1
|
Nie W, Zhao X, Zhang Y, Zeng C, Yang H, Liu B. Chlorogenic acid alleviates DNCB-induced atopic dermatitis by inhibiting the Akt1/NF-κB signaling pathway. Eur J Pharmacol 2025; 998:177534. [PMID: 40118327 DOI: 10.1016/j.ejphar.2025.177534] [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: 11/11/2024] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
OBJECTIVE Atopic dermatitis (AD) is a prevalent chronic inflammatory skin disease that significantly impacts patients' quality of life. Chlorogenic acid (CGA), a polyphenol present in various dietary sources and plants, has been shown to reduce skin inflammation. However, its efficacy and mechanisms of action in AD have not been thoroughly investigated. This study aimed to evaluate the therapeutic effect of CGA on AD in mice and explored its mechanism. METHODS To establish a BALB/c mouse model of AD induced by 2,4-dinitrochlorobenzene (DNCB) to evaluate the therapeutic potential of CGA. The anti-inflammatory effects of CGA were assessed by measuring IL-1β and IL-6 levels in TNF-α-stimulated HaCaT cells. The phosphorylation levels of PI3K, Akt, Akt1, NF-κB, and IκB-α were analyzed using Western blotting. Molecular docking was conducted to evaluate the binding affinity of CGA to Akt1. RESULTS Topical application of CGA significantly reduced dermatitis scores, spleen index, epidermal thickness, mast cell infiltration, and skin fibrosis. CGA reversed DNCB-induced increases in IgE, histamine, TNF-α, IL-1β, IL-6, and IL-8 levels. Western blot analysis showed that CGA inhibited the PI3K/Akt and NF-κB signaling pathways. In vitro, CGA exerts its anti-inflammatory effects by inhibiting the Akt1/NF-κB pathway, and the Akt activator (SC79) can counteract this effect. Molecular docking and dynamics simulations suggest that CGA may inhibit Akt1 activity by interacting with specific residues (ALA-50, GLY-37, TYR-326, ASP-323). CONCLUSIONS CGA improves AD by inhibiting the Akt1/NF-κB pathway, suggesting its potential as a natural treatment for AD.
Collapse
Affiliation(s)
- Wenkai Nie
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xuan Zhao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yan Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Cheng Zeng
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Huiwen Yang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Bing Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510699, China.
| |
Collapse
|
2
|
Li A, Liu P, Gan J, Fang W, Liu A. Phellodendrine Exerts Protective Effects on Intra-abdominal Sepsis by Inactivating AKT/NF-kB Signaling. Cell Biochem Biophys 2025; 83:2489-2497. [PMID: 39953352 DOI: 10.1007/s12013-024-01658-2] [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] [Accepted: 12/19/2024] [Indexed: 02/17/2025]
Abstract
Acute kidney injury (AKI) and acute lung injury (ALI) are major complications of intra-abdominal sepsis, leading to increased mortality. Phellodendrine (PHE) is a characteristic and important active ingredient of Phellodendri Cortex, possessing multiple pharmacological properties. This study intends to explore the effect of PHE on intra-abdominal sepsis-induced AKI and ALI. An intra-abdominal infection-induced rat model of sepsis was established by fecal intraperitoneal injection, followed by the administration of PHE. ELISA was used to determine plasma levels of inflammatory cytokines. Hematoxylin-eosin, Periodic acid Schiff, and Masson trichrome staining were employed for histopathological analysis of rat kidney and lung tissues. Western blotting was used to estimate the AKT/NF-κB signaling-related protein levels. The results showed that PHE improved the survival rate of septic rats and reduced plasma levels of proinflammatory cytokines. PHE administration attenuated pathological lesions in the kidneys and lungs of septic rats. Mechanistically, PHE treatment blocked AKT/NF-κB signaling in septic rats' kidneys and lungs. In conclusion, PHE ameliorates intra-abdominal sepsis-induced kidney and lung injury possibly by inactivating AKT/NF-kB signaling.
Collapse
Affiliation(s)
- Ang Li
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Peng Liu
- Department of Emergency, Wuhan Fourth Hospital, Wuhan, China
| | - Jiaohong Gan
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weijun Fang
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Anjie Liu
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
3
|
Dong H, Zhang Y, Zhou Y, Zhang M, Zhang L, Feng J, Wu W, Liu Y, Wang T. Gastrodia elata Blume extract alleviates sepsis-induced lung injury by suppressing IL-23/IL-17 A axis. Fitoterapia 2025; 184:106624. [PMID: 40398516 DOI: 10.1016/j.fitote.2025.106624] [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: 12/08/2024] [Revised: 05/03/2025] [Accepted: 05/11/2025] [Indexed: 05/23/2025]
Abstract
The Compendium of Materia Medica recorded that the traditional Chinese medicine Gastrodia elata Blume (G. elata) has applications in antibacterial and antiviral. GE05 was prepared from the rhizome of G. elata and its active ingredients have been proven to alleviate inflammatory response. However, the protective effect and the underlying mechanism of G. elata in sepsis remain unclear. Our study aims to uncover the efficacy and molecular mechanisms of GE05 in ameliorating sepsis-induced lung injury. Here, we have shown that GE05 could inhibit the expression of inflammatory cytokines in peritoneal macrophages. Bioinformatics analysis showed the activation of interleukin (IL)-17 signaling pathway in acute lung injury (ALI) mice. We established septicemia models and showed that GE05 improves survival rates and protects against sepsis-induced lung injury by downregulating the IL-23/IL-17 A axis. Quantitative real- time PCR (qPCR) analysis and immunohistochemistry have indicated that IL-17 A inhibition reduces the release of chemokine ligand (Cxcl) 1, Cxcl2, granulocyte-macrophage colony stimulating factor (GM-CSF), and granulocyte colony-stimulating factor (G-CSF), mitigating neutrophil infiltration-induced lung tissue damage. Meanwhile, GE05 could inhibit the activation of the IL-17 A-related phosphatidylinositol 3-kinase/ c-Jun N-terminal kinase (PI3K/JNK) signaling pathway, suppressing the expression of tumor necrosis factor-alpha (TNF-α), IL-1β, and IL-6. These results demonstrated that GE05 is a promising agent that targets the IL-23/IL-17 A axis, providing new way for preventing and treating sepsis-induced lung injury.
Collapse
Affiliation(s)
- Huiqing Dong
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China
| | - Yun Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yang Zhou
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Menghui Zhang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China; School of Pharmacy, Henan University, Kaifeng 475001, China
| | - Lei Zhang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China
| | - Jing Feng
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Wanying Wu
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yifei Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China
| | - Ting Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 101408, China; Yunnan Engineering Research Center of Green Planting and Processing of Gastrodia, Zhaotong University, Zhaotong 657000, Yunnan, China; Yunnan Key Laboratory of Gastrodia and Fungi Symbiotic Biology, Zhaotong University, Zhaotong 657000, Yunnan, China.
| |
Collapse
|
4
|
Yu W, Jie S, Su G, Zhuangwei N, Yiqing Z, Suhong C, Guiyuan L. The ultrafine powder of atractylodis macrocephalae rhizoma improves immune function in naturally aging rats by regulating the PI3K/Akt/NF-κB signaling pathway. Front Pharmacol 2025; 16:1550357. [PMID: 40255567 PMCID: PMC12006087 DOI: 10.3389/fphar.2025.1550357] [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: 12/23/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025] Open
Abstract
Background The phenomenon of population aging presents a significant global challenge, with the aging population in China steadily increasing. As individuals progress in age, there is a gradual deterioration of human organs and systems, as well as a decline in the immune system, referred to as immunosenescence. Atractylodis macrocephalae rhizoma (BZ) has been historically used in China for its medicinal properties, including gastrointestinal improvement, immunomodulation, anti-aging, antioxidant effects, and anti-tumor effects. Nevertheless, there remains a gap in understanding the pharmacological and molecular mechanisms underlying its anti-immunosenescence effects. Methods This study employed UPLC-ESI-MS and network pharmacology to create a network map of BZ ultrafine powder (BZU) and its aging targets. Enrichment analysis was then used to identify the primary mechanistic pathways underlying BZU's anti-immunosenescence effects. The primary components of BZU were quantitatively analyzed using high-performance liquid chromatography (HPLC). Naturally aging rats were used to examine the effects of different oral doses (0.25, 0.5, and 1 g/kg) of BZU over 5 weeks on aging performance, peripheral blood immunophenotyping and cell count, and splenic lymphocyte proliferation rate. To validate the findings of network pharmacology, quantitative RT-PCR, Western blotting, and immunofluorescence analyses were conducted. Results Our analyses demonstrated that BZU improved various indicators of aging in naturally aging rats, such as increasing the number of voluntary activities, enhance grip strength and fatigue resistance, increasing the microcirculatory blood flow and improving hematological levels. The BZU administration enhanced T and B lymphocyte proliferation and significantly improved the lymphocyte-to-T cell subpopulation ratio. It can elevate serum IL-2 and IL-4 levels while reducing IL-6, IFN-γ and TNF-α levels in naturally aging rats. Finally, it increased CD3 protein expression in the spleen while decreasing protein levels of PI3K, p-AKT, IKKα/β, and NF-κB. It also decreased the mRNA expression of Pik3cg, Akt1, Pdk1 and Nfκb1. Conclusion These findings suggest that BZU may enhance lymphocyte proliferation by inhibiting the PI3K/Akt/NF-κB signaling pathway, correcting immune cell imbalances, reducing inflammatory responses, and ultimately enhancing immune function and potentially delaying aging.
Collapse
Affiliation(s)
| | | | | | | | | | - Chen Suhong
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lv Guiyuan
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
5
|
Selvaraju V, Babu SR, Judd RL, Geetha T. Lupeol Attenuates Palmitate-Induced Hypertrophy in 3T3-L1 Adipocytes. Biomolecules 2025; 15:129. [PMID: 39858523 PMCID: PMC11763665 DOI: 10.3390/biom15010129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
Obesity is characterized by the enlargement of adipose tissue due to an increased calorie intake exceeding the body's energy expenditure. Changes in the size of adipose tissue can lead to harmful consequences, with excessive fat accumulation resulting in adipocyte hypertrophy and promoting metabolic dysfunction. These adiposity-associated pathologies can be influenced by dietary components and their potential health benefits. Lupeol, a pharmacologically active pentacyclic triterpenoid found in medicinal plants, vegetables, and fruits, has been shown to exhibit antioxidant and anti-inflammatory properties. This study investigated the role of lupeol on adipocyte hypertrophy by evaluating key adipogenic regulators in vitro. First, 3T3-L1 MBX mouse embryonic cells were differentiated into adipocytes and hypertrophy was induced using 500 µM palmitic acid. The treated adipocytes showed a significantly increased lipid droplet size, confirming adipocyte hypertrophy. Both adipocytes and hypertrophied adipocytes were then treated with or without 60 µM lupeol, following a dose-dependent study. Lipid droplet size was assessed and validated by Oil Red O staining. Western blot analysis was performed to measure the expression of adipogenic and inflammatory markers. Differentiated adipocytes showed increased fatty acid-binding protein 4 (FABP4) expression and Oil Red O staining, indicating an increased lipid content. Western blot analysis revealed that lupeol treatment reduced the expression of FABP4, peroxisome proliferator-activated receptor-γ (PPARγ), and adipokines. In conclusion, the results suggest that lupeol reverts the inflammatory and adipogenic markers that are enhanced in adipocyte hypertrophy. Through its anti-inflammatory effects, lupeol offers protective effects against adipocyte hypertrophy and contributes to reducing hypertrophic adiposity.
Collapse
Affiliation(s)
| | - Shivani R. Babu
- Department of Anatomy, Physiology and Pharmacology, Auburn University, Auburn, AL 36849, USA
| | - Robert L. Judd
- Department of Anatomy, Physiology and Pharmacology, Auburn University, Auburn, AL 36849, USA
| | - Thangiah Geetha
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
6
|
Lü J, Jiang C, Drabick JJ, Joshi M, Perimbeti S. Angelica gigas Nakai (Korean Dang-gui) Root Alcoholic Extracts in Health Promotion and Disease Therapy - active Phytochemicals and In Vivo Molecular Targets. Pharm Res 2025; 42:25-47. [PMID: 39779619 PMCID: PMC11785709 DOI: 10.1007/s11095-024-03809-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
Angelica gigas Nakai (AGN) root is a medicinal herbal widely used in traditional medicine in Korea. AGN root ethanolic extracts have been marketed as dietary supplements in the United States for memory health and pain management. We have recently reviewed the pharmacokinetics (PK) and first-pass hepatic metabolism of ingested AGN supplements in humans for the signature pyranocoumarins decursin (D, Cmax 1x), decursinol angelate (DA, Cmax ~ 10x) and their common botanical precursor and hepatic metabolite decursinol (DOH, Cmax ~ 1000x). Here we update in vivo medicinal activities of AGN and/or its pyranocoumarins and furanocoumarin nodakenin in cancer, pain, memory loss, cerebral ischemia reperfusion stroke, metabolic syndrome and vascular endothelial dysfunctions, anxiety, sleep disorder, epilepsy, inflammatory bowel disease, osteoporosis and osteoarthritis. Given their polypharmacology nature, the pertinent mechanisms of action are likely misrepresented by many cell culture studies that did not consider the drug metabolism knowledge. We report here Rho-associated protein kinases (ROCK1/2) as novel targets for DA and DOH. Combining with published inhibitory activity of DOH on acetylcholinesterase, agonist activity of DOH and antagonist/degrader activity of DA/D on androgen and estrogen receptors, D/DA promoting activity for glutamic acid decarboxylase (GAD)- gamma-aminobutyric acid (GABA) inhibitory axis and inhibition of glutamate dehydrogenase (GDH), monoamine oxidase-A (MAO-A) and transient receptor potential vanilloid 1 (TRPV1), we postulate their contributions to neuro-cognitive, metabolic, oncologic, vascular and other beneficial bioactivities of AGN extracts. A clinical trial is being planned for an AGN extract to manage side effects of androgen deprivation therapy in prostate cancer patients.
Collapse
Affiliation(s)
- Junxuan Lü
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Penn State Cancer Institute, Pennsylvania State University, Hershey, PA, 17033, USA.
