People's Republic of China.
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Bahauddin Zakariya University, Multan, 60000, Pakistan.
| | - Wing-Fu Lai
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, People's Republic of China
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Saba Ashraf
- Nishtar Medical University and Hospital, Multan, 60000, Pakistan
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore, Pakistan
| | - Reyaj Mikrani
- School of Basic Medicine, and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, People's Republic of China
| | - Muhammad Naveed
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Farhan Siddique
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Bahauddin Zakariya University, Multan, 60000, Pakistan
| | - Abdoh Taleb
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Jahanzeb Mudassir
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Bahauddin Zakariya University, Multan, 60000, Pakistan
| | - Ghulam Jilany Khan
- Faculty of Pharmacy, University of Central Punjab, Lahore, 54570, Pakistan
| | - Muhammad Tayyab Ansari
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Bahauddin Zakariya University, Multan, 60000, Pakistan
| |
Collapse
|
12
|
DT-13 induced apoptosis and promoted differentiation of acute myeloid leukemia cells by activating AMPK-KLF2 pathway. Pharmacol Res 2020; 158:104864. [PMID: 32416217 DOI: 10.1016/j.phrs.2020.104864] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/08/2020] [Accepted: 04/24/2020] [Indexed: 02/08/2023]
Abstract
Acute myeloid leukemia (AML) is a malignant disease originating from hematopoietic stem cells (HSC). Chemotherapy and/or HSC transplantation is unsatisfactory due to serious side effects, multidrug resistance, and high relapse rate. Thus, alternative strategies are urgently needed to develop more effective therapies. Liriope muscari baily saponins C (DT-13) is a novel compound isolated from Liriope muscari (Decne.) Baily, and exhibited a potent cytotoxicity against several solid tumors. However, the anti-AML activity of DT-13 and the potential mechanisms are still unknown. This study is the first to demonstrate that DT-13 had preferential cytotoxicity against AML cells, and remarkably inhibited proliferation and colony forming ability. Moreover, DT-13 induced the death receptor pathway-dependent apoptosis of HL-60 and Kasumi-1 cells by up-regulating Fas, FasL, DR5 and TRAIL as well as promoted the cleavage of caspase 8, caspase 3 and PARP. Meanwhile, DT-13 induced the differentiation with morphological change related to myeloid differentiation, elevated NBT and α-NAE positive cell rates, differentiation markers CD11b and CD14 as well as level of transcription factors C/EBPα and C/EBPβ. RNA-sequencing analysis revealed that KLF2 may be one of the potential targets regulated by DT-13. Further studies indicated that KLF2 played a critical role in DT-13-induced apoptosis and differentiation. Moreover, activation of AMPK-FOXO was proved to be the upstream of KLF2 pathway that contributed to the induction of apoptosis and differentiation by DT-13. Additionally, restoration of KLF2 by DT-13 was highly correlated with the AMPK-related histone acetylation mechanisms. Finally, DT-13 exhibited an obvious anti-AML effect in NOD/SCID mice with the engraftment of HL-60 cells. Our study suggests that DT-13 may serve as a novel agent for AML by AMPL-KLF2-mediated apoptosis and differentiation.
Collapse
|
13
|
Synthesis of Ligand Functionalized ErbB-3 Targeted Novel DNA Nano-Threads Loaded with the Low Dose of Doxorubicin for Efficient In Vitro Evaluation of the Resistant Anti-Cancer Activity. Pharm Res 2020; 37:75. [PMID: 32232574 DOI: 10.1007/s11095-020-02803-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 03/18/2020] [Indexed: 12/25/2022]
Abstract
PURPOSE Doxorubicin (Dox) being a hydrophobic drug needs a unique carrier for the effective encapsulation with uniformity in the aqueous dispersion, cell culture media and the biological-fluids that may efficiently target its release at the tumor site. METHODS Circular DNA-nanotechnology was employed to synthesize DNA Nano-threads (DNA-NTs) by polymerization of triangular DNA-tiles. It involved circularizing a linear single-stranded scaffold strand to make sturdier and rigid triangles. DNA-NTs were characterized by the AFM and Native-PAGE tests. Dox binding and loading to the Neuregulin1 (NRG1) functionalized DNA based nano-threads (NF-DBNs) was estimated by the UV-shift analysis. The biocompatibility of the blank NRG-1/DNA-NTs and enhanced cytotoxicity of the NF-DBNs was assessed by the MTT assay. Cell proliferation/apoptosis was analyzed through the Flow-cytometry experiment. Cell-surface binding and the cell-internalization of the NF-DBNs was captured by the double-photon confocal microscopy (DPCM). RESULTS The AFM images revealed uniform DNA-NTs with the diameter 30 to 80 nm and length 400 to 800 nm. PAGE native gel was used for the further confirmation of the successful assembly of the strands to synthesize DNA-NTs that gave one sharp band with the decreased electrophoretic mobility down the gel. MTT assay showed that blank DNA-NTs were biocompatible to the cells with less cytotoxicity even at elevated concentrations with most of the cells (94%) remaining alive compared to the dose-dependent enhanced cytotoxicity of NF-DBNs further evidenced by the Flow-cytometry analysis. CONCLUSION Uniform and stiffer DNA-NTs for the potential applications in targeted drug delivery was achieved through circular DNA scaffolding.
