1
|
Xue Z, Li N, Du K, Shu J, Huang Z, Gao Z, Xie X, Li Q, Lu Y. Inhibiting synovial inflammation and promoting cartilage repair in rheumatoid arthritis using a matrix metalloproteinase-binding hydrogel. Mater Today Bio 2025; 32:101792. [PMID: 40343163 PMCID: PMC12059346 DOI: 10.1016/j.mtbio.2025.101792] [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: 02/25/2025] [Revised: 04/11/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Originating from synovial tissue, matrix metalloproteinase-9 (MMP-9) is a key inflammatory factor that promotes the formation and invasion of synovial pannus, leading to cartilage matrix destruction in rheumatoid arthritis (RA). However, clinical trials of systemic use of MMP-9 inhibitors are not successful due to severe side effects. Thus, locally inhibiting MMP-9 may be an alternative in the treatment of RA. Herein, we developed MMP-9 binding peptide-functionalized copper sulfide nanoparticles (CuS-T NPs) and delivered them with light crosslinking chondroitin sulfate methacrylate (ChSMA) hydrogel. We found that the CuS NP-doped hydrogels could inhibit synovial inflammation. Specifically, the CuS-T/ChSMA hydrogel could rapidly bind to MMP-9, thereby inhibiting not only the invasion of RA fibroblast-like synoviocytes but also the polarization of inflammatory M1-type macrophages. The underlying mechanism involved the inhibition of the MAPK pathway. Moreover, ChSMA hydrogel provided a cartilage matrix-mimic microenvironment and synergistically promoted the generation of collagen-2 and aggrecan with CuS NPs. In an adjuvant-induced arthritis mouse model, the intra-articular injection of ChSMA/CuS-T hydrogel significantly alleviated synovial inflammation and accelerated cartilage repair without causing any side effects, killing two birds with one stone in RA therapy.
Collapse
Affiliation(s)
- Zhanpeng Xue
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Nan Li
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Kaijun Du
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Jianxiong Shu
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Zhenwen Huang
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Zhifei Gao
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Xiaobo Xie
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Qi Li
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Yao Lu
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Southern Medical University, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
2
|
Taherkhani S, Sheibani M, Mohammadkhanizadeh A, Virag JAI, de Castro Braz L, Azizi Y. Metalloproteinases (MMPs) in hypertensive disorders: role, function, pharmacology, and potential strategies to mitigate pathophysiological changes. Front Pharmacol 2025; 16:1559288. [PMID: 40492135 PMCID: PMC12146787 DOI: 10.3389/fphar.2025.1559288] [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: 01/12/2025] [Accepted: 04/29/2025] [Indexed: 06/11/2025] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of enzymes that play an important role in the pathophysiology of hypertensive disorders, particularly through their involvement in extracellular matrix (ECM) remodeling and vascular dysfunction. Their activity is closely linked to hypertension-mediated organ damage, which affects the vascular and cardio-renal systems. MMPs are responsible for degrading various components of the ECM, which is crucial for maintaining vascular structure and function. In hypertensive patients, several MMPs, including MMP-1, MMP-3, and MMP-9, are often found at elevated levels. This is associated with vascular remodeling and dysfunction due to chronic high blood pressure. The activation of MMPs in hypertension can be triggered by several factors, such as oxidative stress, inflammatory cytokines, and vasoactive agents like angiotensin II. In addition to increasing MMP activity, these variables cause an imbalance between MMPs and tissue inhibitors of metalloproteinases (TIMPs), which are the MMPs' natural inhibitors. This imbalance contributes to excessive degradation of the ECM and promotes pathological changes in vascular smooth muscle cells (VSMCs), leading to their transition from a contractile to a synthetic phenotype. This shift facilitates cell growth and migration, exacerbating vascular remodeling. Given their critical roles in hypertension-related organ damage, MMPs are being explored as potential pharmacological targets. Inhibitors of MMPs may help mitigate the adverse effects of hypertension by restoring balance in ECM remodeling processes. Understanding their mechanisms opens avenues for targeted therapies that could significantly improve outcomes for individuals suffering from hypertension-related complications.
Collapse
Affiliation(s)
- Soroush Taherkhani
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sheibani
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Mohammadkhanizadeh
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Jitka A. I. Virag
- Department of Physiology, East Carolina University, Greenville, NC, United States
| | | | - Yaser Azizi
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Mei LN, Wang ZJ, Duan Y, Shen JS, Ye HB, Zhu YY, Luo XD. 4-Hydroxyboesenbergin B of Alpinia japonica protected gastrointestinal tract by inhibiting vancomycin-resistant enterococcus and balancing intestinal microbiota. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119737. [PMID: 40179999 DOI: 10.1016/j.jep.2025.119737] [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: 12/29/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/05/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alpinia japonica, a traditional herb utilized in Miao medicine in southwestern China, has been employed to alleviate symptoms such as stomachache, diarrhea, and abdominal pain, some of these symptoms may be associated with bacterial infections of the gastrointestinal tract. AIM OF THE STUDY To explore antimicrobial compounds related to traditional uses of A. japonica and its potential pathway in vitro and in vivo. MATERIALS AND METHODS Bioactive components of A. japonica were isolated by bioguide separation method. The antibacterial bioactivity of 4-hydroxyboesenbergin B (4-HB) was evaluated by time-kill curve and drug resistance induction. The pathway of 4-HB against VRE was investigated through network pharmacological analysis and validated by in vitro experiments and RT-qPCR assays. Moreover, a mouse gastrointestinal tract model was established to validate the antibacterial bioactivity of 4-HB in vivo. RESULTS 4-HB from A. japonica inhibited VRE (MIC = 16 μg/mL), rapidly killed the bacteria within 4 h at the 4 MIC concentration and exhibited low susceptibility to drug resistance. 4-HB specifically targeted VRE biofilms by down-regulating the expression of AtlA, SgrA, GelE, and Ace. As a result, 4-HB diminished the adhesion and aggregation ability of VRE, reduced the extracellular matrix content, disrupted biofilm structure and morphology, thereby reducing VRE resistance and virulence. Additionally, 4-HB significantly reduced VRE colonization, enhanced intestinal microbiota diversity, and promoted the restoration of intestinal microbiota balance in vivo. Notably, 4-HB enhanced the abundance of beneficial bacteria genera, such as Lactobacillus and Limosilactobacillus. CONCLUSIONS 4-HB has a significant ability to destroy VRE biofilms and balance intestinal microbiota, which might be responsible for the traditional use of A. japonica partly.
Collapse
Affiliation(s)
- Li-Na Mei
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Zhao-Jie Wang
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Yu Duan
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Jia-Shan Shen
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Hong-Bo Ye
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Yan-Yan Zhu
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences Kunming, 650201, China.
| |
Collapse
|
4
|
Ibrahim NS, Shoukry EH, Sharaky M, Diab HM, Elwahy AHM, Abdelhamid IA. Synthesis, cytotoxicity, oxidative stress, anti-metastatic and anti-inflammatory effects of novel 2-methylene-1H-indene-1,3-dione tethered 2-(2-methoxyphenoxy)-N-arylacetamide: induction of apoptosis in HCT116 and HeLa cells. Chem Biol Interact 2025; 416:111549. [PMID: 40355039 DOI: 10.1016/j.cbi.2025.111549] [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: 03/02/2025] [Revised: 04/20/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025]
Abstract
Six novel chalcones were synthesized, and their structures were confirmed using various spectral tools. All the prepared compounds were subjected to SRB cytotoxic screening against nine cancer and two normal cell lines. Compound 7a showed the highest impact against colorectal carcinoma (HCT116) and cervical cancer (HeLa) with IC50 values of 4.6 ± 0.03 and 5.5 ± 0.1 μg/mL, respectively, compared to doxorubicin (4.8 ± 0.4 and 5.7 ± 0.4 μg/mL, respectively). ELISA assay revealed that the apoptotic proteins (P53, Bax, caspases-3, -8, and -9) and the oxidative marker (Malondialdehyde (MDA)) were significantly activated in 7a treated HCT116 and HeLa cells. However, the anti-metastatic markers (Matrix metalloproteinase 2 (MMP2) and Matrix metalloproteinase-9 (MMP9)), anti-apoptotic Bcl2, antioxidant Glutathione (GSH), and anti-inflammatory (interleukin (IL)-6, and IL-1β) were inhibited in HCT116 and HeLa cells treated with 7a. Flow-cytometric analysis of the cell cycle revealed that the percentage of cells in S and G2/M phases in 7a treated HCT116 cells was increased. After 24 h of treatment, Hela-treated cells had a slightly higher proportion of G0/G1 cells. Comet assay demonstrated that compound 7a caused DNA damage with a percentage of 26.22 ± 1.1 % in HCT116 compared to the untreated cells (6.18 ± 0.88 %). Theoretical molecular modeling against P53 cancer mutant Y220C and Bcl2 showed binding energies of -22.7 and -23.3 kcal/mol, respectively, which confirmed our ELISA results.
Collapse
Affiliation(s)
- Nada S Ibrahim
- Department of Chemistry (Biochemistry Division), Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Eman Hatem Shoukry
- Department of Chemistry (Biochemistry Division), Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Marwa Sharaky
- Pharmacology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Hadeer M Diab
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Ahmed H M Elwahy
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| | - Ismail A Abdelhamid
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
5
|
Timoshenko O, Kugaevskaya E, Gureeva T, Morozevich G, Lupatov A, Mekhtiev A, Rudovich A, Zhabinskii V, Khripach V, Lisitsa A. Isoxazolyl steroid blocks the Shh signaling pathway and the expression of MMP-2 and MMP-9 in cervical carcinoma cell lines. Steroids 2025; 217:109599. [PMID: 40101884 DOI: 10.1016/j.steroids.2025.109599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Cervical cancer is the fourth leading cause of cancer death among women worldwide. Matrix metalloproteinases MMP-2 and MMP-9 play a leading role in the processes of invasion and metastasis in cervical cancer. Research on the development of MMP inhibitors not yielded the expected results due to their serious side effects. Study of signaling pathways involved in regulation of MMPs expression is of great importance for search of new classes of therapeutic drugs. Aberrant activation of the Sonic Hedgehog (Shh) signaling pathway is associated with increased MMPs in many types of human cancer. This study investigated the inhibitory action of 17β-((3-butylisoxazol-5-yl)methyl)-androst-5-en-3β-ol on the Shh signaling pathway key genes (Ptch, Smo, Gli) expression and MMP-2, MMP-9 genes expression in human cervical carcinoma cell lines (SiHa and CaSki) and keratinocytes (HaCaT). Cyclopamine was used for comparative analysis. Gene expression analysis was performed using real-time PCR; the effects on survival and cell cycle were studied using the MTT test and flow cytometry method. 17β-((3-butylisoxazol-5-yl)methyl)-androst-5-en-3β-ol had higher cytotoxicity and more effectively blocked the Shh signaling pathway genes and MMP-2 and MMP-9 genes compared to cyclopamine in all cell lines. The results obtained demonstrate potential of 17β-((3-butylisoxazol-5-yl)methyl)-androst-5-en-3β-ol as the anticancer drug that simultaneously block the Shh signaling pathway and MMP expression. We are confident that the search for substances capable of simultaneously affecting several key components involved in tumor progression is of great importance for the creation of next-generation therapeutic agents.
Collapse
Affiliation(s)
- Olga Timoshenko
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Str., 119121 Moscow, Russia.
| | - Elena Kugaevskaya
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Str., 119121 Moscow, Russia
| | - Tatiana Gureeva
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Str., 119121 Moscow, Russia
| | - Galina Morozevich
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Str., 119121 Moscow, Russia
| | - Alexey Lupatov
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Str., 119121 Moscow, Russia
| | - Arif Mekhtiev
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Str., 119121 Moscow, Russia
| | - Anton Rudovich
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich Str., 5/2, 220084 Minsk, Belarus
| | - Vladimir Zhabinskii
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich Str., 5/2, 220084 Minsk, Belarus
| | - Vladimir Khripach
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich Str., 5/2, 220084 Minsk, Belarus
| | - Andrey Lisitsa
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Str., 119121 Moscow, Russia
| |
Collapse
|
6
|
Xiao R, Xu P, Li X, Shen F, Tao S, Zhu X, Cai Y, Feng Z, Liu Z, Xiao H, Ding F, Zhu M. The LysoPS/GPR174 axis drives metastatic progression in esophageal squamous cell carcinoma through cAMP-PKA-CREB signaling activation. J Transl Med 2025; 23:438. [PMID: 40229851 PMCID: PMC11995483 DOI: 10.1186/s12967-025-06419-0] [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: 11/08/2024] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a highly lethal malignancy with a 5-year survival rate of less than 20%, largely due to its high propensity for metastasis and recurrence. There is an urgent need to identify targeted therapeutic agents for this disease. While lysophosphatidylserine (LysoPS) and its receptor GPR174 are known regulators of immune and inflammatory processes, their mechanistic role in ESCC progression remains unexplored. This study investigates the LysoPS/GPR174 axis in driving ESCC metastasis and its underlying molecular pathways. METHODS LC-MS was used to measure LysoPS concentration, and Western blotting was performed for protein quantification. The correlation between GPR174 expression and ESCC prognosis was analyzed using ESCC tissue microarrays, immunohistochemistry, and Kaplan-Meier survival analysis. Wound healing and Transwell assays were carried out to evaluate the migratory and invasive capacities of cells. The proliferative ability of ESCC cell lines was assessed with the CCK-8 assay. Nuclear-cytoplasmic extraction assay was conducted to separate the nucleus and cytoplasm. Metastasis model of nude mouse was employed to investigate the metastasis of ESCC cell lines. RESULTS We found that the levels of LysoPS were significantly increased in metastatic ESCC tissues compared to nonmetastatic ESCC tissues. Moreover, a correlation was established between LysoPS-mediated tumor metastasis and GPR174 expression in ESCC. Our results also revealed that high expression of GPR174 in ESCC is associated with tumor metastasis and poor survival outcomes in ESCC patients. Further exploration of the underlying mechanism showed that LysoPS stimulates the up- regulation of GPR174 expression. The increased GPR174 then activates the cAMP-PKA signaling pathway. Subsequently, the active subunit of PKA translocates into the nucleus, where it phosphorylates CREB, thereby promoting the metastasis of ESCC. In vivo, GPR174 overexpression increased metastasis burden. CONCLUSIONS Our study demonstrates that the LysoPS/GPR174 axis, through the cAMP-PKA-CREB pathway, plays a crucial role in promoting the invasion and metastasis of ESCC. This highlights its potential as a novel target for predicting ESCC progression and may offer new insights for the development of targeted therapies for this deadly disease.
