1
|
Wang J, Tang X, Liu F, Mao B, Zhang Q, Zhao J, Chen W, Cui S. Sources, metabolism, health benefits and future development of saponins from plants. Food Res Int 2024; 197:115226. [PMID: 39593311 DOI: 10.1016/j.foodres.2024.115226] [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: 07/03/2024] [Revised: 09/29/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024]
Abstract
Saponins are a class of glycoside compounds whose aglycones are triterpenoids or spirostanes, widely exist in a variety of Chinese herbs. Saponins are one of the important active components of medicinal plants and have a wide range of bioactivities. In order to promote the better development and utilization of saponins, the process of digestion, absorption and metabolism of saponins in vivo was reviewed in this paper. At the same time, the main bioactivities of common saponins and their potential mechanisms for alleviating diseases were summarized. Finally, the potential of saponins as functional food has been pointed out, and microbial transformation can make saponins better play this potential in the future.
Collapse
Affiliation(s)
- Jiang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Xin Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Fei Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
2
|
Zhang M, Zhang J, Ma Y, Jin Y, Li Y, Wu X. Nephropathy induced by cisplatin results from mitochondrial disruption, impaired energy metabolism, altered expression of renal transporters, and accumulation of urinary toxins. J Trace Elem Med Biol 2024; 86:127553. [PMID: 39427559 DOI: 10.1016/j.jtemb.2024.127553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 09/09/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND The administration of platinum-based drugs such as cisplatin and its derivatives, which are frequently used during clinical chemotherapy, is highly restricted due to the incidence of nephrotoxicity. The present study focused on investigating cisplatin-induced nephrotoxicity from the perspective of energy metabolism, renal transporter expression and urinary toxin accumulation. METHODS This study investigated cisplatin's toxic effects, including nephrotoxicity, cardiotoxicity, hepatotoxicity, pulmonary toxicity, and splenotoxicity. We used transmission electron microscopy (TEM) and scanning electron microscopy (SEM) to characterize the accumulation of cisplatin in the kidney and the structure of renal mitochondria. The production of reactive oxygen species (ROS) induced by cisplatin in renal tubular epithelial cells was evaluated by in vitro experiments, and apoptosis of renal tubular epithelial cells and alterations to the renal microvasculature were assessed. Metabolites associated with the glycolytic and tricarboxylic acid pathways were measured, and renal transporters expression, autophagy, and urinary toxins (UTs) accumulation were also assessed. RESULTS Our results reveal that cisplatin-induced varying degrees of damage to the heart, liver, spleen, lungs, and kidneys, including inflammatory and fibrotic damage. Accumulation of cisplatin in renal mitochondria disrupted mitochondrial structure and mitochondrial function, as evidenced by decreased levels of glucose 6-phosphate and ribose 5-phosphate and elevated levels of isocitric acid. Cisplatin-induced accumulation of ROS in renal tubular epithelial cells led to apoptosis and, ultimately, constriction or loss of renal microvasculature. Furthermore, dysregulation of renal transporter expression, activation of autophagy and increased accumulation of UTs was observed. CONCLUSION Accumulation of cisplatin in the kidney led to damage to mitochondrial structure and function, apoptosis of renal tubular epithelial cells, constriction or loss of renal microvasculature, dysfunction of renal transporters, activation of autophagy, and accumulation of UTs.
Collapse
Affiliation(s)
- Mingkang Zhang
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Jianping Zhang
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Yanrong Ma
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Yongwen Jin
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Yile Li
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Xin'an Wu
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China.
| |
Collapse
|
3
|
Zhang D, Jin R, Li G, Zhang C, Zhou Y. Ameliorative effect of rutecarpine supplementation against cisplatin-induced nephrotoxicity in rats via inhibition of monocyte chemoattractant protein-1, intercellular adhesion molecule-1, high-mobility group box 1, and nuclear factor kappa B. Biotechnol Appl Biochem 2024. [PMID: 39506243 DOI: 10.1002/bab.2692] [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: 08/06/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024]
Abstract
Cisplatin, the pioneering heavy metal compound, stands out as a potent drug for the treatment of various solid tumors. However, its clinical utility is hampered by notable toxicity and adverse effects, particularly nephrotoxicity. The potency of rutecarpine, a phytochemical, in mitigating cisplatin-induced nephrotoxicity was assessed in the present study. In this experimental setup, healthy male Wistar rats were grouped into four and Group I rats served as the control group, receiving only vehicle control. Group II rats were subjected to cisplatin treatment alone, administered intraperitoneally at a dosage of 7 mg/kg body weight on the 19th, 20th, and 21st days. Group III and IV rats were orally administered with rutecarpine at doses of 10 and 20 mg/kg body weight, respectively, starting from Day 1 and continuing daily for 21 days. Additionally, they were injected intraperitoneally with cisplatin at the same dosage and schedule as Group II. Relative kidney weight and renal biochemical markers blood urea nitrogen, lactate dehydrogenase, serum urea, and creatinine were measured to assess rutecarpine inhibitory potency against cisplatin toxicity. Markers of oxidative damage and antioxidants levels were quantified in the ruteacarpine- and cisplatin-treated rats. The study investigated the anti-inflammatory property of rutecarpine in cisplatin-induced nephrotoxicity by analyzing inflammatory cytokines. Renal tissue levels of monocyte chemoattractant protein-1, intercellular adhesion molecule-1, high-mobility group box 1, and nuclear factor kappa B, key markers of nephrotoxicity, were quantified to assess rutecarpine's potential to mitigate cisplatin-triggered damage. Histopathological examinations were performed to confirm the impact of rutecarpine against cisplatin-induced nephrotoxicity. Treatment with rutecarpine notably reduced renal biochemical markers, prevented renal edema, and attenuated oxidative stress-induced damage in cisplatin-treated rats. Both inflammatory and nephrotoxicity markers showed significant decreases in rats treated with rutecarpine along with cisplatin. Histological analysis affirmed that rutecarpine pretreatment effectively prevented cisplatin-induced nephrotoxicity. The study findings demonstrate that rutecarpine ameliorates cisplatin-triggered nephrotoxicity through its antioxidant and anti-inflammatory properties, suggesting that rutecarpine supplementation alongside cisplatin treatment could potentially reduce nephrotoxicity in cancer patients.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Emergency, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Rui Jin
- Department of Emergency, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu, China
| | - Guoxing Li
- Outpatient Department, General Hospital of Western Theater of Chinese People's Liberation Army, Chengdu, Sichuan, China
| | - CaiFeng Zhang
- Department of Encephalopathy, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Yanhong Zhou
- Department of Emergency, Banan Hospital Affiliated to Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Fawzy MH, Moustafa YM, Khodeer DM, Saeed NM, El-Sayed NM. Doxepin as OCT2 inhibitor ameliorates inflammatory response and modulates PI3K/Akt signaling associated with cisplatin-induced nephrotoxicity in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03473-1. [PMID: 39400714 DOI: 10.1007/s00210-024-03473-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/18/2024] [Indexed: 10/15/2024]
Abstract
Organic cationic transporter 2 (OCT2) was identified as the main transporter involved in the accumulation of cisplatin (CP) in the proximal tubular renal cells, resulting in nephrotoxicity. Doxepin (DOX) is a tricyclic agent with an inhibitory effect on OCT2. This study aimed to explore the possible mechanisms of the renoprotective role of DOX toward CP-induced nephrotoxicity. Rats were randomly divided into six groups: group 1, control; group 2, CP; groups 3, 4, and 5 were treated with graded doses of DOX (5, 10, and 20 mg/kg, respectively) intraperitoneally (ip) once daily for 10 consecutive days and group 6 was treated only with DOX (20 mg/kg). On the seventh day, a single injected dose of CP (10 mg/kg, ip) was given to the rats in groups 2-5. Seventy-two hours after CP injection, rats were sacrificed, and the kidneys were removed for histological and biochemical measurements. DOX ameliorated the CP-induced histopathological alterations. DOX significantly reduced the expression of OCT2, lipid peroxidation marker (MDA), and inflammatory cytokines, including TNF-α, IL-6, IL-1, IL-2, and IL-1β, and increased the activity of antioxidant enzymes. In addition, pre- and co-treatment with DOX significantly reduced the CP-mediated apoptotic effect by reducing the renal tissue expression of BAX and caspase-3 levels, upregulating the expression of Bcl-2, and modulating the phosphorylation of PI3K/Akt signaling cascade. DOX exerts a nephroprotective impact against CP-mediated nephrotoxicity via the inhibition of OCT2, suppression of inflammation, oxidative stress, and apoptotic markers, and modulation of PI3K/Akt signaling cascade.