Collapse
|
14
|
Gao Y, Khan GJ, Wei X, Zhai KF, Sun L, Yuan S. DT-13 inhibits breast cancer cell migration via non-muscle myosin II-A regulation in tumor microenvironment synchronized adaptations. Clin Transl Oncol 2020; 22:1591-1602. [PMID: 32056128 DOI: 10.1007/s12094-020-02303-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/18/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Tumor metastasis is a terrifying characteristic of cancer. Numerous studies have been conducted to overcome metastasis by targeting tumor microenvironment (TME). However, due to complexity of tumor microenvironment, it remained difficult for accurate targeting. Dwarf-lillytruf tuber monomer-13 (DT-13) possess good potential against TME. OBJECTIVE As TME is supportive for tumor metastasis, alternatively it is a challenging for therapeutic intervention. In our present study, we explored molecular mechanism through which TME induced cell migration and how DT-13 interferes in this mechanism. METHODS We used a novel model of co-culture system which is eventually developed in our lab. Tumor cells were co-cultured with hypoxia induced cancer-associated fibroblasts (CAF) or with chemically induced cancer-associated adipocytes (CAA). The effect of hypoxia in conditioned medium for CAF was assessed through expression of α-SMA and HIF by western blotting while oil red staining was done to assess the successful chemical induction for adipocytes (CAA), the effect of TME through conditioned medium on cell migration was analyzed by trans-well cell migration, and cell motility (wound healing) analyses. The expression changes in cellular proteins were assessed through western blotting and immunofluorescent studies. RESULTS AND CONCLUSION Our results showed that tumor microenvironment has a direct role in promoting breast cancer cell migration by stromal cells; moreover, we found that DT-13 restricts this TME regulated cell migration via targeting stromal cells in vitro. Additionally we also found that DT-13 targets NMII-A for its effect on breast cancer cell migration for the regulation of stromal cells in TME.
Collapse
Affiliation(s)
- Y Gao
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing, 210009, China
| | - G J Khan
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing, 210009, China.,State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China.,Faculty of Pharmacy (FOP), University of Central Punjab, Lahore, Pakistan
| | - X Wei
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing, 210009, China
| | - K-F Zhai
- Engineering Research Center of Natural Medicine and Functional Food, Institute of Pharmaceutical Biotechnology, School of Biological and Food Engineering, Suzhou University, 49, Bianhe Road, Suzhou, 234000, People's Republic of China.
| | - L Sun
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing, 210009, China.