Collapse
Affiliation(s)
- Rong Xiao
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Pei Xu
- Department of Cardiothoracic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiangyuan Li
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Feng Shen
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Shuangfen Tao
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiaocen Zhu
- Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu Cai
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhuowei Feng
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhiyi Liu
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Haibo Xiao
- Department of Cardiothoracic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Fangbao Ding
- Department of Cardiothoracic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Meiling Zhu
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
7
|
Kast RE. Potential Benefits of Adding Alendronate, Celecoxib, Itraconazole, Ramelteon, and Simvastatin to Endometrial Cancer Treatment: The EC5 Regimen. Curr Issues Mol Biol 2025; 47:153. [PMID: 40136407 PMCID: PMC11941490 DOI: 10.3390/cimb47030153] [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: 01/26/2025] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Metastatic endometrial cancer continues to be a common cause of death as of 2024, even after maximal use of all currently available standard treatments. To address this problem of metastatic cancer generally in 2025, the drug repurposing movement within oncology identifies medicines in common general medical use that have clinical or preclinical experimental data indicating that they interfere with or inhibit a specific growth driving element identified in a given cancer. The drug repurposing movement within oncology also uses data from large scale in vitro screens of thousands of drugs, looking for simple empirical growth inhibition in a given cancer type. This paper outlines the data showing that five drugs from general medical practice meet these evidence criteria for inhibition of endometrial cancer growth, the EC5 regimen. The EC5 regimen uses the osteoporosis treatment drug, alendronate; the analgesic drug, celecoxib; the antifungal drug, itraconazole; the sleep aid, ramelteon; and the cholesterol lowering drug, simvastatin. Side effects seen with these drugs are usually minimal and easily tolerated by patients.
Collapse
|
8
|
Oselusi SO, Sibuyi NR, Martin DR, Meyer M, Madiehe AM. Potential matrix metalloproteinase 2 and 9 inhibitors identified from Ehretia species for the treatment of chronic wounds - Computational drug discovery approaches. Comput Biol Med 2025; 185:109487. [PMID: 39637455 DOI: 10.1016/j.compbiomed.2024.109487] [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: 04/26/2024] [Revised: 10/29/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Matrix metalloproteinases (MMPs) serve as prognostic factors in several pathophysiological conditions, including chronic wounds. Therefore, they are considered important therapeutic targets in the intervention and treatment of these conditions. In this study, computational tools such as molecular docking and molecular dynamics simulations were used to gain insight into protein‒ligand interactions and determine the free binding energy between Ehretia species phytoconstituents and gelatinases (MMP2 and MMP9). A total of 74 phytoconstituents from Ehretia species were compiled from the literature, and 46 of these compounds were identified as potential inhibitors of at least one type of MMP. Molecular docking revealed that lithospermic acid B, rosmarinic acid, and danshensu had stronger binding affinities against the two enzymes than the reference ligands. Furthermore, (9S, 10E, 12Z, 15Z)-9-hydroxy-10,12,15-octadecatrienoic (∗-octadecatrienoic) had a higher binding energy for MMP2, whereas caffeic anhydride and caffeic acid established stronger binding energy with MMP9 than the reference ligand. These complexes also demonstrated relatively stable, favourable, and comparable conformational changes with those of unbound proteins at 500 ns. The free energy decomposition results further provide detailed insights into the contributions of active site residues and different types of interactions to the overall binding free energy. Finally, most of the hit phytoconstituents (rosmarinic acid, caffeic anhydride, caffeic acid, and danshensu) had good physicochemical, drug-likeness, and pharmacokinetic properties. Collectively, our findings showed that phytoconstituents from Ehretia species could be beneficial in the search for novel MMP inhibitors as therapeutic agents for the treatment of chronic wounds.
Collapse
Affiliation(s)
- Samson O Oselusi
- Nanobiotechnology Research Group, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa; DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa
| | - Nicole Rs Sibuyi
- DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa; Health Platform, Advanced Materials Division, Mintek, 200 Malibongwe Drive, Randburg, South Africa
| | - Darius R Martin
- DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa
| | - Mervin Meyer
- DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa
| | - Abram M Madiehe
- Nanobiotechnology Research Group, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa; DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa.
| |
Collapse
|
9
|
Sreesada P, Vandana, Krishnan B, Amrutha R, Chavan Y, Alfia H, Jyothis A, Venugopal P, Aradhya R, Suravajhala P, Nair BG. Matrix metalloproteinases: Master regulators of tissue morphogenesis. Gene 2025; 933:148990. [PMID: 39393432 DOI: 10.1016/j.gene.2024.148990] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/13/2024]
Abstract
The matrix metalloproteinases (MMPs) are a class of zinc proteases that aid in breaking most of the extracellular matrix's (ECM) constituents. Additionally, MMPs play a part in processing elements that affect inflammation, cell development and proliferation, and many more. In vivo genetic study of the Drosophila MMPs Mmp1 and Mmp2 reveals they are essential for tissue remodeling but not embryonic development. The canonical and conserved MMP domain organization is present in both fly MMPs. Because Mmp2 appeared to be membrane-anchored and Mmp1 appeared to be released, the pericellular localization of Drosophila MMPs has been used to classify them. This suggests that the protein's localization is the critical distinction in this small MMP family. The signal sequence, the propeptide, the catalytic domain, and the hemopexin-like domain are among the numerous domains found in MMPs. Following secretion from the extracellular environment to the endoplasmic reticulum, the pre-domain, also known as the signal sequence, serves to direct MMP production. MMPs of the secretory and membrane types (MT-MMPs) are two groups of MMPs that have been widely recognized. Subgroups of MMPs are categorized based on their structure and function. While analysis of the intracellular activity of human MMPs is challenging because the human genome contains around 23 distinct MMPs with overlapping functions, only two MMPs, dMMP1 and dMMP2, are encoded by the Drosophila melanogaster genome. On the other hand, the balance between MMPs and the family members are implicated in various pathophysiology/progression of diseases, but whether or not the mechanisms of MMP inhibition are not clearly understood as master regulators. In this review, we outline the role of MMPs as master regulators of tissue morphogenesis.
Collapse
Affiliation(s)
- P Sreesada
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO 690525, Kerala, India
| | - Vandana
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO 690525, Kerala, India
| | - Bhagath Krishnan
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO 690525, Kerala, India
| | - R Amrutha
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO 690525, Kerala, India
| | - Yash Chavan
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO 690525, Kerala, India
| | - Hasanath Alfia
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO 690525, Kerala, India
| | - Anjali Jyothis
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO 690525, Kerala, India
| | - Parvathy Venugopal
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO 690525, Kerala, India
| | - Rajaguru Aradhya
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO 690525, Kerala, India.
| | - Prashanth Suravajhala
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO 690525, Kerala, India.
| | - Bipin G Nair
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO 690525, Kerala, India
| |
Collapse
|
10
|
Das S, Patel T, Himaja A, Regula S, Banerjee S, De AK, Qureshi IA, Gayen S, Ghosh B, Adhikari N, Jha T. Derivatives of D(-)-glutamine-based MMP-2 inhibitors as an effective remedy for the management of chronic myeloid leukemia-Part-III: Synthesis, biological screening and in silico binding interaction analysis. Bioorg Chem 2025; 154:108057. [PMID: 39708552 DOI: 10.1016/j.bioorg.2024.108057] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Tyrosine kinase inhibitors (TKIs) have markedly improved the overall survival rate of patients with chronic myeloid leukemia (CML), enabling them to achieve a normal life expectancy. However, toxicity, relapse, and drug resistance continue to pose major challenges in the clinical treatment of CML. The progression of leukemia is directly connected to higher expression levels and enzymatic actions of matrix metalloproteinase-2 (MMP-2). It is also associated with increased expression and enzymatic actions of matrix metalloproteinase-9 (MMP-9). From this perspective, MMP-2 and MMP-9 offers a promising strategy for developing novel therapeutic molecules that could be effective in treating CML. This study is the Part-III of D(-)-glutamine-based MMP-2 inhibitors series for the management of chronic myeloid leukemia. Fourteen newly synthesized p-tosyl-D(-)-glutamine derivatives were examined in cell culture-based antileukemic assays and also evaluated for their ability to inhibit MMPs. The lead compounds 5g and 5j demonstrated the most promising antileukemic potential. Compounds 5g and 5j are safe for normal cells and effectively block gelatinases (MMP-2 and MMP-9). The best active molecule 5g induced significant apoptosis. Compound 5g reduced MMP-2 levels in the K562 cell line. It also had strong antiangiogenic effects in the ACHN cell line. The strongest MMP-2 inhibitor, 5g, had stable binding at the MMP-2 active site, which is linked to its effective inhibition of MMP-2. In conclusion, these p-tosyl-D(-)-glutamine derivatives are promising MMP-2 inhibitors. They have strong anti-CML effects and should be studied more for future CML treatment.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Matrix Metalloproteinase 2/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Matrix Metalloproteinase Inhibitors/pharmacology
- Matrix Metalloproteinase Inhibitors/chemistry
- Matrix Metalloproteinase Inhibitors/chemical synthesis
- Glutamine/chemistry
- Glutamine/metabolism
- Glutamine/pharmacology
- Structure-Activity Relationship
- Molecular Structure
- Drug Screening Assays, Antitumor
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Molecular Docking Simulation
Collapse
Affiliation(s)
- Sanjib Das
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India; School of Pharmacy, Sister Nivedita University, DG Block (New Town), Action Area 1, 1/2, Newtown, Chakpachuria, Kolkata 700156, India
| | - Tarun Patel
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Ambati Himaja
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Sanjeev Regula
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Suvankar Banerjee
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Asit Kumar De
- Department of Chemistry, Jadavpur University, Kolkata, India
| | - Insaf Ahmed Qureshi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500 046, Telangana, India
| | - Shovanlal Gayen
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India.
| | - Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| |
Collapse
|
11
|
Reddy RA, Varshini MS, Kumar RS. Matrix Metalloproteinase-2 (MMP-2): As an Essential Factor in Cancer Progression. Recent Pat Anticancer Drug Discov 2025; 20:26-44. [PMID: 37861020 PMCID: PMC11826896 DOI: 10.2174/0115748928251754230922095544] [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: 03/03/2023] [Revised: 07/21/2023] [Accepted: 08/24/2023] [Indexed: 10/21/2023]
Abstract
The development of cancer has been a multistep process involving mutation, proliferation, survival, invasion, and metastasis. Of all the characteristics of cancer, metastasis is believed to be the hallmark as it is responsible for the highest number of cancer-related deaths. In connection with this, Matrix metalloproteinases (MMPs), that has a role in metastasis, are one of the novel therapeutic targets. MMPs belong to the family of zinc-dependent endopeptidases and are capable of degrading the components of the extracellular matrix (ECM). The role of MMPs in ECM remodeling includes tissue morphogenesis, uterine cycling, growth, tissue repair, and angiogenesis. During pathological conditions, MMPs play a critical role in the excessive degradation of ECM which includes arthritis, tumour invasion, tumour metastasis, and several other autoimmune disorders. Moreover, they are believed to be involved in many physiological aspects of the cell, such as proliferation, migration, differentiation, angiogenesis, and apoptosis. It is reported that dysregulation of MMP in a variety of cancer subtypes have a dual role in tumour growth and metastasis processes. Further, multiple studies suggest the therapeutic potential of targeting MMP in invading cancer. The expression of MMP-2 correlates with the clinical characteristics of cancer patients, and its expression profile is a new diagnostic and prognostic biomarker for a variety of human diseases. Hence, manipulating the expression or function of MMP-2 may be a potential treatment strategy for different diseases, including cancers. Hence, the present review discusses the therapeutic potential of targeting MMP in various types of cancers and their recent patents.
Collapse
|
12
|
van der Net A, Rahman Z, Bordoloi AD, Muntz I, ten Dijke P, Boukany PE, Koenderink GH. EMT-related cell-matrix interactions are linked to states of cell unjamming in cancer spheroid invasion. iScience 2024; 27:111424. [PMID: 39717087 PMCID: PMC11665421 DOI: 10.1016/j.isci.2024.111424] [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: 05/13/2024] [Revised: 09/25/2024] [Accepted: 11/15/2024] [Indexed: 12/25/2024] Open
Abstract
Epithelial-to-mesenchymal transitions (EMT) and unjamming transitions provide two distinct pathways for cancer cells to become invasive, but it is still unclear to what extent these pathways are connected. Here, we addressed this question by performing 3D spheroid invasion assays on epithelial-like (A549) and mesenchymal-like (MV3) cancer cell lines in collagen-based hydrogels, where we varied both the invasive character of the cells and matrix porosity. We found that the onset time of invasion was correlated with the matrix porosity and vimentin levels, while the spheroid expansion rate correlated with MMP1 levels. Spheroids displayed solid-like (non-invasive) states in small-pore hydrogels and fluid-like (strand-based) or gas-like (disseminating cells) states in large-pore hydrogels or for mesenchymal-like cells. Our findings are consistent with different unjamming states as a function of cell motility and matrix confinement predicted in recent models for cancer invasion, but show that cell motility and matrix confinement are coupled via EMT-related matrix degradation.