- Center for Cannabis and Natural Product Pharmaceutics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | - Cheng Jiang
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
- Center for Cannabis and Natural Product Pharmaceutics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Joseph J Drabick
- Penn State Cancer Institute, Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Medicine Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Monika Joshi
- Penn State Cancer Institute, Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Medicine Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Stuthi Perimbeti
- Penn State Cancer Institute, Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Medicine Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| |
Collapse
|
7
|
Lv H, Wang J, Wan Y, Zhou Y. Exploration of the Key Pathways and Genes Involved in Osteoarthritis Genesis: Evidence from Multiple Platforms and Real-World Validation. J Inflamm Res 2024; 17:10223-10237. [PMID: 39649419 PMCID: PMC11625429 DOI: 10.2147/jir.s488935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/14/2024] [Indexed: 12/10/2024] Open
Abstract
Background Osteoarthritis (OA), a degenerative and chronic joint disease, is essential for identifying novel biomarkers for the clinical diagnosis of OA. Methods We collected 35 OA patients and 32 healthy controls from four clinical cohorts and 8 real-world samples from our institute. The activation status of 7530 signalling pathways was calculated via the gene set enrichment analysis (GSEA) algorithm. Ten machine learning algorithms and 101 algorithm combinations were further applied to recognize the most diagnostic genes. KDELR3 was chosen for further validation via immunohistochemical staining to determine its diagnostic value in real-world samples. Results Sixteen pathways, namely, the cellular respiration chain, protein transport, lysosomal and endocytosis pathways, were activated in OA patients. A total of 101 types of algorithm combinations were considered for the diagnostic model, and 58 were successfully output. The two-step model of glmBoost plus RF had the highest average AUC value of 0.95 and was composed of LY86, SORL1, KDELR3, CSK, PTGS1, and PTGS2. Preferable consistency of the diagnostic mole and real conditions was observed in all four cohorts (GSE55235: Kappa=1.000, P<0.001; GSE55457: Kappa=0.700, P<0.001; GSE82107: Kappa=0.643, P=0.004; GSE1919: Kappa=1.000, P<0.001). KDELR3 was expressed at higher levels in OA patients than were the other genes, and with the help of immunohistochemistry (IHC), we confirmed that OA patients presented high levels of KDELR3 in synovial tissues. The infiltration of immunocytes, macrophages, and natural killer T cells was high in OA patients. KDELR3 might be involved in the activation and infiltration of effector memory CD4 T cells (Rpearson = 0.58, P < 0.001) and natural killer T cells (Rpearson = 0.53, P < 0.001). Conclusion We constructed and validated a six-gene diagnostic model for OA patients via machine learning, and KDELR3 emerged as a novel biomarker for OA.
Collapse
Affiliation(s)
- Hao Lv
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People’s Republic of China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People’s Republic of China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People’s Republic of China
| | - Jingkun Wang
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People’s Republic of China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People’s Republic of China
| | - Yang Wan
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People’s Republic of China
| | - Yun Zhou
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People’s Republic of China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People’s Republic of China
| |
Collapse
|
8
|
Qian Y, Dong J, Zhang W, Xue X, Xiong Z, Zeng W, Wang Q, Fan Z, Zuo Z, Huang Z, Jiang Y. Deguelin inhibits the glioblastoma progression through suppressing CCL2/NFκB signaling pathway. Neuropharmacology 2024; 259:110109. [PMID: 39128581 DOI: 10.1016/j.neuropharm.2024.110109] [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: 03/25/2024] [Revised: 07/01/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Glioblastoma multiforme (GBM) is the most common primary intracranial tumor with characteristics of high aggressiveness and poor prognosis. Deguelin, a component from the bark of Leguminosae Mundulea sericea (African plant), displays antiproliferative effects in some tumors, however, the inhibitory effect and mechanism of deguelin on GBM were still poorly understood. At first, we found that deguelin reduced the viability of GBM cells by causing cell cycle arrest in G2/M phase and inducing their apoptosis. Secondly, deguelin inhibited the migration of GBM cells. Next, RNA-seq analysis identified that CCL2 (encoding chemokine CCL2) was downregulated significantly in deguelin-treated GBM cells. As reported, CCL2 promoted the cell growth, and CCL2 was associated with regulating NFκB signaling pathway, as well as involved in modulating tumor microenvironment (TME). Furthermore, we found that deguelin inactivated CCL2/NFκB signaling pathway, and exougous CCL2 could rescue the anti-inhibitory effect of deguelin on GBM cells via upregulating NFκB. Finally, we established a syngeneic intracranial orthotopic GBM model and found that deguelin regressed the tumor growth, contributed to an anti-tumorigenic TME and inhibited angiogenesis of GBM by suppressing CCL2/NFκB in vivo. Taken together, these results suggest the anti-GBM effect of deguelin via inhibiting CCL2/NFκB pathway, which may provide a new strategy for the treatment of GBM.
Collapse
Affiliation(s)
- Yiming Qian
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Jianhong Dong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Wei Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Xiumin Xue
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Zhenrong Xiong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Weiquan Zeng
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Qian Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Ziwei Fan
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zhenxing Zuo
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhihui Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| | - Yuanyuan Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
9
|
Du MD, He KY, Fan SQ, Li JY, Liu JF, Lei ZQ, Qin G. The Mechanism by Which Cyperus rotundus Ameliorates Osteoarthritis: A Work Based on Network Pharmacology. J Inflamm Res 2024; 17:7893-7912. [PMID: 39494203 PMCID: PMC11531273 DOI: 10.2147/jir.s483652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024] Open
Abstract
Background Cyperus rotundus (CR) is widely used in traditional Chinese medicine to prevent and treat a variety of diseases. However, its functions and mechanism of action in osteoarthritis (OA) has not been elucidated. Here, a comprehensive strategy combining network pharmacology, molecular docking, molecular dynamics simulation and in vitro experiments was used to address this issue. Methods The bioactive ingredients of CR were screened in TCMSP database, and the potential targets of these ingredients were obtained through Swiss Target Prediction database. Genes in OA pathogenesis were collected through GeneCards, OMIM and DisGeNET databases. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed using DAVID database. STRING database and Cytoscape 3.10 software were used to construct "component-target-pathway" network, and predict the core targets affected by CR. The binding affinity between bioactive components and the core targets was evaluated by molecular docking and molecular dynamics simulation. The therapeutic activity of kaempferol on chondrocytes in inflammatory conditions was verified by in vitro experiments. Results Fifteen CR bioactive ingredients were obtained, targeting 192 OA-related genes. A series of biological processes, cell components, molecular functions and pathways were predicted to be modulated by CR components. The core targets of CR in OA treatment were AKT serine/threonine kinase 1 (AKT1), interleukin 1 beta (IL1B), SRC proto-oncogene, non-receptor tyrosine kinase (SRC), BCL2 apoptosis regulator (BCL2), signal transducer and activator of transcription 3 (STAT3), epidermal growth factor receptor (EGFR), hypoxia-inducible factor 1 subunit alpha (HIF1A), matrix metallopeptidase 9 (MMP9), estrogen receptor 1 (ESR1) and PPARG orthologs from vertebrates (PPARG), and the main bioactive ingredients of CR showed good binding affinity with these targets. In addition, kaempferol, one of the CR bioactive components, weakens the effects of IL-1β on the viability, apoptosis and inflammation of chondrocytes. Conclusion Theoretically, CR has great potential to ameliorate the symptoms and progression of OA, via multiple components, multiple targets, and multiple downstream pathways.
Collapse
Affiliation(s)
- Min-Dong Du
- Department of Osteoarthrosis, Xing-An Jieshou Orthopedics Hospital, Guilin, People’s Republic of China
- Department of Orthopaedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Kai-Yi He
- Department of Osteoarthrosis, The First Affiliated Hospital of Guangxi Traditional Chinese Medical University, Nanning, People’s Republic of China
| | - Si-Qi Fan
- Department of Osteoarthrosis, The First Affiliated Hospital of Guangxi Traditional Chinese Medical University, Nanning, People’s Republic of China
| | - Jin-Yi Li
- Department of Osteoarthrosis, The First Affiliated Hospital of Guangxi Traditional Chinese Medical University, Nanning, People’s Republic of China
| | - Jin-Fu Liu
- Department of Osteoarthrosis, The First Affiliated Hospital of Guangxi Traditional Chinese Medical University, Nanning, People’s Republic of China
| | - Zi-Qiang Lei
- Department of Osteoarthrosis, The First Affiliated Hospital of Guangxi Traditional Chinese Medical University, Nanning, People’s Republic of China
| | - Gang Qin
- Department of Osteoarthrosis, The First Affiliated Hospital of Guangxi Traditional Chinese Medical University, Nanning, People’s Republic of China
| |
Collapse
|
10
|
Yu P, Peng X, Sun H, Xin Q, Kang H, Wang P, Zhao Y, Xu X, Zhou G, Xie J, Li J. Inspired by lubricin: a tailored cartilage-armor with durable lubricity and autophagy-activated antioxidation for targeted therapy of osteoarthritis. MATERIALS HORIZONS 2024; 11:5352-5365. [PMID: 39143938 DOI: 10.1039/d4mh00812j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Osteoarthritis (OA), which disables articular cartilage, affects millions of people. The self-healing capacity is inhibited by internal oxidative stress and external lubrication deficiency and enzymatic degradation. To overcome these challenges, a tailored cartilage-armor is designed to ameliorate the inflamed cartilage, which is implemented by a novel collagen type II (Col II)-binding peptide conjugated zwitterionic polymer (PSB-b-PColBP, PSP). By mimicking natural lubricin, PSP specifically targets the cartilage surface and forms an in situ hydration armor. This engineered cartilage-armor can prevent enzymatic cartilage degradation (nearly 100% resistance to catabolic enzymes) and provide durable lubrication properties (COF < 0.013 for 500 cycles). An autophagy-activation process, absent in previous biomimetic lubricants, enhances the enzymatic activity of the tailored cartilage-armor, offering effective anti-oxidant properties to suppress oxidative stress. By inhibiting the PI3K-Akt/NF-κB signaling pathway, chondrocytes protected by the tailored armor can secrete a cartilage matrix even in inflammatory microenvironments. In OA rat models, osteophyte formation and the inflammatory response have been inhibited by the cartilage-armor, demonstrating a therapeutic effect comparable to most drug-loaded systems. This study underscores the potential of tailoring cartilage-armor with the cartilage targeting and autophagy-activating properties in integrating offensive-defensive mechanisms for cartilage remodeling. This represents an alternative strategy for clinical OA therapy.
Collapse
Affiliation(s)
- Peng Yu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Xu Peng
- Experimental and Research Animal Institute, Sichuan University, Chengdu 610207, P. R. China
| | - Hui Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Qiangwei Xin
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Han Kang
- Life Science Core Facilities, College of Life Sciences, Sichuan University, Chengdu 610065, P. R. China
| | - Peng Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Yao Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Xinyuan Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Guangwu Zhou
- School of Aeronautics and Astronautics, Sichuan University, Chengdu 610207, P. R. China
| | - Jing Xie
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| |
Collapse
|
11
|
Lee J, Heo JB, Cho S, Ryu CW, Heo HJ, Yun MY, Nam G, Song GY, Bae JS. Inhibitory Effects of Decursin Derivative against Lipopolysaccharide-Induced Inflammation. Pharmaceuticals (Basel) 2024; 17:1337. [PMID: 39458978 PMCID: PMC11509908 DOI: 10.3390/ph17101337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND This study aims to explore the protective role of JB-V-60-a novel synthetic derivative of decur-sin-against lipopolysaccharide (LPS)-induced inflammation. METHODS We examined the effects of JB-V-60 on heme oxygenase (HO)-1, cyclooxygenase (COX)-2, and inducible nitric oxide synthase (iNOS) in LPS-activated human pulmonary artery endothelial cells (HPAECs). Additionally, we assessed its effects on iNOS, tumor necrosis factor (TNF)-α, and interleukin (IL)-1β in LPS-exposed mice. RESULTS JB-V-60 enhanced HO-1 levels, inhibited NF-κB activation, reduced COX-2/PGE2 and iNOS/NO concentra-tions, and lowered phosphorylation of signal transducer and activator of transcription 1. It also promoted the translocation of Nrf2 into the nucleus, allowing its binding to antioxidant response elements and resulting in reduced IL-1β in LPS-stimulated HPAECs. The reduction in iNOS/NO levels by JB-V-60 was reversed when HO-1 was inhibited via RNAi. In the animal model, JB-V-60 sig-nificantly decreased iNOS expression in lung tissues and TNF-α levels in bronchoalveolar lavage fluid. CONCLUSIONS These findings highlight the anti-inflammatory effects of JB-V-60 and its potential as a treat-ment for inflammatory disorders.
Collapse
Affiliation(s)
- Jinhee Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea; (J.L.); (S.C.); (C.-W.R.)
| | - Jong-Beom Heo
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejon 34134, Republic of Korea; (J.-B.H.); (H.-J.H.)
| | - Sanghee Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea; (J.L.); (S.C.); (C.-W.R.)
| | - Chang-Woo Ryu
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea; (J.L.); (S.C.); (C.-W.R.)
| | - Hae-Joon Heo
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejon 34134, Republic of Korea; (J.-B.H.); (H.-J.H.)
| | - Mi-Young Yun
- Department of Beauty Science, Kwangju Women’s University, Gwangju 62396, Republic of Korea;
| | - Gaewon Nam
- Department of Bio-Cosmetic Science, Seowon University 377-3, Musimseoro, Seowon-gu, Cheongju, Chungbuk 28674, Republic of Korea;
| | - Gyu-Yong Song
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejon 34134, Republic of Korea; (J.-B.H.); (H.-J.H.)
| | - Jong-Sup Bae
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea; (J.L.); (S.C.); (C.-W.R.)
| |
Collapse
|
12
|
Wang L, Shao T, Liu C, Han Z, Zhang S, Dong Y, Han T, Cheng B, Ren W. Liensinine inhibits IL-1β-stimulated inflammatory response in chondrocytes and attenuates papain-induced osteoarthritis in rats. Int Immunopharmacol 2024; 138:112601. [PMID: 38971106 DOI: 10.1016/j.intimp.2024.112601] [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: 12/12/2023] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/08/2024]
Abstract
Osteoarthritis (OA) is a joint disease caused by inflammation of cartilage and synovial tissue. Suppressing the process of inflammatory reaction and the generation of oxidative stress is an effective strategy to alleviate the progression of OA. Liensinine is one of the main components of lotus seeds, which has anti-hypertensive and anti-arrhythmia activities. In this study, we aimed to determine the anti-inflammatory effect of liensinine in an OA. Here, we found that liensinine significantly inhibited the inflammatory response of SW1353 cells and primary chondrocytes by inhibiting the release of inflammatory cytokines and oxidative stress. Moreover, we showed that liensinine was able to inhibit the activation of the NF-κB signaling pathway in IL-1β-induced SW1353 cells. Lastly, we found that liensinine significantly ameliorated cartilage damage and inflammatory response in papain-induced rats. Our study demonstrated a significant protective effect of liensinine against OA, which might be by inhibiting the activation of the NF-κB signaling pathway, and provide a new insight for the treatment of OA using liensinine.