| | - S Yuan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
15
|
Baig MMFA, Lai WF, Mikrani R, Jabeen M, Naveed M, Abbas M, Farooq MA, Ahsan A, Kassim SA, Khan GJ, Ansari MT. Synthetic NRG-1 functionalized DNA nanospindels towards HER2/neu targets for in vitro anti-cancer activity assessment against breast cancer MCF-7 cells. J Pharm Biomed Anal 2020; 182:113133. [PMID: 32004770 DOI: 10.1016/j.jpba.2020.113133] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/14/2020] [Accepted: 01/24/2020] [Indexed: 02/08/2023]
Abstract
DNA based nano-carriers synthesized from short circular scaffolds (circular DNA nanotechnology) attains stiffer topology for ligand functionalization (neuregulin-1/NRG-1 ligand) and biological applications (targeted drug delivery). Daunorubicin (DR) is a hydrophobic chemical that requires robust vectors to efficiently encapsulate and avoid its free dispersion in water, biological media and cell culture. Here we design DNA nanospindels (DNA-NS) to efficiently load DR and target the (highly expressed) HER2/neu receptors on the plasma membrane of drug-resistant MCF-7 (breast cancer) cells. DNA-NS were synthesized by polymerizing the DNA-triangles (utilizing 84-nt short circular scaffold strand) into larger DNA nano-ribbons characterized by the native-PAGE testing. AFM results revealed the spinning of DNA nanoribbons on its (own) axis because of the intrinsic curvature of the DNA double helix resulting in the formation of the firm and twisted DNA-NS with the diameter (50-70 nm) and length (0.5-4 μm). DA loading onto DNA-NS was confirmed by the UV shift analysis. The MTT results with the blank DNA-NS evidenced its biocompatibility (remained value of 93%) compared to the decreased viability of the MCF-7 cells after treatment with DNA-NS (DR loaded). These findings were further supported by the analysis of cell proliferation/apoptosis through flow cytometry showing 64% apoptosis after treating with the DR loaded DNA-NS. Hence, through the short circular DNA nanotechnology, we have achieved a stiffer, uniform, and biocompatible DNA-NS for applications in the targeted therapy.
Collapse
Affiliation(s)
- Mirza Muhammad Faran Ashraf Baig
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, PR China; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60000, Pakistan.
| | - Wing-Fu Lai
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, PR China; School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, PR China
| | - Reyaj Mikrani
- Department of Pharmaceutics, Basic medicine, and Clinical Pharmacy, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - Mehreen Jabeen
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60000, Pakistan
| | - Muhammad Naveed
- School of Pharmacy, Nanjing Medical University, Jiangsu Province, Nanjing 211166, PR China
| | - Muhammad Abbas
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Muhammad Asim Farooq
- Department of Pharmaceutics, Basic medicine, and Clinical Pharmacy, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - Anam Ahsan
- College of Animal Science & Veterinary Medicine, Shanxi Agricultural University, Taigu, PR China
| | - Said Abasse Kassim
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, PR China
| | - Ghulam Jilany Khan
- Faculty of Pharmacy, University of Central Punjab, Lahore, 54570, Pakistan
| | - Muhammad Tayyab Ansari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60000, Pakistan
| |
Collapse
|
16
|
Xu Z, Han X, Ou D, Liu T, Li Z, Jiang G, Liu J, Zhang J. Targeting PI3K/AKT/mTOR-mediated autophagy for tumor therapy. Appl Microbiol Biotechnol 2019; 104:575-587. [PMID: 31832711 DOI: 10.1007/s00253-019-10257-8] [Citation(s) in RCA: 404] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/05/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022]
Abstract
Autophagy is a highly conserved catabolic process and participates in a variety of cellular biological activities. The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway, as a critical regulator of autophagy, is involved in the initiation and promotion of a series of pathological disorders including various tumors. Autophagy also participates in regulating the balance between the tumor and the tumor microenvironment. Natural products have been considered a treasure of new drug discoveries and are of great value to medicine. Mounting evidence has suggested that numerous natural products are targeting PI3K/AKT/mTOR-mediated autophagy, thereby suppressing tumor growth. Furthermore, autophagy plays a "double-edged sword" role in different tumors. Targeting PI3K/AKT/mTOR-mediated autophagy is an important therapeutic strategy for a variety of tumors, and plays important roles in enhancing the chemosensitivity of tumor cells and avoiding drug resistance. Therefore, we summarized the roles of PI3K/AKT/mTOR-mediated autophagy in tumorigenesis, progression, and drug resistance of tumors, which may be utilized to design preferably therapeutic strategies for various tumors.
Collapse
Affiliation(s)
- Zhenru Xu
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xu Han
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Daming Ou
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Ting Liu
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Zunxiong Li
- University of South China, Hengyang, Hunan, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jing Liu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
| | - Ji Zhang
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China.
| |
Collapse
|
17
|
Khan GJ, Sun L, Abbas M, Naveed M, Jamshaid T, Baig MMFA, Yuan S. In-vitro Pre-Treatment of Cancer Cells with TGF-β1: A Novel Approach of Tail Vein Lung Cancer Metastasis Mouse Model for Anti-Metastatic Studies. Curr Mol Pharmacol 2019; 12:249-260. [PMID: 30848226 DOI: 10.2174/1874467212666190306165703] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/10/2019] [Accepted: 02/12/2019] [Indexed: 02/06/2023]
Abstract
Background:
Aggressive behavior of tumor metastasis comes from certain mutations,
changes in cellular metabolic and signaling pathways that are majorly altered by tumor microenvironment
(TME), its other components and growth factors like transforming growth factor-β1 (TGF-β1)
which is chiefly known for its epithelial to mesenchymal transformation (EMT). EMT is a critical step
of metastasis cascade in actual human lung cancer scenario.