Collapse
Affiliation(s)
- Anouk van der Net
- Delft University of Technology, Department of Bionanoscience, Kavli Institute of Nanoscience, Delft 2629 HZ, the Netherlands
| | - Zaid Rahman
- Delft University of Technology, Department of Chemical Engineering, Delft 2629 HZ, the Netherlands
| | - Ankur D. Bordoloi
- Delft University of Technology, Department of Chemical Engineering, Delft 2629 HZ, the Netherlands
| | - Iain Muntz
- Delft University of Technology, Department of Bionanoscience, Kavli Institute of Nanoscience, Delft 2629 HZ, the Netherlands
| | - Peter ten Dijke
- Leiden University Medical Center, Department of Cell and Chemical Biology and Oncode Institute, Leiden 2333 ZC, the Netherlands
| | - Pouyan E. Boukany
- Delft University of Technology, Department of Chemical Engineering, Delft 2629 HZ, the Netherlands
| | - Gijsje H. Koenderink
- Delft University of Technology, Department of Bionanoscience, Kavli Institute of Nanoscience, Delft 2629 HZ, the Netherlands
| |
Collapse
|
13
|
Ibrahim NS, Sayed HA, Sharaky M, Diab HM, Elwahy AHM, Abdelhamid IA. Synthesis, cytotoxicity, anti-inflammatory, anti-metastatic and anti-oxidant activities of novel chalcones incorporating 2-phenoxy-N-arylacetamide and thiophene moieties: induction of apoptosis in MCF7 and HEP2 cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:10091-10107. [PMID: 38980411 PMCID: PMC11582173 DOI: 10.1007/s00210-024-03255-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/22/2024] [Indexed: 07/10/2024]
Abstract
Eight Novel chalcones were synthesized and their structures were confirmed by different spectral tools. All the prepared compounds were subjected to SRB cytotoxic screening against several cancer cell lines. Compound 5c exerted the most promising effect against MCF7 and HEP2 cells with IC50 values of 9.5 and 12 µg/mL, respectively. Real-time PCR demonstrated the inhibitory effect of compound 5c on the expression level of Antigen kiel 67 (KI-67), Survivin, Interleukin-1beta (IL-1B), Interleukin-6 (IL-6), Cyclooxygenase-2 (COX-2) and Protein kinase B (AKT1) genes. Flow-cytometric analysis of the cell cycle indicated that compound 5c stopped the cell cycle at the G0/G1 and G2/M phases in MCF7 and HEP2 treated cells, respectively. ELISA assay showed that Caspase 8, Caspase 9, P53, BAX, and Glutathione (GSH) were extremely activated and Matrix metalloproteinase 2 (MMP2), Matrix metalloproteinase 9 (MMP9), BCL2, Malondialdehyde (MDA), and IL-6 were deactivated in 5c treated MCF7 and HEP2 cells. Wound healing revealed that chalcone 5c reduced the ability to close the scrape wound and decreased the number of migrating MCF7 and HEP2 cells compared to the untreated cells after 48 h. Theoretical molecular modeling against P53 cancer mutant Y220C and Bcl2 showed binding energies of -22.8 and -24.2 Kcal/mole, respectively, which confirmed our ELISA results.
Collapse
Affiliation(s)
- Nada S Ibrahim
- Department of Chemistry (Biochemistry Division), Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Hager Ahmed Sayed
- Department of Chemistry (Biochemistry Division), Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Marwa Sharaky
- Pharmacology unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Hadeer M Diab
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Ahmed H M Elwahy
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| | - Ismail A Abdelhamid
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
14
|
Doppelt-Flikshtain O, Asbi T, Younis A, Ginesin O, Cohen Z, Tamari T, Berg T, Yanovich C, Aran D, Zohar Y, Assaraf YG, Zigdon-Giladi H. Inhibition of osteosarcoma metastasis in vivo by targeted downregulation of MMP1 and MMP9. Matrix Biol 2024; 134:48-58. [PMID: 39278602 DOI: 10.1016/j.matbio.2024.09.002] [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: 04/01/2024] [Revised: 09/08/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Osteosarcoma (OS) mortality stems from lung metastases. Matrix metalloproteinases (MMPs) facilitate metastatic dissemination by degrading extracellular matrix components. Herein we studied the impact of targeted MMP downregulation on OS metastasis. Differential gene expression analysis of human OS cell lines revealed high MMP9 expression in the majority of OS cell lines. Furthermore, 143B, a metastatic OS cell line, exhibited increased MMP1 and MMP9 mRNA levels. Gene set enrichment analysis on metastatic and non-metastatic OS patient specimens indicated epithelial-mesenchymal transition as the most enriched gene set, with MMP9 displaying strong association to genes in this network. Using the same dataset, Kaplan-Meier analysis revealed a correlation between MMP1 expression and dismal patient survival. Hence, we undertook targeted suppression of MMP1 and MMP9 gene expression in OS cell lines. The ability of OS cells to migrate and form colonies was markedly reduced upon MMP1 and MMP9 downregulation, whereas their cell proliferation capacity remained intact. MMP9 downregulation decreased tumor growth and lung metastases area in an orthotopic mouse OS model. Consistently, human OS lung metastasis specimens displayed marked MMP9 protein expression. Our findings highlight the role of MMP1 and MMP9 in OS metastasis, warranting further exploration of simultaneous inhibition of MMPs for future OS therapeutics.
Collapse
Affiliation(s)
- Ofri Doppelt-Flikshtain
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Haifa, Israel
| | - Thabet Asbi
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Haifa, Israel; Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel
| | - Amin Younis
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Haifa, Israel; Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel
| | - Ofir Ginesin
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Haifa, Israel; Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel
| | - Ziv Cohen
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tal Tamari
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Haifa, Israel
| | - Tal Berg
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Haifa, Israel
| | - Chen Yanovich
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Dvir Aran
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yaniv Zohar
- Department of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Hadar Zigdon-Giladi
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Haifa, Israel; Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel.
| |
Collapse
|
15
|
Yao Y, Shen G, Luo J, Wang J, Xu Z, Wang H, Cui L. Research Progress with Atractylone as an Antitumor Agent. Molecules 2024; 29:5450. [PMID: 39598839 PMCID: PMC11597220 DOI: 10.3390/molecules29225450] [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: 09/06/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Atractylone is a sesquiterpenoid compound extracted from Rhizoma Atractylodis. As one of the main active components in the volatile oil of the Atractylodes genus, it has exhibited certain therapeutic effects, including anti-inflammatory, antiviral, antioxidant, antiallergic, antiangiogenic, and neuroprotective activities, among others. With further research on the chemical constituents and pharmacology of sesquiterpenes, research on the antitumor activity of Atractylone has also been further expanded. Much of the current literature pays particular attention to the antitumor activity of Atractylone, which was found to inhibit the apoptosis of tumor cells and prevent growth, invasion, and migration through different apoptosis pathways and signaling pathways. Due to its promising potential for cancer prevention, it may play a role in reducing the incidence of malignant tumors. In this paper, the antitumor activity and mechanism of Atractylone are reviewed, providing a reference to inform future research on the tumor treatment, clinical application, and further development and utilization of this plant genus.
Collapse
Affiliation(s)
- Ying Yao
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
| | - Guanghuan Shen
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
- Postdoctoral Programme of Meteria Medica Institute, Harbin University of Commerce, Harbin 150076, China
| | - Jianghan Luo
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
| | - Jinhong Wang
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
| | - Zheng Xu
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
| | - Hao Wang
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
| | - Linlin Cui
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
| |
Collapse
|
16
|
Liu Y, Sun X, Wang L, Dou Y, Tian Y, Yu T, Zhang Y, Zhao Q, Lu J, Feng Y, Wang J, Liu X, Shang Y, Li C, Yang Q. Sequential Targeted Enzyme-Instructed Self-Assembly Supramolecular Nanofibers to Attenuate Intervertebral Disc Degeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408678. [PMID: 39221659 DOI: 10.1002/adma.202408678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/06/2024] [Indexed: 09/04/2024]
Abstract
As an age-related disease, intervertebral disc degeneration is closely related to inflammation and aging. Inflammatory cytokines and cellular senescence collectively contribute to the degradation of intervertebral disc. Blocking this synergy reduces disc extracellular matrix damage caused by inflammation and aging. In this study, drug-loaded nanofibers with sequential targeting functions are constructed through intelligent response, hydrophilicity, and in situ self-assembly empowerment of flurbiprofen. The peptide precursor responds to the cleavage of overexpressed MMP-2 in the degenerative intervertebral disc microenvironment (intracellular and extracellular), resulting in the formation of self-assembled nanofibers that enable the on-demand release of flurbiprofen and COX-2 response. In vitro, Comp. 1 (Flurbiprofen-GFFYPLGLAGEEEERGD) reduces the expression of inflammation-related genes and proteins and the polarization of M1 macrophages by competitively inhibiting COX-2 and increases the expression of extracellular matrix proteins COL-2 and aggrecan. Additionally, it can reduce the expression of Senescence-Associated Secretory Phenotype and DNA damage in aged nucleus pulposus cells and promote the recovery of proliferation and cell cycle. In vivo, drug-loaded nanofibers delay intervertebral disc degeneration by inhibiting inflammation and preventing the accumulation of senescent cells. Therefore, the sequentially targeted self-assembled drug-loaded nanofibers can delay intervertebral disc degeneration by blocking the synergistic effect of inflammatory cytokines and cellular senescence.
Collapse
Affiliation(s)
- Yang Liu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, P. R. China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, P. R. China
| | - Lianlei Wang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Yiming Dou
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, P. R. China
| | - Ye Tian
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, P. R. China
| | - Tianyu Yu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, P. R. China
| | - Yiming Zhang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, P. R. China
| | - Qingqian Zhao
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, P. R. China
| | - Jiayi Lu
- Academy of Interdisciplinary Studies on Intelligent Molecules, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin, 300387, P. R. China
| | - Yinyin Feng
- Academy of Interdisciplinary Studies on Intelligent Molecules, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin, 300387, P. R. China
| | - Jiayu Wang
- Academy of Interdisciplinary Studies on Intelligent Molecules, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin, 300387, P. R. China
| | - Xinyu Liu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Yuna Shang
- Academy of Interdisciplinary Studies on Intelligent Molecules, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin, 300387, P. R. China
| | - Chunju Li
- Academy of Interdisciplinary Studies on Intelligent Molecules, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin, 300387, P. R. China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, P. R. China
| |
Collapse
|
17
|
Cavalu S, Saber S, Ramadan A, Elmorsy EA, Hamad RS, Abdel-Reheim MA, Youssef ME. Unveiling citicoline's mechanisms and clinical relevance in the treatment of neuroinflammatory disorders. FASEB J 2024; 38:e70030. [PMID: 39221499 DOI: 10.1096/fj.202400823r] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/07/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Citicoline, a compound produced naturally in small amounts in the human body, assumes a pivotal role in phosphatidylcholine synthesis, a dynamic constituent of membranes of neurons. Across diverse models of brain injury and neurodegeneration, citicoline has demonstrated its potential through neuroprotective and anti-inflammatory effects. This review aims to elucidate citicoline's anti-inflammatory mechanism and its clinical implications in conditions such as ischemic stroke, head trauma, glaucoma, and age-associated memory impairment. Citicoline's anti-inflammatory prowess is rooted in its ability to stabilize cellular membranes, thereby curbing the excessive release of glutamate-a pro-inflammatory neurotransmitter. Moreover, it actively diminishes free radicals and inflammatory cytokines productions, which could otherwise harm neurons and incite neuroinflammation. It also exhibits the potential to modulate microglia activity, the brain's resident immune cells, and hinder the activation of NF-κB, a transcription factor governing inflammatory genes. Clinical trials have subjected citicoline to rigorous scrutiny in patients grappling with acute ischemic stroke, head trauma, glaucoma, and age-related memory impairment. While findings from these trials are mixed, numerous studies suggest that citicoline could confer improvements in neurological function, disability reduction, expedited recovery, and cognitive decline prevention within these cohorts. Additionally, citicoline boasts a favorable safety profile and high tolerability. In summary, citicoline stands as a promising agent, wielding both neuroprotective and anti-inflammatory potential across a spectrum of neurological conditions. However, further research is imperative to delineate the optimal dosage, treatment duration, and underlying mechanisms. Moreover, identifying specific patient subgroups most likely to reap the benefits of citicoline as a new therapy remains a critical avenue for exploration.
Collapse
Affiliation(s)
- Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Asmaa Ramadan
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Central Laboratory, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt
| | - Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
18
|
Baidya SK, Patel T, Himaja A, Banerjee S, Das S, Ghosh B, Jha T, Adhikari N. Biphenylsulfonamides as effective MMP-2 inhibitors with promising antileukemic efficacy: Synthesis, in vitro biological evaluation, molecular docking, and MD simulation analysis. Drug Dev Res 2024; 85:e22255. [PMID: 39233391 DOI: 10.1002/ddr.22255] [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: 05/21/2024] [Revised: 07/17/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024]
Abstract
Overexpression of matrix metalloproteinase-2 (MMP-2) possesses a correlation with leukemia especially chronic myeloid leukemia (CML). However, no such MMP-2 inhibitor has come out in the market to date for treating leukemia. In this study, synthesis, biological evaluation, and molecular modeling studies of a set of biphenylsulfonamide derivatives as promising MMP-2 inhibitors were performed, focusing on their potential applications as antileukemic therapeutics. Compounds DH-18 and DH-19 exerted the most effective MMP-2 inhibition (IC50 of 139.45 nM and 115.16 nM, respectively) with potent antileukemic efficacy against the CML cell line K562 (IC50 of 0.338 µM and 0.398 µM, respectively). The lead molecules DH-18 and DH-19 reduced the MMP-2 expression by 21.3% and 17.8%, respectively with effective apoptotic induction (45.4% and 39.8%, respectively) in the K562 cell line. Moreover, both these compounds significantly arrested different phases of the cell cycle. Again, both these molecules depicted promising antiangiogenic efficacy in the ACHN cell line. Nevertheless, the molecular docking and molecular dynamics (MD) simulation studies revealed that DH-18 formed strong bidentate chelation with the catalytic Zn2+ ion through the hydroxamate zinc binding group (ZBG). Apart from that, the MD simulation study also disclosed stable binding interactions of DH-18 and MMP-2 along with crucial interactions with active site amino acid residues namely His120, Glu121, His124, His130, Pro140, and Tyr142. In a nutshell, this study highlighted the importance of biphenylsulfonamide-based novel and promising MMP-2 inhibitors to open up a new avenue for potential therapy against CML.