Collapse
Affiliation(s)
- Mariam H Fawzy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Yasser M Moustafa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University in Cairo, Badr City, Egypt
| | - Dina M Khodeer
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Noha M Saeed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Norhan M El-Sayed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
5
|
Gao M, Zhu H, Xu H, Jin X, Zheng G, Zhu J, Gu C, Wang X. PGK1 can affect the prognosis and development of bladder cancer. Cancer Med 2024; 13:e70242. [PMID: 39315723 PMCID: PMC11420942 DOI: 10.1002/cam4.70242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 08/05/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Previous studies have demonstrated that the glycolytic enzyme phosphoglycerate kinase 1 (PGK1) can promote tumor development. This study sought to investigate the specific role of PGK1 in bladder cancer (BLCA). METHODS Public databases and immunohistochemistry assays were utilized to analyze the expression of PGK1 in BLCA and its prognostic significance. Cell proliferation was assessed through CCK-8 and colony formation assays, while the level of metastasis was evaluated using transwell migration experiments. Additionally, IC50 experiments were conducted to assess the impact of PGK1 on cisplatin sensitivity. RESULTS The mRNA and protein expression levels of PGK1 were significantly upregulated in BLCA. Cox proportional hazards model analysis revealed that PGK1 and T stage were independent prognostic factors for BLCA patients. Both CCK-8 and colony assays demonstrated that PGK1 promotes proliferation. Furthermore, a positive correlation was observed between PGK1 and Ki67, a proliferation index. Transwell migration assays confirmed the ability of PGK1 to enhance metastasis. Finally, PGK1 increased the IC50 associated with cisplatin treatment in BLCA. CONCLUSION Collectively, these findings suggest that PGK1 may hold clinical value in predicting BLCA prognosis and improving the outcomes of this patient population.
Collapse
Affiliation(s)
- Mingde Gao
- Department of Urology, Affiliated Tumor Hospital of Nantong University and Nantong Tumor Hospital, Nantong, People's Republic of China
| | - Haixia Zhu
- Department of Central Laboratory, Affiliated Tumor Hospital of Nantong University and Nantong Tumor Hospital, Nantong, People's Republic of China
| | - Haifei Xu
- Department of Urology, Affiliated Tumor Hospital of Nantong University and Nantong Tumor Hospital, Nantong, People's Republic of China
| | - Xiaoxia Jin
- Department of Pathology, Affiliated Tumor Hospital of Nantong University and Nantong Tumor Hospital, Nantong, People's Republic of China
| | - Guihua Zheng
- Department of Pathology, Affiliated Tumor Hospital of Nantong University and Nantong Tumor Hospital, Nantong, People's Republic of China
| | - Jinfeng Zhu
- Department of Urology, Affiliated Tumor Hospital of Nantong University and Nantong Tumor Hospital, Nantong, People's Republic of China
| | - Chunyan Gu
- Department of Pathology, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, People's Republic of China
| | - Xiaolin Wang
- Department of Urology, Affiliated Tumor Hospital of Nantong University and Nantong Tumor Hospital, Nantong, People's Republic of China
| |
Collapse
|
6
|
Khairnar SI, Kulkarni YA, Singh K. Mitigation of cisplatin-induced nephrotoxicity by chelidonic acid in Wistar rats. J Trace Elem Med Biol 2024; 81:127321. [PMID: 37918276 DOI: 10.1016/j.jtemb.2023.127321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/03/2023] [Accepted: 10/13/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION Cisplatin, an anti-cancer drug is used to treat a wide range of solid tumors. Nevertheless, nephrotoxicity is the major adverse effect that restricts its clinical application. The present study focuses on the effect of chelidonic acid in cisplatin-induced nephrotoxicity. METHODS Wistar rats were injected with cisplatin (5 mg/kg, intraperitoneally (i.p.), once in a week for 4 weeks) and chelidonic acid (10, 20, and 40 mg/kg, per oral (p.o.) for 4 weeks). Body weight, urine, biochemical, and oxidative stress parameters were performed to evaluate the effect of chelidonic acid in cisplatin-induced nephrotoxicity in rats. Pro-inflammatory cytokines and nuclear factor erythroid 2-related factor 2 (Nrf2) concentrations were determined. Expression of phospho-AMP activated protein kinase (phospho-AMP) and hypoxia-inducible factor 1-alpha (HIF-1α) was studied with western blot. Haematoxylin and eosin, periodic acid-Schiff, and Masson's trichrome staining were used to study kidney tissues. RESULTS Relative kidney weight and urine output were significantly increased in cisplatin-administered rats. Whereas, albumin, and creatinine concentration were decreased, and treatment with chelidonic acid reverses these deleterious effects of cisplatin significantly. Kidney functions were improved by chelidonic acid treatment with a reduction in tumor necrosis factor-alpha (TNF-α), Interleukin-6 (IL-6), and transforming growth factor-beta (TGF-β1) concentration. The oxidative stress was decreased as compared to the cisplatin group. Furthermore, Nrf2 was significantly increased by chelidonic acid treatment. Chelidonic acid treatment significantly increased the expression of phospho-AMPK and HIF-1α in kidney tissue. Histopathological studies revealed that chelidonic acid reduced kidney damage. CONCLUSION The findings showed that chelidonic acid increases phospho-AMPK and HIF-1α in the kidney tissue and significantly lowers the inflammatory cytokines, thus it is an effective molecule for providing protection against cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Shraddha I Khairnar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| | - Kavita Singh
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai 400056, India.
| |
Collapse
|
7
|
Li B, Yang J, Wang P, Li X, Li M, Zhang Y. Exercise performance reduction and preventive measures in highland sports. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:1464-1478. [PMID: 38432877 PMCID: PMC10929890 DOI: 10.11817/j.issn.1672-7347.2023.230074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Indexed: 03/05/2024]
Abstract
The plateau is a special environment with low pressure, low oxygen, low temperature, and high ultraviolet radiation. The exercise performance of people on the plateau is generally reduced, which seriously affects the life and health of people living in the plateau and entering the plateau. In recent years, the prevention and treatment of injury caused by high altitude hypoxia has attracted wide attention. It has shown that the higher the altitude with the longer the duration of exercise, the faster the stationing, the greater the impact on people's sports performance. Rapid entry into the plateau and long-term stay in the plateau have an impact on people's explosive power, endurance and fine operation. Advances in medical technology enable various prevention methods to be used to acclimate to high altitude environments. However, in vitro intervention methods are costly, easy to rebound and possess limited effects. Therefore, drug prevention and treatment is obviously a more economical choice. Chemical drugs increase the efficiency of high altitude exercise by improving the ischemic and hypoxic symptoms of the heart and brain, increasing lung ventilation and arterial oxygenation capacity, and accelerating the elimination of adverse product accumulation after exercise. Single Chinese medicine, Chinese patent medicine, and compound preparations can improve exercise performance by promoting body metabolism, improving muscle endurance, enhancing immunity, and other mechanisms. Traditional Chinese medicine has unique advantage and application prospect in improving plateau sports performance damage.