Objective:
Our present study is focused on unveiling the in-vivo metastatic behavior of TGF-β1 treated
lung cancer cells that undergo EMT.
Methods:
The lung cancer epithelial A549 cells were treated in-vitro with TGF-β1 (3-5ng/ml for 72 h)
for EMT. After confirming the transformation of cells by phenotype modifications, wound healing and
cell migration assay and qRT-PCR analyses of EMT biomarkers including E. Cadherin, Vimentin,
Snail, Slug, MMP2 and MMP9; those TGF-β1 modified cells were probed with fluorescent trackers and
were injected into the tail vein of BALB/c nude mice for metastatic dissemination studies.
Results:
Our findings indicate that the distribution of TGF-β1 treated A549 cells as compared to W.T
A549 towards lungs is less in terms of total relative fluorescent cluster count, however, the difference is
insignificant (52±4, 60±5 respectively). Additionally, we show that TGF-β1 treated cells tend to metastasize
almost 2, 3, 1.5, 2 and 1.7 times more than W.T towards liver, brain, ovaries, bones and adrenal
gland, respectively, which is very much like human lung cancer metastasis.
Conclusion:
Conclusively, it is the first study ever reporting that a pre-treatment of cells with TGF-β1
for experimental lung cancer metastasis mouse model may portray a more precise approach for the
development of potential therapeutic treatments. Additional pre-treatment studies with the application
of other TME conditions like hypoxia and factors like NFκB, VEGF etc. may be a future prospect to
develop a better understanding.
Collapse
Affiliation(s)
- Ghulam Jilany Khan
- Jiangsu key laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Li Sun
- Jiangsu key laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Muhammad Abbas
- State key Laboratory of Pharmaceutical Biotechnology, Nanjing University, 210023, Nanjing, China
| | - Muhammad Naveed
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China
| | - Talha Jamshaid
- Department of Pharmaceutics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | | | - Shengtao Yuan
- Jiangsu key laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
18
|
Baig MMFA, Zhang QW, Younis MR, Xia XH. A DNA Nanodevice Simultaneously Activating the EGFR and Integrin for Enhancing Cytoskeletal Activity and Cancer Cell Treatment. NANO LETTERS 2019; 19:7503-7513. [PMID: 31515999 DOI: 10.1021/acs.nanolett.9b03325] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cell-surface receptors (e.g., EGFR and integrin) and their interactions play determining roles in signal transduction and cytoskeletal activation, which affect cell attachment/detachment, invasion, motility, metastasis (intracellular), and cell-cell signaling. For instance, the interactions between the EGFR and integrin (α6β4) may cause increased mechanical force and shear stress via enhanced cytoskeleton activation. Here, we design a DNA nanodevice (DNA-ND) that can simultaneously target the EGFR and integrin receptors on the caveolae. The piconewton (pN) forces in response to the EGFR-integrin coactivation can be sensed upon the unfolding of the DNA hairpin structure on the side arm of the device via changes of the fluorescence and plasmonic signals. We find that simultaneous activation of EGFR-integrin receptors causes enhanced signal transduction, contractions of the cells, and initiation of the biochemical pathways, thus resulting in a change of the cell division and endocytosis/exocytosis processes that affect the cell proliferation/apoptosis. The DNA-ND further enables us to visualize the cointernalization and degradation of the receptors by lysosomes, providing a novel approach toward bioimaging and mechano-pharmacology.