Collapse
Affiliation(s)
- Sandip K Baidya
- Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Natural Science Laboratory, Jadavpur University, Kolkata, India
- School of Pharmacy, Sister Nivedita University, Kolkata, India
| | - Tarun Patel
- Department of Pharmacy, Epigenetic Research Laboratory, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Ambati Himaja
- Department of Pharmacy, Epigenetic Research Laboratory, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Suvankar Banerjee
- Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Natural Science Laboratory, Jadavpur University, Kolkata, India
| | - Sanjib Das
- Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Natural Science Laboratory, Jadavpur University, Kolkata, India
- School of Pharmacy, Sister Nivedita University, Kolkata, India
| | - Balaram Ghosh
- Department of Pharmacy, Epigenetic Research Laboratory, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Tarun Jha
- Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Natural Science Laboratory, Jadavpur University, Kolkata, India
| | - Nilanjan Adhikari
- Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Natural Science Laboratory, Jadavpur University, Kolkata, India
| |
Collapse
|
19
|
Das S, Mondal S, Patel T, Himaja A, Adhikari N, Banerjee S, Baidya SK, De AK, Gayen S, Ghosh B, Jha T. Derivatives of D(-) glutamine-based MMP-2 inhibitors as an effective remedy for the management of chronic myeloid leukemia-Part-I: Synthesis, biological screening and in silico binding interaction analysis. Eur J Med Chem 2024; 274:116563. [PMID: 38843586 DOI: 10.1016/j.ejmech.2024.116563] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/23/2024] [Accepted: 05/31/2024] [Indexed: 06/17/2024]
Abstract
Chronic myeloid leukemia (CML) is a global issue and the available drugs such as tyrosine kinase inhibitors (TKIs) comprise various toxic effects as well as resistance and cross-resistance. Therefore, novel molecules targeting specific enzymes may unravel a new direction in antileukemic drug discovery. In this context, targeting gelatinases (MMP-2 and MMP-9) can be an alternative option for the development of novel molecules effective against CML. In this article, some D(-)glutamine derivatives were synthesized and evaluated through cell-based antileukemic assays and tested against gelatinases. The lead compounds, i.e., benzyl analogs exerted the most promising antileukemic potential showing nontoxicity in normal cell line including efficacious gelatinase inhibition. Both these lead molecules yielded effective apoptosis and displayed marked reductions in MMP-2 expression in the K562 cell line. Not only that, but both of them also revealed effective antiangiogenic efficacy. Importantly, the most potent MMP-2 inhibitor, i.e., benzyl derivative of p-tosyl D(-)glutamine disclosed stable binding interaction at the MMP-2 active site correlating with the highly effective MMP-2 inhibitory activity. Therefore, such D(-)glutamine derivatives might be explored further as promising MMP-2 inhibitors with efficacious antileukemic profiles for the treatment of CML in the future.
Collapse
Affiliation(s)
- Sanjib Das
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Subha Mondal
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Tarun Patel
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Ambati Himaja
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | - Suvankar Banerjee
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Sandip Kumar Baidya
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Asit Kumar De
- Department of Chemistry, Jadavpur University, Kolkata, India
| | - Shovanlal Gayen
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India.
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| |
Collapse
|
20
|
Zeng T, Ren W, Zeng H, Wang D, Wu X, Xu G. TFAP2A Activates S100A2 to Mediate Glutamine Metabolism and Promote Lung Adenocarcinoma Metastasis. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e13825. [PMID: 39187936 PMCID: PMC11347387 DOI: 10.1111/crj.13825] [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: 03/15/2024] [Revised: 07/03/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a fatal disease with metabolic abnormalities. The dysregulation of S100 calcium-binding protein A2 (S100A2), a member of the S100 protein family, is connected to the development of various cancers. The impact of S100A2 on the LUAD occurrence and metastasis, however, has not yet been reported. The functional mechanism of S100A2 on LUAD cell metastasis was examined in this article. METHODS The expression of TFAP2A and S100A2 in LUAD tissues and cells was analyzed by bioinformatics and qRT-PCR, respectively. The enrichment pathway analysis was performed on S100A2. Bioinformatics analysis determined the binding relationship between TFAP2A and S100A2, and their interaction was validated through dual-luciferase and chromatin immunoprecipitation experiments. Cell viability was determined using cell counting kit-8 (CCK-8). A transwell assay was performed to analyze the invasion and migration of cells. Immunofluorescence was conducted to obtain vimentin and E-cadherin expression, and a western blot was used to detect the expression of MMP-2, MMP-9, GLS, and GLUD1. The kits measured the NADPH/NADP ratio, glutathione (GSH)/glutathione disulfide (GSSG) levels, and the contents of glutamine, α-KG, and glutamate. RESULTS S100A2 was upregulated in LUAD tissues and cells, and S100A2 mediated glutamine metabolism to induce LUAD metastasis. Additionally, the transcriptional regulator TFAP2A was discovered upstream of S100A2, and TFAP2A expression was upregulated in LUAD, which indicated that TFAP2A promoted the S100A2 expression. The rescue experiment found that upregulation of S100A2 could reverse the inhibitory effects of silencing TFAP2A on glutamine metabolism and cell metastasis. CONCLUSION In conclusion, by regulating glutamine metabolism, the TFAP2A/S100A2 axis facilitated LUAD metastasis. This suggested that targeting S100A2 could be beneficial for LUAD treatment.
Collapse
Affiliation(s)
- Tao Zeng
- Department of Cardiothoracic SurgerySichuan Mianyang 404 HospitalMianyang CitySichuan ProvinceChina
| | - Wangsheng Ren
- Department of Cardiothoracic SurgerySichuan Mianyang 404 HospitalMianyang CitySichuan ProvinceChina
| | - Hang Zeng
- Department of Cardiothoracic SurgerySichuan Mianyang 404 HospitalMianyang CitySichuan ProvinceChina
| | - Dachun Wang
- Department of Cardiothoracic SurgerySichuan Mianyang 404 HospitalMianyang CitySichuan ProvinceChina
| | - Xianyu Wu
- Department of Cardiothoracic SurgerySichuan Mianyang 404 HospitalMianyang CitySichuan ProvinceChina
| | - Guo Xu
- Department of Cardiothoracic SurgerySichuan Mianyang 404 HospitalMianyang CitySichuan ProvinceChina
| |
Collapse
|
21
|
Wang Y, Wang Y, Sun T, Xu J. Bacteriocins in Cancer Treatment: Mechanisms and Clinical Potentials. Biomolecules 2024; 14:831. [PMID: 39062544 PMCID: PMC11274894 DOI: 10.3390/biom14070831] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Cancer poses a severe threat to human health. Although conventional chemotherapy remains a cornerstone of cancer treatment, its significant side effects and the growing issue of drug resistance necessitate the urgent search for more efficient and less toxic anticancer drugs. In recent years, bacteriocins, antimicrobial peptides of microbial origin, have garnered significant attention due to their targeted antitumor activity. This unique activity is mainly attributed to their cationic and amphiphilic nature, which enables bacteriocins to specifically kill tumor cells without harming normal cells. When involving non-membrane-disrupting mechanisms, such as apoptosis induction, cell cycle blockade, and metastasis inhibition, the core mechanism of action is achieved by disrupting cell membranes, which endows bacteriocins with low drug resistance and high selectivity. However, the susceptibility of bacteriocins to hydrolysis and hemolysis in vivo limits their clinical application. To overcome these challenges, structural optimization of bacteriocins or their combination with nanotechnology is proposed for future development. This review aims to study the mechanism of action and current research status of bacteriocins as anticancer treatments, thus providing new insights for their clinical development and application.
Collapse
Affiliation(s)
- Yiwen Wang
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China; (Y.W.); (Y.W.)
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China
| | - Yue Wang
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China; (Y.W.); (Y.W.)
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China
| | - Tao Sun
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China; (Y.W.); (Y.W.)
- Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang 110042, China
| | - Junnan Xu
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China; (Y.W.); (Y.W.)
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China
- Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang 110042, China
| |
Collapse
|
22
|
Nieradko-Iwanicka B, Piasecki J, Borzęcki A. Treatment with bestatin (the exogenous synthetic inhibitor of metalloproteinases) reduces the activity of metalloproteinase 2 and 12 in the spleen and lung tissues of rats in a model of lipopolysaccharide-induced sepsis. Biomed Pharmacother 2024; 174:116480. [PMID: 38547765 DOI: 10.1016/j.biopha.2024.116480] [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: 12/28/2023] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024] Open
Abstract
Sepsis is caused by an inadequate or dysregulated host response to infection. Enzymes causing cellular degradation are matrix metalloproteinases (MMPs). Lipopolysaccharide (LPS) is used in models of sepsis in laboratory settings The aim of the study was to measure MMP 2 and 12 concentrations in spleen and lungs in rats in which septic shock was induced by LPS. The experiment was carried out on 40 male Wistar rats (5 groups of 8): 0. controls 1. administered LPS 2. administered bestatin 3. LPS and bestatin 4.bestatin and after 6 hours LPS Animals were decapitated. Lungs and spleens were collected. Concentrations of MMP-2 and MMP-12 were determined using immunoenzymatic methods. Mean (±SD) MMP-2 in the controls was 43.57 ± 20.53 ng/ml in the lungs and 1.7 ± 0.72 ng/ml in the spleen; Group 1: 31.28 ± 13.13 ng/ml, 0.83 ± 0.8 ng/ml; Group 2: 44.24 ± 22.75 ng /ml, 1.01 ± 0.32 ng/ml; Group 3: 35.94 ± 15.13 ng/ml, 0.41 ± 0.03 ng/ml; Group 4:79.42 ± 44.70 ng/ml, 0.45 ± 0.15, respectively. Mean MMP-12 in controls was 19.79 ± 10.01 ng/ml in lungs and 41.13 ± 15.99 ng/ml in the spleen; Group 1:27.97 ± 15.1 ng/ml; 40.44 ± 11.2 ng/ml; Group 2: 37.93 ± 25.38 ng/ml 41.05 ± 18.08 ng/ml; Group 3: 40.59 ± 11.46 ng/ml, 35.16 ± 12.89 ng/ml; Group 4: 39.4 ± 17.83 ng/ml, 42.04 ± 12.35 ng/ml, respectively. CONCLUSIONS: 1. Bestatin reduces MMP 2 and 12 levels in spleen and lungs. 2. Treatment with bestatin minimizes the effect of LPS.
Collapse
Affiliation(s)
- Barbara Nieradko-Iwanicka
- Hygiene and Epidemiology Department, Medical University of Lublin, Poland Medical University of Lublin, Hygiene and Epidemiology Department, Chodzki 7 Street, Lublin 20-093, Poland.
| | - Jarosław Piasecki
- Doctoral School, Medical University of Lublin, Chodzki 7 Street, Lublin 20-093, Poland
| | - Andrzej Borzęcki
- Hygiene and Epidemiology Department, Medical University of Lublin, Poland Medical University of Lublin, Hygiene and Epidemiology Department, Chodzki 7 Street, Lublin 20-093, Poland
| |
Collapse
|
23
|
Zuo Q, Xu DQ, Yue SJ, Fu RJ, Tang YP. Chemical Composition, Pharmacological Effects and Clinical Applications of Cinobufacini. Chin J Integr Med 2024; 30:366-378. [PMID: 38212503 DOI: 10.1007/s11655-024-3708-6] [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: 08/09/2023] [Indexed: 01/13/2024]
Abstract
Chinese medicine cinobufacini is an extract from the dried skin of Bufo bufo gargarizans Cantor, with active ingredients of bufadienolides and indole alkaloids. With further research and clinical applications, it is found that cinobufacini alone or in combination with other therapeutic methods can play an anti-tumor role by controlling proliferation of tumor cells, promoting apoptosis, inhibiting formation of tumor neovascularization, reversing multidrug resistance, and regulating immune response; it also has the functions of relieving cancer pain and regulating immune function. In this paper, the chemical composition, pharmacological effects, clinical applications, and adverse reactions of cinobufacini are summarized. However, the extraction of monomer components of cinobufacini, the relationship between different mechanisms, and the causes of adverse reactions need to be further studied. Also, high-quality clinical studies should be conducted.
Collapse
Affiliation(s)
- Qian Zuo
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Ding-Qiao Xu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Shi-Jun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Rui-Jia Fu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi'an, 712046, China.
| |
Collapse
|
24
|
Akinsipe T, Mohamedelhassan R, Akinpelu A, Pondugula SR, Mistriotis P, Avila LA, Suryawanshi A. Cellular interactions in tumor microenvironment during breast cancer progression: new frontiers and implications for novel therapeutics. Front Immunol 2024; 15:1302587. [PMID: 38533507 PMCID: PMC10963559 DOI: 10.3389/fimmu.2024.1302587] [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: 09/26/2023] [Accepted: 02/16/2024] [Indexed: 03/28/2024] Open
Abstract
The breast cancer tumor microenvironment (TME) is dynamic, with various immune and non-immune cells interacting to regulate tumor progression and anti-tumor immunity. It is now evident that the cells within the TME significantly contribute to breast cancer progression and resistance to various conventional and newly developed anti-tumor therapies. Both immune and non-immune cells in the TME play critical roles in tumor onset, uncontrolled proliferation, metastasis, immune evasion, and resistance to anti-tumor therapies. Consequently, molecular and cellular components of breast TME have emerged as promising therapeutic targets for developing novel treatments. The breast TME primarily comprises cancer cells, stromal cells, vasculature, and infiltrating immune cells. Currently, numerous clinical trials targeting specific TME components of breast cancer are underway. However, the complexity of the TME and its impact on the evasion of anti-tumor immunity necessitate further research to develop novel and improved breast cancer therapies. The multifaceted nature of breast TME cells arises from their phenotypic and functional plasticity, which endows them with both pro and anti-tumor roles during tumor progression. In this review, we discuss current understanding and recent advances in the pro and anti-tumoral functions of TME cells and their implications for developing safe and effective therapies to control breast cancer progress.