Collapse
Affiliation(s)
- Boshen Li
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000.
- PLA Highland Medical Laboratory, 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou 730050.
| | - Jun Yang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000
| | - Peng Wang
- PLA Highland Medical Laboratory, 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou 730050
| | - Xiaolin Li
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000
- PLA Highland Medical Laboratory, 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou 730050
| | - Maoxing Li
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000.
- PLA Highland Medical Laboratory, 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou 730050.
- Institute of Radiation Medicine Sciences, Academy of Military Medicine, Academy of Military Sciences, Beijing 100850, China.
| | - Yuxuan Zhang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000
- PLA Highland Medical Laboratory, 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou 730050
| |
Collapse
|
8
|
Xie Y, Wang C. Herb-drug interactions between Panax notoginseng or its biologically active compounds and therapeutic drugs: A comprehensive pharmacodynamic and pharmacokinetic review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116156. [PMID: 36754189 DOI: 10.1016/j.jep.2023.116156] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/24/2022] [Accepted: 01/06/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Herbs, along with the use of herb-drug interactions (HDIs) to combat diseases, are increasing in popularity worldwide. HDIs have two effects: favorable interactions that tend to improve therapeutic outcomes and/or minimize the toxic effects of drugs, and unfavorable interactions aggravating the condition of patients. Panax notoginseng (Burk.) F.H. Chen is a medicinal plant that has long been commonly used in traditional Chinese medicine to reduce swelling, relieve pain, clear blood stasis, and stop bleeding. Numerous studies have demonstrated the existence of intricate pharmacodynamic (PD) and pharmacokinetic (PK) interactions between P. notoginseng and conventional drugs. However, these HDIs have not been systematically summarized. AIM OF THE REVIEW To collect the available literature on the combined applications of P. notoginseng and drugs published from 2005 to 2022 and summarize the molecular mechanisms of interactions to circumvent the potential risks of combination therapy. MATERIALS AND METHODS This work was conducted by searching PubMed, Scopus, Web of Science, and CNKI databases. The search terms included "notoginseng", "Sanqi", "drug interaction," "synergy/synergistic", "combination/combine", "enzyme", "CYP", and "transporter". RESULTS P. notoginseng and its bioactive ingredients interact synergistically with numerous drugs, including anticancer, antiplatelet, and antimicrobial agents, to surmount drug resistance and side effects. This review elaborates on the molecular mechanisms of the PD processed involved. P. notoginseng shapes the PK processes of the absorption, distribution, metabolism, and excretion of other drugs by regulating metabolic enzymes and transporters, mainly cytochrome P450 enzymes and P-glycoprotein. This effect is a red flag for drugs with a narrow therapeutic window. Notably, amphipathic saponins in P. notoginseng act as auxiliary materials in drug delivery systems to enhance drug solubility and absorption and represent a new entry point for studying interactions. CONCLUSION This article provides a comprehensive overview of HDIs by analyzing the results of the in vivo and in vitro studies on P. notoginseng and its bioactive components. The knowledge presented here offers a scientific guideline for investigating the clinical importance of combination therapies. Physicians and patients need information on possible interactions between P. notoginseng and other drugs, and this review can help them make scientific predictions regarding the consequences of combination treatments.
Collapse
Affiliation(s)
- Yujuan Xie
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China.
| |
Collapse
|
9
|
Li J, Li T, Li Z, Song Z, Gong X. Potential therapeutic effects of Chinese meteria medica in mitigating drug-induced acute kidney injury. Front Pharmacol 2023; 14:1153297. [PMID: 37077810 PMCID: PMC10106589 DOI: 10.3389/fphar.2023.1153297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023] Open
Abstract
Drug-induced acute kidney injury (DI-AKI) is one of the leading causes of kidney injury, is associated with high mortality and morbidity, and limits the clinical use of certain therapeutic or diagnostic agents, such as antineoplastic drugs, antibiotics, immunosuppressants, non-steroidal anti-inflammatory drugs, and contrast media. In recent years, numerous studies have shown that many Chinese meteria medica, metabolites derived from botanical drugs, and Chinese medicinal formulas confer protective effects against DI-AKI by targeting a variety of cellular or molecular mechanisms, such as oxidative stress, inflammatory, cell necrosis, apoptosis, and autophagy. This review summarizes the research status of common DI-AKI with Chinese meteria medica interventions, including cisplatin, gentamicin, contrast agents, methotrexate, and acetaminophen. At the same time, this review introduces the metabolites with application prospects represented by ginseng saponins, tetramethylpyrazine, panax notoginseng saponins, and curcumin. Overall, this review provides a reference for the development of promising nephroprotectants.
Collapse
|
10
|
Lu J, Li H, Yu D, Zhao P, Liu Y. Heat stress inhibits the proliferation and differentiation of myoblasts and is associated with damage to mitochondria. Front Cell Dev Biol 2023; 11:1171506. [PMID: 37113771 PMCID: PMC10126414 DOI: 10.3389/fcell.2023.1171506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/24/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction: Heat stress is harmful to the health of humans and animals, more and more common, as a consequence of global warming, while the mechanism that heat stress modulates skeletal development remains unknown. Hence, we conducted a model of heat stress in vitro. Methods: We used Hu sheep myoblasts as the research object, real-time quantitative PCR (RT-qPCR) and western blot (WB) were conducted to detect the expression of mRNA and protein in heat-stressed myoblasts. The would-healing assay was used to detect the migration of myoblasts. The mitochondria were observed by a transmission electron microscope. Results: mRNA and protein expression of HSP60 was significantly enriched in the heat-stressed myoblasts during proliferation and differentiation (p < 0.05). In our study, we indicated that heat stress enriched the intracellular ROS of the myoblasts (p < 0.001), leading to an increase in autophagy in the myoblasts to induce apoptosis. The results demonstrated that the protein expression of LC3B-1 and BCL-2 was significantly increased in myoblasts under heat stress during proliferation and differentiation (p < 0.05). Additionally, heat stress inhibited mitochondrial biogenesis and function and reduced the mitochondrial membrane potential and downregulated the expression of mtCo2, mtNd1 and DNM1L (p < 0.05) in myoblasts during proliferation and differentiation. Consequently, heat stress inhibited the proliferation and differentiation of the myoblasts, in accordance with the downregulation of the expression of PAX7, MYOD, MYF5, MYOG and MYHC (p < 0.05). Moreover, heat stress also inhibited the cell migration of the myoblasts. Discussion: This work demonstrates that heat stress inhibits proliferation and differentiation, and accelerates apoptosis by impairing mitochondrial function and promoting autophagy, which provides a mechanism to understand heat stress affects the development of the skeletal muscle.