Collapse
Affiliation(s)
- Lei Wang
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, Henan, China; Xinxiang Key Laboratory of Cellular Stress Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Tianci Shao
- Xinxiang Key Laboratory of Cellular Stress Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Chen Liu
- Xinxiang Key Laboratory of Cellular Stress Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Ziyu Han
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Shenghui Zhang
- Xinxiang Key Laboratory of Cellular Stress Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Yuqian Dong
- Xinxiang Key Laboratory of Cellular Stress Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Tao Han
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Binfeng Cheng
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China; Xinxiang Key Laboratory of Cellular Stress Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Wenjie Ren
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| |
Collapse
|
13
|
Tang Z, Feng H, Chen X, Shao S, Li C. SNORC knockdown alleviates inflammation, autophagy defect and matrix degradation of chondrocytes in osteoarthritis development. Mol Cell Biochem 2024; 479:2323-2335. [PMID: 37659033 DOI: 10.1007/s11010-023-04842-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/23/2023] [Indexed: 09/05/2023]
Abstract
Excessive inflammation and autophagy defect of chondrocytes play important roles in the pathological process of osteoarthritis (OA). The present study aimed to clarify the roles of small novel rich in cartilage (SNORC) in these pathological changes of chondrocytes in OA. Bioinformatics analysis of GEO dataset GSE207881 displayed that SNORC was a potential biomarker for OA. As confirmed by quantitative real-time PCR, immunohistochemical staining and western blotting, SNORC was significantly up-regulated in cartilage of OA rat model and interleukin (IL)-1β-stimulated primary rat articular chondrocytes in contrast to their corresponding normal control. Knocking down SNORC in IL-1β-induced chondrocytes obviously suppressed the production of nitric oxide (NO), IL-6, tumor necrosis factor (TNF)-α and prostaglandin E2 (PGE2) to alleviate inflammation, and reduced the protein levels of a disintegrin and metalloproteinase with thrombospondin 5 (ADAMTS5) and matrix metallopeptidase (MMP)13 and elevated collagen type 2 alpha 1 (COL2A1) level to improve matrix degradation. Down-regulation of SNORC increased Beclin1 expression and LC3II/LC3I ratio, but suppressed p62 expression to restore impaired autophagy in IL-1β-induced chondrocytes. Moreover, down-regulating SNORC mitigated mitochondrial dysfunction and apoptosis in IL-1β-stimulated chondrocytes. Mechanically, SNORC simultaneously activated the phosphatidylinositol-3-kinase/serine threonine kinase (PI3K/AKT) and c-Jun N-terminal kinase (JNK)/c-Jun signaling pathway in the IL-1β-induced chondrocyte, while re-activating the PI3K and JNK signals abolished the suppressive effect of down-regulating SNORC on IL-1β-induced chondrocyte damage. In a word, SNORC knockdown alleviates inflammation, matrix degradation, autophagy defect and excessive apoptosis of chondrocytes during OA development via suppressing the PI3K and JNK signaling pathway.
Collapse
Affiliation(s)
- Zhifang Tang
- Clinical Medical College of Dali University, Dali, 671000, China
| | - Hanzhen Feng
- Clinical Medical College of Dali University, Dali, 671000, China
| | - Xusheng Chen
- Kunming Medical University, Kunming, 650500, China
| | - Shuiyan Shao
- Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Chuan Li
- Institute of Traumatology and Orthopedics, 920th Hospital of Joint Logistics Support Force, PLA, No.212 Daguan Road, Xishan District, Kunming, Yunnan, 650000, China.
| |
Collapse
|
14
|
Jo HG, Baek CY, Hwang Y, Baek E, Song HS, Lee D. Pain Relief, Functional Recovery, and Chondroprotective Effects of Angelica gigas Nakai in Osteoarthritis Due to Its Anti-Inflammatory Property: An In Vitro and In Vivo Study. Nutrients 2024; 16:2435. [PMID: 39125316 PMCID: PMC11314059 DOI: 10.3390/nu16152435] [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: 06/26/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Osteoarthritis (OA), characterized by chronic pain and joint degradation, is a progressive joint disease primarily induced by age-related systemic inflammation. Angelica gigas Nakai (AG), a medicinal plant widely used in East Asia, exhibits promising results for such conditions. This study aimed to evaluate the potential of AG as a drug candidate for modulating the multifaceted pathology of OA based on its anti-inflammatory properties. We evaluated the efficacy of AG in pain relief, functional improvement, and cartilage erosion delay using monosodium iodoacetate-induced OA rats and acetic acid-induced writhing mice, along with its anti-inflammatory effects on multiple targets in the serum and cartilage of in vivo models and lipopolysaccharide-stimulated RAW 264.7 cells. In vivo experiments demonstrated significant analgesic and chondroprotective effects of AG, along with functional recovery, in model animals compared with the active controls. AG dose-dependently modulated inflammatory OA pathology-related targets, including interleukin-1β, tumor necrosis factor-α, matrix metalloproteinase-13, and cyclooxygenase-2, both in vitro and in vivo. In conclusion, AG could be a potential drug candidate for modulating the multifaceted pathology of OA. Nevertheless, further comprehensive investigations, involving a broader range of compounds, pathologies, and mechanisms, are warranted to validate these findings.
Collapse
Affiliation(s)
- Hee-Geun Jo
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.); (C.Y.B.)
- Naturalis Inc., 6, Daewangpangyo-ro, Bundang-gu, Seongnam-si 13549, Republic of Korea
| | - Chae Yun Baek
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.); (C.Y.B.)
| | - Yeseul Hwang
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.); (C.Y.B.)
| | - Eunhye Baek
- RexSoft Inc., 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Ho Sueb Song
- Department of Acupuncture & Moxibustion Medicine, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| | - Donghun Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.); (C.Y.B.)
| |
Collapse
|
15
|
Zahran EM, Mohamad SA, Elsayed MM, Hisham M, Maher SA, Abdelmohsen UR, Elrehany M, Desoukey SY, Kamel MS. Ursolic acid inhibits NF-κB signaling and attenuates MMP-9/TIMP-1 in progressive osteoarthritis: a network pharmacology-based analysis. RSC Adv 2024; 14:18296-18310. [PMID: 38863821 PMCID: PMC11165403 DOI: 10.1039/d4ra02780a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 05/30/2024] [Indexed: 06/13/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease, characterized by infiltration of monocytes into the synovial joint which promotes inflammation, stiffness, joint swelling, cartilage degradation and further bone destruction. The leaves of Ocimum forskolei have been used for inflammation-related disease management in traditional medicine. Additionally, the downregulation of NF-κB and the MMP/TIMP-1 ratio has been shown to protect against OA. The LC-HR-MS metabolic analysis of Ocimum yielded 19 putative compounds, among which ursolic acid (UA) was detected. Ursolic acid possesses significant anti-inflammatory effects and has been reported to downregulate oxidative stress and inflammatory biomarkers. It was tested on rats in a model of intra-articular carrageenan injection to investigate its efficacy on osteoarthritis progression. The UA emulgel exerted chondroprotective, analgesic and local anaesthetic efficacies confirmed via histopathological investigation and radiographical imaging. A network pharmacology followed by molecular docking highlighted TNF-α, TGF-β and NF-κB as the top filtered genes. Quantitative real-time PCR analysis showed that UA significantly attenuated serum levels of TNF-α, IL-1β, NF-κB, MMP-9/TIMP-1 and elevated levels of TGF-β. Taken together, these results suggest that UA could serve as a functional food-derived phytochemical with a multi-targeted efficacy on progression of OA, regulating the immune and inflammatory responses, particularly, attenuating chondrocytes degeneration via suppression of NF-κB and MMP-9/TIMP-1. Accordingly, UA might be a promising alternative to conventional therapy for safe, easily applicable and effective management of OA.
Collapse
Affiliation(s)
- Eman Maher Zahran
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University Universities Zone New Minia City 61111 Egypt
| | - Soad A Mohamad
- Department of Pharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Deraya University Universities Zone New Minia City 61111 Egypt
| | - Mohamed M Elsayed
- Faculty of Pharmacy, Deraya University Universities Zone New Minia City 61111 Egypt
| | - Mohamed Hisham
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Deraya University Universities Zone New Minia City 61111 Egypt
| | - Sherif A Maher
- Department of Biochemistry, Faculty of Pharmacy, New Valley University New Valley Elkharga 71511 Egypt
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University Universities Zone New Minia City 61111 Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University 61519 Minia Egypt
| | - Mahmoud Elrehany
- Department of Biochemistry, Faculty of Pharmacy, Deraya University Universities Zone New Minia City 61111 Egypt
| | - Samar Yehia Desoukey
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University 61519 Minia Egypt
| | - Mohamed Salah Kamel
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University 61519 Minia Egypt
| |
Collapse
|
16
|
Fan Y, Yin L, Zhong X, He Z, Meng X, Chai F, Kong M, Zhang Q, Xia C, Tong Y, Bi Q. An integrated network pharmacology, molecular docking and experiment validation study to investigate the potential mechanism of Isobavachalcone in the treatment of osteoarthritis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117827. [PMID: 38310989 DOI: 10.1016/j.jep.2024.117827] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND In many different plants, including Dorstenia and Psoralea corylifolia L., Isobavachalcone (IBC) is a naturally occurring flavonoid chemical having a range of biological actions, including anti-inflammatory, immunomodulatory, and anti-bacterial. The "Theory of Medicinal Properties" of the Tang Dynasty states that Psoralea corylifolia L. has the ability to alleviate discomfort in the knees and waist. One of the most widespread chronic illnesses, osteoarthritis (OA), is characterized by stiffness and discomfort in the joints. However, there hasn't been much research done on the effectiveness and underlying processes of IBC in the treatment of osteoarthritis. AIM OF THE STUDY To investigate the potential efficacy and mechanism of IBC in treating osteoarthritis, we adopted an integrated strategy of network pharmacology, molecular docking and experiment assessment. MATERIALS AND METHODS The purpose of this research was to determine the impact of IBC on OA and the underlying mechanisms. IBC and OA possible targets and processes were predicted using network pharmacology, including the relationship between IBC and OA intersection targets, Cytoscape protein-protein interaction (PPI) to obtain key potential targets, and GO and KEGG pathway enrichment analysis to reveal the probable mechanism of IBC on OA. Following that, in vitro tests were carried out to confirm the expected underlying processes. Finally, in vivo tests clarified IBC's therapeutic efficacy on OA. RESULTS We anticipated and validated that the impact of IBC on osteoarthritis is mostly controlled by the PI3K-AKT-NF-κB signaling pathway by combining the findings of network pharmacology analysis, molecular docking and Experiment Validation. CONCLUSIONS This study reveals the IBC has potential to delay OA development.
Collapse
Affiliation(s)
- Yong Fan
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China; Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Li Yin
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China; Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Xugang Zhong
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Zeju He
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Xiang Meng
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Fang Chai
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Mingxiang Kong
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Qiong Zhang
- Department of Nursing, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Chen Xia
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China.
| | - Yu Tong
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China.
| | - Qing Bi
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China; Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China; Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China.
| |
Collapse
|
17
|
Li L, Fan Q, Zhao Y, Zhang Q, Qin G, Li C, Li W. Gentiopicroside ameliorates the lipopolysaccharide-induced inflammatory response and hypertrophy in chondrocytes. J Orthop Surg Res 2024; 19:198. [PMID: 38528538 DOI: 10.1186/s13018-024-04676-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/14/2024] [Indexed: 03/27/2024] Open
Abstract
PURPOSE This study aimed to evaluate the protective effects of gentiopicroside against lipopolysaccharide-induced chondrocyte inflammation. METHODS SW 1353 chondrosarcoma cells were stimulated with LPS (5 μg/ml) for 24 h and treated with different concentrations of gentiopicroside (GPS) for 24 h. The toxic effects of GPS on chondrocytes were determined using a CCK-8 assay and EdU staining. Western blotting, qPCR, and immunofluorescence analysis were used to examine the protective effect of GPS against the inflammatory response in chondrocytes induced by lipopolysaccharide (LPS). One-way ANOVA was used to compare the differences between the groups (significance level of 0.05). RESULTS The CCK-8 results showed that 10, 20 and 40 μM GPS had no significant toxic effects on chondrocytes; GPS effectively reduced the production of IL-1β and PGE2, reversed LPS-induced extracellular matrix degradation in cartilage by inhibiting the Stat3/Runx2 signaling pathway, and suppressed the hypertrophic transformation of SW 1353 chondrosarcoma cells. CONCLUSION Our study demonstrated that GPS significantly inhibited the LPS-induced inflammatory response and hypertrophic cellular degeneration in SW 1353 chondrosarcoma cells and is a valuable traditional Chinese medicine for the treatment of knee osteoarthritis.