Collapse
Affiliation(s)
- Tosin Akinsipe
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, United States
| | - Rania Mohamedelhassan
- Department of Chemical Engineering, College of Engineering, Auburn University, Auburn, AL, United States
| | - Ayuba Akinpelu
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Satyanarayana R. Pondugula
- Department of Chemical Engineering, College of Engineering, Auburn University, Auburn, AL, United States
| | - Panagiotis Mistriotis
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - L. Adriana Avila
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, United States
| | - Amol Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
25
|
Mi T, Zhang Z, Zhanghuang C, Jin L, Tan X, Liu J, Wu X, Li M, Wang J, Wang Z, Guo P, He D. Doxycycline hydrochloride inhibits the progress of malignant rhabdoid tumor of kidney by targeting MMP17 and MMP1 through PI3K-Akt signaling pathway. Eur J Pharmacol 2024; 964:176291. [PMID: 38158115 DOI: 10.1016/j.ejphar.2023.176291] [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: 08/12/2023] [Revised: 12/09/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE To identify therapeutic targets for malignant rhabdoid tumors of kidney (MRTK) and to investigate the effects and underlying mechanism of doxycycline hydrochloride on these tumors. METHODS Gene expression and clinical data of MRTK were retrieved from the TARGET database. Differentially expressed genes (DEGs) and prognostic-related genes (PRGs) were selected through a combination of statistical analyses. The functional roles of MMP17 and MMP1 were elucidated through RNA overexpression and intervention experiments. Furthermore, in vitro and in vivo studies provided evidence for the inhibitory effect of doxycycline hydrochloride on MRTK. Additionally, transcriptome sequencing was employed to investigate the underlying molecular mechanisms. RESULTS 3507 DEGs and 690 PRGs in MRTK were identified. Among these, we focused on 41 highly expressed genes associated with poor prognosis and revealed their involvement in extracellular matrix regulatory pathways. Notably, MMP17 and MMP1 stood out as particularly influential genes. When these genes were knocked out, a significant inhibition of proliferation, invasion and migration was observed in G401 cells. Furthermore, our study explored the impact of the matrix metalloproteinase inhibitor, doxycycline hydrochloride, on the malignant progression of G401 both in vitro and in vivo. Combined with sequencing data, the results indicated that doxycycline hydrochloride effectively inhibited MRTK progression, due to its ability to suppress the expression of MMP17 and MMP1 through the PI3K-Akt signaling pathway. CONCLUSION Doxycycline hydrochloride inhibits the expression of MMP17 and MMP1 through the PI3K-Akt signaling pathway, thereby inhibiting the malignant progression of MRTK in vivo and in vitro.
Collapse
Affiliation(s)
- Tao Mi
- Children's Urogenital Development and Tissue Engineering Chongqing Key Laboratory, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China
| | - Zhaoxia Zhang
- Children's Urogenital Development and Tissue Engineering Chongqing Key Laboratory, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China
| | - Chenghao Zhanghuang
- Children's Urogenital Development and Tissue Engineering Chongqing Key Laboratory, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China
| | - Liming Jin
- Children's Urogenital Development and Tissue Engineering Chongqing Key Laboratory, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China
| | - Xiaojun Tan
- Children's Urogenital Development and Tissue Engineering Chongqing Key Laboratory, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China
| | - Jiayan Liu
- Children's Urogenital Development and Tissue Engineering Chongqing Key Laboratory, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China
| | - Xin Wu
- Children's Urogenital Development and Tissue Engineering Chongqing Key Laboratory, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China
| | - Mujie Li
- Children's Urogenital Development and Tissue Engineering Chongqing Key Laboratory, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China
| | - Jinkui Wang
- Children's Urogenital Development and Tissue Engineering Chongqing Key Laboratory, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China
| | - Zhang Wang
- Children's Urogenital Development and Tissue Engineering Chongqing Key Laboratory, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China
| | - Peng Guo
- Children's Urogenital Development and Tissue Engineering Chongqing Key Laboratory, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Dawei He
- Children's Urogenital Development and Tissue Engineering Chongqing Key Laboratory, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China; Department of Urology, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China; Key Laboratory of Children's Developmental Diseases Research, Affiliated Children's Hospital of Chongqing Medical University, Ministry of Education, Chongqing, 400014, China; National International Science and Technology Cooperation Base for Major Childhood Developmental Diseases, Children 's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China; National Clinical Research Center for Child Health and Diseases, Children 's Hospital Affiliated to Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
26
|
Liu H, Zheng Q, Li M, Kou J, Wei J, Feng W. Dose-dependent bidirectional pharmacological effects of vinorelbine-based metronomic combination chemotherapy on tumor growth and metastasis and mechanisms in melanoma mouse model. Fundam Clin Pharmacol 2024; 38:99-112. [PMID: 37458143 DOI: 10.1111/fcp.12939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/25/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND There is evidence that the empirical setting of doses and schedules of antineoplastic agents in metronomic chemotherapy (MC) might lead to undesirable outcomes, such as promoting tumor growth or metastasis at certain low doses. However, details about the dose effect of antineoplastic agents in MC have not been fully known yet. OBJECTIVES Vinorelbine combined with cisplatin or fluorouracil (VNR/CDDP or VNR/FU) was selected to investigate its effects on tumor growth or metastasis as well as mechanisms. METHODS Experimental techniques, including immunohistochemistry, western blot, immunofluorescence, and flow cytometry, were used to explore the mechanisms, along with cell proliferation, apoptosis, migration, and invasion. RESULTS The results showed that VNR/CDDP or VNR/FU promoted tumor growth and metastasis at low doses and inhibited them at high ones. Except that expressions of apoptotic proteins were elevated at both low and high doses, low-dose treatments enhanced angiogenesis and promoted the mobilization and recruitment of myeloid-derived suppressor cells (MDSCs), while high-dose treatments reversed these effects. Additionally, low concentrations of VNR/CDDP or VNR/FU stimulated tumor cell functions such as anti-apoptosis, migration, and invasion, but high concentrations only suppressed cell proliferation and increased apoptosis. CONCLUSION This study elucidated a bidirectional action mode regulated by multiple mechanisms at different doses in MC and also highlighted the risks of low-dose metronomic administration of antineoplastic agents in the clinic. More preclinical and clinical studies focusing on the dose-effect of metronomic regimens are urgently needed because an effective therapeutic regimen should be an optimal setting of drugs, doses, schedules, or combinations.
Collapse
Affiliation(s)
- Hua Liu
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qiaowei Zheng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Min Li
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jianrong Kou
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Junsong Wei
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Weiyi Feng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| |
Collapse
|
27
|
Chen Y, Zhu D, Zhong H, Gan Z, Zong S, Wang Z, Cui Y, Wang Y. Ultrasensitive Detection of Matrix Metalloproteinase 2 Activity Using a Ratiometric Surface-Enhanced Raman Scattering Nanosensor with a Core-Satellite Structure. ACS APPLIED MATERIALS & INTERFACES 2024; 16:4160-4168. [PMID: 38204415 DOI: 10.1021/acsami.3c15344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Matrix metalloproteinase 2 (MMP-2) has been considered a promising molecular biomarker for cancer diagnosis due to its related dysregulation. In this work, a core-satellite structure-powered ratiometric surface-enhanced Raman scattering (SERS) nanosensor with high sensitivity and specificity to MMP-2 was developed. The SERS nanosensor was composed of a magnetic bead encapsulated within a 5,5'-dithiobis(2-nitrobenzoic acid) (DTNB)-labeled gold shell as the capture core and a 4-mercaptobenzonitrile (MBN)-encoded silver nanoparticle as the signal satellite, which were connected through a peptide substrate of MMP-2. MMP-2-triggered cleavage of peptides from the core surface resulted in a decrease of the SERS intensity of MBN. Since the SERS intensity of DTNB was used as an internal standard, the reliable and sensitive quantification of MMP-2 activity would be realized by the ratiometric SERS signal, with a limit of detection as low as 2.067 ng/mL and a dynamic range from 5 to 100 ng/mL. Importantly, the nanosensor enabled a precise determination of MMP-2 activity in tumor cell secretions, which may provide an avenue for early diagnosis and classification of malignant tumors.
Collapse
Affiliation(s)
- Yingying Chen
- Jiangsu Key Laboratory on Optoelectronic Technology, School of Computer and Electronic Information/School of Artificial Intelligence, Nanjing Normal University, Nanjing 210023, Jiangsu, People's Republic of China
| | - Dan Zhu
- Jiangsu Key Laboratory on Optoelectronic Technology, School of Computer and Electronic Information/School of Artificial Intelligence, Nanjing Normal University, Nanjing 210023, Jiangsu, People's Republic of China
| | - Hanyan Zhong
- Jiangsu Key Laboratory on Optoelectronic Technology, School of Computer and Electronic Information/School of Artificial Intelligence, Nanjing Normal University, Nanjing 210023, Jiangsu, People's Republic of China
| | - Zhixing Gan
- Jiangsu Key Laboratory on Optoelectronic Technology, School of Computer and Electronic Information/School of Artificial Intelligence, Nanjing Normal University, Nanjing 210023, Jiangsu, People's Republic of China
| | - Shenfei Zong
- Advanced Photonics Center, School of Electronic Science and Engineering, Southeast University, Nanjing 210096, Jiangsu, People's Republic of China
| | - Zhuyuan Wang
- Advanced Photonics Center, School of Electronic Science and Engineering, Southeast University, Nanjing 210096, Jiangsu, People's Republic of China
| | - Yiping Cui
- Jiangsu Key Laboratory on Optoelectronic Technology, School of Computer and Electronic Information/School of Artificial Intelligence, Nanjing Normal University, Nanjing 210023, Jiangsu, People's Republic of China
- Advanced Photonics Center, School of Electronic Science and Engineering, Southeast University, Nanjing 210096, Jiangsu, People's Republic of China
| | - Yiping Wang
- Jiangsu Key Laboratory on Optoelectronic Technology, School of Computer and Electronic Information/School of Artificial Intelligence, Nanjing Normal University, Nanjing 210023, Jiangsu, People's Republic of China
| |
Collapse
|
28
|
Abstract
Abstract
Matrix metalloproteinase-2 (MMP-2) is a gelatinase and is involved in multiple steps of the metastatic cascade. More than a decade ago an increased expression of MMP-2 in tumour cells or higher serum levels was reported to be a prognostic biomarker for a lower disease-free and overall survival rate. In recent years new evidence has indicated that MMP-2 has an important role in the tumour ecosystem. It is one of the many players in the onco-sphere, involved in interacting between tumour cells, host cells and the microenvironment. It plays a role in the dissemination of tumour cells, the epithelial–mesenchymal and mesenchymal–epithelial transitions, the formation of the pre-metastatic and metastatic niches, dormancy of tumour cells and modulating the immune system. The aim of this review is to highlight these multiple roles in the metastatic cascade and how many signalling pathways can up or down-regulate MMP-2 activity in the different stages of cancer progression and the effect of MMP-2 on the onco-sphere. Research in head and neck cancer is used as an example of these processes. The use of non-specific MMP inhibitors has been unsuccessful showing only limited benefits and associated with high toxicity as such that none have progressed past Phase III trials. Preclinical trials are undergoing using antibodies directed against specific matrix metalloproteinases, these targeted therapies may be potentially less toxic to the patients.
Collapse
Affiliation(s)
- Nigel P. Murray
- Minimal Residual Disease Laboratory, Faculty of Medicine , University Finis Terrae , Santiago , Chile
- Department of Haematology , Hospital de Carabineros de Chile , Santiago , Chile
| |
Collapse
|
29
|
Shan Y, Zheng L, Zhang S, Qian B. Abnormal expression of FOXM1 in carcinogenesis of renal cell carcinoma: From experimental findings to clinical applications. Biochem Biophys Res Commun 2024; 692:149251. [PMID: 38056162 DOI: 10.1016/j.bbrc.2023.149251] [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: 09/12/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 12/08/2023]
Abstract
Renal cell carcinoma (RCC) is a prevalent malignancy within the genitourinary system. At present, patients with high-grade or advanced RCC continue to have a bleak prognosis. Mounting research have emphasized the significant involvement of Forkhead box M1 (FOXM1) in RCC development and progression. Therefore, it is imperative to consolidate the existing evidence regarding the contributions of FOXM1 to RCC tumorigenesis through a comprehensive review. This study elucidated the essential functions of FOXM1 in promoting RCC growth, invasion, and metastasis by regulating cell cycle progression, DNA repair, angiogenesis, and epithelial-mesenchymal transition (EMT). Also, FOXM1 might serve as a novel diagnostic and prognostic biomarker as well as a therapeutic target for RCC. Clinical findings demonstrated that the expression of FOXM1 was markedly upregulated in RCC samples, while a high level of FOXM1 was found to be associated with a poor overall survival rate of RCC. Furthermore, it is worth noting that FOXM1 may have a significant impact on the resistance of renal cell carcinoma (RCC) to radiotherapy. This observation suggests that inhibiting FOXM1 could be a promising strategy to impede the progression of RCC and enhance its sensitivity to radiotherapy. The present review highlighted the pivotal role of FOXM1 in RCC development. FOXM1 has the capacity to emerge as not only a valuable diagnostic and prognostic tool but also a viable therapeutic option for unresectable RCC.
Collapse
Affiliation(s)
- Yanmei Shan
- Department of Nephrology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
| | - Liying Zheng
- Postgraduate Department, First Affiliated Hospital of Gannan Medical College, Ganzhou, China
| | - Shilong Zhang
- Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Biao Qian
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China; Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, 341000, Jiangxi, China
| |
Collapse
|
30
|
Cai J, Jiang Y, Chen P, Liang J, Zhang Y, Yuan R, Fan H, Zhong Y, Cai J, Cheng S, Zhang Y. TBC1D1 represses glioma progression by altering the integrity of the cytoskeleton. Aging (Albany NY) 2024; 16:431-444. [PMID: 38189823 PMCID: PMC10817367 DOI: 10.18632/aging.205377] [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: 03/16/2023] [Accepted: 11/20/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Glioma is one of the most aggressive malignant brain tumors and is characterized by invasive growth and poor prognosis. TBC1D1, a member of the TBC family, is associated with the development of various malignancies. However, the role of TBC1D1 in glioma-genesis remains unclear. METHODS The effect of TBC1D1 on the prognosis of glioma patients and related influencing factors were analyzed in the Chinese Glioma Genome Atlas (CGGA) and The Cancer Genome Atlas (TCGA) databases. Expression of TBC1D1 in glioma cell lines was detected by western blotting. Cell viability and proliferation were measured by EdU and Colony formation assays, respectively. Transwell and wound healing assays were performed to determine the cell migration and invasion capacities. Immunofluorescence was used to observe actin morphology in the cytoskeleton. RESULTS We discovered that high TBC1D1 expression in gliomas led to poor prognosis. Downregulation of TBC1D1 in glioma cells significantly inhibited multiple important functions, such as proliferation, migration, and invasion. We further demonstrated that the tumor-inhibitory effect of TBC1D1 might occur through the P-LIMK/cofilin pathway, destroying the cytoskeletal structure and affecting the depolymerization of F-actin, thereby inhibiting glioma migration. CONCLUSION TBC1D1 affects the balance and integrity of the actin cytoskeleton via cofilin, thereby altering the morphology and aggressiveness of glioma cells. This study provides a new perspective on its role in tumorigenesis, thereby identifying a potential therapeutic target for the treatment of gliomas.