Collapse
Affiliation(s)
- Jiawei Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Huixia Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Huixia Li, ; Debing Yu,
| | - Debing Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi, Xizang, China
- *Correspondence: Huixia Li, ; Debing Yu,
| | - Peng Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yuan Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
11
|
Liu Y, Zhai J, Qin F, Gao L, She Y, Wang M. Protective role of polyphenol extract from highland barley against cisplatin-induced renal toxicity and mitochondrial damage in rats. FOOD SCIENCE AND TECHNOLOGY RESEARCH 2023. [DOI: 10.3136/fstr.fstr-d-21-00284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Yunfan Liu
- College of Biochemical Engineering, Beijing Union University
| | - Jiazhou Zhai
- Beijing Municipal Key Laboratory of Biologically Active Substances and Functional Food
| | - Fei Qin
- College of Biochemical Engineering, Beijing Union University
| | - Liping Gao
- College of Biochemical Engineering, Beijing Union University
| | - Yongxin She
- Institute of Quality Standards & Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences
| | - Mengqiang Wang
- Institute of Quality Standards & Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences
| |
Collapse
|
12
|
Sun X, Zhong X, Ma W, Feng W, Huang Q, Ma M, Lv M, Hu R, Han Z, Li J, Zhou X. Germacrone induces caspase-3/GSDME activation and enhances ROS production, causing HepG2 pyroptosis. Exp Ther Med 2022; 24:456. [PMID: 35747157 PMCID: PMC9204551 DOI: 10.3892/etm.2022.11383] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/14/2022] [Indexed: 11/09/2022] Open
Abstract
Liver cancer is a highly lethal malignancy. Despite considerable efforts made in recent years, the prognosis of patients with liver cancer remains poor. Curcuma zedoaria (known as Ezhu in Chinese) is widely prescribed in traditional Chinese medicine. Germacrone (GM) is a sesquiterpene constituent derived from the essential oil of Ezhu, and exerts anti-carcinogenic effects by inducing apoptosis in various cancer cells. The present study investigated the potential mechanism of GM in HepG2 cells. Cell Counting Kit-8, colony-formation and lactate dehydrogenase-release assays, as well as cell death assays using flow cytometry, were performed to evaluate HepG2 cell proliferation following GM treatment. HepG2 cells were transfected with caspase-3 small interfering RNA and then treated with GM. Caspase-3 expression levels were determined by reverse transcription-quantitative PCR and western blotting. The present study showed that GM inhibited the growth of HepG2 cells and induced the proteolytic cleavage of caspase 3, with concomitant cleavage of gasdermin E (GSDME), by markedly increasing the production of reactive oxygen species (ROS). This led to caspase 3-dependent cleavage of GSDME, thereby promoting pyroptosis in HepG2 cells. However, these changes were rescued by ROS scavengers, such as N-acetylcysteine. Furthermore, GM inhibited tumor growth by promoting the cleavage of caspase 3 and GSDME in HepG2 cell xenograft models. These results indicated that GM induced GSDME-dependent pyroptosis through caspase 3 activation, at least in part, by damaging the mitochondria and enhancing ROS production, thereby supporting the possible development of GM as a candidate for the prevention and treatment of liver cancer.
Collapse
Affiliation(s)
- Xinfeng Sun
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, P.R. China.,Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Xin Zhong
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, P.R. China.,Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Wenfeng Ma
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, P.R. China.,Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Wenxing Feng
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, P.R. China.,Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Qi Huang
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, P.R. China.,Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Mengqing Ma
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, P.R. China.,Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Minling Lv
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, P.R. China.,Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Rui Hu
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, P.R. China.,Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Zhiyi Han
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, P.R. China.,Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Jing Li
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, P.R. China.,Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Xiaozhou Zhou
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, P.R. China.,Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| |
Collapse
|
13
|
Jiang W, Tang M, Yang L, Zhao X, Gao J, Jiao Y, Li T, Tie C, Gao T, Han Y, Jiang JD. Analgesic Alkaloids Derived From Traditional Chinese Medicine in Pain Management. Front Pharmacol 2022; 13:851508. [PMID: 35620295 PMCID: PMC9127080 DOI: 10.3389/fphar.2022.851508] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic pain is one of the most prevalent health problems. The establishment of chronic pain is complex. Current medication for chronic pain mainly dependent on anticonvulsants, tricyclic antidepressants and opioidergic drugs. However, they have limited therapeutic efficacy, and some even with severe side effects. We turned our interest into alkaloids separated from traditional Chinese medicine (TCM), that usually act on multiple drug targets. In this article, we introduced the best-studied analgesic alkaloids derived from TCM, including tetrahydropalmatine, aloperine, oxysophocarpine, matrine, sinomenine, ligustrazine, evodiamine, brucine, tetrandrine, Stopholidine, and lappaconitine, focusing on their mechanisms and potential clinical applications. To better describe the mechanism of these alkaloids, we adopted the concept of drug-cloud (dCloud) theory. dCloud illustrated the full therapeutic spectrum of multitarget analgesics with two dimensions, which are “direct efficacy”, including inhibition of ion channels, activating γ-Aminobutyric Acid/opioid receptors, to suppress pain signal directly; and “background efficacy”, including reducing neuronal inflammation/oxidative stress, inhibition of glial cell activation, restoring the balance between excitatory and inhibitory neurotransmission, to cure the root causes of chronic pain. Empirical evidence showed drug combination is beneficial to 30–50% chronic pain patients. To promote the discovery of effective analgesic combinations, we introduced an ancient Chinese therapeutic regimen that combines herbal drugs with “Jun”, “Chen”, “Zuo”, and “Shi” properties. In dCloud, “Jun” drug acts directly on the major symptom of the disease; “Chen” drug generates major background effects; “Zuo” drug has salutary and supportive functions; and “Shi” drug facilitates drug delivery to the targeted tissue. Subsequently, using this concept, we interpreted the therapeutic effect of established analgesic compositions containing TCM derived analgesic alkaloids, which may contribute to the establishment of an alternative drug discovery model.
Collapse
Affiliation(s)
- Wei Jiang
- Zhejiang Zhenyuan Pharmaceutical Co., Ltd., Shaoxing, China
| | - Mingze Tang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Limin Yang
- Zhejiang Zhenyuan Pharmaceutical Co., Ltd., Shaoxing, China
| | - Xu Zhao
- First Clinical Division, Peking University Hospital of Stomatology, Beijing, China
| | - Jun Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences & Peking Union Medical College, Beijing, China
| | - Yue Jiao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment of Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tao Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment of Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cai Tie
- State Key Laboratory of Coal Resources and Safety Mining, China University of Mining and Technology, Beijing, China.,School of Chemical and Environmental Engineering, China University of Mining and Technology, Beijing, China
| | - Tianle Gao
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China.,Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Beijing, China
| | - Yanxing Han
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Jian-Dong Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China.,Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Thévenod F, Schreiber T, Lee WK. Renal hypoxia-HIF-PHD-EPO signaling in transition metal nephrotoxicity: friend or foe? Arch Toxicol 2022; 96:1573-1607. [PMID: 35445830 PMCID: PMC9095554 DOI: 10.1007/s00204-022-03285-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/14/2022] [Indexed: 12/18/2022]
Abstract
The kidney is the main organ that senses changes in systemic oxygen tension, but it is also the key detoxification, transit and excretion site of transition metals (TMs). Pivotal to oxygen sensing are prolyl-hydroxylases (PHDs), which hydroxylate specific residues in hypoxia-inducible factors (HIFs), key transcription factors that orchestrate responses to hypoxia, such as induction of erythropoietin (EPO). The essential TM ion Fe is a key component and regulator of the hypoxia–PHD–HIF–EPO (HPHE) signaling axis, which governs erythropoiesis, angiogenesis, anaerobic metabolism, adaptation, survival and proliferation, and hence cell and body homeostasis. However, inadequate concentrations of essential TMs or entry of non-essential TMs in organisms cause toxicity and disrupt health. Non-essential TMs are toxic because they enter cells and displace essential TMs by ionic and molecular mimicry, e. g. in metalloproteins. Here, we review the molecular mechanisms of HPHE interactions with TMs (Fe, Co, Ni, Cd, Cr, and Pt) as well as their implications in renal physiology, pathophysiology and toxicology. Some TMs, such as Fe and Co, may activate renal HPHE signaling, which may be beneficial under some circumstances, for example, by mitigating renal injuries from other causes, but may also promote pathologies, such as renal cancer development and metastasis. Yet some other TMs appear to disrupt renal HPHE signaling, contributing to the complex picture of TM (nephro-)toxicity. Strikingly, despite a wealth of literature on the topic, current knowledge lacks a deeper molecular understanding of TM interaction with HPHE signaling, in particular in the kidney. This precludes rationale preventive and therapeutic approaches to TM nephrotoxicity, although recently activators of HPHE signaling have become available for therapy.