Collapse
Affiliation(s)
- Longfei Li
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai, Shandong, China
| | - Qianqian Fan
- Department of Rehabilitation, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yixuan Zhao
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai, Shandong, China
| | - Qian Zhang
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai, Shandong, China
| | - Gaofeng Qin
- Department of Rehabilitation, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chen Li
- Department of Rehabilitation, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Wei Li
- Department of Rehabilitation, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
18
|
Jiang Y, Zhong S, Tan H, Fu Y, Lai J, Liu L, Weng J, Chen H, He S. Study on the mechanism of action of Saposhnikovia divaricata and its key phytochemical on rheumatoid arthritis based on network pharmacology and bioinformatics. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117586. [PMID: 38104871 DOI: 10.1016/j.jep.2023.117586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saposhnikovia divaricata (Turcz.) Schischk (SD; called "fangfeng" in China) has been widely used in the clinical treatment of rheumatoid arthritis (RA) and has shown well therapeutic effects, but the specific mechanisms of action of its bioactive phytochemicals remain unclear. AIM OF THE STUDY This study aimed to investigate the molecular biological mechanism of SD in treating RA through a pharmacology-based strategy. The SD-specific core ingredient Prangenidin was screened for further in-depth study. MATERIALS AND METHODS The bioactive phytochemicals of SD and potential targets for the treatment of RA were screened by network pharmacology, and phytochemicals-related parameters such as pharmacology, and toxicology were evaluated. The protein interaction network was established to screen the core targets, and the correlation between the core targets and RA was further validated by bioinformatics strategy. Finally, molecular docking of core components and corresponding targets was performed. The in vitro experiments were performed to elucidate the regulation of Prangenidin on MH7A cells and on the PI3K/AKT pathway, and the in vivo therapeutic effect of Prangenidin was validated in collagen-induced arthritis (CIA) mice. RESULTS A total of 18 bioactive phytochemicals and 66 potential target genes intersecting with the screened RA disease target genes were identified from SD. Finally, core ingredients such as wogonin, beta-sitosterol, 5-O-Methylvisamminol, and prangenidin and core targets such as PTGS2, RELA, and AKT1 were obtained. The underlying mechanism of SD in treating RA might be achieved by regulating pathways such as PI3K/AKT, IL-17 pathway, apoptosis, and multiple biological processes to exert anti-inflammatory and immunomodulatory effects. Molecular docking confirmed that all core ingredients and key targets had great docking activity. Prangenidin inhibited viability, migration, and invasion, and induced apoptosis in MH7A cells. Prangenidin also reduced the production of IL-1β, IL-6, IL-8, MMP-1, and MMP-3. Molecular analysis showed that Prangenidin exerts its regulatory effect on MH7A cells by inhibiting PI3K/AKT pathway. Treatment with Prangenidin ameliorated synovial inflammation in the joints of mice with CIA. CONCLUSION Our findings provide insights into the therapeutic effects of SD on RA, successfully predicting the effective ingredients and potential targets, which could suggest a novel theoretical basis for further exploration of its molecular mechanisms. It also revealed that Prangenidin inhibited viability, migration, invasion, cytokine, and MMPs expression, and induced apoptosis in RA FLSs via the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Yong Jiang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Shuxin Zhong
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Huangsheng Tan
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Yuanfei Fu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Juyi Lai
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Lijin Liu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Juanling Weng
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Hanwei Chen
- Department of Radiology, Panyu Health Management Center (Panyu Rehabilitation Hospital), Guangzhou, 511495, China.
| | - Shenghua He
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China.
| |
Collapse
|
19
|
Lee SJ, Lee AY, Pak SW, Kim WI, Yang YG, Lim JO, Chae SW, Cho YK, Kim JC, Moon BC, Seo YS, Shin IS. Protective effects of Angelica decursiva Franchet & Savatier on allergic responses through enhancement of Nrf2 and suppression of NF-kB/MMP-9 in ovalbumin-exposed mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116863. [PMID: 37423516 DOI: 10.1016/j.jep.2023.116863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Angelica decursiva Franchet & Savatier is a traditional medicinal plant used to treat asthma, cough, headache, pyrexia and thick phlegm in China, Japan and Korea. A. decursiva contains many types of coumarins, which can exert several pharmacological activities including anti-inflammatory and antioxidant properties for treating various diseases such as pneumonitis, atopic dermatitis, diabetes, and Alzheimer's disease. AIM OF THE STUDY In this study, we analyzed the components of A. decursiva ethanol extract (ADE) by high performance liquid chromatography (HPLC) and investigated the therapeutic effects of ADE against allergic asthma using lipopolysaccharide (LPS) stimulated RAW264.7 cells and an ovalbumin (OVA)-exposed allergic asthma model. To elucidate the mechanism of action of ADE, we examined the protein expression through network pharmacological analysis. MATERIALS AND METHODS To establish asthma model, the mice were sensitized on day 0 and 14 via intraperitoneal injection of OVA with aluminum hydroxide. The mice were inhaled with OVA using an ultrasonic nebulizer on day 21, 22 and 23. ADE (50 and 100 mg/kg) was administered to mice by oral gave form day 18-23. On day 24, airway hyperresponsiveness (AHR) was measured using flexivent. On day 25, the mice were sacrificed and collected bronchoalveolar lavage fluids (BALF), serum and lung tissue. In LPS-stimulated RAW264.7 cell, nitric oxide and cytokines were measured. Additionally, expression of nuclear factor erythroid-2-related factor (Nrf2) and suppression of nuclear factor (NF)-κB were detected using double-immunofluorescence. RESULTS We detected the five coumarin components which included nodakenin, umbelliferon, (-)-marmesin (=nodakenetin), bergapten, and decursin, in ADE by high performance liquid chromatography. Treatment with ADE decreased the production of nitric oxide, interleukin (IL)-6 and tumor necrosis factor (TNF)-α in LPS-stimulated RAW264.7 cells accompanied by the enhanced expression of nuclear factor erythroid-2-related factor (Nrf2) and suppression of nuclear factor (NF)-κB. In the asthma model, the administration of ADE reduced inflammatory cell count and airway hyperresponsiveness in OVA-exposed animals with decreased levels of IL-4, IL-13, and OVA-specific immunoglobulin E. These results were accompanied by the reduction of pulmonary inflammation and mucus secretion. Furthermore, ADE administration inhibited the expression of NF-κB and matrix metalloproteinase (MMP)-9 in OVA-exposed animals, which was consistent with the results of network pharmacological analysis. CONCLUSION This study demonstrated that ADE effectively attenuated allergic inflammation induced by OVA inhalation through the enhancement of Nrf2 expression and suppression of NF-κB expression. Therefore, ADE may be a potential therapeutic agent for controlling asthma.
Collapse
Affiliation(s)
- Se-Jin Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - A Yeong Lee
- Herbal Medicine Resources Research Division, Korea Institute of Oriental Medicine, 177 Geonjae-ro, Naju-si, Jeonnam, 58245, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea; Department of BioMedical-Chemical Engineering, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Gyeonggi-do, Bucheon-si, 14662, Republic of Korea
| | - So-Won Pak
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Woong-Il Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Yea-Gin Yang
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Je-Oh Lim
- Herbal Medicine Resources Research Division, Korea Institute of Oriental Medicine, 177 Geonjae-ro, Naju-si, Jeonnam, 58245, Republic of Korea
| | - Sung-Wook Chae
- KM Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea; Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology (KIT), 30 Baekhak1-gil, Jeongeup-si, Jeollabuk-do, 53212, Republic of Korea
| | - Young-Kwon Cho
- College of Health Sciences, Cheongju University, 298 Daesung-ro, Sangdang-gu, Cheongju-si, Chungbuk, 28503, Republic of Korea
| | - Jong-Choon Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Byeong Cheol Moon
- Herbal Medicine Resources Research Division, Korea Institute of Oriental Medicine, 177 Geonjae-ro, Naju-si, Jeonnam, 58245, Republic of Korea
| | - Yun-Soo Seo
- Herbal Medicine Resources Research Division, Korea Institute of Oriental Medicine, 177 Geonjae-ro, Naju-si, Jeonnam, 58245, Republic of Korea; Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology (KIT), 30 Baekhak1-gil, Jeongeup-si, Jeollabuk-do, 53212, Republic of Korea.
| | - In-Sik Shin
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
20
|
Oh E, Jang H, Ok S, Eom J, Lee H, Kim SH, Kim JH, Jeong YM, Kim KJ, Yun SP, Kwon HJ, Lee IC, Park JY, Yang S. WGA-M001, a Mixture of Total Extracts of Tagetes erecta and Ocimum basilicum, Synergistically Alleviates Cartilage Destruction by Inhibiting ERK and NF-κB Signaling. Int J Mol Sci 2023; 24:17459. [PMID: 38139287 PMCID: PMC10743532 DOI: 10.3390/ijms242417459] [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: 10/27/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Tagetes erecta and Ocimum basilicum are medicinal plants that exhibit anti-inflammatory effects against various diseases. However, their individual and combined effects on osteoarthritis (OA) are unknown. Herein, we aimed to demonstrate the effects of T. erecta, O. basilicum, and their mixture, WGA-M001, on OA pathogenesis. The administration of total extracts of T. erecta and O. basilicum reduced cartilage degradation and inflammation without causing cytotoxicity. Although WGA-M001 contained lower concentrations of the individual extracts, it strongly inhibited the expression of pathogenic factors. In vivo OA studies also supported that WGA-M001 had protective effects against cartilage destruction at lower doses than those of T. erecta and O. basilicum. Moreover, its effects were stronger than those observed using Boswellia and Perna canaliculus. WGA-M001 effectively inhibited the interleukin (IL)-1β-induced nuclear factor kappa-light-chain-enhancer of the activated B cell (NF-κB) pathway and ERK phosphorylation. Furthermore, RNA-sequence analysis also showed that WGA-M001 decreased the expression of genes related to the IL-1β-induced NF-κB and ERK signaling pathways. Therefore, WGA-M001 is more effective than the single total extracts of T. erecta and O. basilicum in attenuating OA progression by regulating ERK and NF-κB signaling. Our results open new possibilities for WGA-M001 as a potential therapeutic agent for OA treatment.
Collapse
Affiliation(s)
- Eunjeong Oh
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| | - Hahyeong Jang
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
| | - Subin Ok
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
| | - Jiwon Eom
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
| | - Hyunyong Lee
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
| | - Sung Hun Kim
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
| | - Jong Hwa Kim
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
| | - Yu Mi Jeong
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
| | - Kyeong Jin Kim
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
- Department of Horticulture, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea;
| | - Hyung-Jun Kwon
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea; (H.-J.K.); (I.-C.L.); (J.-Y.P.)
| | - In-Chul Lee
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea; (H.-J.K.); (I.-C.L.); (J.-Y.P.)
| | - Ji-Young Park
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea; (H.-J.K.); (I.-C.L.); (J.-Y.P.)
| | - Siyoung Yang
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
| |
Collapse
|
21
|
Batallé G, Bai X, Balboni G, Pol O. The Impact of UFP-512 in Mice with Osteoarthritis Pain: The Role of Hydrogen Sulfide. Antioxidants (Basel) 2023; 12:2085. [PMID: 38136204 PMCID: PMC10740868 DOI: 10.3390/antiox12122085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
The pain-relieving properties of opioids in inflammatory and neuropathic pain are heightened by hydrogen sulfide (H2S). However, whether allodynia and functional and/or emotional impairments related to osteoarthritis (OA) could be reduced by activating δ-opioid receptors (DOR) and the plausible influence of H2S on these actions has not been completely established. In female C57BL/6J mice with OA pain generated via monosodium acetate (MIA), we analyze: (i) the effects of UFP-512 (a DOR agonist), given alone and co-administered with two H2S donors, on the symptoms of allodynia, loss of grip strength (GS), and anxiodepressive-like comportment; (ii) the reversion of UFP-512 actions with naltrindole (a DOR antagonist), and (iii) the impact of UFP-512 on the expression of phosphorylated NF-kB inhibitor alpha (p-IKBα) and the antioxidant enzymes superoxide dismutase 1 (SOD-1) and glutathione sulfur transferase M1 (GSTM1); and the effects of H2S on DOR levels in the dorsal root ganglia (DRG), amygdala (AMG), and hippocampus (HIP) of MIA-injected animals. Results showed that systemic and local administration of UFP-512 dose-dependently diminished the allodynia and loss of GS caused by MIA, whose effects were potentiated by H2S and reversed by naltrindole. UFP-512 also inhibited anxiodepressive-like behaviors, normalized the overexpression of p-IKBα in DRG and HIP, and enhanced the expression of SOD-1 and GSTM1 in DRG, HIP, and/or AMG. Moreover, the increased expression of DOR triggered by H2S might support the improved analgesic actions of UFP-512 co-administered with H2S donors. This study proposes the use of DOR agonists, alone or combined with H2S donors, as a new treatment for OA pain.
Collapse
Affiliation(s)
- Gerard Batallé
- Grup de Neurofarmacologia Molecular, Institut de Recerca Sant Pau, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Xue Bai
- Grup de Neurofarmacologia Molecular, Institut de Recerca Sant Pau, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Gianfranco Balboni
- Unit of Pharmaceutical, Pharmacological and Nutraceutical Sciences, Department of Life and Environmental Sciences, University of Cagliari, 09042 Cagliari, Italy
| | - Olga Pol
- Grup de Neurofarmacologia Molecular, Institut de Recerca Sant Pau, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
22
|
Wang L, Xu H, Li X, Chen H, Zhang H, Zhu X, Lin Z, Guo S, Bao Z, Rui H, He W, Zhang H. Cucurbitacin E reduces IL-1β-induced inflammation and cartilage degeneration by inhibiting the PI3K/Akt pathway in osteoarthritic chondrocytes. J Transl Med 2023; 21:880. [PMID: 38049841 PMCID: PMC10696753 DOI: 10.1186/s12967-023-04771-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/28/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Osteoarthritis is a degenerative joint disease. Cartilage degeneration is the earliest and most important pathological change in osteoarthritis, and persistent inflammation is one of the driving factors of cartilage degeneration. Cucurbitacin E, an isolated compound in the Cucurbitacin family, has been shown to have anti-inflammatory effects, but its role and mechanism in osteoarthritic chondrocytes are unclear. METHODS For in vitro experiments, human chondrocytes were stimulated with IL-1β, and the expression of inflammatory genes was measured by Western blotting and qPCR. The expression of extracellular matrix proteins was evaluated by immunofluorescence staining, Western blotting and saffron staining. Differences in gene expression between cartilage from osteoarthritis patients and normal cartilage were analysed by bioinformatics methods, and the relationship between Cucurbitacin E and its target was analysed by a cellular thermal shift assay, molecular docking analysis and molecular dynamics simulation. For in vivo experiments, knee osteoarthritis was induced by DMM in C57BL/6 mouse knee joints, and the effect of Cucurbitacin E on knee joint degeneration was evaluated. RESULTS The in vitro experiments confirmed that Cucurbitacin E effectively inhibited the production of the inflammatory cytokine interleukin-1β(IL-1β) and cyclooxygenase-2 (COX-2) by IL-1β-stimulated chondrocytes and alleviates extracellular matrix degradation. The in vivo experiments demonstrated that Cucurbitacin E had a protective effect on the knee cartilage of C57BL/6 mice with medial meniscal instability in the osteoarthritis model. Mechanistically, bioinformatic analysis of the GSE114007 and GSE117999 datasets showed that the PI3K/AKT pathway was highly activated in osteoarthritis. Immunohistochemical analysis of PI3K/Akt signalling pathway proteins in pathological slices of human cartilage showed that the level of p-PI3K in patients with osteoarthritis was higher than that in the normal group. PI3K/Akt were upregulated in IL-1β-stimulated chondrocytes, and Cucurbitacin E intervention reversed this phenomenon. The cellular thermal shift assay, molecular docking analysis and molecular dynamics experiment showed that Cucurbitacin E had a strong binding affinity for the inhibitory target PI3K. SC79 activated Akt phosphorylation and reversed the effect of Cucurbitacin E on IL-1β-induced chondrocyte degeneration, demonstrating that Cucurbitacin E inhibits IL-1β-induced chondrocyte inflammation and degeneration by inhibiting the PI3K/AKT pathway. CONCLUSION Cucurbitacin E inhibits the activation of the PI3K/AKT pathway, thereby alleviating the progression of OA. In summary, we believe that Cucurbitacin E is a potential drug for the treatment of OA.