Collapse
Affiliation(s)
- Jiahong Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi, China
| | - Yong’an Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi, China
| | - Peng Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi, China
| | - Jiawei Liang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi, China
| | - Yi Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi, China
| | - Raorao Yuan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi, China
| | - Hengyi Fan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi, China
| | - Yuefei Zhong
- Department of Neurology, Shang Rao GuangXin District People’s Hospital, Shangrao 334100, Jiangxi, China
| | - Jianhui Cai
- Department of Neurosurgery, Nanchang County People’s Hospital, Nanchang 330200, Jiangxi, China
| | - Shiqi Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi, China
| | - Yan Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi, China
| |
Collapse
|
31
|
Wang X, Jing H. Anticancer Potential of ACEIs/ARBs Administration in Colorectal Cancer. Curr Med Chem 2024; 31:4867-4879. [PMID: 38549531 DOI: 10.2174/0109298673249782231226101119] [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: 03/22/2023] [Revised: 10/03/2023] [Accepted: 10/20/2023] [Indexed: 05/30/2024]
Abstract
BACKGROUND Colorectal cancer (CC) is the fourth most common type of cancer that causes illness and death. Medicines like ACE inhibitors and ARBs, usually used for heart problems, have shown they might help with the growth and development of CC. INTRODUCTION An analysis of ACE inhibitors and colon cancer is conducted in this comprehensive review. The main goal is to see how ACEIs/ARBs affect the chances of getting cancer and dying in patients with CC. METHODS A systematic literature search was conducted to identify relevant studies. Inclusion criteria encompassed studies that evaluated the use of ACEIs/ARBs in patients with CC and reported outcomes related to new cancer incidence and mortality. Data from selected studies were extracted and analyzed using appropriate statistical methods. RESULTS The study showed that fewer cancer cases occurred in patients who took ACEIs/ARBs compared to those who did not (RR 0.962, 95% CI 0.934-0.991, p = 0.010). Furthermore, patients with CC who utilized ACEIs/ARBs exhibited a decreased mortality rate compared to non-users (HR 0.833, 95% CI 0.640-1.085, p = 0.175). CONCLUSION This review suggests that using ACEIs/ARBs medicine could help people with CC live longer and lower their chances of dying. These results highlight the potential benefits of utilizing ACE inhibitors in the management of CC, warranting further investigation and consideration in clinical practice.
Collapse
Affiliation(s)
- Xin Wang
- Department of Cardiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| | - Haiyun Jing
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou, 450000, Henan, People's Republic of China
| |
Collapse
|
32
|
Fujimura T, Ohuchi K, Ikawa T, Kambayashi Y, Amagai R, Furudate S, Asano Y. Possible effects of plasminogen activator inhibitor-1 on promoting angiogenesis through matrix metalloproteinase 9 in advanced mycosis fungoides. Hematol Oncol 2024; 42:e3244. [PMID: 38287534 DOI: 10.1002/hon.3244] [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: 04/28/2023] [Revised: 09/01/2023] [Accepted: 12/07/2023] [Indexed: 01/31/2024]
Abstract
Mycosis fungoides (MF) progresses slowly before advancing to skin tumors followed by lymph node and visceral involvement. Among MF progression, stage IIB is an initial time point of tumor formation in MF. Since MF in tumor stage possess abundant blood vessels, it is important to evaluate the pro-angiogenic factors before and after MF in stage IIB. In this report, we investigated pro-angiogenic soluble factors in MF patients, as well as its pro-angiogenetic effects on tumor cells and stroma cells. We first evaluated the serum levels of pro-angiogenic factors in 9 MF patients without tumor formation and 8 MF patients with tumor formation. Among them, the serum MMP-9 and plasminogen activator inhibitors 1 (PAI-1) was significantly increased in MF with tumor formation compared in MF without tumor formation, leading to favorable formation of human dermal microvascular endothelial cells tube networks. Moreover, PAI-1 stimulation significantly increased the mRNA expression and protein production MMP-9 on monocytes derived M2 macrophages and HUT-78. Furthermore, since MMP-9 production from tumor cells as well as stromal cells is suppressed by bexarotene, we evaluate the baseline serum pro-angiogenic factors including MMP-9 in 16 patients with advanced cutaneous T cell lymphoma treated with bexarotene. The serum levels of MMP-2 and MMP-9 was significantly increased in bexarotene non-responded patients compared to responded patients. Our present study suggested the significance of MMP-9 and PAI-1 for the progression of MF stage toward to the tumor stage, and could be a therapeutic target in future.
Collapse
Affiliation(s)
- Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kentaro Ohuchi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuya Ikawa
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yumi Kambayashi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryo Amagai
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sadanori Furudate
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshihide Asano
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
33
|
Zhong J, Pan R, Gao M, Mo Y, Peng X, Liang G, Chen Z, Du J, Huang Z. Identification and validation of a T cell marker gene-based signature to predict prognosis and immunotherapy response in gastric cancer. Sci Rep 2023; 13:21357. [PMID: 38049463 PMCID: PMC10696024 DOI: 10.1038/s41598-023-48930-8] [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: 05/09/2023] [Accepted: 12/01/2023] [Indexed: 12/06/2023] Open
Abstract
Although the role of T cells in tumor immunity and modulation of the tumor microenvironment (TME) has been extensively studied, their precise involvement in gastric adenocarcinoma remains inadequately explored. In this work, we analyzed the single-cell RNA sequencing data set in GSE183904 and identified 322 T cell marker genes using the "FindAllMarkers" method of the R package "Seurat". STAD patients in the TCGA database were divided into high-risk and low-risk categories based on risk scores. The five-gene prediction signature based on T cell marker genes can predict the prognosis of gastric cancer patients with high accuracy. In the training cohort, the areas under the receiver operating characteristic (ROC) curve were 0.667, 0.73, and 0.818 at 1, 3, and 5 years. External validation of the predictive signature was also performed using multiple clinical subgroups and GEO cohorts. To help with practical application, a diagnostic model was created that shows values of 0.732, 0.752, and 0.816 for the relevant areas under the ROC curve at 1, 3, and 5 years. The T cell marker genes identified in this study may serve as potential therapeutic targets, and the developed predictive signatures and nomograms may aid in the clinical management of gastric cancer.
Collapse
Affiliation(s)
- Jinlin Zhong
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Rongling Pan
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Miao Gao
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Yuqian Mo
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Xin Peng
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Guoxiao Liang
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Zixuan Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Jinlin Du
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Zhigang Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China.
- Key Laboratory of Noncommunicable Diseases Control and Health Data Statistics of Guangdong Medical University, Dongguan, Guangdong, People's Republic of China.
| |
Collapse
|
34
|
Balakina A, Gadomsky S, Kokovina T, Sashenkova T, Mishchenko D, Terentiev A. New Derivatives of N-Hydroxybutanamide: Preparation, MMP Inhibition, Cytotoxicity, and Antitumor Activity. Int J Mol Sci 2023; 24:16360. [PMID: 38003553 PMCID: PMC10671431 DOI: 10.3390/ijms242216360] [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: 09/19/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Using a novel method of N-substituted succinimide ring opening, new N-hydroxybutanamide derivatives were synthesized. These compounds were evaluated for their ability to inhibit matrix metalloproteinases (MMPs) and their cytotoxicity. The iodoaniline derivative of N1-hydroxy-N4-phenylbutanediamide showed the inhibition of MMP-2, MMP-9, and MMP-14 with an IC50 of 1-1.5 μM. All the compounds exhibited low toxicity towards carcinoma cell lines HeLa and HepG2. The iodoaniline derivative was also slightly toxic to glioma cell lines A-172 and U-251 MG. Non-cancerous FetMSC and Vero cells were found to be the least sensitive to all the compounds. In vivo studies demonstrated that the iodoaniline derivative of N1-hydroxy-N4-phenylbutanediamide had low acute toxicity. In a mouse model of B16 melanoma, this compound showed both antitumor and antimetastatic effects, with a 61.5% inhibition of tumor growth and an 88.6% inhibition of metastasis. Our findings suggest that the iodoaniline derivative of N1-hydroxy-N4-phenylbutanediamide has potential as a lead structure for the development of new MMP inhibitors. Our new synthetic approach can be a cost-effective method for the synthesis of inhibitors of metalloenzymes with promising antitumor potential.
Collapse
Affiliation(s)
- Anastasia Balakina
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
| | - Svyatoslav Gadomsky
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
| | - Tatyana Kokovina
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
- Faculty of Fundamental Physical-Chemical Engineering of M.V. Lomonosov MSU, Leninskie Gory, 119991 Moscow, Russia
| | - Tatyana Sashenkova
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
| | - Denis Mishchenko
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
- Faculty of Fundamental Physical-Chemical Engineering of M.V. Lomonosov MSU, Leninskie Gory, 119991 Moscow, Russia
- Scientific and Educational Center in Chernogolovka, State University of Education, 141014 Mytishchi, Russia
| | - Alexei Terentiev
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
- Faculty of Fundamental Physical-Chemical Engineering of M.V. Lomonosov MSU, Leninskie Gory, 119991 Moscow, Russia
- Scientific and Educational Center in Chernogolovka, State University of Education, 141014 Mytishchi, Russia
| |
Collapse
|
35
|
Ji Y, Huang W, Chen Y, Zhang X, Wu F, Tang W, Lu Z, Huang C. Inhibition of MMP-2 and MMP-9 attenuates surgery-induced cognitive impairment in aged mice. Brain Res Bull 2023; 204:110810. [PMID: 37939860 DOI: 10.1016/j.brainresbull.2023.110810] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 10/29/2023] [Accepted: 11/05/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND The inhibition of matrix metalloproteinases (MMPs) has shown potential in the treatment of various neurodegenerative diseases, and perioperative neurocognitive disorders (PND) is accompanied by the increased expression of MMP-2 and MMP-9 in the hippocampus. However, the effect of inhibiting MMP-2 and MMP-9 on PND is not clear. In this study we aimed to evaluate the effects of inhibiting MMP-2 and MMP-9 on cognitive function in the aged mice after surgery, in order to find a possible target for the prevention and treatment of PND METHODS: In this study, 14-month-old C57BL/6 mice were used to establish a PND model by tibial fracture surgery and sevoflurane anesthesia. Three days later, part of the mice were subjected to cognitive assessment and the other was sacrificed for biochemical analysis. We used the Novel object recognition test and Fear conditioning test to evaluate the postoperative cognitive function of mice. The expression of mmp-2 and MMP-9 was detected by western blotting. We also examined the expression of claudin-5 and occludin using Western blotting, and the activation of microglia and astrocytes using immunofluorescence. RESULTS The results showed that surgery increased the expression of MMP-2 and MMP-9 in the hippocampus of mice, accompanied by cognitive impairment, decreased expression of claudin-5 and occludin, and increased activation of microglia and astrocytes. However, inhibition of MMP-2 and MMP-9 expression by SB-3CT reversed these changes. CONCLUSIONS Our study shows that inhibition of MMP-2 and MMP-9 alleviates anesthesia/surgery-induced cognitive decline by increasing BBB integrity and inhibiting glial cell activation.
Collapse
Affiliation(s)
- Yiqin Ji
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, Zhejiang, China
| | - Wanbo Huang
- Ningbo University Health Science Center, Ningbo 315211, Zhejiang, China
| | - Yijun Chen
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, Zhejiang, China
| | - Xincai Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, Zhejiang, China
| | - Fan Wu
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, Zhejiang, China
| | - Wan Tang
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, Zhejiang, China
| | - Zihui Lu
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, Zhejiang, China
| | - Changshun Huang
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, Zhejiang, China.
| |
Collapse
|
36
|
Rezaie H, Alipanah-Moghadam R, Jeddi F, Clark CCT, Aghamohammadi V, Nemati A. Combined dandelion extract and all-trans retinoic acid induces cytotoxicity in human breast cancer cells. Sci Rep 2023; 13:15074. [PMID: 37700002 PMCID: PMC10497591 DOI: 10.1038/s41598-023-42177-z] [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: 04/03/2023] [Accepted: 09/06/2023] [Indexed: 09/14/2023] Open
Abstract
Breast cancer is one of the most prevalent and deadly cancers among women worldwide. Recently, natural compounds have been widely used for the treatment of breast cancer. Present study evaluated antiproliferative and anti-metastasis activities of two natural compounds of dandelion and all-trans-retinoic acid (ATRA) in human MCF-7 and MDA-MB231 breast cancer cells. We also evaluated the expression of MMP-2, MMP-9, IL-1β, p53, NM23 and KAI1 genes. Data showed a clear additive cytotoxic effect in concentrations of 40 μM ATRA with 1.5 and 4 mg/ml of dandelion extract in MCF-7 and MDA-MB231 cells, respectively. In both cell lines, compared with the untreated cells, the expression levels of MMP-9 and IL-1β were significantly decreased while p53 and KAI1 expression levels were increased. Besides, MMP-2 and NM23 had different expressions in the two studied cell lines. In conclusion, dandelion/ATRA co-treatment, in addition to having strong cytotoxic effects, has putative effects on the expression of anti-metastatic genes in both breast cancer cells.