Collapse
Affiliation(s)
- Frank Thévenod
- Institute for Physiology, Pathophysiology and Toxicology, ZBAF, Witten/Herdecke University, Stockumer Strasse 12, 58453, Witten, Germany.
| | - Timm Schreiber
- Institute for Physiology, Pathophysiology and Toxicology, ZBAF, Witten/Herdecke University, Stockumer Strasse 12, 58453, Witten, Germany
| | - Wing-Kee Lee
- Physiology and Pathophysiology of Cells and Membranes, Medical School EWL, Bielefeld University, R.1 B2-13, Morgenbreede 1, 33615 Bielefeld, Germany
| |
Collapse
|
15
|
邹 琼, 伍 晓, 王 进, 夏 谍, 邓 萌, 丁 俞, 代 玉, 赵 嵩, 陈 彤. [Therapeutic effect of Panax notoginseng saponins combined with cyclophosphamide in mice bearing hepatocellular carcinoma H 22 cell xenograft]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:538-545. [PMID: 35527489 PMCID: PMC9085587 DOI: 10.12122/j.issn.1673-4254.2022.04.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To investigate the therapeutic effects of total saponins from Panax notognseng (PNS) combined with cyclophosphamide (CTX) in mice bearing hepatocellular carcinoma H22 cell xenograft. METHODS We examined the effects of treatment with different concentrations of PNS on H22 cell proliferation for 24 to 72 h in vitro using CCK8 colorimetric assay. Annexin V/PI double fluorescence staining was used to detect the effect of PNS on apoptosis of H22 cells. Mouse models bearing H22 cell xenograft were established and treated with CTX (25 mg/kg), PNS (120, 240 or 480 mg/kg), alone or in combinations. After treatments for consecutive 10 days, the mice were euthanized for examinations of carbon clearance ability of the monocytes and macrophages, splenic lymphocyte proliferation, tumor necrosis factor (TNF-α), interleukin-2 (IL-2), serum hemolysin antibody level, blood indicators, and the tumor inhibition rate. RESULTS Treatment with PNS concentration-dependently inhibited the proliferation and significantly promoted apoptosis of cultured H22 cells (P < 0.01). In the tumor-bearing mouse models, PNS alone and its combination with CTX both resulted in obvious enhancement of phagocytosis of the monocyte-macrophages, stimulated the proliferation of splenic lymphocytes, promoted the release of TNF-α and IL-2 and the production of serum hemolysin antibody, and increased the number of white blood cells, red blood cells and lymphocytes in the peripheral blood. Treatment with 480 mg/kg PNS combined with CTX resulted in a tumor inhibition rate of 83.28% (P < 0.01) and a life prolonging rate of 131.25% in the mouse models (P < 0.05). CONCLUSION PNS alone or in combination with CTX can improve the immunity and tumor inhibition rate and prolong the survival time of H22 tumor-bearing mice.
Collapse
Affiliation(s)
- 琼 邹
- 昆明医科大学药学院暨云南省天然药物药理重点实验室,云南 昆明 650500School of Pharmaceutical Science and Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| | - 晓萍 伍
- 昆明医科大学药学院暨云南省天然药物药理重点实验室,云南 昆明 650500School of Pharmaceutical Science and Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| | - 进吉 王
- 昆明医科大学药学院暨云南省天然药物药理重点实验室,云南 昆明 650500School of Pharmaceutical Science and Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| | - 谍 夏
- 昆明医科大学药学院暨云南省天然药物药理重点实验室,云南 昆明 650500School of Pharmaceutical Science and Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| | - 萌玥 邓
- 昆明医科大学药学院暨云南省天然药物药理重点实验室,云南 昆明 650500School of Pharmaceutical Science and Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| | - 俞珍 丁
- 昆明医科大学药学院暨云南省天然药物药理重点实验室,云南 昆明 650500School of Pharmaceutical Science and Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| | - 玉玲 代
- 昆明医科大学药学院暨云南省天然药物药理重点实验室,云南 昆明 650500School of Pharmaceutical Science and Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| | - 嵩月 赵
- 云南省食品药品审核查验中 心,云南 昆明 650228Yunnan Food and Drug Inspection Center, Kunming 650228, China
| | - 彤 陈
- 昆明医科大学药学院暨云南省天然药物药理重点实验室,云南 昆明 650500School of Pharmaceutical Science and Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| |
Collapse
|
16
|
Pei Z, Wu M, Yu H, Long G, Gui Z, Li X, Chen H, Jia Z, Xia W. Isoliquiritin Ameliorates Cisplatin-Induced Renal Proximal Tubular Cell Injury by Antagonizing Apoptosis, Oxidative Stress and Inflammation. Front Med (Lausanne) 2022; 9:873739. [PMID: 35433741 PMCID: PMC9005826 DOI: 10.3389/fmed.2022.873739] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/08/2022] [Indexed: 12/22/2022] Open
Abstract
Acute kidney injury (AKI) is a clinical syndrome characterized by morbidity, mortality, and cost. Cis-diamminedichloroplatinum (cisplatin) is a chemotherapeutic agent used to treat solid tumors and hematological malignancies, but its side effects, especially nephrotoxicity, limit its clinical application. Isoliquiritin (ISL), one of the major flavonoid glycoside compounds in licorice, has been reported to have anti-apoptotic, antioxidant, and anti-inflammatory activities. However, the effect and mechanism of ISL on cisplatin-induced renal proximal tubular cell injury remain unknown. In this study, mouse proximal tubular cells (mPTCs) and human proximal tubule epithelial cells (HK2) were administered increasing concentrations of ISL from 7.8125 to 250 μM. Moreover, mPTC and HK2 cells were pretreated with ISL for 6–8 h, followed by stimulation with cisplatin for 24 h. CCK-8 assay was performed to evaluate the cell viability. Apoptosis and reactive oxygen species (ROS) of cells were measured by using flow cytometer and western blotting. Our results showed that ISL had no obvious effect on cell viability. ISL decreased cisplatin-induced cell injury in a dose-dependent manner. ISL also protected against cisplatin-induced cell apoptosis. Meanwhile, the enhanced protein levels of Bax, cleaved caspase-3/caspase-3 ratio, levels of Pp-65/p-65, levels of IL-6, and the production of ROS induced by cisplatin were significantly attenuated by ISL treatment. Moreover, ISL markedly increased the protein levels of Bcl-2 and SOD2, which were reduced by cisplatin stimulation. These results showed that ISL ameliorated cisplatin-induced renal proximal tubular cell injury by antagonizing apoptosis, oxidative stress and inflammation.
Collapse
Affiliation(s)
- Zhiyin Pei
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Meng Wu
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hanqing Yu
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Guangfeng Long
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhen Gui
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaonan Li
- Department of Children Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hongbing Chen
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Weiwei Xia
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
17
|
Yu L, Wang Y, Guo YH, Wang L, Yang Z, Zhai ZH, Tang L. HIF-1α Alleviates High-Glucose-Induced Renal Tubular Cell Injury by Promoting Parkin/PINK1-Mediated Mitophagy. Front Med (Lausanne) 2022; 8:803874. [PMID: 35186974 PMCID: PMC8850720 DOI: 10.3389/fmed.2021.803874] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
It is well-established that mitophagy leads to Diabetic Nephropathy (DN) and renal failure. Mitophagy mediated by a Hypoxia-inducible factor-1α (HIF-1α) plays a beneficial role in many diseases. Nevertheless, the mechanisms underlying HIF-1α-mediated mitophagy in DN remain unclear. This study defines the role of HIF-1α mediated mitophagy in DN. The expression of HIF-1α was upregulated in HK-2 cells in an High-Glucose (HG) environment, and the YC-1 (a specific inhibitor of HIF-1α) further exacerbated the hypoxia-induced mitochondrial dysfunction. Conversely, the HIF-1α-mediated protective effect was strengthened by scavenger N-acetylcysteine (NAC), a type of reactive oxygen species. Moreover, HIF-1α-Parkin/PINK1-mediated mitophagy prevented apoptosis and ROS production in HK-2 cells subjected to HG exposure. In summary, HIF-1α mediated mitophagy on HK-2 cells under HG conditions could alleviate DN, suggesting that it has huge prospects for DN treatment.