Collapse
Affiliation(s)
- Lin Wang
- Department of Orthopaedics, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Hui Xu
- Department of Orthopaedics, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Xin Li
- School of Basic Medicine Sciences, Anhui Medical University, Hefei, Anhui Province, China
| | - Hongwei Chen
- Department of Orthopaedics, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Haigang Zhang
- Department of Orthopaedics, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Xunpeng Zhu
- Department of Orthopaedics, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zhijie Lin
- Anhui Medical University, Hefei, Anhui Province, China
| | - Shilei Guo
- Anhui Medical University, Hefei, Anhui Province, China
| | - Zhibo Bao
- Anhui Medical University, Hefei, Anhui Province, China
| | - Haicheng Rui
- Anhui Medical University, Hefei, Anhui Province, China
| | - Wei He
- School of Basic Medicine Sciences, Anhui Medical University, Hefei, Anhui Province, China.
| | - Hui Zhang
- Department of Orthopaedics, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China.
| |
Collapse
|
23
|
Zhou W, Yang G, Wen Y, Xiao Q, Sun L, Li Y, Gong Z, Wang Y. Metabolites-Based Network Pharmacology to Preliminarily Verify In Vitro Anti-Inflammatory Effect of Ardisiacrispin B. Int J Mol Sci 2023; 24:17059. [PMID: 38069381 PMCID: PMC10707123 DOI: 10.3390/ijms242317059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Ardisiae Crenatae Radix is an ethnic medicinal herb with good anti-inflammatory activity. Ardisiacrispin B is one of the main components in Ardisiae Crenatae Radix extract, with a content of up to 16.27%, and it may be one of the pharmacological components through which Ardisiae Crenatae Radix exerts anti-inflammatory activity. At present, reports on ardisiacrispin B mainly focus on anti-tumor effects, and there have been no reports on anti-inflammatory activities. As a triterpenoid saponin, due to its large molecular weight and complex structure, the composition of substances that function in the body may include other forms after metabolism, in addition to compounds with original structures. Exploring the anti-inflammatory effects on the prototypes and metabolites of the compound may provide a more comprehensive response to the characteristics of ardisiacrispin B's anti-inflammatory action. In this study, ardisiacrispin B was analyzed for metabolites to explore its metabolic processes in vivo. Subsequently, the anti-inflammatory effects of the prototypes and metabolites were further analyzed through network pharmacology, with the expectation of discovering the signaling metabolic pathways through which they may act. Finally, the anti-inflammatory effects of ardisiacrispin B in vitro and the effects on key signaling pathways at the protein level were explored. The results of this study showed that the isolated compounds were confirmed to be ardisiacrispin B. After the metabolite analysis, a total of 26 metabolites were analyzed, and the metabolism process in rats mainly involves oxidation, dehydration, glucuronide conjugation, and others. Speculation as to the anti-inflammatory molecular mechanisms of the prototypes and metabolites of ardisiacrispin B revealed that it may exert its anti-inflammatory effects mainly by affecting the PI3K-AKT pathway. Further anti-inflammatory mechanisms demonstrated that ardisiacrispin B had a good anti-inflammatory effect on LPS-induced RAW264.7 cells and a strong inhibitory effect on NO, TNF-α, and IL-1β release in cells. Furthermore, it had significant inhibitory effects on the expression of PI3K, P-PI3K, AKT, and P-AKT. This study supplements the gaps in the knowledge on the in vivo metabolic process of ardisiacrispin B and explores its anti-inflammatory mechanism, providing an experimental basis for the development and utilization of pentacyclic triterpenoids.
Collapse
Affiliation(s)
- Wen Zhou
- School of Basic Medicine, Guizhou Medical University, Guiyang 550004, China;
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550004, China; (G.Y.); (Y.W.); (Q.X.); (L.S.)
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
| | - Guixiang Yang
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550004, China; (G.Y.); (Y.W.); (Q.X.); (L.S.)
| | - Yushuang Wen
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550004, China; (G.Y.); (Y.W.); (Q.X.); (L.S.)
| | - Qian Xiao
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550004, China; (G.Y.); (Y.W.); (Q.X.); (L.S.)
| | - Le Sun
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550004, China; (G.Y.); (Y.W.); (Q.X.); (L.S.)
| | - Yongjun Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China;
| | - Zipeng Gong
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
| | - Yonglin Wang
- School of Basic Medicine, Guizhou Medical University, Guiyang 550004, China;
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550004, China; (G.Y.); (Y.W.); (Q.X.); (L.S.)
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
| |
Collapse
|
24
|
Cao N, Wang D, Liu B, Wang Y, Han W, Tian J, Xiang L, Wang Z. Silencing of STUB1 relieves osteoarthritis via inducing NRF2-mediated M2 macrophage polarization. Mol Immunol 2023; 164:112-122. [PMID: 37992540 DOI: 10.1016/j.molimm.2023.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
OBJECTIVES Shifting macrophages towards an anti-inflammatory state is key in treating osteoarthritis (OA) by reducing inflammation and tissue damage. However, the underlying mechanisms guiding this shift remain largely undefined. STUB1, an E3 ubiquitin ligase, known for its regulatory role in macrophage polarization. This study aims to explore the function and underlying action mechanisms of STUB1 in OA. METHODS An in vivo OA model was established in rats. Hematoxylin-Eosin and safranin O-fast green staining were performed to reveal the hispathological injuries in knee-joint tissues. Immunohistochemistry and flow cytometry were performed to detect the distribution of M1 and M2 macrophages. The inflammatory response (TNF-α and IL-6 levels) was evaluated by ELISA. In vitro, the interaction between STUB1 and NFR2 was determined by CO-IP and pull-down assays. After treated with LPS (an in vitro model of OA), the viability and apoptosis of chondrocytes were measured by CCK-8 and flow cytometry, respectively. RESULTS Silencing STUB1 alleviated OA in rats, as indicated by reduced subchondral bone thickness, knee synovitis score, histopathological damages, and inflammatory response. STUB1 silencing also decreased M1 macrophages and increased M2 macrophages in both in vivo and in vitro settings. NRF2 was identified as a target of STUB1, with STUB1 mediating its ubiquitination. Silencing NRF2 reversed the effects of STUB1 silencing on inducing M2 macrophage polarization. Furthermore, silencing STUB1 upregulated NRF2 expression in LPS-treated chondrocytes, promoting cell viability and inhibiting apoptosis. CONCLUSION Silencing STUB1 induces M2 macrophage polarization by inhibiting NRF2 ubiquitination, thereby contributing to the mitigation of OA.
Collapse
Affiliation(s)
- Nan Cao
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Danni Wang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Bin Liu
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Yu Wang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Wenfeng Han
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Jing Tian
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Liangbi Xiang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China.
| | - Zheng Wang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China.
| |
Collapse
|
25
|
Zhang W, Chen H, Xu Z, Zhang X, Tan X, He N, Shen J, Dong J. Liensinine pretreatment reduces inflammation, oxidative stress, apoptosis, and autophagy to alleviate sepsis acute kidney injury. Int Immunopharmacol 2023; 122:110563. [PMID: 37392573 DOI: 10.1016/j.intimp.2023.110563] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
Liensinine is mainly derived from alkaloids extracted and isolated from lotus seeds (Nelumbo nucifera Gaertn). It possesses anti-inflammatory, and antioxidant, according to contemporary pharmacological investigations. However, the effects and therapeutic mechanisms of liensinine on acute kidney injury (AKI) models of sepsis are unclear. To gain insight into these mechanisms, we established a sepsis kidney injury model by LPS injection of mice treated with liensinine, and stimulation of HK-2 with LPS in vitro and treated with liensinine and inhibitors of p38 MAPK, JNK MAPK. We first found that liensinine significantly reduced kidney injury in sepsis mice, while suppressing excessive inflammatory responses, restoring renal oxidative stress-related biomarkers, reducing increased apoptosis in TUNEL-positive cells and excessive autophagy, and that this process was accompanied by an increase in JNK/ p38-ATF 2 axis. In vitro experiments further demonstrated that lensinine reduced the expression of KIM-1, NGAL, inhibited pro- and anti-inflammatory secretion disorders, regulated the activation of the JNK/p38-ATF 2 axis, and reduced the accumulation of ROS, as well as the reduction of apoptotic cells detected by flow cytometry, and that this process played the same role as that of p38 MAPK, JNK MAPK inhibitors. We speculate that liensinine and p38 MAPK, JNK MAPK inhibitors may act on the same targets and could be involved in the mechanism of alleviating sepsis kidney injury in part through modulation of the JNK/p38-ATF 2 axis. Our study demonstrates that lensinine is a potential drug and thus provides a potential avenue for the treatment of AKI.
Collapse
Affiliation(s)
- Wei Zhang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Huizhen Chen
- Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang 222000, China
| | - Zhaoyun Xu
- Blood Transfusion Department, Ganyu District People's Hospital of Lianyungang City, Lianyungang 222100, China
| | - Xiao Zhang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xuelian Tan
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Nana He
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jinyang Shen
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| | - Jingquan Dong
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| |
Collapse
|
26
|
Lee JA, Ngo TH, Shin MR, Choi JW, Choi H, Nam JW, Roh SS. Efficacy of Veronica incana for Treating Osteoarthritis Induced by Monosodium Iodoacetate in Rats. J Med Food 2023; 26:379-389. [PMID: 37319312 DOI: 10.1089/jmf.2023.k.0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023] Open
Abstract
The aim of this study is to investigate the efficacy and the underlying mechanism of Veronica incana in osteoarthritis (OA) induced by intraarticular injection of monosodium iodoacetate (MIA). The selected major four compounds (A-D) of V. incana were found from fractions 3 and 4. Its structure elucidation was determined by liquid chromatography-electrospray ionization-mass spectrometry (LC-ESI-MS) data analysis and nuclear magnetic resonance (NMR) data comparison with literature. MIA (50 μL with 80 mg/mL) for the animal experiment was injected into the right knee joint. The V. incana was administered orally every day to rats for 14 days from 7 days after MIA treatment. Finally, we confirmed the four compounds: (A) verproside; (B) catalposide; (C) 6-vanilloylcatapol; and (D) 6-isovanilloylcatapol. When we evaluated the effect of V. incana on the MIA injection-induced knee OA model, there were a noticeable initial decreased in hind paw weight-bearing distribution compared to the Normal group (P < .001), but V. incana supplementation resulted in a significant increase in the weight-bearing distribution to the treated knee (P < .001). Moreover, the V. incana treatment led to a decrease in the levels of liver function enzymes and tissue malondialdehyde (P < .05 and .01). The V. incana significantly suppressed the inflammatory factors through the nuclear factor-kappa B signaling pathway and downregulated the expression of matrix metalloproteinases, which are involved in the degradation of the extracellular matrix (P < .01 and .001). In addition, we confirmed the alleviation of cartilage degeneration through tissue stains. In conclusion, this study confirmed the major four compounds of V. incana and suggested that V. incana could serve as an anti-inflammatory candidate agent for patients with OA.
Collapse
Affiliation(s)
- Jin A Lee
- Department of Herbology, College of Korean Medicine, Daegu Haany University, Daegu, Korea
- Research Center for Herbal Convergence on Liver Disease, Daegu Haany University, Gyeongsan-si, Korea
| | - Trung Huy Ngo
- College of Pharmacy, Yeungnam University, Gyeongsan-si, Korea
| | - Mi-Rae Shin
- Department of Herbology, College of Korean Medicine, Daegu Haany University, Daegu, Korea
| | - Jeong Won Choi
- Department of Forest Science, Andong National University, Andong, Korea
| | - Hyukjae Choi
- College of Pharmacy, Yeungnam University, Gyeongsan-si, Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan-si, Korea
| | - Joo-Won Nam
- College of Pharmacy, Yeungnam University, Gyeongsan-si, Korea
| | - Seong-Soo Roh
- Department of Herbology, College of Korean Medicine, Daegu Haany University, Daegu, Korea
| |
Collapse
|
27
|
Wang X, Wang D, Deng B, Yan L. Syringaresinol attenuates osteoarthritis via regulating the NF-κB pathway. Int Immunopharmacol 2023; 118:109982. [PMID: 36989902 DOI: 10.1016/j.intimp.2023.109982] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/30/2023]
Abstract
Osteoarthritis (OA) is a now regarded as a worldwide whole joint disease with synovial inflammation, cartilage degeneration, and subchondral sclerosis. Non-steroidal anti-inflammatory drugs (NSAIDs) are commonly used drugs for OA treatment which only relieve the symptoms and restrain the progression of OA. However, various severe adverse effects often occur in patients with long-term NSAIDs use, which heavily burdens the healthcare system and impacts the quality of life. Therefore, it is much imperative to identify alternative drugs with increased efficacy. Syringaresinol (Syr), a naturally occurring phytochemical which belonging to the lignan group of polyphenols, shows anti-tumor and anti-oxidant activities, which to benefit human health. Studies has shown Syr can regulate the inflammatory response by modulating the secretion and expression level of cytokines IL-6, IL-8, and tumor necrosis factor (TNF)-α. it also shows the inhibitory effect on NF-κB pathway in mouse cells. In the present study, we aimed to demonstrate the anti-inflammatory effects of Syr in OA. In vitro Syr treatment in IL-1β-activated mouse chondrocytes significantly restrained the expression of NO, PGE2, IL-6, TNF-α, INOS, COX-2 and MMP-13. Moreover, it considerably ameliorated the degradation of aggrecan and collagen II. Furthermore, the phosphorylation of the NF-kB signaling pathway was significantly suppressed by Syr. Moreover, in vivo, the cartilage degeneration was attenuated and the increased Osteoarthritis Research Society International (OARSI) scores were reversed in the DMM + Syr group, comprared to those in the DMM group. In sum, our study demonstrated that Syr can attenuate the inflammation in vitro and further verified its effect on OA in vivo. Thus, Syr might be a potent therapeautic alternative for OA treatment.