Collapse
Affiliation(s)
- Hamed Rezaie
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Reza Alipanah-Moghadam
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Farhad Jeddi
- Department of Genetics and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Cain C T Clark
- Centre for Intelligent Healthcare, Coventry University, Coventry, CV1 5FB, UK
| | | | - Ali Nemati
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
37
|
Almutairi S, Kalloush HM, Manoon NA, Bardaweel SK. Matrix Metalloproteinases Inhibitors in Cancer Treatment: An Updated Review (2013-2023). Molecules 2023; 28:5567. [PMID: 37513440 PMCID: PMC10384300 DOI: 10.3390/molecules28145567] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/09/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are identifiable members of proteolytic enzymes that can degrade a wide range of proteins in the extracellular matrix (ECM). MMPs can be categorized into six groups based on their substrate specificity and structural differences: collagenases, gelatinases, stromelysins, matrilysins, metalloelastase, and membrane-type MMPs. MMPs have been linked to a wide variety of biological processes, such as cell transformation and carcinogenesis. Over time, MMPs have been evaluated for their role in cancer progression, migration, and metastasis. Accordingly, various MMPs have become attractive therapeutic targets for anticancer drug development. The first generations of broad-spectrum MMP inhibitors displayed effective inhibitory activities but failed in clinical trials due to poor selectivity. Thanks to the evolution of X-ray crystallography, NMR analysis, and homology modeling studies, it has been possible to characterize the active sites of various MMPs and, consequently, to develop more selective, second-generation MMP inhibitors. In this review, we summarize the computational and synthesis approaches used in the development of MMP inhibitors and their evaluation as potential anticancer agents.
Collapse
Affiliation(s)
- Shriefa Almutairi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan
| | - Hanin Moh'd Kalloush
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Nour A Manoon
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan
| | - Sanaa K Bardaweel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan
| |
Collapse
|
38
|
Amin SA, Khatun S, Gayen S, Das S, Jha T. Are inhibitors of histone deacetylase 8 (HDAC8) effective in hematological cancers especially acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL)? Eur J Med Chem 2023; 258:115594. [PMID: 37429084 DOI: 10.1016/j.ejmech.2023.115594] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 07/12/2023]
Abstract
Histone deacetylase 8 (HDAC8) aberrantly deacetylates histone and non-histone proteins. These include structural maintenance of chromosome 3 (SMC3) cohesin protein, retinoic acid induced 1 (RAI1), p53, etc and thus, regulating diverse processes such as leukemic stem cell (LSC) transformation and maintenance. HDAC8, one of the crucial HDACs, affects the gene silencing process in solid and hematological cancer progressions especially on acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL). A specific HDAC8 inhibitor PCI-34051 showed promising results against both T-cell lymphoma and AML. Here, we summarize the role of HDAC8 in hematological malignancies, especially in AML and ALL. This article also introduces the structure/function of HDAC8 and a special attention has been paid to address the HDAC8 enzyme selectivity issue in hematological cancer especially against AML and ALL.
Collapse
Affiliation(s)
- Sk Abdul Amin
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India; Department of Pharmaceutical Technology, JIS University, 81, Nilgunj Road, Agarpara, Kolkata, West Bengal, India.
| | - Samima Khatun
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Shovanlal Gayen
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | - Sanjib Das
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| |
Collapse
|
39
|
Surówka A, Prowans P, Żołnierczuk M, Miśkiewicz M, Wawrowski T, Skodda M, Markowska M, Kędzierska-Kapuza K. The Effect of Calcineurin Inhibitors on MMPs Activity in Heart and Their Side Effects-A Review of Literature. Int J Mol Sci 2023; 24:10291. [PMID: 37373446 DOI: 10.3390/ijms241210291] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
This review focuses on the role of metalloproteinases in the pathogenesis of myocardial injury in various disease entities. It reveals how the expression and serum levels of metalloproteinases and their inhibitors change in many disease states. At the same time, the study offers a review of the impact of immunosuppressive treatment on this relationship. Modern immunosuppressive treatment is based mainly on the use of calcineurin inhibitors, including cyclosporine A and tacrolimus. The use of these drugs may carry a number of side effects, specifically to the cardiovascular system. The scale and degree of long-term influence on the organism remains unclear, but a significant risk of complications for transplant recipients who take immunosuppressive drugs as part of their daily treatment is to be expected. Therefore, the knowledge on this subject should be expanded and the negative effects of post-transplant therapy minimized. Immunosuppressive therapy plays an important role in the expression and activation of tissue metalloproteinases and their specific inhibitors, which leads to many tissue changes. The presented study is a collection of research results on the effects of calcineurin inhibitors on the heart, with particular emphasis placed on the participation of MMP-2 and MMP-9. It is also an analysis of the effects of specific heart diseases on myocardial remodeling through inductive or inhibitory effects on matrix metalloproteinases and their inhibitors.
Collapse
Affiliation(s)
- Anna Surówka
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Piotr Prowans
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Michał Żołnierczuk
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Marek Miśkiewicz
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Tomasz Wawrowski
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Marika Skodda
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Marta Markowska
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
- Department of Plastic and Reconstructive Surgery, 109 Military Hospital, 70-111 Szczecin, Poland
| | - Karolina Kędzierska-Kapuza
- Department of Gastroenterological Surgery and Transplantology, National Medical Institute of the Ministry of Interior Affairs and Administration, 02-507 Warsaw, Poland
- Department of Gastroenterological Surgery and Transplantology, Centre of Postgraduate, Medical Education in Warsaw, 02-507 Warsaw, Poland
| |
Collapse
|
40
|
Zhu X, Wu X, Yang H, Xu Q, Zhang M, Liu X, Lv K. m 6A-mediated upregulation of LINC01003 regulates cell migration by targeting the CAV1/FAK signaling pathway in glioma. Biol Direct 2023; 18:27. [PMID: 37270527 DOI: 10.1186/s13062-023-00386-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 05/30/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) play important roles in the progression of glioma. Here, we examined the potential functions of a lncRNA, LINC01003, in glioma and characterized the underlying molecular mechanisms. METHODS The GEIPA2 and Chinese Glioma Genome Atlas (CCGA) databases were employed to analyze gene expression and the overall survival curve in patients with glioma. The functions of LINC01003 in glioma growth and migration were assessed by loss-of-function experiments in vitro and in vivo. RNA sequencing was used to determine the signaling pathways effected by LINC01003. Bioinformatics analysis and RNA immunoprecipitation (RIP) assays were used to explore the mechanism underlying the N6-methyladenine (m6A) modification-dependent upregulation of LINC01003 in glioma. RESULTS LINC01003 expression was upregulated in glioma cell lines and tissues. Higher LINC01003 expression predicted shorter overall survival time in glioma patients. Functionally, LINC01003 knockdown inhibited the cell cycle and cell proliferation and migration in glioma cells. Mechanistically, RNA sequencing revealed that LINC01003 mediated the focal adhesion signaling pathway. Furthermore, LINC01003 upregulation is induced by m6A modification regulated by METTL3. CONCLUSION This study characterized LINC01003 as a lncRNA that contributes to tumorigenesis in glioma and demonstrated that the LINC01003-CAV1-FAK axis serves as a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Xiaolong Zhu
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, People's Republic of China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, 241001, People's Republic of China
- Non-Coding RNA Research Center of Wannan Medical College, Wuhu, 241001, People's Republic of China
- Anhui Provincial Clinical Research Center for Critical Respiratory Disease, Wuhu, 241001, People's Republic of China
| | - Xingwei Wu
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, People's Republic of China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, 241001, People's Republic of China
- Non-Coding RNA Research Center of Wannan Medical College, Wuhu, 241001, People's Republic of China
| | - Hui Yang
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, People's Republic of China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, 241001, People's Republic of China
- Non-Coding RNA Research Center of Wannan Medical College, Wuhu, 241001, People's Republic of China
- Anhui Provincial Clinical Research Center for Critical Respiratory Disease, Wuhu, 241001, People's Republic of China
| | - Qiancheng Xu
- Anhui Provincial Clinical Research Center for Critical Respiratory Disease, Wuhu, 241001, People's Republic of China
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241001, People's Republic of China
| | - Mengying Zhang
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, People's Republic of China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, 241001, People's Republic of China
- Non-Coding RNA Research Center of Wannan Medical College, Wuhu, 241001, People's Republic of China
- Anhui Provincial Clinical Research Center for Critical Respiratory Disease, Wuhu, 241001, People's Republic of China
| | - Xiaocen Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, People's Republic of China
| | - Kun Lv
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, People's Republic of China.
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, 241001, People's Republic of China.
- Non-Coding RNA Research Center of Wannan Medical College, Wuhu, 241001, People's Republic of China.
- Anhui Provincial Clinical Research Center for Critical Respiratory Disease, Wuhu, 241001, People's Republic of China.
| |
Collapse
|
41
|
Singh AK, Kumar S. Naringin dihydrochalcone potentially binds to catalytic domain of matrix metalloproteinase-2: molecular docking, MM-GBSA, and molecular dynamics simulation approach. Nat Prod Res 2023; 37:1851-1855. [PMID: 36047975 DOI: 10.1080/14786419.2022.2118746] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 10/14/2022]
Abstract
Matrix metalloproteinase-2 (MMP2), an extracellular matrix remodulating protein's increased activity causes cancer-metastasis. Potential MMP2 inhibitors showed sever side-effects in clinical trials. Present study is focused on identification natural MMP2 inhibitor by applying molecular docking, MM-GBSA binding energy estimation and molecular dynamics (MD) simulations. Commercially available flavonoid compound library was used to screen the molecules potentially binding with catalytic domain of MMP2 protein compared to standard MMP2 inhibitor ARP100. Naringin dihydrochalcone (NDC) showed interaction with the important residues (His120, Leu82 and Val117) present at the MMP2 catalytic domain in comparison to known inhibitor ARP100 (dock score ≈ -13 and -8 kcal/mole respectively). Lower ligand-protein binding energy (-67.31 kcal/mole) obtained in MM-GBSA and the MD simulation trajectory analysis showed significant stable and energetically favourable binding of NDC at the catalytic site of MMP2. In conclusion, anti-metastatic potential of NDC should be validated in in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Atul Kumar Singh
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, Punjab, India
| | - Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
42
|
Xu HB, Chen XZ, Wang X, Pan J, Yi-Zhuo Z, Zhou CH. Xihuang pill in the treatment of cancer: TCM theories, pharmacological activities, chemical compounds and clinical applications. JOURNAL OF ETHNOPHARMACOLOGY 2023:116699. [PMID: 37257709 DOI: 10.1016/j.jep.2023.116699] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/17/2023] [Accepted: 05/28/2023] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xihuang pill as a famous traditional Chinese formula has long been used as an adjuvant therapy for cancer. AIM OF THE STUDY This study is aimed at summarizing recent advances in research of Xihuang pill's anti-cancer efficacies from the theoretical basis of traditional Chinese medicine, pharmacological activities, chemical components and its clinical application. MATERIALS AND METHODS The literature information was obtained from several authoritative databases including PubMed, Embase, Cochrane Library, CNKI and Wan Fang before April 30, 2023. We also analyzed the representatively chemical compounds of Xihuang pill in vivo experiments using HPLC-Q/TOF-MS. RESULTS The present study indicated that Xihuang pill, a classic anti-tumor prescription, had efficacies of strengthening body resistance, clearing heat and detoxification, and promoting blood circulation for removing blood stasis. Modern basic researches showed that Xihuang pill played anti-cancer roles through inducing cancer cell apoptosis, inhibiting cell proliferation, migration, invasion and angiogenesis, improving immune function and tumor microenvironment, and regulating related signaling pathways. Its chemical components are primarily consisted of amino acids, terpenoids, fatty acids, fatty acid esters, phenolics, bile acids, bile pigments and volatile oil. Clinically, Xihuang pill, as an adjuvant drug for cancer treatment, was mostly combined with chemotherapy, which could prolong survival, enhance response rate, improve patients' life quality, regulate immune function and alleviate chemotherapy-induced toxicities. CONCLUSIONS This present study suggests that Xihuang pill may be a promising adjuvant therapy for cancer, and proposes the possibility of future research directions for Xihuang pill based on the current research status.
Collapse
Affiliation(s)
- Hong-Bin Xu
- Department of Pharmacy, Ningbo First Hospital, Ningbo University, Ningbo, China; Department of Pharmacy, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Xian-Zhen Chen
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xia Wang
- Department of Oncology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Pan
- Department of Pharmacy, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhao Yi-Zhuo
- Department of Pharmacy, Ningbo First Hospital, Ningbo University, Ningbo, China
| | - Chen-Hui Zhou
- Department of Neurosurgery, Ningbo First Hospital, Ningbo University, Ningbo, China
| |
Collapse
|
43
|
Recent progress in nanocarrier-based drug delivery systems for antitumour metastasis. Eur J Med Chem 2023; 252:115259. [PMID: 36934485 DOI: 10.1016/j.ejmech.2023.115259] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023]
Abstract
Tumour metastasis is one of the major factors leading to poor prognosis as well as lower survival among cancer patients. A number of studies investigating the inhibition of tumour metastasis have been conducted. It is difficult to achieve satisfactory results with surgery alone for distant metastatic tumours, and chemotherapy can boost the healing rate and prognosis of patients. However, the poor therapeutic efficacy of chemotherapy drugs due to their low solubility, lack of tumour targeting, instability in vivo, high toxicity and multidrug resistance hinder their application. Immunotherapy is beneficial to the treatment of metastatic cancers, but it also has disadvantages such as adverse reactions and acquired resistance. Fortunately, delivery of chemotherapeutic drugs with nanocarriers can reduce systemic reactions caused by chemotherapeutic agents and inhibit metastasis. This review discusses the underlying mechanisms of metastasis, therapeutic approaches for antitumour metastasis, the advantages of nanodrug delivery systems and their application in reducing metastasis.