Collapse
Affiliation(s)
- Lu Yu
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yulin Wang
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yan Hong Guo
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liuwei Wang
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zijun Yang
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zi Han Zhai
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin Tang
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
18
|
LRG1 expression reduced inflammation of sepsis-renal injury via activation of NLRP3 inflammasome by HIF-1 alpha. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-021-00214-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
Dark Side of Cancer Therapy: Cancer Treatment-Induced Cardiopulmonary Inflammation, Fibrosis, and Immune Modulation. Int J Mol Sci 2021; 22:ijms221810126. [PMID: 34576287 PMCID: PMC8465322 DOI: 10.3390/ijms221810126] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/07/2021] [Accepted: 09/15/2021] [Indexed: 12/15/2022] Open
Abstract
Advancements in cancer therapy increased the cancer free survival rates and reduced the malignant related deaths. Therapeutic options for patients with thoracic cancers include surgical intervention and the application of chemotherapy with ionizing radiation. Despite these advances, cancer therapy-related cardiopulmonary dysfunction (CTRCPD) is one of the most undesirable side effects of cancer therapy and leads to limitations to cancer treatment. Chemoradiation therapy or immunotherapy promote acute and chronic cardiopulmonary damage by inducing reactive oxygen species, DNA damage, inflammation, fibrosis, deregulation of cellular immunity, cardiopulmonary failure, and non-malignant related deaths among cancer-free patients who received cancer therapy. CTRCPD is a complex entity with multiple factors involved in this pathogenesis. Although the mechanisms of cancer therapy-induced toxicities are multifactorial, damage to the cardiac and pulmonary tissue as well as subsequent fibrosis and organ failure seem to be the underlying events. The available biomarkers and treatment options are not sufficient and efficient to detect cancer therapy-induced early asymptomatic cell fate cardiopulmonary toxicity. Therefore, application of cutting-edge multi-omics technology, such us whole-exome sequencing, DNA methylation, whole-genome sequencing, metabolomics, protein mass spectrometry and single cell transcriptomics, and 10 X spatial genomics, are warranted to identify early and late toxicity, inflammation-induced carcinogenesis response biomarkers, and cancer relapse response biomarkers. In this review, we summarize the current state of knowledge on cancer therapy-induced cardiopulmonary complications and our current understanding of the pathological and molecular consequences of cancer therapy-induced cardiopulmonary fibrosis, inflammation, immune suppression, and tumor recurrence, and possible treatment options for cancer therapy-induced cardiopulmonary toxicity.
Collapse
|
20
|
Li Q, Zhang Y, Yang Y, Huang S, Zou X, Wei C, Liang T, Zhong X. Panax notoginseng saponins reduces the cisplatin-induced acute renal injury by increasing HIF-1α/BNIP3 to inhibit mitochondrial apoptosis pathway. Biomed Pharmacother 2021; 142:111965. [PMID: 34385105 DOI: 10.1016/j.biopha.2021.111965] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/22/2021] [Accepted: 07/22/2021] [Indexed: 11/28/2022] Open
Abstract
Cisplatin (CDDP) may induce apoptosis of renal tubular epithelial cells (RTEC) and cause CDDP-induced acute kidney injury (CAKI) during cancer treatment, but yet lack of preventive measures and effective treatment. As a new Chinese herbal preparation, Panax notoginseng saponins (PNS) has been found to mitigate CDDP-induced CAKI through elevating the expression of HIF-1α in the rat model, according to the data from our previous works. However, the underlying link between HIF-1α and apoptosis has not been well elucidated. The current study as a follow-up work, was aimed to reveal if PNS improves CAKI through HIF-1α-dependent apoptosis. A stably HIF-1α-knockdown human proximal tubular epithelial cell (HK-2) line was established by transfecting a HIF-1α-siRNA into HK-2 cells. Cell viability, mitochondrial function, cell apoptosis ratio and the expression of apoptosis-associated proteins (Cyt C, Bcl2, Bax, caspases 3) were determined. In order to elucidate the underlying mechanism, the expression of HIF-1α and BNIP3 were assessed. Our results showed that treatment of PNS rescued the cell viability of CDDP-injured HK-2 or HIF-1α-knockdown HK-2 cells, and increased the expression levels of ATP and MMP in HK-2 or HIF-1α-knockdown HK-2 cells which were reduced by CDDP. Moreover, PNS treatment decreased the CDDP or CDDP plus HIF-1α-knockdown-induced elevation of apoptosis and apoptosis-associated protein expressions. These findings demonstrate that PNS reduces CAKI through increasing HIF-1α to inhibit mitochondrial apoptosis pathway. Hence, we suggest PNS as a protective and therapeutic new drug for CDDP treatment of cancers, which might have significant meaning of further research and application potential.
Collapse
Affiliation(s)
- Qingqing Li
- Postgraduate, Pharmacy Department, the first affiliated hospital of Guangxi Medical University, Nanning, China
| | - Yansong Zhang
- Postgraduate, Pharmacy Department, the first affiliated hospital of Guangxi Medical University, Nanning, China
| | - Yufang Yang
- Pharmacy Department, the first affiliated hospital of Guangxi Medical University, Nanning, China.
| | - Songqing Huang
- Postgraduate, Pharmacy Department, the first affiliated hospital of Guangxi Medical University, Nanning, China
| | - Xiaoqin Zou
- Pharmacy Department, the first affiliated hospital of Guangxi Medical University, Nanning, China
| | - Congying Wei
- Postgraduate, Pharmacy Department, the first affiliated hospital of Guangxi Medical University, Nanning, China
| | - Taolin Liang
- Postgraduate, Pharmacy Department, the first affiliated hospital of Guangxi Medical University, Nanning, China
| | - Xiaobin Zhong
- Regenerative Medicine Research Center of Guangxi Medical University, Nanning, China
| |
Collapse
|
21
|
PINK1/Parkin-mediated mitophagy inhibits warangalone-induced mitochondrial apoptosis in breast cancer cells. Aging (Albany NY) 2021; 13:12955-12972. [PMID: 33929971 PMCID: PMC8148507 DOI: 10.18632/aging.202965] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/14/2021] [Indexed: 12/19/2022]
Abstract
Breast cancer is the most common malignancy in women all around the world, especially in many countries in Asia. However, antitumor drugs with unique curative effects and low toxic side-effects have not been found yet. Warangalone is an isoflavone extracted from the Cudrania tricuspidata fruit, and is reported to possess anti-inflammatory and anti-cancer activity. The purpose of this study was to determine the effects of warangalone on breast cancer cells. In this study, we found that warangalone decreased the viability of breast cancer cells by increasing the generation of reactive oxygen species (ROS) resulting in mitochondrial damage and decreased mitochondrial membrane potential (MMP). Warangalone induced mitochondrial apoptosis by increasing the BAX/BCL-2 ratio. Warangalone activated mitophagy via upregulation of PINK1 and Parkin expression and co-localization. The combination of warangalone and autophagy inhibitors or PINK1 siRNA increased the degree of cell apoptosis compared to treatment with warangalone alone. Warangalone damages mitochondria via ROS, thereby triggering PINK1/Parkin-mediated mitophagy and inducing mitochondrial apoptosis. However, autophagy/mitophagy protects against warangalone-induced mitochondrial apoptosis. A combination of warangalone and autophagy/mitophagy inhibitors may be a potential treatment for breast cancer.