Collapse
Affiliation(s)
- Xianfeng Wang
- Department of Orthopedic Surgery, Guizhou Provincial Orthopedics Hospital, China
| | - Dangrang Wang
- Department of Gastroenterology, The Second Hospital of Dalian Medical University, China
| | - Biyong Deng
- Department of Orthopedic Surgery, Guizhou Provincial Orthopedics Hospital, China.
| | - Litao Yan
- Department of Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, China.
| |
Collapse
|
28
|
Zhang R, Deng X, Liu Q, Zhang X, Bai X, Weng S, Chen M. Global research trends and hotspots of PI3K/Akt signaling pathway in the field of osteoarthritis: A bibliometric study. Medicine (Baltimore) 2023; 102:e33489. [PMID: 37058031 PMCID: PMC10101318 DOI: 10.1097/md.0000000000033489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/20/2023] [Indexed: 04/15/2023] Open
Abstract
The phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathway has gradually become a new target for the treatment of osteoarthritis (OA). Numerous studies of PI3K/Akt signaling in OA have been published in the past few years. By analyzing these research characteristics and qualities, we aimed to reveal the current research focus and emerging trends in PI3K/Akt signaling in OA. We searched the Web of Science database for relevant articles concerning the PI3K/Akt signaling pathway in OA published from inception to October 31, 2022. The following data were extracted: author name, article title, keywords, topic, publication country/region, institution, publication journal, journal impact factor, number of times cited, and H-index. VOSviewer and Excel 2019 were used to conduct the bibliometric study and visualize the analysis. A total of 374 publications were included in this study. In all selected articles, "orthopedics" was the dominant topic (252 of 374, 67.38%). The most productive year was 2021. Frontiers in Pharmacology published the most articles. The People's Republic of China has published the most articles worldwide. The top 5 keywords were "OA," "expression," "apoptosis," "chondrocytes," and "inflammation." The keywords "autophagy," "mitochondrial dysfunction," "inflammatory response," "cartilage degeneration," and "network pharmacology" have increased in recent years. Our study showed a growing trend in published articles related to the PI3K/Akt signaling pathway in OA. Inflammatory response, cartilage degeneration, and apoptosis remain central topics in the field. Research on autophagy, mitochondrial dysfunction, and network pharmacology is on the rise, and the focus on PI3K/Akt will continue to increase.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Orthopedic, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoqin Deng
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Quan Liu
- Department of Orthopedic, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xintian Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xinxin Bai
- Department of Orthopedic, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shaohuang Weng
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Min Chen
- Department of Orthopedic, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
29
|
Lu R, Wang YG, Qu Y, Wang SX, Peng C, You H, Zhu W, Chen A. Dihydrocaffeic acid improves IL-1β-induced inflammation and cartilage degradation via inhibiting NF-κB and MAPK signalling pathways. Bone Joint Res 2023; 12:259-273. [PMID: 37492935 PMCID: PMC10076109 DOI: 10.1302/2046-3758.124.bjr-2022-0384.r1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
Aims Osteoarthritis (OA) is a prevalent joint disorder with inflammatory response and cartilage deterioration as its main features. Dihydrocaffeic acid (DHCA), a bioactive component extracted from natural plant (gynura bicolor), has demonstrated anti-inflammatory properties in various diseases. We aimed to explore the chondroprotective effect of DHCA on OA and its potential mechanism. Methods In vitro, interleukin-1 beta (IL-1β) was used to establish the mice OA chondrocytes. Cell counting kit-8 evaluated chondrocyte viability. Western blotting analyzed the expression levels of collagen II, aggrecan, SOX9, inducible nitric oxide synthase (iNOS), IL-6, matrix metalloproteinases (MMPs: MMP1, MMP3, and MMP13), and signalling molecules associated with nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. Immunofluorescence analysis assessed the expression of aggrecan, collagen II, MMP13, and p-P65. In vivo, a destabilized medial meniscus (DMM) surgery was used to induce mice OA knee joints. After injection of DHCA or a vehicle into the injured joints, histological staining gauged the severity of cartilage damage. Results DHCA prevented iNOS and IL-6 from being upregulated by IL-1β. Moreover, the IL-1β-induced upregulation of MMPs could be inhibited by DHCA. Additionally, the administration of DHCA counteracted IL-1β-induced downregulation of aggrecan, collagen II, and SOX9. DHCA protected articular cartilage by blocking the NF-κB and MAPK pathways. Furthermore, DHCA mitigated the destruction of articular cartilage in vivo. Conclusion We present evidence that DHCA alleviates inflammation and cartilage degradation in OA chondrocytes via suppressing the NF-κB and MAPK pathways, indicating that DHCA may be a potential agent for OA treatment.
Collapse
Affiliation(s)
- Rui Lu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying-Guang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunkun Qu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan-Xi Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Peng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongbo You
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wentao Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anmin Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Mi XJ, Le HM, Lee S, Park HR, Kim YJ. Silymarin-Functionalized Selenium Nanoparticles Prevent LPS-Induced Inflammatory Response in RAW264.7 Cells through Downregulation of the PI3K/Akt/NF-κB Pathway. ACS OMEGA 2022; 7:42723-42732. [PMID: 36467957 PMCID: PMC9713780 DOI: 10.1021/acsomega.2c04140] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/17/2022] [Indexed: 06/17/2023]
Abstract
Silymarin exhibits an anti-inflammatory property in various cancers and inflammatory diseases. In our previous work, silymarin-mediated selenium nanoparticles (SeNPs) (Si-SeNPs) were developed using a green synthesis technique, and its potential as an anticancer agent was confirmed. In order to further examine the extended comprehensive potential of Si-SeNPs, this investigation focuses on studying the enhanced anti-inflammatory effect of Si-SeNPs in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Enzyme-linked immunosorbent assay and quantitative reverse transcription-polymerase chain reaction were used to evaluate the expression of pro-inflammatory mediators and cytokines. Western blotting and immunofluorescence assays were conducted to assess the protein expression of p-PI3K, p-Akt, p-NF-κB, and p-IκBα. Compared to silymarin, Si-SeNPs exhibited a significantly increased inhibitory effect on LPS-induced release of nitric oxide and the expression of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin 1β (IL-1β) in RAW264.7 cells. A western blot assay indicated that Si-SeNPs downregulated the PI3K/Akt and NF-κB signaling pathways. The immunofluorescence assay suggested that Si-SeNPs inhibited the nuclear translocation and the activation of NF-κB. In addition, 740 Y-P (PI3K agonist) was used to demonstrate that activating the PI3K/Akt signal could partially reverse the inflammatory response, suggesting a causal role of the PI3K/Akt signaling pathway in the anti-inflammatory effect of Si-SeNPs. Consequently, these findings indicate that Si-SeNPs could be a functional agent of the attenuation of LPS-induced inflammatory responses in RAW264.7 macrophages through inhibiting the PI3K/Akt/NF-κB signaling pathway. In addition, biosynthesized Si-SeNPs could be more effective at reducing inflammation than only silymarin extracts. Thus, this study lays an experimental foundation for the clinical application of using biosynthesized SeNPs as a novel candidate in the field of inflammation-associated diseases.
Collapse
Affiliation(s)
- Xiao-jie Mi
- Graduate
School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea
| | - Ha-Minh Le
- Graduate
School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea
| | - Sanghyun Lee
- Department
of Plant Science and Technology, Chung Ang
University, Anseong 17546, Republic of Korea
| | - Hye-Ryung Park
- Graduate
School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea
| | - Yeon-Ju Kim
- Graduate
School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea
| |
Collapse
|
31
|
Shi X, Jie L, Wu P, Zhang N, Mao J, Wang P, Yin S. Calycosin mitigates chondrocyte inflammation and apoptosis by inhibiting the PI3K/AKT and NF-κB pathways. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115536. [PMID: 35843413 DOI: 10.1016/j.jep.2022.115536] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 07/03/2022] [Accepted: 07/10/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shaoyao Gancao Decoction (SG-Tang), originated from the Treatise on Febrile Diseases, is often used to treat OA pain symptoms. Whereas its efficacy has been verified by several clinical studies, the underlying mechanism remained unclear. Network pharmacology and UPLC-QTOF-MS analysis found that calycosin could be regarded as the active components of SG-Tang in treating OA. However, the effect of calycosin on cartilage destruction and the pathogenesis of OA are not known. Therefore, we evaluated the benefits of calycosin for OA and revealed the underlying mechanisms. AIM OF STUDY Using network pharmacology, UPLC-QTOF-MS analysis and experiments, the active components of SG-Tang were analyzed to explore their potential therapeutic mechanism in OA. MATERIALS AND METHODS The components of SG-Tang were detected by UPLC-QTOF-MS, and the possible active components and mechanism of SG-Tang in the treatment of OA were screened by network pharmacology. The OA mouse model was constructed by DMM. In total, 30 mice were randomly divided into three groups: Sham, DMM, and DMM + Calycosin. H&E, safranin O/fast green staining and the OARSI scores were used to evaluate joint injury in mice. In addition, OA models were established using chondrocytes treated with 10 ng/mL IL-1β. Treatment groups were treated with 100, 200 or 400 μM calycosin. CCK-8 assay was used for assessing the cytotoxic effects of calycosin. TUNEL staining and Western blotting were used to detect chondrocyte apoptosis. In addition, PI3K/Akt and NF-κB signaling pathway-related markers and cartilage matrix-related indicators were also detected. RESULTS In vivo studies showed that calycosin inhibited IL-1β-induced IL-6 and TNF-α production, as well as iNOS and COX-2 expression. Meanwhile, calycosin could inhibit IL-1β-induced degradation of cartilage matrix, including downregulation of MMP3, MMP-13, collagen II and aggrecan. NF-κB and PI3K/AKT were also inhibited by calycosin in OA chondrocytes. Furthermore, calycosin inhibited IL-1β-induced apoptosis in mouse chondrocytes. In a mouse model of OA, our results suggest that calycosin has a chondroprotective effect. CONCLUSIONS According to this study, calycosin may act as a protective agent against OA by inhibiting the PI3K/AKT and NF-κB pathways. Furthermore, this study suggested that calycosin is a potential candidate for the treatment of OA.
Collapse
Affiliation(s)
- Xiaoqing Shi
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lishi Jie
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peng Wu
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Nongshan Zhang
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jun Mao
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peimin Wang
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Songjiang Yin
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
32
|
Zheng M, Okawa S, Bravo M, Chen F, Martínez-Chantar ML, del Sol A. ChemPert: mapping between chemical perturbation and transcriptional response for non-cancer cells. Nucleic Acids Res 2022; 51:D877-D889. [PMID: 36200827 PMCID: PMC9825489 DOI: 10.1093/nar/gkac862] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/08/2022] [Accepted: 09/25/2022] [Indexed: 01/30/2023] Open
Abstract
Prior knowledge of perturbation data can significantly assist in inferring the relationship between chemical perturbations and their specific transcriptional response. However, current databases mostly contain cancer cell lines, which are unsuitable for the aforementioned inference in non-cancer cells, such as cells related to non-cancer disease, immunology and aging. Here, we present ChemPert (https://chempert.uni.lu/), a database consisting of 82 270 transcriptional signatures in response to 2566 unique perturbagens (drugs, small molecules and protein ligands) across 167 non-cancer cell types, as well as the protein targets of 57 818 perturbagens. In addition, we develop a computational tool that leverages the non-cancer cell datasets, which enables more accurate predictions of perturbation responses and drugs in non-cancer cells compared to those based onto cancer databases. In particular, ChemPert correctly predicted drug effects for treating hepatitis and novel drugs for osteoarthritis. The ChemPert web interface is user-friendly and allows easy access of the entire datasets and the computational tool, providing valuable resources for both experimental researchers who wish to find datasets relevant to their research and computational researchers who need comprehensive non-cancer perturbation transcriptomics datasets for developing novel algorithms. Overall, ChemPert will facilitate future in silico compound screening for non-cancer cells.