Collapse
|
44
|
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S, Xiao M. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer 2023; 22:48. [PMID: 36906534 PMCID: PMC10007858 DOI: 10.1186/s12943-023-01744-8] [Citation(s) in RCA: 326] [Impact Index Per Article: 163.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/11/2023] [Indexed: 03/13/2023] Open
Abstract
The malignant tumor is a multi-etiological, systemic and complex disease characterized by uncontrolled cell proliferation and distant metastasis. Anticancer treatments including adjuvant therapies and targeted therapies are effective in eliminating cancer cells but in a limited number of patients. Increasing evidence suggests that the extracellular matrix (ECM) plays an important role in tumor development through changes in macromolecule components, degradation enzymes and stiffness. These variations are under the control of cellular components in tumor tissue via the aberrant activation of signaling pathways, the interaction of the ECM components to multiple surface receptors, and mechanical impact. Additionally, the ECM shaped by cancer regulates immune cells which results in an immune suppressive microenvironment and hinders the efficacy of immunotherapies. Thus, the ECM acts as a barrier to protect cancer from treatments and supports tumor progression. Nevertheless, the profound regulatory network of the ECM remodeling hampers the design of individualized antitumor treatment. Here, we elaborate on the composition of the malignant ECM, and discuss the specific mechanisms of the ECM remodeling. Precisely, we highlight the impact of the ECM remodeling on tumor development, including proliferation, anoikis, metastasis, angiogenesis, lymphangiogenesis, and immune escape. Finally, we emphasize ECM "normalization" as a potential strategy for anti-malignant treatment.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - He Dou
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xi Yu
- Department of Gynecological Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, 150001, China
| | - Shanshan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150000, China.
| | - Min Xiao
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
45
|
Shaikh AS, Sethi A, Makhal PN, Rathi B, Kaki VR. Quest for selective MMP9 inhibitors: a computational approach. J Biomol Struct Dyn 2023; 41:15053-15066. [PMID: 36905674 DOI: 10.1080/07391102.2023.2186710] [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: 11/24/2022] [Accepted: 02/23/2023] [Indexed: 03/13/2023]
Abstract
Matrix Metalloproteinases-9 (MMP-9) is one of the important targets that play a vital role in various diseases such as cancer, Alzheimer's, arthritis, etc. Traditionally, MMP-9 inhibitors have been unable to achieve selectivity to get around this target; thereby, novel mechanisms such as inhibition of activated MMP-9 zymogen (pro-MMP-9) have been discovered. The JNJ0966 was one of the few compounds that attained the requisite selectivity by inhibiting the activation of MMP-9 zymogen (pro-MMP-9). Since JNJ0966, no other small molecules have been identified. Herein, extensive in silico studies were called upon to bolster the prospect of exploring potential candidates. The key objective of this research is to identify the potential hits from the ChEMBL database via molecular docking and dynamics approach. Protein with PDB ID: 5UE4, having a unique inhibitor in an allosteric binding pocket of MMP-9, was chosen for the study. Structure-based virtual screening and MMGBSA binding affinity calculations were performed, and five potential hits were finalized. Detailed analysis of the best-scoring molecules was performed with ADMET analysis and molecular dynamics (MD) simulation. All five hits outperformed JNJ0966 in the docking assessment, ADMET analysis, and molecular dynamics simulation. Accordingly, our research findings imply that these hits can be investigated for in vitro and in vivo studies against proMMP9 and might be explored as potential anticancer drugs. The outcome of our research might contribute in expediting the exploration of drugs that inhibits proMMP-9.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Arbaz Sujat Shaikh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Aaftaab Sethi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, Delhi, India
- HeteroChem InnoTech Pvt. Ltd., Hansraj College Campus, University of Delhi, Delhi, India
| | - Priyanka N Makhal
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, Delhi, India
- HeteroChem InnoTech Pvt. Ltd., Hansraj College Campus, University of Delhi, Delhi, India
| | - Venkata Rao Kaki
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
46
|
Borges MADH, Passos FRS, Quintans JDSS, Azeredo FJ. Hecogenin and its derivates: A pharmacology review. Biomed Pharmacother 2023; 159:114251. [PMID: 36641922 DOI: 10.1016/j.biopha.2023.114251] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
Despite the several uses of drugs from natural compounds in the pharmaceutical industry, new molecules have been discovered and associated with pharmacological activities over the years. Hecogenin, a steroidal saponin, has been the subject of several studies due to reports of pharmacological activities. This study combines the articles published to date that show the pharmacological activity and the mechanism of action of hecogenin, its acetate, and its derivates. This compilation shows that the compounds can act in different pathologies that affect many systems of the human body. They showed pharmacological properties in inflammation, mediating cytokines, cells, and environment. Also, it participated in tumoral processes by pathways like PPGARγ, ERK½, and MMP-2 and showed antimicrobial effects against organisms like Candida and Aedes aegypti's larvae. This review indicates that continuing studies with these molecules are essential once they have the potential to be a future drug.
Collapse
Affiliation(s)
| | | | | | - Francine Johansson Azeredo
- Pharmacy Graduate Program, Faculty of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil; Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA.
| |
Collapse
|
47
|
Zhao F, Zhao P, Chang J, Sun X, Ma X, Shi B, Yin M, Wang Y, Yang Y. Identification and vitro verification of the potential drug targets of active ingredients of Chonglou in the treatment of lung adenocarcinoma based on EMT-related genes. Front Genet 2023; 14:1112671. [PMID: 36824434 PMCID: PMC9942681 DOI: 10.3389/fgene.2023.1112671] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/16/2023] [Indexed: 02/10/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is the main histological type of lung cancer with an unfavorable survival rate. Metastasis is the leading LUAD-related death with Epithelial-Mesenchymal Transition (EMT) playing an essential role. The anticancer efficacies of the active ingredients in Chonglou have been widely reported in various cancers. However, the potential therapeutic targets of the Chonglou active ingredients in LUAD patients remain unknown. Here, the network pharmacology and bioinformatics were performed to analyze the associations of the clinical characteristics, immune infiltration factors and m6A-related genes with the EMT-related genes associated with LUAD (EMT-LUAD related genes), and the molecular docking, STRING, GO, and KEGG enrichment for the drug targets of Chonglou active ingredients associated with EMT (EMT-LUAD-Chonglou related genes). And, cell viability analysis and cell invasion and infiltration analysis were used to confirm the theoretical basis of this study. A total of 166 EMT-LUAD related genes were identified and a multivariate Cox proportional hazards regression model with a favorable predictive accuracy was constructed. Meanwhile, the immune cell infiltration, immune cell subsets, checkpoint inhibitors and the expression of m6A-related genes were significantly associated with the risk scores for EMT-LUAD related genes with independent significant prognostic value of all included LUAD patients. Furthermore, 12 EMT-LUAD-Chonglou related genes with five core drug targets were identified, which participated in LUAD development through extracellular matrix disassembly, collagen metabolic process, collagen catabolic process, extracellular matrix organization, extracellular structure organization and inflammatory response. Moreover, we found that the active ingredients of Chonglou could indeed inhibit the progression of lung adenocarcinoma cells. These results are oriented towards EMT-related genes to achieve a better understanding of the role of Chonglou and its targets in osteosarcoma development and metastasis, thus guiding future preclinical studies and facilitating clinical translation of LUAD treatment.
Collapse
Affiliation(s)
- Fulai Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Peng Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Junli Chang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Xingyuan Sun
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Xiaoping Ma
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Binhao Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Mengchen Yin
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China,*Correspondence: Yongjun Wang, ; Yanping Yang,
| | - Yanping Yang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China,*Correspondence: Yongjun Wang, ; Yanping Yang,
| |
Collapse
|
48
|
Inhibition of Macropinocytosis Enhances the Sensitivity of Osteosarcoma Cells to Benzethonium Chloride. Cancers (Basel) 2023; 15:cancers15030961. [PMID: 36765917 PMCID: PMC9913482 DOI: 10.3390/cancers15030961] [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: 01/08/2023] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Osteosarcoma (OS) is a primary malignant tumor of bone. Chemotherapy is one of the crucial approaches to prevent its metastasis and improve prognosis. Despite continuous improvements in the clinical treatment of OS, tumor resistance and metastasis remain dominant clinical challenges. Macropinocytosis, a form of non-selective nutrient endocytosis, has received increasing attention as a novel target for cancer therapy, yet its role in OS cells remains obscure. Benzethonium chloride (BZN) is an FDA-approved antiseptic and bactericide with broad-spectrum anticancer effects. Here, we described that BZN suppressed the proliferation, migration, and invasion of OS cells in vitro and in vivo, but simultaneously promoted the massive accumulation of cytoplasmic vacuoles as well. Mechanistically, BZN repressed the ERK1/2 signaling pathway, and the ERK1/2 activator partially neutralized the inhibitory effect of BZN on OS cells. Subsequently, we demonstrated that vacuoles originated from macropinocytosis and indicated that OS cells might employ macropinocytosis as a compensatory survival mechanism in response to BZN. Remarkably, macropinocytosis inhibitors enhanced the anti-OS effect of BZN in vitro and in vivo. In conclusion, our results suggest that BZN may inhibit OS cells by repressing the ERK1/2 signaling pathway and propose a potential strategy to enhance the BZN-induced inhibitory effect by suppressing macropinocytosis.
Collapse
|
49
|
Hashemi M, Mirdamadi MSA, Talebi Y, Khaniabad N, Banaei G, Daneii P, Gholami S, Ghorbani A, Tavakolpournegari A, Farsani ZM, Zarrabi A, Nabavi N, Zandieh MA, Rashidi M, Taheriazam A, Entezari M, Khan H. Pre-clinical and clinical importance of miR-21 in human cancers: Tumorigenesis, therapy response, delivery approaches and targeting agents. Pharmacol Res 2023; 187:106568. [PMID: 36423787 DOI: 10.1016/j.phrs.2022.106568] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022]
Abstract
The field of non-coding RNA (ncRNA) has made significant progress in understanding the pathogenesis of diseases and has broadened our knowledge towards their targeting, especially in cancer therapy. ncRNAs are a large family of RNAs with microRNAs (miRNAs) being one kind of endogenous RNA which lack encoded proteins. By now, miRNAs have been well-coined in pathogenesis and development of cancer. The current review focuses on the role of miR-21 in cancers and its association with tumor progression. miR-21 has both oncogenic and onco-suppressor functions and most of the experiments are in agreement with the tumor-promoting function of this miRNA. miR-21 primarily decreases PTEN expression to induce PI3K/Akt signaling in cancer progression. Overexpression of miR-21 inhibits apoptosis and is vital for inducing pro-survival autophagy. miR-21 is vital for metabolic reprogramming and can induce glycolysis to enhance tumor progression. miR-21 stimulates EMT mechanisms and increases expression of MMP-2 and MMP-9 thereby elevating tumor metastasis. miR-21 is a target of anti-cancer agents such as curcumin and curcumol and its down-regulation impairs tumor progression. Upregulation of miR-21 results in cancer resistance to chemotherapy and radiotherapy. Increasing evidence has revealed the role of miR-21 as a biomarker as it is present in both the serum and exosomes making them beneficial biomarkers for non-invasive diagnosis of cancer.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Motahare Sadat Ayat Mirdamadi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Yasmin Talebi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Biology, Islamic Azad University Central Tehran Branch, Tehran, Iran
| | - Nasrin Khaniabad
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Gooya Banaei
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Pouria Daneii
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Sadaf Gholami
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Amin Ghorbani
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Alireza Tavakolpournegari
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Zoheir Mohammadian Farsani
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H3Z6, Canada
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| |
Collapse
|
50
|
Jo HW, Kim MM. β-Caryophyllene oxide inhibits metastasis by downregulating MMP-2, p-p38 and p-ERK in human fibrosarcoma cells. J Food Biochem 2022; 46:e14468. [PMID: 36190169 DOI: 10.1111/jfbc.14468] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/17/2022] [Accepted: 09/23/2022] [Indexed: 01/14/2023]
Abstract
When cancer cells transform into malignant tumors, they gain the ability to ignore growth-inhibiting signals, have endless reproduction potential, resist apoptosis, and induce angiogenesis and invade other tissues. Matrix metalloproteinases (MMPs) allow tumor cells to move into surrounding tissues in many malignancies, but metastasis is blocked by MMPs inhibitors. Therefore, the effect of β-caryophyllene oxide (CPO) contained in Piper nigrum on Mitogen-activated protein kinase (MAPKs) related to MMPs signaling pathways in human fibrosarcoma was examined in HT1080 cells. The effect of CPO on cell viability was performed using the MTT assay. Cytotoxicity was observed in the presence of CPO above 16 μM. Next, gelatin zymography was performed in the cells activated with phorbol-12-myristate-13-acetate (PMA). It was found that CPO at 32 μM reduced MMP-9 activity by 28% and MMP-2 activity by 60%. To confirm the effect of CPO on MMPs, Western blot analyses for MMP-2, MAPKs were carried out in this study. The expression level of MMP-2 was reduced by 45% in the presence of CPO at 32 μM, but those of p-p38 and p-ERK were reduced by 50% and 40%, respectively. CPO decreased the expression levels of MMP-2 and MMP-9 in the immunofluorescence staining assay. Finally, an invasion assay was performed in PMA-treated human fibrosarcoma cells. It was demonstrated that CPO reduced cell invasion of HT1080 cells in a dose-dependent manner starting at a concentration of 2 μM. The above results suggest that CPO could be used as a potential candidate for the treatment of metastasis by inhibiting MMP-2, p-p38 and p-ERK. PRACTICAL APPLICATIONS: Cancer makes it easier for cells to spread to other tissue via blood and lymph systems. Tumor cells deplete nutrients and induce angiogenesis, which penetrates and spreads to other parts of the body. As a result, the effect of CPO against cell invasion was evaluated in this study. CPO reduced cancer cell invasion by inactivating p-ERK and p-p38, according to the findings. MMP-2 and MMP-9 activation and protein expression were also decreased by CPO. As a result, CPO might be used as an alternate treatment agent for preventing metastasis.
Collapse
Affiliation(s)
- Hyun Woo Jo
- Department of Applied Chemistry, Dong-Eui University, Busan, Republic of Korea.,Department of Food Science and Technology, Dong-Eui University, Busan, Republic of Korea
| | - Moon-Moo Kim
- Department of Applied Chemistry, Dong-Eui University, Busan, Republic of Korea
| |
Collapse
|