Collapse
|
22
|
Deng F, Zheng X, Sharma I, Dai Y, Wang Y, Kanwar YS. Regulated cell death in cisplatin-induced AKI: relevance of myo-inositol metabolism. Am J Physiol Renal Physiol 2021; 320:F578-F595. [PMID: 33615890 PMCID: PMC8083971 DOI: 10.1152/ajprenal.00016.2021] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
Regulated cell death (RCD), distinct from accidental cell death, refers to a process of well-controlled programmed cell death with well-defined pathological mechanisms. In the past few decades, various terms for RCDs were coined, and some of them have been implicated in the pathogenesis of various types of acute kidney injury (AKI). Cisplatin is widely used as a chemotherapeutic drug for a broad spectrum of cancers, but its usage was hampered because of being highly nephrotoxic. Cisplatin-induced AKI is commonly seen clinically, and it also serves as a well-established prototypic model for laboratory investigations relevant to acute nephropathy affecting especially the tubular compartment. Literature reports over a period of three decades have indicated that there are multiple types of RCDs, including apoptosis, necroptosis, pyroptosis, ferroptosis, and mitochondrial permeability transition-mediated necrosis, and some of them are pertinent to the pathogenesis of cisplatin-induced AKI. Interestingly, myo-inositol metabolism, a vital biological process that is largely restricted to the kidney, seems to be relevant to the pathogenesis of certain forms of RCDs. A comprehensive understanding of RCDs in cisplatin-induced AKI and their relevance to myo-inositol homeostasis may yield novel therapeutic targets for the amelioration of cisplatin-related nephropathy.
Collapse
Affiliation(s)
- Fei Deng
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, Northwestern University, Chicago, Illinois
- Department of Medicine, Northwestern University, Chicago, Illinois
| | - Xiaoping Zheng
- Department of Pathology, Northwestern University, Chicago, Illinois
- Department of Medicine, Northwestern University, Chicago, Illinois
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Isha Sharma
- Department of Pathology, Northwestern University, Chicago, Illinois
- Department of Medicine, Northwestern University, Chicago, Illinois
| | - Yingbo Dai
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Urology, The Fifth Affiliated Hospital of Sun Yet-Sen University, Zhuhai, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yashpal S Kanwar
- Department of Pathology, Northwestern University, Chicago, Illinois
- Department of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
23
|
Liang T, Wei C, Lu S, Qin M, Qin G, Zhang Y, Zhong X, Zou X, Yang Y. Ginaton injection alleviates cisplatin-induced renal interstitial fibrosis in rats via inhibition of apoptosis through regulation of the p38MAPK/TGF-β1 and p38MAPK/HIF-1α pathways. Biomed Rep 2021; 14:38. [PMID: 33692901 PMCID: PMC7938297 DOI: 10.3892/br.2021.1414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/04/2021] [Indexed: 11/06/2022] Open
Abstract
Ginaton injection (Ginkgo biloba extract; GBE) has been reported to protect against cisplatin-induced acute renal failure in rats. In the present study, the effects and molecular mechanisms of GBE on cisplatin-induced renal interstitial fibrosis were evaluated using a rat model. The rats were intraperitoneally injected with cisplatin once on the first day and a subset of rats were treated with GBE or SB203580 (SB; a specific p38 MAPK inhibitor) daily from days 22 to 40. The levels of N-acetyl-β-D-Glucosaminidase (NAG) in the urine, and of urea nitrogen (BUN) and creatinine (Scr) in the blood were assessed. The damage and fibrosis of renal tissues were evaluated using hematoxylin and eosin staining, as well as Masson's trichrome staining, respectively. Apoptosis in renal tissues was detected using a TUNEL assay. The protein expression levels of α-smooth muscle actin (SMA), collagen 1 (Col I), Bax, Bcl-2, caspase-3/cleaved caspase-3, hypoxia-inducible factor-1α (HIF-1α), TGF-β1 and p38MAPK, as well as the mRNA levels of p38MAPK in renal tissues were investigated. The results showed that GBE markedly reduced the levels of urinary NAG, Scr and BUN, and renal expression of α-SMA and Col I levels were also reduced. Furthermore, GBE significantly reduced renal tissue injury and the relative area of renal interstitial fibrosis induced by cisplatin. GBE effectively reduced the apoptotic rate of renal tissues, the protein expression levels of Bax, cleaved caspase-3, phospho-p38MAPK, TGF-β1 and HIF-1α, as well as the mRNA expression levels of p38MAPK in renal tissues induced by cisplatin, whereas GBE significantly increased Bcl-2 protein expression. SB exhibited similar effects to GBE, although it was not as effective. In summary, the present study is the first to show that GBE significantly alleviated renal interstitial fibrosis following cisplatin-induced acute renal injury. The mechanisms by which GBE exhibited its effects were associated with the inhibition of apoptosis via downregulation of the p38MAPK/TGF-β1 and p38MAPK/HIF-1α signaling pathways.
Collapse
Affiliation(s)
- Taolin Liang
- Postgraduate Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chongying Wei
- Postgraduate Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Sisi Lu
- Postgraduate Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Mengyuan Qin
- Postgraduate Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Guiming Qin
- Postgraduate Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yansong Zhang
- Postgraduate Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaobin Zhong
- Regenerative Medicine Research Center of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaoqin Zou
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yufang Yang
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
24
|
Chen J, Li L, Bai X, Xiao L, Shangguan J, Zhang W, Zhang X, Wang S, Liu G. Inhibition of Autophagy Prevents Panax Notoginseng Saponins (PNS) Protection on Cardiac Myocytes Against Endoplasmic Reticulum (ER) Stress-Induced Mitochondrial Injury, Ca 2+ Homeostasis and Associated Apoptosis. Front Pharmacol 2021; 12:620812. [PMID: 33762943 PMCID: PMC7982947 DOI: 10.3389/fphar.2021.620812] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/18/2021] [Indexed: 12/20/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is often closely linked to autophagy, hypoxia signaling, mitochondrial biogenesis and reactive oxygen species (ROS) responses. Understanding the interaction between ER stress, mitochondrial function and autophagy is of great importance to provide new mechanisms for the pathology, prevention and treatment of cardiovascular diseases. Our previous study has reported that Panax notoginseng saponins (PNS) protection against thapsigargin (TG)-induced ER stress response and associated cell apoptosis in cardiac myocytes is calcium dependent and mediated by ER Ca2+ release through RyR2. However, whether its protection upon ER stress and associated apoptosis is related to mitochondrial function and autophagy remains largely unknown. Here, we investigated the roles of PNS played in TG-induced mitochondrial function, ROS accumulation and autophagy. We also assessed its effects on Ca2+ homeostasis, ER stress response and associated cell death in the presence of autophagy inhibition. PNS-pretreated primary cultured neonatal rat cardiomyocytes were stimulated with TG to induce ER stress response. Mitochondrial potential (Δψm) was measured by JC-1. The general and mitochondrial ROS were measured by DCFH-DA and MitoSOX Red, respectively. Autophagy was evaluated by immunofluorescence of LC3, and immunoblots of LC3, p62, ATG7 and PINK1. In addition, mRFP-GFP-LC3 labeling was used to assess the autophagic influx. SiATG7 transfected H9c2 cells were generated to inhibit autophagy. Cytosolic and ER Ca2+ dynamics were investigated by calcium imaging. RyR2 oxidation was tested by oxyblot. Cell viability was examined by TUNEL assay. ER stress response and cell apoptosis were detected by immunoblots of BiP, CHOP, Cleaved Caspase-3 and Caspase-12. The results demonstrated that firstly, PNS protects against TG-induced mitochondrial injury and ROS accumulation. Secondly, PNS enhances autophagy in TG-induced cardiac myocytes. Thirdly, inhibition of autophagy diminishes PNS prevention of TG-induced mitochondrial injury, ROS accumulation and disruption of Ca2+ homeostasis. Last but not least, inhibition of autophagy abolishes PNS protection against TG-induced ER stress response and associated apoptosis. In summary, PNS protection against ER stress response and associated apoptosis is related to the regulation of mitochondrial injury and ROS overproduction via modulation of autophagy. These data provide new insights for molecular mechanisms of PNS as a potential preventive approach to the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Jun Chen
- Vasculocardiology Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Li
- Vasculocardiology Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xueyang Bai
- Vasculocardiology Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lili Xiao
- Vasculocardiology Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiahong Shangguan
- Vasculocardiology Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenjing Zhang
- Vasculocardiology Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiangqin Zhang
- Vasculocardiology Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shen Wang
- Vasculocardiology Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gangqiong Liu
- Vasculocardiology Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
25
|
Li RL, He LY, Zhang Q, Liu J, Lu F, Duan HXY, Fan LH, Peng W, Huang YL, Wu CJ. HIF-1α is a Potential Molecular Target for Herbal Medicine to Treat Diseases. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4915-4949. [PMID: 33235435 PMCID: PMC7680173 DOI: 10.2147/dddt.s274980] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022]
Abstract
HIF-1α is an important factor regulating oxygen balance in mammals, and its expression is closely related to various physiological and pathological conditions of the body. Because HIF-1α plays an important role in the occurrence and development of cancer and other diseases, it has become an enduring research hotspot. At the same time, natural medicines and traditional Chinese medicine compounds have amazing curative effects in various diseases related to HIF-1 subtype due to their unique pharmacological effects and more effective ingredients. Therefore, in this article, we first outline the structure of HIF-1α and the regulation related to its expression, then introduce various diseases closely related to HIF-1α, and finally focus on the regulation of natural medicines and compound Chinese medicines through various pathways. This will help us understand HIF-1α systematically, and use HIF-1α as a target to discover more natural medicines and traditional Chinese medicines that can treat related diseases.