Collapse
Affiliation(s)
- Mirza Muhammad Faran Ashraf Baig
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210023 , China
| | - Qian-Wen Zhang
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210023 , China
| | - Muhammad Rizwan Younis
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210023 , China
| | - Xing-Hua Xia
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210023 , China
| |
Collapse
|
19
|
Evaluation of chitosan/eudragit hybrid coating over cubic DNA nanospheres with superior stability and therapeutic outcomes. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.05.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
20
|
Design, synthesis and evaluation of DNA nano-cubes as a core material protected by the alginate coating for oral administration of anti-diabetic drug. J Food Drug Anal 2019; 27:805-814. [PMID: 31324296 PMCID: PMC9307041 DOI: 10.1016/j.jfda.2019.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/27/2019] [Accepted: 03/29/2019] [Indexed: 02/06/2023] Open
Abstract
Poor control towards glycemic levels among diabetic patients may lead to severe micro/ macro-vascular and neuropathic complexities. Proper functioning of alpha-beta cells of pancreases is required to attain long term glycemic control among type 2 diabetics. The recent developments to manage diabetes are focused on controlling the insulin-glucagon secretions from the pancreases. DPP-4 inhibitors class of drugs after elevating GLP-1/GIP (incretins) levels in the blood, not only raise the insulin levels but also suppress the glucagon level. Vildagliptin (VI) is a potent DPP-4 inhibitor with least adverse events compared to other DPP-4 inhibitors. We encapsulated VI into 3D nanocube that gets bind to the DNA due to secondary amine in its chemical structure. DNA-nanocube being negatively charged was incubated with the PLL to attain positive surface. Ultimately VI loaded nanocubes were coated with the negatively charged Na-alginate via electrostatic attraction method to get stable spherical nanospheres for oral delivery of VI. Nanospheres were evaluated physically through native PAGE analysis, DSC, TGA, dissolution testing, XRD and FTIR. We attained uniformed and spherical nanospheres with stable topology, nanoscale size precision (40–150 nm in diameter), Entrapment efficiency (up to 90%), prolonged drug release (13 ± 4 h) at basic pH, and superior oral antidiabetic effects with improved GLP1 and glycemic levels. The formulated nanospheres attained size uniformity and better therapeutic outcomes in terms of reduced adverse events and better control of glycemic levels than previously reported methods with decreased dosage frequency tested in Db/Db mice.
Collapse
|
21
|
PLL-alginate and the HPMC-EC hybrid coating over the 3D DNA nanocubes as compact nanoparticles for oral administration. APPLIED NANOSCIENCE 2019. [DOI: 10.1007/s13204-019-01075-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
He J, Wei X, Li S, Quan X, Li R, Du H, Yuan S, Sun L. DT-13 suppresses breast cancer metastasis by modulating PLOD2 in the adipocytes microenvironment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 59:152778. [PMID: 31005809 DOI: 10.1016/j.phymed.2018.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 11/30/2018] [Accepted: 12/01/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Metastasis is the main cause of death in breast cancer and previous researches have indicated the pivotal role of adipocytes in breast cancer metastasis. DT-13, the saponin monomer 13 of the Dwarf lilyturf tuber, has been proved to exert potential anti-metastatic effect, the detailed mechanisms have not been well elucidated and the role of DT-13 in modulating adipocyte-breast cancer microenvironment has been given little attention. PURPOSE This study aims to explore the mechanisms of DT-13 in inhibiting breast cancer metastasis and whether DT-13 inhibit breast cancer metastasis via modulating the interactions between adipocytes and breast cancer cells. METHODS The cytotoxic effect of DT-13 on breast cancer cell viability was detected by MTT assay. Migration assays was used to conduct the effect of DT-13 on breast cancer cells migration. Orthotopic xenograft tumor model was used to test the effect of DT-13 on breast cancer metastasis. qRT-PCR and Western blot were used to investigate the mechanisms of DT-13 inhibiting breast cancer metastasis. RESULTS DT-13 inhibited breast cancer cells migration at the concentration without cytotoxicity. Furthermore, DT-13 decreased PLOD2 expression through modulating JAK/STAT3 and PI3K/AKT signaling pathways directly or indirectly in the adipocyte-breast cancer microenvironment. Orthotopic implantation mouse model of breast cancer further confirmed that DT-13 inhibited breast cancer metastasis via downregulating PLOD2 in vivo. CONCLUSION DT-13 suppressed breast cancer metastasis via reducing the expression of PLOD2.