Collapse
Affiliation(s)
| | | | - Miren Bravo
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Bizkaia, Spain
| | - Fei Chen
- German Research Center for Artificial Intelligence (DFKI), 66123 Saarbrücken, Germany
| | - María-Luz Martínez-Chantar
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Bizkaia, Spain
| | - Antonio del Sol
- To whom correspondence should be addressed. Tel: +352 46 66 44 6982;
| |
Collapse
|
33
|
Zhang T, Dong Z, Liu F, Pan E, He N, Ma F, Wang G, Wang Y, Dong J. Avermectin induces carp neurotoxicity by mediating blood-brain barrier dysfunction, oxidative stress, inflammation, and apoptosis through PI3K/Akt and NF-κB pathways. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 243:113961. [PMID: 35969982 DOI: 10.1016/j.ecoenv.2022.113961] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 06/15/2023]
Abstract
Avermectin, a "low toxicity insecticide", has been widely used in recent years, but its non-target toxicity, especially to aquatic organisms, has been neglected. In this study, we evaluated the neurotoxic effects of avermectin on carp by establishing a 96 h avermectin acute toxicity test, and its possible mechanism was discussed. The 96 h LC50 of avermectin in carp was found to be 24.04 μg/L. Therefore, 3.005 μg/L and 12.02 μg/L were used as the low-dose and high-dose groups, respectively, to investigate the neurotoxic effects of avermectin on carp. The results of high-performance liquid chromatography (HPLC) analysis showed that avermectin accumulated in the carp brain. Histopathological observation and immunohistochemical analysis (IHC) of TNF-α and Bax showed that avermectin exposure led to inflammatory cell infiltration and neuronal necrosis. The mRNA levels of tight junction genes and the IHC results of ZO-1 and Occludin showed that the structure of the blood-brain barrier (BBB) was destroyed. Biochemical analysis showed that avermectin induced the accumulation of MDA in the brain and decreased the activity of antioxidant enzymes CAT and SOD, leading to oxidative stress. In addition, avermectin induces brain inflammation by activating NF-κB pathway and releasing inflammatory factors IL-1β, IL-6, TNF-α and iNOS. TEM and TUNEL assays showed that exposure to avermectin induced apoptosis in brain. what is more, the expression of apoptosis-related genes and proteins suggested that avermectin-induced apoptosis may be associated with inhibition of the PI3K/Akt signaling pathway. This study also showed that avermectin-induced NF-κB signaling activation was partially dependent on its upstream PI3K/Akt signaling pathway. Therefore, this study concludes that avermectin can induce neurotoxicity in carp by disrupting the blood-brain barrier structure and generating oxidative stress, inflammation, and apoptosis and that NF-κB and PI3K/Akt signaling pathways are involved in this process.
Collapse
Affiliation(s)
- Tianmeng Zhang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China; Department of Medicine Laboratory, Department of Cardiac Function Examination, The Second People's Hospital of Lianyungang City, Lianyungang Hospital Affiliated to Jiangsu University, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222000, China
| | - Zhuhua Dong
- Deapartment of Economics and Related Studies, University of York, York, YO10 5DD, United Kingdom
| | - Feixue Liu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China
| | - Enzhuang Pan
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China
| | - Nana He
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China
| | - Fenfen Ma
- Department of Medicine Laboratory, Department of Cardiac Function Examination, The Second People's Hospital of Lianyungang City, Lianyungang Hospital Affiliated to Jiangsu University, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222000, China
| | - Guanglu Wang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yan Wang
- Department of Medicine Laboratory, Department of Cardiac Function Examination, The Second People's Hospital of Lianyungang City, Lianyungang Hospital Affiliated to Jiangsu University, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222000, China.
| | - Jingquan Dong
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China.
| |
Collapse
|
34
|
Dihydroartemisinin Attenuated Intervertebral Disc Degeneration via Inhibiting PI3K/AKT and NF-κB Signaling Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8672969. [PMID: 36120596 PMCID: PMC9481359 DOI: 10.1155/2022/8672969] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/13/2022] [Indexed: 11/24/2022]
Abstract
Intervertebral disc degeneration (IDD) is the leading cause of low back pain (LBP). However, effective therapeutic drugs for IDD remain to be further explored. Inflammatory cytokines play a pivotal role in the onset and progression of IDD. Dihydroartemisinin (DHA) has been well reported to have powerful anti-inflammatory effects, but whether DHA could ameliorate the development of IDD remained unclear. In this study, the effects of DHA on extracellular matrix (ECM) metabolism and cellular senescence were firstly investigated in nucleus pulposus cells (NPCs) under tumor necrosis factor alpha (TNFα)-induced inflammation. Meanwhile, AKT agonist sc-79 was used to determine whether DHA exerted its actions through regulating PI3K/AKT and NF-κB signaling pathways. Next, the therapeutic effects of DHA were tested in a puncture-induced rat IDD model. Finally, we detected the activation of PI3K/AKT and NF-κB signaling pathways in clinical degenerative nucleus pulposus specimens. We demonstrated that DHA ameliorated the imbalance between anabolism and catabolism of extracellular matrix and alleviated NPCs senescence induced by TNFα in vitro. Further, we illustrated that DHA mitigated the IDD progression in a puncture-induced rat model. Mechanistically, DHA inhibited the activation of PI3K/AKT and NF-κB signaling pathways induced by TNFα, which was undermined by AKT agonist sc-79. Molecular docking predicted that DHA bound to the PI3K directly. Intriguingly, we also verified the activation of PI3K/AKT and NF-κB signaling pathways in clinical degenerative nucleus pulposus specimens, suggesting that DHA may qualify itself as a promising drug for mitigating IDD.
Collapse
|
35
|
Xiao J, Zhang G, Mai J, He Q, Chen W, Li J, Ma Y, Pan Z, Yang J, Li S, Li M, Chen B, Wang H. Bioinformatics analysis combined with experimental validation to explore the mechanism of XianLing GuBao capsule against osteoarthritis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 294:115292. [PMID: 35447200 DOI: 10.1016/j.jep.2022.115292] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/17/2022] [Accepted: 04/10/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE XianLing GuBao Capsule (XLGB) is often used to treat osteoarthritis (OA), osteoporosis, fractures, and other musculoskeleton disorders. However, the molecular mechanism of XLGB for treating OA is still unclear. AIM OF THE STUDY This study set out to uncover the molecular mechanism underlying the treatment of osteoarthritis with XLGB. MATERIALS AND METHODS Disease genes were obtained from CTD, DisGeNET, and GeneCards databases, and XLGB drug targets were obtained from ETCM and target genes predicted by XLGB metabolic components reported in the literature. Then we used the Venn diagram viewer to extract disease and drug intersection genes as potential therapeutic genes for Protein-protein interaction (PPI), GO terminology, and KEGG pathway analysis. Subsequently, we performed qRT-PCR, Western blot and histological analysis to validate the therapeutic effect of XLGB against OA and its molecular mechanism. RESULTS A total of 1039 OA genes and 949 XLGB target genes were collected, and finally 188 potential therapeutic target genes were obtained. PPI network analysis indicated that the main target genes for XLGB to treat OA include Akt1, Mapk3, Il-6, Il-1β, Ptgs2, Mmp9, etc. The results of KEGG and GO enrichment analysis suggested that XLGB may treat OA by anti-inflammatory and reducing extracellular matrix degradation. In vitro, XLGB down-regulated the expressions of Mmp3, Mmp9, Mmp12, Mmp13, Cox-2, Il-6, increased the expression of Collagen II and Sox9. Mechanistically, XLGB inhibits the activation of PI3K/AKT/NF-κB and MAPK pathways. Moreover, the results of animal experiments indicated that XLGB reduced cartilage destruction, bone resorption, and synovitis in osteoarthritic rats. CONCLUSIONS XLGB has a protective effect against OA by suppressing PI3K/AKT/NF-κB and MAPK signaling. Our study provides a theoretical basis for XLGB in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Jiacong Xiao
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Gangyu Zhang
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Jiale Mai
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Qi He
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Weijian Chen
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Jianliang Li
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Yanhuai Ma
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Zhaofeng Pan
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Junzheng Yang
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Shaocong Li
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Miao Li
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Bohao Chen
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Haibin Wang
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, PR China.
| |
Collapse
|
36
|
Qu Y, Shen Y, Teng L, Huang Y, Yang Y, Jian X, Fan S, Wu P, Fu Q. Chicoric acid attenuates tumor necrosis factor-α-induced inflammation and apoptosis via the Nrf2/HO-1, PI3K/AKT and NF-κB signaling pathways in C28/I2 cells and ameliorates the progression of osteoarthritis in a rat model. Int Immunopharmacol 2022; 111:109129. [PMID: 35961266 DOI: 10.1016/j.intimp.2022.109129] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/29/2022]
Abstract
Osteoarthritis (OA) is the most common arthritis, and is characterized by inflammation and cartilage degradation. Chicoric acid (CA), a bioactive caffeic acid derivative isolated from the root of Taraxacum mongolicumHand. - Mazz., has been reported to have anti-inflammatory effects. However, the therapeutic effects of CA on chondrocyte inflammation remain unknown. Our study aimed to explore the effect of CA on OA both in vivo and in vitro. In vitro, CA treatment significantly suppressed the overproduction of nitric oxide (NO), prostaglandin E2 (PGE2), inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), interleukin-1 beta (IL-1β), interleukin-6 (IL-6), and IL-12 in tumor necrosis factor alpha (TNF-α)-induced human C28/I2 chondrocytes. Moreover, CA attenuated TNF-α induced degradation of the extracellular matrix (ECM) by upregulating the expression of collagen Ⅱ and aggrecan, and downregulating ADAMTS-5 and matrix metalloproteinases (MMPs). Additionally, CA treatment inhibited apoptosis in C28/I2 cells by upregulating of Bcl-2 levels, downregulating Bax and ROS levels, and activating the Nrf2/HO-1 pathway. Mechanistically, CA exerted an anti-inflammatory effect by inhibiting the PI3K/AKT and NF-κB signaling pathways, enhancing Nrf-2/HO-1 to limit the activation of NF-κB. In vivo experiments also proved the therapeutic effects of CA on OA in rats. These findings indicate that CA may become a new drug for the treatment of OA.
Collapse
Affiliation(s)
- Yuhan Qu
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Yue Shen
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Li Teng
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Yuehui Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Yuting Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Xi Jian
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Shengli Fan
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Ping Wu
- Department of Pharmacy, Chengdu Integrated TCM and Western Medicine Hospital, Chengdu 610041, China.
| | - Qiang Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China.
| |
Collapse
|
37
|
Lupeol protects against cardiac hypertrophy via TLR4-PI3K-Akt-NF-κB pathways. Acta Pharmacol Sin 2022; 43:1989-2002. [PMID: 34916609 PMCID: PMC9343642 DOI: 10.1038/s41401-021-00820-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/09/2021] [Indexed: 12/11/2022]
Abstract
Inflammation and apoptosis are main pathological processes that lead to the development of cardiac hypertrophy. Lupeol, a natural triterpenoid, has shown anti-inflammatory and anti-apoptotic activities as well as potential protective effects on cardiovascular diseases. In this study we investigated whether lupeol attenuated cardiac hypertrophy and fibrosis induced by pressure overload in vivo and in vitro, and explored the underlying mechanisms. Cardiac hypertrophy was induced in mice by transverse aortic constriction (TAC) surgery, and in neonatal rat cardiomyocytes (NRCMs) by stimulation with phenylephrine (PE) in vitro. We showed that administration of lupeol (50 mg ·kg-1· d-1, i.g., for 4 weeks) prevented the morphological changes and cardiac dysfunction and remodeling in TAC mice, and treatment with lupeol (50 μg/mL) significantly attenuated the hypertrophy of PE-stimulated NRCMs, and blunted the upregulated hypertrophic markers ANP, BNP, and β-MHC. Furthermore, lupeol treatment attenuated the apoptotic and inflammatory responses in the heart tissue. We revealed that lupeol attenuated the inflammatory responses including the reduction of inflammatory cytokines and inhibition of NF-κB p65 nuclear translocation, which was mediated by the TLR4-PI3K-Akt signaling. Administration of a PI3K/Akt agonist 740 Y-P reversed the protective effects of lupeol in TAC mice as well as in PE-stimulated NRCMs. Moreover, pre-treatment with a TLR4 agonist RS 09 abolished the protective effects of lupeol and restored the inhibition of PI3K-Akt-NF-κB signaling by lupeol in PE-stimulated NRCMs. Collectively, our results demonstrate that the lupeol protects against cardiac hypertrophy via anti-inflammatory mechanisms, which results from inhibiting the TLR4-PI3K-Akt-NF-κB signaling.
Collapse
|
38
|
Lü J, Jiang C, Schell TD, Joshi M, Raman JD, Xing C. Angelica gigas: Signature Compounds, In Vivo Anticancer, Analgesic, Neuroprotective and Other Activities, and the Clinical Translation Challenges. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:1475-1527. [PMID: 35876033 DOI: 10.1142/s0192415x2250063x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Angelica gigas Nakai (AGN) root is a medicinal herbal widely used in traditional medicine in Korea. AGN root ethanolic extract dietary supplements are marketed in the United States for memory health and pain management. We comprehensively reviewed the anticancer, analgesic, pro-memory and other bio-activities of AGN extract and its signature phytochemicals decursin, decursinol angelate, and decursinol a decade ago in 2012 and updated their anticancer activities in 2015. In the last decade, significant progress has been made for understanding the pharmacokinetics (PK) and metabolism of these compounds in animal models and single dose human PK studies have been published by us and others. In addition to increased knowledge of the known bioactivities, new bioactivities with potential novel health benefits have been reported in animal models of cerebral ischemia/stroke, anxiety, sleep disorder, epilepsy, inflammatory bowel disease, sepsis, metabolic disorders, osteoporosis, osteoarthritis, and even male infertility. Herein, we will update PK and metabolism of pyranocoumarins, review in vivo bioactivities from animal models and human studies, and critically appraise the relevant active compounds, the cellular and molecular pharmacodynamic targets, and pertinent mechanisms of action. Knowledge gaps include whether human pyranocoumarin PK metrics are AGN dose dependent and subjected to metabolic ceiling, or metabolic adaptation after repeated use. Critical clinical translation challenges include sourcing of AGN extracts, product consistency and quality control, and AGN dose optimization for different health conditions and disease indications. Future research directions are articulated to fill knowledge gaps and address these challenges.