Collapse
Affiliation(s)
- Ruo-Lan Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, People's Republic of China
| | - Li-Ying He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, People's Republic of China
| | - Qing Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, People's Republic of China
| | - Jia Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, People's Republic of China
| | - Feng Lu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, People's Republic of China
| | - Hu-Xin-Yue Duan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, People's Republic of China
| | - Lin-Hong Fan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, People's Republic of China
| | - Wei Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, People's Republic of China
| | - Yong-Liang Huang
- Pharmacy Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, People's Republic of China
| | - Chun-Jie Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, People's Republic of China
| |
Collapse
|
26
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Kalantari M, Mohammadinejad R, Javaheri T, Sethi G. Association of the Epithelial-Mesenchymal Transition (EMT) with Cisplatin Resistance. Int J Mol Sci 2020; 21:E4002. [PMID: 32503307 PMCID: PMC7312011 DOI: 10.3390/ijms21114002] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 02/08/2023] Open
Abstract
Therapy resistance is a characteristic of cancer cells that significantly reduces the effectiveness of drugs. Despite the popularity of cisplatin (CP) as a chemotherapeutic agent, which is widely used in the treatment of various types of cancer, resistance of cancer cells to CP chemotherapy has been extensively observed. Among various reported mechanism(s), the epithelial-mesenchymal transition (EMT) process can significantly contribute to chemoresistance by converting the motionless epithelial cells into mobile mesenchymal cells and altering cell-cell adhesion as well as the cellular extracellular matrix, leading to invasion of tumor cells. By analyzing the impact of the different molecular pathways such as microRNAs, long non-coding RNAs, nuclear factor-κB (NF-ĸB), phosphoinositide 3-kinase-related protein kinase (PI3K)/Akt, mammalian target rapamycin (mTOR), and Wnt, which play an important role in resistance exhibited to CP therapy, we first give an introduction about the EMT mechanism and its role in drug resistance. We then focus specifically on the molecular pathways involved in drug resistance and the pharmacological strategies that can be used to mitigate this resistance. Overall, we highlight the various targeted signaling pathways that could be considered in future studies to pave the way for the inhibition of EMT-mediated resistance displayed by tumor cells in response to CP exposure.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran;
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417414418, Iran;
- Kazerun Health Technology Incubator, Shiraz University of Medical Sciences, Shiraz 1433671348, Iran
| | - Mahshad Kalantari
- Department of Genetic Science, Tehran Medical Science Branch, Islamic Azad University, Tehran 19168931813, Iran;
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 1355576169, Iran
| | - Tahereh Javaheri
- Health Informatics Lab, Metropolitan College, Boston University, Boston, MA 02215, USA
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
| |
Collapse
|
27
|
Xiong LL, Qiu DL, Xiu GH, Al-Hawwas M, Jiang Y, Wang YC, Hu Y, Chen L, Xia QJ, Wang TH. DPYSL2 is a novel regulator for neural stem cell differentiation in rats: revealed by Panax notoginseng saponin administration. Stem Cell Res Ther 2020; 11:155. [PMID: 32299503 PMCID: PMC7164273 DOI: 10.1186/s13287-020-01652-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/04/2020] [Accepted: 03/13/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The limited neuronal differentiation of the endogenous or grafted neural stem cells (NSCs) after brain injury hampers the clinic usage of NSCs. Panax notoginseng saponins (PNS) were extensively used for their clinical value, such as in controlling blood pressure, blood glucose, and inhibiting neuronal apoptosis and enhancing neuronal protection, but whether or not it exerts an effect in promoting neuronal differentiation of the endogenous NSCs is completely unclear and the potential underlying mechanism requires further exploration. METHODS Firstly, we determined whether PNS could successfully induce NSCs to differentiate to neurons under the serum condition. Mass spectrometry and quantitative polymerase chain reaction (Q-PCR) were then performed to screen the differentially expressed proteins (genes) between the PNS + serum and serum control group, upon which dihydropyrimidinase-like 2 (DPYSL2), a possible candidate, was then selected for the subsequent research. To further investigate the actual role of DPYSL2 in the NSC differentiation, DPYSL2-expressing lentivirus was employed to obtain DPYSL2 overexpression in NSCs. DPYSL2-knockout rats were constructed to study its effects on hippocampal neural stem cells. Immunofluorescent staining was performed to identify the differentiation direction of NSCs after 7 days from DPYSL2 transfection, as well as those from DPYSL2-knockout rats. RESULTS Seven differentially expressed protein spots were detected by PD Quest, and DPYSL2 was found as one of the key factors of NSC differentiation in a PNS-treated condition. The results of immunostaining further showed that mainly Tuj1 and GFAP-positive cells increased in the DPYSL2-overexpressed group, while both were depressed in the hippocampal NSCs in the DPYSL2-knockout rat. CONCLUSIONS The present study revealed that the differentiation direction of NSCs could be enhanced through PNS administration, and the DPYSL2 is a key regulator in promoting NSC differentiation. These results not only emphasized the effect of PNS but also indicated DPYSL2 could be a novel target to enhance the NSC differentiation in future clinical trials.
Collapse
Affiliation(s)
- Liu-Lin Xiong
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - De-Lu Qiu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guang-Hui Xiu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mohammed Al-Hawwas
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Ya Jiang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650031, China
| | - You-Cui Wang
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yue Hu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Chen
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing-Jie Xia
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting-Hua Wang
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Institute of Neuroscience, Kunming Medical University, Kunming, 650031, China.
| |
Collapse
|