Collapse
Affiliation(s)
- Jinyong He
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xiaohui Wei
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Sijing Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xingping Quan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Ruiming Li
- Tasly Research Institute, Tianjin Tasly Hodling Group Co., Ltd., Tianjin, China
| | - Hongzhi Du
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Shengtao Yuan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China.
| | - Li Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
23
|
DNA scaffold nanoparticles coated with HPMC/EC for oral delivery. Int J Pharm 2019; 562:321-332. [PMID: 30928213 DOI: 10.1016/j.ijpharm.2019.03.054] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/11/2022]
|
24
|
Baig MMFA, Naveed M, Abbas M, Kassim SA, Khan GJ, Ullah S, Sohail M, Nawaz W, Younis MR, Ansari MT. Chitosan-coated rectangular DNA nanospheres for better outcomes of anti-diabetic drug. JOURNAL OF NANOPARTICLE RESEARCH 2019; 21:98. [DOI: 10.1007/s11051-019-4534-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/17/2019] [Indexed: 05/12/2025]
|
25
|
Wang Z, Wang Y, Zhu S, Liu Y, Peng X, Zhang S, Zhang Z, Qiu Y, Jin M, Wang R, Zhong Y, Kong D. DT-13 Inhibits Proliferation and Metastasis of Human Prostate Cancer Cells Through Blocking PI3K/Akt Pathway. Front Pharmacol 2018; 9:1450. [PMID: 30581390 PMCID: PMC6292965 DOI: 10.3389/fphar.2018.01450] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/26/2018] [Indexed: 12/24/2022] Open
Abstract
DT-13, a saponin monomer 13 from the dwarf lilyturf tuber, was reported to exhibit anti-inflammatory, hepatoprotective, cardioprotective as well as antitumor activities in a number of tumor cells. Prostate cancer is the second leading cause of cancer death in males, discovery of novel antitumor drug for therapy of prostate cancer is expected. Aiming to evaluate whether DT-13 could become a candidate to treat prostate cancer, we recently investigated the antitumor effect of DT-13 on human prostate cancer cells and the underlying mechanism. DT-13 was found to effectively inhibit proliferation and metastasis of prostate cancer PC3 and DU145 cell lines in a dose-dependent manner. Treatment by DT-13 resulted in a mitochondria-mediated apoptosis, which was accompanied by the chromatin condensation and nuclear shrinkage in the prostate cancer cells. Moreover, DT-13 caused remarkable upregulation of Bax, Bad, Cytochrome C, cleaved -caspase 3, -caspase 9 and -PARP, in contrast to the downregulation of Bcl-2. Nevertheless, no obvious change in intracellular ROS level was observed after DT-13 treatment. We further demonstrated that DT-13 could inhibit PC3 cell metastasis in which suppression of Integrinβ1 and MMP2/9 might be involved. Western blot analysis indicated DT-13 significantly decreased the phosphorylation of PDK1, Akt, mTOR as well as p70S6K, suggesting the pro-apoptotic and anti-metastatic effects of DT-13 on prostate cancer cells might be attributed to the blockade of PI3K/Akt pathway. Collectively, our findings suggest DT-13 is worthy of further investigation as a drug candidate for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Zhengming Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yingying Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Shan Zhu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yao Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xin Peng
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Shaolu Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhe Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yuling Qiu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Meihua Jin
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ran Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yuxu Zhong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
26
|
Abbas M, Ahmed A, Khan GJ, Baig MMFA, Naveed M, Mikrani R, Cao T, Naeem S, Shi M, Dingding C. Clinical evaluation of carcinoembryonic and carbohydrate antigens as cancer biomarkers to monitor palliative chemotherapy in advanced stage gastric cancer. Curr Probl Cancer 2018; 43:5-17. [PMID: 30172422 DOI: 10.1016/j.currproblcancer.2018.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 07/19/2018] [Accepted: 08/01/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Carcinoembryonic antigen (CEA), carbohydrate antigen (CA)-125, CA19-9, and CA72-4 are often found modulated parameters in gastric cancer. OBJECTIVE Our present study is focused to evaluate the synchronization of these biomarkers in response to palliative chemotherapy. METHOD A retrospective study was conducted on 216 gastric cancer patients undergoing first-line cisplatin chemotherapy along with antiangiogenic regimen. Blood samples were taken and analyzed biochemically and statistically. RESULTS Progression occurred in 78 of 216 patients and the median progression-free survival (PFS) was 5 months. For serum CEA, the median PFS was 4 versus 7 months for elevated and normal groups respectively (P = 0.01). The median PFS for normal and elevated CA19-9 and CA72-4 was 6 vs 4 months respectively (P = 0.001). In the multivariate Cox regression model, elevated pretreatment level of CEA, CA19-9, and distant metastases were independent factors associated with increased risk of progression (P = 0.021, P = 0.000, P = 0.006, respectively). CONCLUSIONS Conclusively, elevated pretreatment level of CEA and CA19-9 is correlated with high risk of progression and worse prognosis. Moreover, an additional antiangiogenic therapy is more effective in decreasing cancer biomarker level after palliative chemotherapy that may be correlated with therapeutic triumph.