Collapse
Affiliation(s)
- Junxuan Lü
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Cheng Jiang
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Todd D Schell
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Monika Joshi
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Medicine Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Jay D Raman
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Urology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Chengguo Xing
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
39
|
Xian S, Lin Z, Zhou C, Wu X. The Protective Effect of Evodiamine in Osteoarthritis: An In Vitro and In Vivo Study in Mice Model. Front Pharmacol 2022; 13:899108. [PMID: 35795554 PMCID: PMC9251407 DOI: 10.3389/fphar.2022.899108] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/16/2022] [Indexed: 12/22/2022] Open
Abstract
Osteoarthritis (OA) is a chronic disease with high economic burden characterized by cartilage degradation and joint inflammation. Evodiamine (EV), which can be extracted from Evodia rutaecarpa (Rutaceae), is a traditional Chinese medicine to treat inflammation, cardiovascular disorders, infection, and obesity. Studies have shown that EV can suppress the activation of immune cells and restrain the secretion of pro-inflammatory cytokines. However, it is still not well known about its role in the treatment of OA. In this study, we utilized interleukin-1β (IL-1β)–stimulated mouse chondrocytes in vitro and the destabilization of the medial meniscus (DMM) model in vivo to demonstrate the anti-inflammatory properties of EV in OA. The results suggested that EV decreased the generation of NO, IL-6, TNF-α, and PGE2. Meanwhile, the increased expression of iNOS, COX-2, and MMP-13 and the degradation of aggrecan and Col-II were significantly alleviated by EV in IL-1β–activated mouse chondrocytes. Moreover, EV can inhibit the considerable IL-1β–stimulated phosphorylation of the NF-κB signaling pathway and nuclear translocation of p65, compared with the control group. Furthermore, EV alleviated cartilage degeneration and reversed the increased Osteoarthritis Research Society International (OARSI) scores in the OA model in vivo. Our study demonstrates that EV can suppress inflammation in vitro and cartilage degeneration in vivo in OA, which implies that EV may be a potential candidate for the treatment of OA.
Collapse
Affiliation(s)
- Shuyuan Xian
- Department of Orthopedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zeng Lin
- Department of Orthopedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chao Zhou
- Department of Orthopaedics, Yinshanhu Hospital of Wuzhong District, Suzhou, China
- *Correspondence: Chao Zhou, ; Xing Wu,
| | - Xing Wu
- Department of Orthopedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Chao Zhou, ; Xing Wu,
| |
Collapse
|
40
|
Govoni M, Danesi F. Do Pomegranate Hydrolyzable Tannins and Their Derived Metabolites Provide Relief in Osteoarthritis? Findings from a Scoping Review. Molecules 2022; 27:1033. [PMID: 35164312 PMCID: PMC8840395 DOI: 10.3390/molecules27031033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 11/16/2022] Open
Abstract
Osteoarthritis (OA) is the most common form of arthritis affecting both the elderly and the middle-aged population. Although various therapeutics have been developed to arrest the structural deterioration of cartilage, the current treatments are limited to delay the progress of OA clinically. Therefore, it is pivotal to study new therapeutic agents for chondroprotection and the prevention of cartilage degeneration. Hydrolyzable tannin (HT)-containing foods aroused considerable interest in recent years for their relevant anti-inflammatory effects. The focus of this scoping review is to provide an overview of the evidence of the therapeutic potential of HTs and their metabolites in preventing or alleviating the course of OA. A broad search of PubMed and Scopus databases on this topic resulted in 156 articles. After the exclusion of reviews and not relevant records, 31 articles were retrieved. Although only some papers did not consider the biotransformation of HTs, most recent studies also have investigated the effect of HT metabolites. Further larger clinical trials, with an in-deep analysis of HT metabolization, are still needed to unravel the potential benefits of these compounds in OA, paving the way towards the development of a dietary strategy for the improvement of pro-inflammatory cytokine-induced chondrocyte dysfunctions and injuries.
Collapse
Affiliation(s)
- Marco Govoni
- Reconstructive Orthopedic Surgery and Innovative Techniques—Musculoskeletal Tissue Bank, IRCCS Istituto Ortopedico Rizzoli, Via G.C. Pupilli 1, 40136 Bologna, Italy;
| | - Francesca Danesi
- Human Nutrition Unit, Department of Agricultural and Food Sciences (DISTAL), University of Bologna, Piazza Goidanich 60, 47521 Cesena, Italy
| |
Collapse
|
41
|
Chen X, Zhou B, Gao Y, Wang K, Wu J, Shuai M, Men K, Duan X. Efficient Treatment of Rheumatoid Arthritis by Degradable LPCE Nano-Conjugate-Delivered p65 siRNA. Pharmaceutics 2022; 14:pharmaceutics14010162. [PMID: 35057057 PMCID: PMC8780552 DOI: 10.3390/pharmaceutics14010162] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/22/2021] [Accepted: 01/06/2022] [Indexed: 02/05/2023] Open
Abstract
Rheumatoid arthritis (RA) is one of the most common autoimmune diseases worldwide, causing severe cartilage damage and disability. Despite the recent progress made in RA treatment, limitations remain in achieving early and efficient therapeutic intervention. Advanced therapeutic strategies are in high demand, and siRNA-based therapeutic technology with a gene-silencing ability represents a new approach for RA treatment. In this study, we created a cationic delivery micelle consisting of low-molecular-weight (LMW) polyethylenimine (PEI)–cholesterol–polyethylene glycol (PEG) (LPCE) for small interfering RNA (siRNA)-based RA gene therapy. The carrier is based on LMW PEI and modified with cholesterol and PEG. With these two modifications, the LPCE micelle becomes multifunctional, and it efficiently delivered siRNA to macrophages with a high efficiency greater than 70%. The synthesized LPCE exhibits strong siRNA protection ability and high safety. By delivering nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 siRNA, the p65 siRNA/LPCE complex efficiently inhibited macrophage-based cytokine release in vitro. Local administration of the p65 siRNA/LPCE complex exhibited a fast and potent anti-inflammatory effect against RA in a mouse model. According to the results of this study, the functionalized LPCE micelle that we prepared has potential gene therapeutic implications for RA.
Collapse
Affiliation(s)
- Xiaohua Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (X.C.); (Y.G.); (K.W.); (J.W.); (K.M.)
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China;
| | - Bailing Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (X.C.); (Y.G.); (K.W.); (J.W.); (K.M.)
- Correspondence: (B.Z.); (X.D.)
| | - Yan Gao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (X.C.); (Y.G.); (K.W.); (J.W.); (K.M.)
| | - Kaiyu Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (X.C.); (Y.G.); (K.W.); (J.W.); (K.M.)
| | - Jieping Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (X.C.); (Y.G.); (K.W.); (J.W.); (K.M.)
| | - Ming Shuai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China;
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (X.C.); (Y.G.); (K.W.); (J.W.); (K.M.)
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China;
- Correspondence: (B.Z.); (X.D.)
| |
Collapse
|
42
|
Cheng Y, Li F, Zhang WS, Zou GY, Shen YX. Silencing BLNK protects against interleukin-1β-induced chondrocyte injury through the NF-κB signaling pathway. Cytokine 2021; 148:155686. [PMID: 34521030 DOI: 10.1016/j.cyto.2021.155686] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/30/2021] [Accepted: 08/17/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Osteoarthritis (OA) is the most common joint disease in the elderly and is characterized by the progressive degeneration of articular cartilage. It is necessary to study the molecular pathology of OA. This study aimed to explore the role and mechanism of BLNK in regulating interleukin-1β (IL-1β)-induced chondrocyte injury and OA progression. METHODS GSE1919 (5 normal samples and 5 OA samples) was downloaded from the Gene Expression Omnibus (GEO) database. The limma package in R software was used to identify differentially expressed genes (DEGs) between control and OA-affected cartilage. Gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of the differentially expressed genes were also performed. Apoptosis was assessed by flow cytometry. An OA rat model was established, and the relative expression of BLNK was assessed by real time quantitative PCR (qRT-PCR) and immunohistochemical staining. The expression of collagen II, MMP9, p65 and p-p65 was measured by Western blot analysis. Moreover, inflammatory factors (TNF-α and IL-18) were assessed by ELISA. The NF-κB inhibitor JSH-23 was used to assess the impact of BLNK on the NF-κB signaling pathway. RESULTS In total, 1318 DEGs were identified between normal and OA-affected cartilage according to the criteria (P-value <0.05 and |logFC > 1|). These DEGs were mainly enriched in the NF-κB pathway. BLNK was highly expressed in OA cartilage tissue and injured chondrocytes. Silencing BLNK significantly downregulated the IL-1β-induced apoptosis of chondrocytes. Silencing BLNK partially increased collagen II expression and downregulated MMP13 expression. Moreover, silencing BLNK partially decreased TNF-α and IL-18 expression. BLNK silencing inhibited the activation of NF-κB in OA. Silencing BLNK delayed OA progression through the NF-κB signaling pathway. CONCLUSION Silencing BLNK delayed OA progression and IL-1β-induced chondrocyte injury by regulating the NF-κB pathway.
Collapse
Affiliation(s)
- Yi Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, PR China; Department of Orthopaedics, The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng 224005, PR China
| | - Feng Li
- Department of Orthopaedics, The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng 224005, PR China
| | - Wen-Sheng Zhang
- Department of Orthopaedics, The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng 224005, PR China
| | - Guo-You Zou
- Department of Orthopaedics, The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng 224005, PR China
| | - Yi-Xin Shen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, PR China.
| |
Collapse
|
43
|
Network Pharmacology Study and Experimental Confirmation Revealing the Ameliorative Effects of Decursin on Chemotherapy-Induced Alopecia. Pharmaceuticals (Basel) 2021; 14:ph14111150. [PMID: 34832932 PMCID: PMC8618121 DOI: 10.3390/ph14111150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022] Open
Abstract
Decursin, a pyranocoumarin compound from the root of Angelica gigas Nakai as a main constituent, has been reported to have various biological activities, including anti-inflammatory, anticancer, and antioxidant effects. This study aimed to predict and confirm the pharmacological relevance of Decursin on chemotherapy-induced alopecia (CIA) with the underlying molecular mechanisms. Decursin-targeted genes were compared with the gene set of alopecia and investigated through functional enrichment analysis. CIA was induced in C57BL/6J mice by injection of cyclophosphamide, and 1, 10, and 100 μM of Decursin were topically treated to depilated dorsal skin. KGF+ expression was detected in the dorsal skin tissues. Based on the predicted results, caspase, PIK3/AKT, and MAPKs protein expressions by Decursin were analyzed in the TNF-α-induced keratinocytes. The Decursin network had 60.20% overlapped genes with the network of alopecia. Biological processes, such as cellular response to chemical stimulus, apoptosis, PI3K-AKT signaling pathway, and MAPK signaling pathway, were derived from the Decursin network. In the Decursin-treated skin, there was morphological hair growth and histological restoration of hair follicles in the CIA mice. The KGF+ fluorescence and protein expressions were significantly increased by Decursin treatment. In addition, caspase-3, -7, and -8 expressions, induced by TNF-α, were dose-dependently decreased along with the inhibition of PI3K, AKT, ERK, and p38 expressions in Decursin-treated keratinocytes. These findings indicated that Decursin would be a potent therapeutic option for hair loss, in response to chemotherapy.
Collapse
|
44
|
Xu Y, Zhang M, Yang W, Xia B, Wang W, Pan X. Nootkatone protects cartilage against degeneration in mice by inhibiting NF-κB signaling pathway. Int Immunopharmacol 2021; 100:108119. [PMID: 34492535 DOI: 10.1016/j.intimp.2021.108119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/24/2021] [Accepted: 08/29/2021] [Indexed: 12/31/2022]
Abstract
Osteoarthritis is a common chronic disease associated with chondrocyte inflammation and cartilage matrix hydrolyzation. Studies report that IL-1β plays a critical role in osteoarthritis. Anti-inflammatory effect of nootkatone has been explored in acute and chronic inflammatory disease, thus the current study sought to explore its therapeutic effect in osteoarthritis. Notably, the effect of nootkatone in osteoarthritis has not been elucidated. Therefore, murine primary chondrocytes were extracted and ACLT induced OA mouse model was established in the current study to explore the therapeutic effect of nootkatone in OA both in vitro and in vivo. The findings showed that nootkatone inhibited inflammatory response and protected cartilage balance in murine primary chondrocyte. Further analysis showed that nootkatone suppressed inflammation and protected cartilage against degeneration induced by ACLT surgery in mice. The cellular mechanism of the protective effect of nootkatone in osteoarthritis and associated signaling pathway was identified as the NF-κB signaling pathway. In summary, the findings of the current study indicated that nootkatone is a potential therapeutic agent for OA.
Collapse
Affiliation(s)
- Yue Xu
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pediatric Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Jinan, Shandong 250012, China
| | - Minfa Zhang
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Wanliang Yang
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Bowei Xia
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wenhan Wang
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xin Pan
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
45
|
Pourbagher-Shahri AM, Farkhondeh T, Talebi M, Kopustinskiene DM, Samarghandian S, Bernatoniene J. An Overview of NO Signaling Pathways in Aging. Molecules 2021; 26:4533. [PMID: 34361685 PMCID: PMC8348219 DOI: 10.3390/molecules26154533] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Nitric Oxide (NO) is a potent signaling molecule involved in the regulation of various cellular mechanisms and pathways under normal and pathological conditions. NO production, its effects, and its efficacy, are extremely sensitive to aging-related changes in the cells. Herein, we review the mechanisms of NO signaling in the cardiovascular system, central nervous system (CNS), reproduction system, as well as its effects on skin, kidneys, thyroid, muscles, and on the immune system during aging. The aging-related decline in NO levels and bioavailability is also discussed in this review. The decreased NO production by endothelial nitric oxide synthase (eNOS) was revealed in the aged cardiovascular system. In the CNS, the decline of the neuronal (n)NOS production of NO was related to the impairment of memory, sleep, and cognition. NO played an important role in the aging of oocytes and aged-induced erectile dysfunction. Aging downregulated NO signaling pathways in endothelial cells resulting in skin, kidney, thyroid, and muscle disorders. Putative therapeutic agents (natural/synthetic) affecting NO signaling mechanisms in the aging process are discussed in the present study. In summary, all of the studies reviewed demonstrate that NO plays a crucial role in the cellular aging processes.
Collapse
Affiliation(s)
- Ali Mohammad Pourbagher-Shahri
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand 9717853577, Iran;
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand 9717853577, Iran
| | - Marjan Talebi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran 1991953381, Iran;
| | - Dalia M. Kopustinskiene
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu Pr. 13, LT-50161 Kaunas, Lithuania;
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur 9318614139, Iran
| | - Jurga Bernatoniene
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu Pr. 13, LT-50161 Kaunas, Lithuania;
- Department of Drug Technology and Social Pharmacy, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu Pr. 13, LT-50161 Kaunas, Lithuania
| |
Collapse
|