Collapse
Affiliation(s)
- Muhammad Abbas
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, PR China; Department of Oncology, Jiangsu Cancer Hospital, Jiangsu institute of cancer research, Nanjing medical university affiliated cancer hospital Nanjing 210009, Jiangsu, PR China
| | - Abrar Ahmed
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, PR China; Department of Oncology, Jiangsu Cancer Hospital, Jiangsu institute of cancer research, Nanjing medical university affiliated cancer hospital Nanjing 210009, Jiangsu, PR China
| | - Ghulam Jilany Khan
- Jiangsu key laboratory of Drug Screening, Evaluation and Pharmacodynamics Research, China Pharmaceutical University, Nanjing, PR China; Department of Pharmacology and Therapeutics, Faculty of Pharmacy (FOP), University of Central Punjab, Lahore, Pakistan; State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, PR China.
| | - Mirza Muhammad Faran Ashraf Baig
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, PR China
| | - Muhammad Naveed
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, PR China
| | - Reyaj Mikrani
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, PR China
| | - Tengli Cao
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, PR China
| | - Shagufta Naeem
- Department of Pathology, Ayub Medical College, Abbottabad, Pakistan.
| | - Meiqi Shi
- Department of Oncology, Jiangsu Cancer Hospital, Jiangsu institute of cancer research, Nanjing medical university affiliated cancer hospital Nanjing 210009, Jiangsu, PR China.
| | - Chen Dingding
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, PR China.
| |
Collapse
|
27
|
Abbas M, Faggian A, Sintali DN, Khan GJ, Naeem S, Shi M, Dingding C. Current and future biomarkers in gastric cancer. Biomed Pharmacother 2018; 103:1688-1700. [DOI: 10.1016/j.biopha.2018.04.178] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/24/2018] [Accepted: 04/24/2018] [Indexed: 02/06/2023] Open
|
28
|
Rwibasira Rudinga G, Khan GJ, Kong Y. Protease-Activated Receptor 4 (PAR4): A Promising Target for Antiplatelet Therapy. Int J Mol Sci 2018; 19:E573. [PMID: 29443899 PMCID: PMC5855795 DOI: 10.3390/ijms19020573] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/09/2018] [Accepted: 02/09/2018] [Indexed: 12/29/2022] Open
Abstract
Cardiovascular diseases (CVDs) are currently among the leading causes of death worldwide. Platelet aggregation is a key cellular component of arterial thrombi and major cause of CVDs. Protease-activated receptors (PARs), including PAR1, PAR2, PAR3 and PAR4, fall within a subfamily of seven-transmembrane G-protein-coupled receptors (GPCR). Human platelets express PAR1 and PAR4, which contribute to the signaling transduction processes. In association with CVDs, PAR4 not only contributes to platelet activation but also is a modulator of cellular responses that serve as hallmarks of inflammation. Although several antiplatelet drugs are available on the market, they have many side effects that limit their use. Emerging evidence shows that PAR4 targeting is a safer strategy for preventing thrombosis and consequently may improve the overall cardiac safety profile. Our present review summarizes the PAR4 structural characteristics, activation mechanism, role in the pathophysiology of diseases and understanding the association of PAR4 targeting for improved cardiac protection. Conclusively, this review highlights the importance of PAR4 antagonists and its potential utility in different CVDs.
Collapse
Affiliation(s)
- Gamariel Rwibasira Rudinga
- School of Life Science & Technology, China Pharmaceutical University, 24 Tong Jia Street, Nanjing 210009, China.
| | - Ghulam Jilany Khan
- Jiangsu Center for Pharmacodynamics Research, Evaluation and Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| | - Yi Kong
- School of Life Science & Technology, China Pharmaceutical University, 24 Tong Jia Street, Nanjing 210009, China.
| |
Collapse
|